Speaker: James R. Heath, Ph.D., Gilloon Professor & Professor of Chemistry, CalTech
Single cell barcode chips (SCBC) are microchip platforms designed to quantitatively analyze up to 20 functional proteins from single cells. SCBC can assay up to 10,000 single cells in parallel. Dr. Heath will highlight the application of SCBC to monitor a cohort of cancer patients participating in immunotherapy trials. He will also describe a functional analysis of oncogenic signaling pathways in single cancer cells extracted from tumors at different stages of tumor progression.
Speaker: Dean Sproles, Founder, Chairman, CEO & President, Iverson Genetic Diagnostics
WARFARIN Study (Warfarin Adverse Event Reduction for Adults Receiving Genetic Testing at Therapy Initiation) Major hospital systems nationwide participate in landmark study to assess the utility of genetic testing to determine a personalized warfarin dose. Previous studies have shown that DNA testing can dramatically improve the safety and effectiveness of warfarin, with estimated savings to the U.S. healthcare system of approximately $1.1 billion.
Speaker: Paul J. Utz, Stanford University
Genomics, transcriptomics, and epigenetics hold great promise for realizing personalized medicine. However, the proteome, particularly the antibody repertoire, is orders of magnitude more complex and has been underappreciated in personalized medicine. Dr. Utz will describe the challenges of personalized medicine in human immune diseases and will describe multiplexed proteomics platforms that have been developed to address these challenges.
Speaker: Peter Tonellato, Harvard
The advent of NGS has placed us at the cusp of the use of real-time sequencing for clinical diagnosis and prognosis. Yet, the technologies are outpacing the ability of hospitals to adapt, assimilate and implement clinical whole genome analysis as a routine test for common maladies. The efforts in cancer demonstrate the most promising opportunity. We will present a review of current and planned efforts to improve current research-based WGA to address the unique barriers and requirements of Clinical WGA.
Speaker: Atul Butte, Stanford University
There is an urgent need to translate genome-era discoveries into clinical utility, but the difficulties in making bench-to-bedside translations have been well described. The nascent field of translational bioinformatics may help. Dr. Butte, a pediatric endocrinologist and computer scientist, will show how bioinformatics tools applied to big public data has yielded new uses for drugs and new prototype drugs and diagnostics for type 2 diabetes.
Speaker: Jonathan Sheldon, Oracle
This presentation will focus on our work to provide a scalable secure platform for personalized medicine that accelerates biomarker discovery, validation and delivery to the point of care. This session will discuss in detail our metholody to integrate in a manner that is agnostic to scientific approach and technology cross platform 'omics' data with high quality clinical data to provide an integrated view across genotype and phenotype whilst still ensuring scalability at hundreds of thousands of whole genome sequences. We will also discuss how the current flat file paradigm is no longer fit for purpose as translational medicine moves into 'production' and the false dichotomy that exists between enterprise and open source software and how both need to coexist somewhat uniquely in this area.
Speaker: Catherine Polizzi, Ph.D., Partner, Morrison & Foerster LLP
Patent law continues to evolve in ways that significantly impact personalized medicine companies. This two part talk provides a concise synopsis of recent cases decided by the Federal Circuit and Supreme Court, and their impact on patent strategies available for protecting personalized medicine inventions. Part I focuses on a set of decided cases focused on the patent eligibility of diagnostic method claims. Part II continues with a discussion of the Myriad case, now at the Supreme Court, and its impact on personalized medicine companies. Both talks will highlight thematic elements of the case law and provide context for personalized medicine executives seeking to understanding their implications for developing effective patent strategies.
Speaker: Michael Shuster, Ph.D., Partner, Fenwick & West
Patent law continues to evolve in ways that significantly impact personalized medicine companies. This two part talk provides a concise synopsis of recent cases decided by the Federal Circuit and Supreme Court, and their impact on patent strategies available for protecting personalized medicine inventions. Part I focuses on a set of decided cases focused on the patent eligibility of diagnostic method claims. Part II continues with a discussion of the Myriad case, now at the Supreme Court, and its impact on personalized medicine companies. Both talks will highlight thematic elements of the case law and provide context for personalized medicine executives seeking to understanding their implications for developing effective patent strategies.
Speaker: Ira Klein, M.D., MBA, Medical Director, Aetna
Every good steward of health care resources want the same thing: right treatment at the right time for the right patient with the right diagnosis. Payers are no different. The explosion of genetic testing scope and complexity, along with the multiplicity of “panels”, increases the difficulty in achieving the above stated goals. The regulatory environment for genetic testing is not ideal for reliably achieving reproducible and reliable results. Furthermore, comparative effectiveness research in the field of oncology, now with most new drugs having companion biologic tests, is still in its infancy. These are the challenges.
Speaker: Leroy Hood, M.D., Ph.D., President, Institute for Systems Biology
Personalized medicine is being transformed by emerging technologies that permit one to explore new dimensions of patient data space. In this session you will hear about a wide range of technologies ranging from 3rd generation DNA sequencing, single-cell proteomics and protein arrays on the one had to new strategies for genetic testing and the use of stem cells to delineate normal and disease mechanisms. I will talk about the power of single-cell analyses for understanding biology and disease and discuss powerful systems strategies for identifying blood biomarkers and then talk about applications of this approach.
Speaker: David Pearce, Sanford Health
Sanford Health is the largest rural health provider in the USA having a 220,00-square mile service area over 5 states. Through development of our clinical research programs we are learning to apply personalized medicine across our footprint. Through use of a global consenting process patients have the option to participate in research. Through integration of an electronic medical record with biobanking we are prepared for the advent of genomic medicine through having a patients DNA sequence available for targeted therapies and interventions.
Speaker: David de Graaf, Selventa
With current FDA-approved drugs having a response rate of only 40-50%, the promise of personalized healthcare remains largely unfulfilled. As we further understand how the drivers of disease are manifested across thousands of interrelated biochemical pathways, it becomes clear that current molecular diagnostic tests are often limited by their inability to capture the majority of disease-relevant biomarkers. As the industry creates more data that will lead to better biomarkers and treatment algorithms, Big Data solutions present the opportunity to replace the standard of care with data-driven algorithms based on an individual’s disease characteristics. These "systems diagnostic" tests will enable physicians to determine which treatments are going to work for which patients ahead of time.
Speaker: David Fryburg, Selventa
Good scientific practice requires that the results of an experiment be conclusive. As such, most experiments require the manipulation of a single variable and therefore are reductionist by design. Similarly, most new therapeutics are developed to specifically manipulate a single molecular target. Most diseases, despite phenotypic similarity, are a mixture of underlying molecular mechanisms driving the disease. It is predictable, therefore, that the application of a fine molecular scalpel to a blend of mechanisms will often yield false negative Phase II results. These issues will be discussed as well as their implications for personal medicine approaches early in R&D.
Speaker: Ian Walker, Ph.D., MBA, Head of Stratified Medicine and Combinations Alliance, Cancer Research, UK
The CRUK Stratified Medicine program is a combined research and clinical delivery initiative to conduct detailed tumor profiling for 9,000 patients with 6 cancers. Several big pharma companies and Oracle Health Sciences are involved.
Speaker: Amy J. Sehnert, M.D., Vice President of Clinical Affairs, Verinata Health
Noninvasive prenatal testing represents a major paradigm shift in maternal-fetal medicine. The verifi® test is a noninvasive prenatal test that was initially introduced to detect trisomy 21 (Down syndrome), trisomy 18, and trisomy 13 using cell-free DNA in maternal blood. The test was recently expanded with an option to test for sex chromosome aneuploidies. Verinata’s approach to advancing safe, accurate, and more comprehensive tests for physicians and pregnant women will be discussed.
Speaker: Michael F. Christman, Ph.D., President and CEO, Coriell Institute for Medical Research
Advances in understanding genomic variation and associated clinical phenotypes continue to increase while the cost of full genome sequencing rapidly declines. Having access to your genomic information will become increasingly important as physicians are progressively receptive to incorporating genomics into routine clinical practice. When you need a new prescription, it will be necessary for your physician to quickly and securely access your genetic data to understand drug efficacy prior to dosing. Who will patients and medical professionals trust to store and interpret the data? Coriell is positioned to significantly contribute to the research needed to accelerate the adoption and routine use of genomics in medicine. Dr. Christman will speak to various challenges approached in a large cohort study, the Coriell Personalized Medicine Collaborative (CPMC), in which personal genomic information is reported to study participants. A new partnership with the United States Air Force (USAF), which brings personalized medicine to 2,000 USAF Medical Service personnel, will also be discussed.
Speaker: Myla Lai-Goldman, M.D., Managing Partner Personalized Science, LLC
In 2007, a pharmacogenetic marker for abacavir hypersensitivity was rapidly incorporated into routine medical practice following demonstration of strong clinical utility in pivotal clinical studies. Historic test ordering patterns were studied leading to the development of a three-phase test adoption model. This model indicated the specific activities, which need to be accomplished during test development to facilitate test acceptance. Using this model, we will review pharmacogenetic markers that have not yet been integrated into routine clinical care and suggest reasons for their lack of acceptance. Additionally, we will extend the model to evaluate its potential to guide future test development.
Speaker: Badri Rengarajan, M.D., Medical Director, Archimedes
Product sponsors often have to negotiate reimbursement when they do not yet have a complete dataset (including long-term outcomes and real-world usage). Unfortunately, this makes it challenging to secure a high reimbursement price. Full-scale simulation modeling with virtual patients proceeding through a virtual healthcare system can generate longitudinal data in real-world settings in a matter of days, thereby changing the tone of the negotiation and potentially supporting a higher reimbursement.
Speaker: Peter Hirth, Ph.D., Co-founder & CEO, Plexxikon
With the approval of Zelboraf last year, a new area began in the treatment for patients with metastatic melanoma. This clearly established a new standard for clinical care while simultaneously opening the door for additional treatment options that are currently tested, many of which follow the personalized medicine strategy. This talk will address the impact and challenges of personalized treatment for metastatic melanoma.
Speaker: Michael Snyder, Ph.D., Professor & Chair, Stanford Center of Genomics & Personalized Medicine
We used a combination of omics methods to assess health states in a single individual over the course of almost three years. Genome sequencing was used to determine disease risk. Longitudinal personal omits profiling of transcriptome, proteome and metabolome was used to monitor disease assess, including viral infections and the onset of diabetes. These approaches are elected to transform personalized medicine.
Speaker: Anil Sethi
Personalized Medicine currently refers to diagnostic and clinical interventions, primarily from institutions, providers, and researchers. However, patient involvement from Personalized Healthcare is set to explode, by way of Personal Health Record accounts (PHRs) – under patient control. PHRs capture health info from the white space, the data between clinical encounters. Starting in 2014, ONC/CMS mandates that under Meaningful Use Stage 2 – a patient has amazing new rights to View/Download/Transmit detailed clinical summaries, out of institutions and private practices and into their PHRs. This "Blue Button" initiative triggers a new era of patient-controlled health records. For the first time, patients expect to curate data about them, data aggregated from clinical settings, both inpatient and outpatient, but also self-reported health-and-wellness data, combined with lifestyle medicine, direct-to-consumer genetic testing, and Quantified-Self data from Fitbit-like sensors, and iPhone apps. Real health progress happens only when advanced clinical therapeutics leverages this PHR data, incorporating patients' holistic health info, to inform PM. We'll layout the federal standards, mandate adoption timelines, privacy, and business opportunities.
Speaker: James S. Burns, President & CEO, AssureRx
AssureRx Health provides treatment support solutions to help MDs select appropriate medications for patients with neuropsychiatric disorders. Pharmacogenomic-guided prescribing improves patient outcomes and reduces side effects, ineffective drug trials, and medical utilization costs.
Speaker: Lawrence Corey, M.D., President & Director, Fred Hutchinson Cancer Research Center
Chronic viral and bacterial infections are a leading cause of morbidity worldwide. Many of these pathogens, such as hepatitis B and C, HIV, EBV, HHV-8, and heliobacter pylori, are associated with cancer. Other chronic viral infections, such as HSV, cause often lifelong relapsing morbidity. The interplay by which an organism persists long term in the human for years is being unraveled. Technology to elucidate host viral interactions on a single cell level in vivo are providing insights into how one can alter this milieu to tip the host-pathogen balance toward clearance of the pathogen. Currently, shingles vaccine is the lone therapeutic vaccine. However, inroads in the development of vaccines for eradicating these organisms and hence, interrupting their role in cancer, are in development.
Speaker: Dave Kronlage, SVP Pipeline Development and Pre-commercialization, Pamlab Inc.
Errors of metabolism have been found in subgroups of patients with certain chronic conditions related to the underlying disease pathophysiology. Some of these biomarkers predict an inadequate response to standard treatment and a poorer prognosis. Bioactive nutrients that by-pass errors in metabolism or address specific metabolic processes have been shown to enhance patient outcomes. Pamlab is a biomedical company that specializes in natural personalized medicine by offering health care providers high quality prescription medical foods to safely target the distinct nutritional requirements of patients suffering from a neurologic or metabolic disorder.
Speaker: Mara Aspinall, CEO, Ventana Medical Systems, a member of the Roche Group
Diagnostics has emerged – everyone is talking about them. Pharma is paying for them, scientists are improving them, venture capitalists are funding them, and conference organizers are constantly focusing on them. Efficacy as well as sensitivity and specificity have improved dramatically with old and new technologies.
Yes medical schools are not teaching them, physicians are not using them regularly, regulators are perplexed by them and payors are constantly looking for ways to pay less for them.
Why is happening? How can innovation improve the situation? What can do about it?
Speaker: Anil Sethi, CEO, Pinch Bio
Personalized Medicine refers to diagnostic and clinical interventions. However, patient involvement from Personalized Healthcare is exploding, by way of Patient-Controlled Health Records (PCHRs). PCHRs capture health info from the white space, between clinical encounters. In 2014, ONC/CMS Meaningful Use Stage 2 mandates that patients have amazing new rights to View/Download/Transmit clinical summaries (into their PCHRs), after every encounter. Patients will curate data: clinical and self-reported, direct-to-consumer genetic testing, Quantified-Self data from Fitbit-like sensors, plus iPhone apps. Meaningful health progress happens only when advanced clinical therapeutics leverages this PCHR data, to inform PM. We'll map-out federal mandates, timelines, and opportunities.
Speaker: Robert Jesse, M.D., Ph.D., Principal Deputy Under Secretary for Health, Department of Veterans Affairs
The Veterans Health Administration is committed to transitioning its model for health care delivery to one that it Personalized, Proactive, and Patient Driven. This is instantiated across the organization at the highest level through VHA’s strategic plan. Operationalizing this broad goal requires commitment and training across all levels of the organization, including the resources to ensure success, and, it absolutely requires the engagement of the patients. This will be supported by a foundation based on both team-based care and connected-health capabilities that ensure patients can optimize their health and well-being.
Speaker: Joe W. Gray, Ph.D., Director, Spatial Systems Biomedicine Center & Chair of Biomedical Engineering, OHSU
International breast cancer analysis efforts have now completed a first draft of the breast cancer "ome". These studies suggest the existence of approximately 10 defined subtypes defined by recurrent transcriptional and genomic features plus tens to hundreds of genomic aberrations that occur rarely or may be unique to an individual tumor. This presentation will focus on (a) computational strategies to identify pathways controlling aspects of cancer pathophysiology that are deregulated by the diverse genomic aberrations, (b) development of cell model systems that mirror these deregulated pathways, (c) high throughput image analysis based analysis strategies for elucidation of the aspects of cancer pathophysiology enabled by specific deregulated pathways and (d) strategies to prioritize and target the most promising deregulated pathways.
Speaker: Mavis L. Yee, Partner, Nixon Peabody LLP
With the onset of electronic medical records and the explosion of mobile apps, there is a fast approaching convergence of legal issues that will impact how healthcare providers interact with patients and other providers through the use of mobile technology. The session will briefly describe emerging legal issues and federal law governing the use of mobile technology for the transmission and storage of personal medical information.
Speaker: Rowan Chapman, Ph.D., Head of Precision Diagnostics at GE Healthcare
With aging populations and unsustainable increases in healthcare spending, the need for personalized medicine has never been greater. As scientific innovations and technologies proliferate, how can healthcare providers manage the overwhelming amount of diagnostic data that is being generated from multiple sources and collaborate across specialties to select the right treatment for the right patient at the right time? Rowan Chapman will discuss the opportunity to digitize and integrate data – to make diagnostic information actionable – and the impact this could have on patients, providers and payers across the world.
Speaker: Evian Gordon, BSc, Ph.D., MBBCh Chairman and CEO Brain Resource
The complementary benefit of predicting treatment response using cognition, genomics and EEG is presented in 1000 patients with Depression and 336 patients with ADHD. In predicting treatment response to Escitalopram, Sertraline and Venlafaxine XR, included cognition interaction between thinking and emotion, genomics BDNF and EEG biomarkers of frontal theta. In ADHD response to stimulant medication, cognition attention and response variability, DRD4 and EEG theta (4-7Hz)-beta (SMR 12-15Hz) ratio provided different clinical insights into treatment prediction.
Speaker: Jeffrey D. Marrazzo, Chief Business Officer, MolecularHealth
The falling cost of sequencing, growing number of available cancer therapies and trials, and rapidly expanding body of biomedical knowledge together are enabling the interpretation of NGS for those patients who can afford it. However, the reality of interpreting NGS data within the context of drug data and biomedical knowledge, even for expert clinicians like molecular pathologists and geneticists, requires the implementation of new technologies. These technical capabilities, along with the growing responsibilities of clinical pharmacologists in the NGS-informed treatment decision-making process, will continue to evolve and become adopted as genetic sequence-informed Personalized Cancer Therapy moves into the community oncology setting.
Speaker: Mathias Ehrich, M.D., VP, Research and Development, Sequenom
Noninvasive prenatal testing (NIPT) has recently been introduced for testing of women at high risk for carrying a baby with trisomy 21. Saldivar, et al have presented their overall experience in >30,000 patients undergoing NIPT for fetal aneuploidy. Of these high-risk patients the indications for testing included age-related risk (73.3%), ultrasound abnormalities (26.4%), positive serum screening results (12.6%), and history of aneuploidy (4.3%). We will present data from our experience in a high-throughput testing environment.
Speaker: Walter Koch, Ph.D., Vice President & Head of Global Research, Roche Molecular Systems, Inc.
The advent of high throughput gene sequencing and other genomic characterization technologies is facilitating discovery of the underlying molecular basis of many diseases at an unprecedented pace. In particular, efforts of The Cancer Genome Atlas project and others are identifying a plethora of potential drivers of aberrant cancer cell behavior that could be targeted therapeutically. As companies and academic centers race to establish Next Generation Sequencing-based characterization of tumors into routine clinical diagnostic use, there are a number of challenges for making this genomic information clinically actionable. First, newly discovered mutated or overexpressed proteins must be proven to be critical “drivers” of cancer cell growth and/or metastasis. Additionally, there is a dearth of approved drugs that target the multitude of oncoproteins and pathways that are either known or still being discovered. To increase the pace of translation from discovery to routine implementation in the clinic, novel clinical development strategies will be needed, moving from the current model of co-development of one drug with one companion diagnostic, to differential diagnosis of molecularly-defined subtypes of disease using biomarker panels, many of which will have implications for existing therapies, as well as evolving use for drugs entering into development.
Speaker: John Hornberger, M.D., CEO and President, CEDAR Associates LLC
Much of the future of personalized medicine is based on development of sophisticated algorithms predicting diagnosis, outcome, or response to therapy. Medicare recently stated an unwillingness to reimburse for such algorithms. The sophistication of these algorithms – developed based on solid principles of biostatistical and artificial intellegence - are such that even the most mathematically capable human cannot replicate the computations. Establishing policies not to reimburse for such algorithms lacks face validity, akin to assigning all the value of a computer to the hardware and assigning no value to the software, and creates disincentive to invest in this nascent field. This talk will discuss theoretically credible and proven methods for assigning a value, and thereby a price, to such innovations in health care.
Speaker: Ralph Snyderman, M.D., Chancellor Emeritus, Duke University
The concept of personalized medicine, spurred by the genomic revolution, was proposed a decade ago as the next transformation of medicine. Despite the development of exciting technical innovations, the practice of medicine has largely remained unchanged. However, the adoption of personalized health care is now rapidly gaining ground through continued advances in technology and the understanding of the power of personalized health care (PHC) as a new practice model. PHC uses personalized medicine applications, along with personalized health planning, to improve how care is delivered by making it more predictable, proactive, and strategic, thereby providing the best care for each patient. The transformation of medicine by PHC has indeed begun.
Speaker: Randy Scott, Ph.D., Chairman and CEO, InVitae Corporation
The power, speed and low cost of next generation DNA sequencing is rapidly changing the paradigm of genetic testing by making it economically feasible to aggregate all genetic testing into a single assay at very low cost. How we manage and incorporate that information into routine medical practice and empower that data for the research community may be the largest informatics business opportunity of the 21st century -- with the power to dramatically alter how we define, research, treat, and prevent human disease.
Speaker: John Mattison, M.D., CMIO & Assistant Medical Director, Kaiser-Permanente, SCAL
Much of the current healthcare crisis derives from diseases of lifestyle that result in obesity, diabetes and their numerous comorbities. "Information Therapy" has been remarkably ineffective in addressing these epidemics. I will focus on how personalizing the behavioral approaches for each consumer is required to support healthier lifestyles.
Speaker: Colin Hill, CEO, President, Chairman & Co-Founder, GNS Healthcare
Despite numerous breakthroughs in biotechnology, the quality of healthcare has improved only incrementally, while costs have soared. This is due in part to our one-size-fits-all approach to medicine. Thus, there is a huge opportunity to reduce costs while improving care for patients by harnessing Big Data to discover what works and for whom. We are doing this today using Real World Outcomes data: medical claims, pharmacy claims, and electronic medical records. Using high performance supercomputing and sophisticated machine learning algorithms, we are building causal models of disease at the individual patient level. We are using these models to predict individual patient risk and disease trajectories in IBD, and to match interventions to individual patients at risk of developing diabetes and serious heart conditions. Our partners in the health insurance industry are using these models to optimize benefit design and disease modification programs to reduce costs and improve patient outcomes. Using similar models, we are helping our partners in the pharmaceutical industry match treatments to patients by identifying predictive biomarkers for diseases such as cancer and rheumatoid arthritis using genomic and proteomic data. As these types of molecular data become combined with Real World Outcomes data and Electronic Health Records, even more opportunities will open up, allowing Big Data analytics to help usher in a new era of personalized medicine.
Speaker: Stephen M. Stahl, M.D., Ph.D., UCSD
Major depression is a debilitating illness that affects over 121 million people worldwide and is considered by the WHO to be the leading cause of disability worldwide. The current paradigm of antidepressant "trial and error" results in a poorer prognosis and greater cost for patients. There is a high unmet medical need for therapies linked to biomarkers that predict a greater chance of treatment success in mental illness. Emerging therapies that that target specific biomarkers in major depression hold promise for personalized medicine in mental illness.
Speaker: Ann Kapoun, Ph.D., Vice President of Translational Medicine, OncoMed Pharmaceuticals
Dr. Kapoun will discuss OncoMed’s approach and implementation of predictive biomarkers into their clinical trials for patient selection. OncoMed Pharmaceuticals is a clinical-stage company that discovers and develops novel therapeutics targeting cancer stem cells, the cells shown to be capable of driving tumor growth, recurrence and metastasis. OncoMed has advanced five anti-cancer therapeutics into the clinic, including demcizumab (OMP-21M18, Anti-DLL4), OMP-59R5 (Anti-Notch2/3), OMP-52M51 (Anti-Notch1), OMP-18R5 (Anti-Fzd7), and OMP-54F28 (Fzd8-Fc), which target key cancer stem cell signaling pathways including Notch and Wnt.
Speaker: Gil Omenn, M.D., Ph.D., Director, University of Michigan Center for Computational Medicine & Bioinformatics
Molecularly-targeted therapies require companion diagnostic tests to identify and stratify patients for most appropriate choices of therapy. Few biomarker candidates have been developed to clinical utility. The Institute of Medicine created a Committee on Omics-Based Predictive Tests; its 2012 report is “Evolution of Translational Omics: Lessons Learned and Path Forward”. Its framework for test development has three phases: discovery, validation, and demonstration of clinical utility. Numerous lessons are drawn from case studies, notably including now-retracted publications from Nevins, Potti, and collaborators at Duke and premature launch of companies and clinical trials. Guidance is provided for the responsible parties: all co-authors, host institution, funders, regulators, and journals. Hopefully these recommendations will help accelerate progress toward personalized medicine.
Speaker: Yael Mosse, M.D., Perelman School of Medicine, University of Pennsylvania
Advances in the treatment of children with high-risk neuroblastoma have, until recently, involved the addition of cytotoxic therapy to already dose-intensive regimens. The discovery of hereditary and germline mutations in the oncogene ALK places neuroblastoma among other cancers such as melanoma and non-small cell lung cancer which benefit from targeted therapies with small molecule tyrosine kinase inhibitors. However, inhibition of mutated ALK has proven to be complex and remains a therapeutic challenge. This presentation will focus on (a) biology of ALK in neuroblastoma compared to ALK-translocated human malignancies, (b) preclinical progress with ALK inhibition, (c) integration of functional and computational modeling to stratify patients to ALKi therapy, (d) Combining therapeutic interventions with molecular diagnostics for patient selection and clinical trial design.
Speaker: Bruce R.Conklin, M.D., Departments of Medicine & Pharmacology, Gladstone Institute of Cardiovascular Disease.
Dr. Conklin is using induced pluripotent stem (iPS) cells to model human disease. His major focus is on genes that cause “sudden death” due to abnormal heart rhythm, as well as genes involved in heart failure from cardiomyopathy. He uses iPS cells from patients who have genetic diseases, and is also engineering iPS cells to have specific mutations. Engineering iPS cells allowing direct comparison of many gene variants with the exact same (isogenic) background. Recent genetic studies such as GWAS, provide gene variant associations that largely remain untested. The use of iPS cells with discrete mutations in an isogenic background provides an experimental system to directly test gene variants that have been implicated in genetic studies. Personalized medicine can benefit from experimental testing of gene variants to prove (or disprove) hypothetical associations that GWAS and other genetic studies.
Speaker: Stephan Brock, Ph.D., Chief Technology Officer, President, Molecular Health
The practice of medicine has historically emphasized the role of guidelines – standards set by oversight bodies reviewing clinical studies, published literature and anecdotal evidence. With the advent of gene sequencing, powerful technologies to analyze immense volumes of data, and diminishing barriers to accessing patient data, novel clinico-molecular analyses are enabling increasingly precise treatment decision-making. Considered together in a real-world case study, Genomic, Pharmacological safety, and “Population” level integrated data analysis provides health practitioners with a compelling toolkit to optimize medical decision-making.
Speaker: Nigam H. Shah, MBBS, Ph.D., Assistant Professor of Medicine (Biomedical Informatics), Stanford University
We present data mining methods that transform unstructured patient notes taken by doctors, nurses and other clinicians into a de-identified, temporally ordered, patient-feature matrix using standardized medical terminologies. We demonstrate how to use the resulting high-throughput data to monitor for adverse drug events and profile the safety of specific drugs based on the clinical notes in the EHR. We will discuss a proof of principle study which shows the potential of text-analytics to uncover ‘natural experiments’, which profile the safety of Cilostazol in patients with Congestive Heart Failure. Such data-mining methods have applications in post-marketing drug safety surveillance and in obtaining personalized insight into difficult-to-test clinical hypotheses.
Speaker: Janet Woodcock, M.D., Director, Center for Drug Evaluation & Research, FDA
By fast-tracking individualized treatments through the FDA approval process and encouraging collaboration between the regulatory and industry arenas, Dr. Woodcock has shown dedication to personalized medicine. In 2011 during her tenure, Xalkori and Zelboraf, individualized treatments both paired with companion diagnostics, received FDA approval.
Speaker: Laura Esserman, M.D., Director, Carol Franc Buck Breast Care Center, Professor of Surgery and Radiology, UCSF
Dr. Esserman will review the need for new approaches to clinical trials. Give an example of an approach to clinical trials that will allow us to screen Phase II agents and identify the tumor subtypes for which the agents are most effective. Also she will discuss the eventual need to develop therapeutic switching if we want to harness emerging targeted therapies.
Speaker: Steffan Ho, M.D., Ph.D., Director, Translational Oncology, Pfizer Inc.
Lung cancer has undergone a remarkable transformation from a histology-defined malignancy treated with chemotherapy to a disease in which molecular characterization directs treatment with an increasingly diverse spectrum of highly effective targeted therapeutic agents. The evolution of diagnostic testing in lung cancer provides a broadly relevant view of the real world application of Personalized Medicine in the clinic, and offers important insights into future challenges.
Speaker: Susan Garfield, Lou Hochheiser, Naomi Aronson, Rina Wolf, Jeanne De Sa, Ira Klein
During this panel discussion, payers, health technology assessment leaders and leaders from medical groups will discuss how personalized medicine technologies will be evaluated and paid for. Each will describe the key factors in assessing value from their constituent perspectives. The session will provide audience members with insights into payer perspectives on personalized medicine, evidence requirements to achieve coverage and payment, how health technology assessments are evolving to assess personalized medicine technologies, and how healthcare provider groups are considering the economics of practice change related to adopting personalized medicine.
Speaker: James Kobielus, Big Data Evangelist, IBM
Healthcare providers and payers require analytics-driven decision support tools to personalize treatment delivery, improve patient outcomes, and lower operational costs. A key new technology to support these objectives is similarity analytics, which identifies patients who are similar to a given patient for decision support and comparative effectiveness research analysis. In this presentation, Jianying Hu and James Kobielus from IBM will discuss the treatment personalization applications of patient similarity analytics and complementary big-data analytics technologies.
Speaker: Cliff Reid, Ph.D., CEO, Complete Genomic
Whole human genome sequencing is becoming so accurate and inexpensive that it will soon be less expensive to sequence the entire genome than to run even a few genetic tests. Once a person’s genome is sequenced and securely stored, it acts as the ultimate genetic test: all subsequent genetic tests are nothing more than database lookups, which cost approximately nothing. This offers a promising path to reducing healthcare costs, with national health care systems well positioned to benefit, and private insurance systems struggling to adapt. But the biggest winners from this disruptive technology may be an entirely new category of diagnostics companies.
Speaker: Brett Davis & Deven Atnoor, Oracle
The volume, variety and velocity of new genomic data, along with dramatic declining cost of obtaining it are having a profound impact on the prospects for personalized healthcare. However, gains in genomic data are only part of the journey toward personalized healthcare and new data management platforms will be required to bring a variety of currently siloed data types together to truly achieve value based, personalized healthcare. This presentation will discuss the opportunities and challenges in the quest for personalized healthcare as well as the new informatics platforms that will be required to support not just translational research but the personalization of care. Specifically, Oracle will demonstrate its Translational Research Center (TRC) platform that is being implemented by many of the world’s leading academic medical centers, integrated delivery networks and cancer centers to answer the “hard questions” in healthcare – what works, for whom, why, in what context and at what cost?
Speaker: Mark Trusheim, Visiting Scientist & Executive-In-Residence, MIT Sloan School of Management
Science leading to ever smaller patient sub-populations, regulatory and payer evidence standards increasing, patents weakening, reimbursement under pressure globally - is personalized medicine facing an economic crossroads? Through the challenges, there exists a path to sustainable, viable economics for all. It will require some change by all to realize the potential for patients and unlock incentives for all. Pulling together recent quantitative research, we’ll move beyond talking points to evidence based rationale for that change.
Speaker: Hasso Plattner, Prof. Dr. h.c. mult. Co-founder, SAP
SAP and the Hasso Plattner Institute Potsdam have pioneered the use of SAP HANA, an in-memory database, to provide an integrated platform for processing and real-time analysis of large volumes of genomic data. The platform supports a wide variety of alignment and variant calling algorithms. Variants are stored in a large in-memory database and seamlessly aggregated with numerous international scientific databases (including NCBI, PubMed, 1000 genomes project and dbSNP). This novel storage mechanism gives the researcher an unprecedented ability to perform sophisticated real-time analyses of variants across large cohorts.
Speaker: Joyce Peng, Marketing Director, BGI Americas
BGI, the largest genomics organization in the world, is legendary for its extensively published research. This talk will cover recent medical research by BGI, including the application of metagenomics in diabetes and single-cell sequencing in cancer research. With the goal of translating medical research breakthroughs into global healthcare solutions, BGI is bringing state-of-the-art multi-omic technologies to the clinical setting. Building on extensive clinical testing experience in China, BGI today is enabling medical providers and patients worldwide realize the promise of genomics-based medical diagnostics and personalized healthcare.
Speaker: Michael Kamerick, DC Specialist Leader, Recombinant By Deloitte
This session will explore multiple real world deployments where organizations leverage data as an asset to support a personalized medicine mission. The technology and expertise for these projects is provided by Recombinant, a software/analytics unit of Deloitte that is 100% focused on enabling the secondary use of healthcare data.
Speaker: Jeanne De Sa, Senior VP, UnitedHealth Center for Health Reform & Modernization, UnitedHealth Group
The presentation will review recent claims data from UnitedHealthcare for genetic testing and molecular diagnostic services as well as estimates of national spending. The speaker will discuss implications of their findings for the future of personalized medicine and its role in an improved delivery system. The presentation will cover trends in Medicare, Medicaid and the commercial sector and discuss issues relevant to the use of diagnostics in those populations.
Speaker: Corey S. Goodman, Ph.D., Managing Partner & Co-founder, venBio LLC
Discoveries from biomedical research have reached heights but despite that tools, technologies and targets for drug discovery are greater than ever, Big Pharma is having problems with their internal pipelines. Additionally, many biotech companies are having problems getting funded while Life science venture capital firms are closing. To fix this ecosystem, we need all three legs of the stool – biotech, venture capital, and pharma – working together in new collaborative models to deliver on the promise of bringing important new medicines forward for human health.
Speaker: Yusra Hussain, M.D., Clinical Assistant Professor & Medical Director, Aging Adult Services, Stanford Hospital & Clinics
Much of what we know today is based on studies in centenarians and super-centenarians. Through research we are finding that aging itself is a risk factor for many diseases such as heart disease, cancer, and neurodegenerative diseases. The percentage of people with these illnesses is projected to increase considerably in the coming decades. Findings from animal studies have demonstrated that aging is a plastic process that can be manipulated by both genetic and environmental factors. I will discuss some of the genetic variants that protect centenarians from increased risk of age associated diseases. I will also touch base on the concept of compression of morbidity presented first by Dr. Fries in the mid nineties. I will also go over some of the known environmental mechanisms that promote longevity.
Speaker: Mehrdad Ayati, M.D., Faculty Stanford University School of Medicine
The geriatric population is prescribed the highest proportion of medications in relation to their percentage of the U.S population-13% of current geriatric population purchase 33% of all prescription drugs and this number will increase to 50% by 2040. Prevalence of both adverse drug reactions and treatment failures increase in the older patients. Adverse Drug Events (ADEs) are responsible for 5-28% of acute geriatric medical admissions. It has been estimated that for every one dollar we spend on medications at nursing home facilities, we spend $1.33 in health care resources for the treatment of drug related morbidity and mortality.
The genetics of drug metabolizing enzymes (DMEs), P450 or CYP, and drug transporters (DTs) play a major role in their clinical response. Our knowledge of pharmacogenomics started back in the 6th century BC by Pythagoras who found that eating fava beans caused illness in few but not all individuals. This knowledge has been built upon until our current understanding of the relationship between xenobiotic metabolism with the CYP system (examples are Clopidogrel metabolism related to CYP2C19; the impact of CYP2C9 polymorphic variants and functional genetic variants in the VKORC1 gene on the pharmacokinetics and pharmacodynamics of warfarin; and similar effects on Statin, Beta Blocker, SSRIs, Antiepileptic, etc.). Bringing the knowledge of pharmacogenomics into clinical practice will have a substantial impact in minimizing the adverse drug events and improving the quality of care in the elderly population.
Speaker: Don Morris, Ph.D., VP, Scientific Product & Technology Development, Archimedes Inc
Use of complex personal genomic information to inform routine clinical decisions requires a new approach to treatment guidelines and new tools to evaluate and then communicate the options. Archimedes IndiGO, a decision support tool already in use at Kaiser Permanente and other organizations, generates personalized guidelines that can take complex data into account. Its new risk-based decision paradigm can easily be extended allowing for a seamless integration of a variety of complex information in the clinic.
Speaker: Yaniv Erlich, Ph.D., Whitehead Fellow, Whitehead Institute for Biomedical Research
Sharing sequencing datasets without identifiers has become a common practice in genomics. We developed a novel technique that uses entirely free, publicly accessible Internet resources to fully identify individuals in these studies. I will present quantitative analysis about the probability of identifying US individuals by this technique. In addition, I will demonstrate the power of our approach by tracing back the identities of multiple whole genome datasets in public sequencing repositories.
Speaker: Kim Popovits, Chairman, CEO & President, Genomic Health
Often we hear personalized medicine as getting the right drug to the right patient in the right dose at the right time. In cancer it is really about patients understanding their individual disease since one tumor is not like any other tumor. Therefore, it is critical for physicians and patients to understand the genomic makeup, or molecular signature, of a particular tumor in order to better direct treatment. All cancer patients deserve to have access to personalized medicine regardless of where they are treated; however, several barriers stand in the way of this goal including physician education and the ability to translate an unprecedented amount of data into clinically relevant information. The knowledge and tools are here; however, industry, academia, community physicians and payors must work together to deliver THE PROMISE OF personalized medicine to all CANCER patients.
Speaker: Marty Tenenbaum, Sarah Green, Tony Blau, Frank McCormick
Cancer Commons is an open science initiative founded to fast-track research on personalized oncology by matching patients to the best available therapies for their tumors, and learning as much as possible from the results. The rapid learning process continually updates a consensus knowledge base of biomarkers and targeted therapies through data analysis and discussion of the latest laboratory and clinical findings. By fostering the free flow of data and knowledge among physicians, researchers and patients, and across cancers and institutions, we aim to propel fresh lines of research, suggest novel treatment approaches, and ultimately offer new hope to millions of cancer patients. Join a conversation with four of the principals and invited industry discussants to learn how you and your organization can participate.
Speaker: Steven C Quay, M.D., Ph.D., FCAP, Founder, Atossa Genetics, Inc.
How to obtain routine, repeated, painless breast biopsy samples non-invasively for cytopathology, NGS, proteome, and transcriptome analysis of precursors to breast cancer; Identifying the earliest “-omic” changes in reversible precursor lesions of the breast, such as atypical ductal hyperplasia; The use of breast specimens obtained non-invasively for biomarker discovery, clinical trial support, and patient selection, and to inform personalized medical therapy; Cancer prevention using intraductal treatment of reversible hyperplastic lesions
Speaker: Deepak Srivastava, M.D., Director of the Gladstone Institute of Cardiovascular Disease and Professor at UCSF
The convergence of stem cell technology and advances in genomics promise to pave the way for tailored therapies based on individual genetic background. Reprogramming skin cells into induced pluripotent stem (iPS) cells is beginning to allow prediction of drug efficacy and toxicity based on individual genetic variation. We have generated iPS cells from patients with genetically defined cardiovascular disorders and used cells derived from them for understanding the role of modifier genes and disease mechanisms. Finally, we have succeeded in reprogramming endogenous non-myocytes in the heart into new cardiomyocytes, harnessing ones own cells for regenerating tissue in situ.
Speaker: Ken Song, M.D., Chief Executive Officer, Ariosa Diagnostics.
Ariosa’s Harmony PrenatalTM Test offers pregnant women an early, accurate and personalized option for detecting common fetal genetic conditions via a blood test. This presentation will discuss Ariosa’s propriety and efficient approach to analyze cell-free DNA in blood and how this new approach is transforming prenatal care.
Speaker: Amir Dan Rubin, President & CEO, Stanford Hospital & Clinics
Stanford Hospital & Clinics seeks to deliver personalized care solutions through its Stanford Health Partners array of offerings.
Speaker: Cesar Duarte, M.D., MSc, Ph.D., Genoprimer Molecular Diagnostic and Personalized Medicine, Brazil
Genoprimer is a molecular diagnostic laboratory dedicated to diagnosis of human genetic diseases, infectious diseases and pharmacogenomics. Their mission is to provide the medical community with genomic information in order to improve Brazil’s health services
Speaker: John Adler, M.D., Founder, Editor-in-Chief, Cureus
How Cureus is fueling a revolution in peer-reviewed medical publishing. Peer reviewed medical journals have been a critical part of the scientific discourse for over 200 years, but medical science is stymied by a peer review system that has not changed since its inception. Over 5500 medical journals exist today, and information delivery remains fragmented, balkanized and one-sided. Cureus, an emerging, socially interactive medical journal, is changing this dynamic. Cureus is publishing a variety of new content across all medical disciplines, has an Editorial Board made up of leaders in their respective medical specialties from all over the world, and has a growing following, particularly among young, tech-savvy physicians.
Speaker: Garry Nolan, Ph.D., Professor in the Department of Microbiology and Immunology, Stanford University
Few would be surprised that molecular indicators of cell physiology represent a “status report” of cell history, health, and even future cellular function. Given this, key indicators in cells can act PK/PD “barometers” of in vivo function. However, it is often overlooked that the focus upon a “target” can ignore that such targets exist within the framework of responsive and dynamic networks. As such, metabolic elements in networks far from the originating pathology of a cell can act as surrogates of the pathology, reflections of how a drug might be correcting the pathology, or by themselves those surrogates might be modulated en masse to force corrective action upon the pathology. Often, the network is “silent” about the pathology until it is perturbed to reveal how it thinks (much like when you interview a candidate for a job position—you don’t look at their CV alone, you invite them in for questioning). The story today in pharmacology and biomedicine should not be about the lone “target”—it’s not even about a pathway. It’s about the network—describable as an inter-cellular and intra-cellular system of responsive and compensatory metabolisms that are the “body language” of systems biology.
Speaker: Steven C Quay, M.D., Ph.D., FCAP, Founder, Atossa Genetics, Inc.
As medical practice moves toward more ‘protocol’ driven treatment paradigms, the ability of genomics-based diagnostic companies to launch innovative tests becomes ever more difficult. Using the CarePath business model and the understanding of practice patterns to design products and services to address the entire CarePath of a disease or condition provides an innovative way to distinguish a start-up company from the crowd, establish synergies across all parts of the business, and build stakeholder value for the organization. The Breast Health CarePath established by Atossa Genetics, comprising five FDA-cleared medical devices, four CLIA-validated Laboratory Developed Tests, and a pharmaceutical/ biotech treatment program, will be provided as an example.
Speaker: Yuval Itan, Ph.D., Postdoctoral Fellow, The Rockefeller University
The human gene connectome (HGC) is the full set of predicted biologically plausible routes, distances, and degrees of separation between all possible pairs of human genes, identified by a shortest distance algorithm on the full human genome network. The HGC can be applied across all genetic fields, including the identification of novel disease causing gene(s) by estimating the biological proximity of patients’ genetic variants to experimentally validated disease causing genes.
Speaker: Peter J. van der Spek, Ph.D., Head of the Department of Bioinformatics, Erasmus Medical Center, Netherlands
Medicine is more and more becoming evidence based; our work at the Erasmus Medical Center focuses on the development of the skull and the brain in relation to transcriptional programs and epigenetic regulation during development and cancer. We will present mutations in various genes expressed in stem cells, during proliferation, differentiation and apoptosis and address the IT strategy used for storing whole genome sequencing data. Also illustrated will be our molecular imaging genomics approach, used for validation of our candidate mutations.
Speaker: Stefan Roever, CEO, Genia Technologies
DNA Sequencing is used in the clinic today, but what does it take to make sequencing part of routine clinical care? Genia’s mission is to make genetic information universally available. Its integrated circuits and proprietary NanoTag chemistry enable massively parallel single-molecule DNA sequencing. This versatile nanopore-based platform allows for single molecule, electrical, real-time analysis without the need for complicated optics, labels, amplification, or fluidics. By developing a true integrated circuit on standard semiconductor process technology, Genia's sequencing platform enables the use of molecular diagnostics in everyday clinical care.
Speaker: Jeff Wisotzkey, Ph.D., HCLD, CC, Director, Molecular Pathology and Technical Director of Laboratory Operations, Central Pennsylvania Alliance Laboratory
Dr. Wisotzkey's presentation will focus on the day to day issues inherent to the ever changing landscape of companion diagnostics in the clinical molecular pathology laboratory with specific emphasis on providing a synopsis of the FDA approval and subsequent implementation of the QIAGEN therascreenR KRAS molecular assay from the perspective of the clinical laboratory. Dr. Wisotzkey also will touch briefly on several other companion diagnostic applications in his laboratory.
Speaker: Michael Thaler, Professor, UCSC
‘Personalized’ or ‘Precision’ medicine is informed by associations between genomic variation, clinical phenotypes, and disease. The utilization of genetic data in clinical decision-making generates novel challenges that require reinterpretations of bioethics principles as occurred with the social and scientific transformations of the 1970’s, yet the ethical and legal questions raised by the new paradigm remain largely unaddressed. This presentation will focus on informed consent issues in the era of personalized medicine.
Speaker: Mark Monane, M.D., Chief Medical Officer, CardioDx, Inc.
Every day, approximately 10,000 patients present to primary care clinicians with signs and symptoms suggestive of coronary artery disease. Despite this large number, the diagnostic work-up of these patients remains highly variable and poses an enormous burden on the healthcare system, including $5 billion in testing costs, as well as patient risks associated with the ionizing radiation and contrast dye used in diagnostic tests. Corus® CAD is the first cardiovascular genomic test designed to help clinicians safely, accurately, and conveniently determine, from a blood sample, whether their patients’ symptoms are due to blockages in the heart arteries, also known as obstructive coronary artery disease.
Speaker: Ian Walker, Ph.D., MBA, Head of Stratified Medicine and Combinations Alliance, Cancer Research, UK
In December 2012, Prime Minister David Cameron announced plans to undertake whole-genome sequencing for cancer and rare disease patients in the UK and be the first country to introduce the technology within a mainstream health system.
Dr. Ian Walker will discuss lessons learned from another personalized medicine initiative that combines clinical practice and research objectives. Cancer Research UK, government, and two leading pharmaceutical companies have been working in partnership to standardize genetic testing of tumors across the UK and build a national database linking tumor profiles, treatments and outcomes.
Speaker: Carol Berry, Senior VP and GM, Pharmacogenomic Services Division & CLIA Laboratory, Asuragen
Thyroid cancer is on the rise. Statistics show there are many unnecessary surgeries being performed based on cytology results alone. The case study will present a real world study incorporating molecular testing to aid in resolving indeterminant cytology and how it changed the surgical approach in many cases.
Speaker: Janet Woodcock, M.D., Director, Center for Drug Evaluation & Research, FDA
Genetically-targeted drugs and individualized diagnostics are now being incorporated into mainstream medicine: personalized medicine has reached a “tipping point”-- it is no longer a prediction, but a reality that has arrived much sooner than expected and represents a major scientific accomplishment. Nevertheless, given the current financial problems in healthcare, additional challenges can be foreseen. Can use of these technologies make medicine more cost-effective? Will personalized medicine reach this next milestone, and if so, when?
Speaker: David Levison, President and CEO, CardioDx, Inc.
There’s been a historical disconnect between payers and diagnostic test manufacturers regarding data required for positive-coverage decisions. New test makers claim they are held to higher standards than existing competitive modalities. The launch of CMS’ MolDx Program is a step towards clarifying the process and requirements for coverage of tests. However, challenges remain in navigating the complex paths to coverage. Industry must sustain momentum for solutions with a dialogue more focused on risk-sharing and common objectives, enabling providers, payers and test manufacturers to succeed.
Speaker: Matthieu-P. Schapranow, Ph.D., Principal Investigator of In-Memory Technology for Life Sciences, Hasso Plattner Institute
This presentation will outline how SAP and the Hasso Plattner Institute Potsdam have applied in-memory technology to combine high-performance alignment, annotation and analysis of full genome data. We will provide you with detailed background information about the technology foundation SAP HANA presented in the keynote of Prof. Hasso Plattner. In addition, we will outline how to integrate existing genome data processing pipelines into and how to design and implement specific scientific applications on top of SAP HANA. Furthermore, the technical background of optimized alignment algorithms for SAP HANA will be outlined.
Speaker: Edward Abrahams, Sheila Walcoff, Alberto Gutierrez, Jeffrey Marrazzo, Risa Stack, Paul Billings
The regulatory pathways that govern personalized medicine products have evolved over time. Multiple agencies, centers within agencies, and different stakeholders, many of which employ different business models, all play a role in determining how our emerging understanding of individual variation will transform health care. This panel will review the current regulatory environment for diagnostic and other personalized medicine products as well as outline current proposals to reform the process.
Speaker: Masato Mitsuhashi, M.D., Ph.D., Chief Scientific Officer, Hitachi Chemical Research Center
In order to simulate pathological or pharmacological events in test tube, whole blood is immediately exposed to specific agents, and the induction of early responder mRNAs is quantified by Hem(A)+ system we developed. This ex vivo assay is a powerful biomarker discovery platform by using numerous combinations of stimulations and target mRNA species. The resultant biomarkers are used to predict individual variations in drug responses/toxicities. Moreover, Hem(A)+ system is now capable of quantifying poly(A)+ RNA in exosome/microvesicles in plasma, urine, culture supernatants, etc. without ultracentrifugation. By measuring cell/tissue specific- or disease-specific mRNAs in plasma and urine exosome, post-treatment clinical course can be monitored quantitatively. Two platforms (ex vivo and exosome) will revolutionize current personalized medicine diagnostics into next dimension.
Speaker: Karthikeshwar Kasirajan, M.D., Chief Medical Officer, Natural Molecular
For decades many have often debated and written about the great promise of pharmacogenomics. However, the concept has not become the standard of care for drug delivery for a variety of reasons. The greatest impediment to routine use is that it remains a complex clinical pathway to implement, with a lack of physician education that allows seamless integration into clinical practice. In order for pharmacogenomic tests to be actionable and user friendly the final product should consist of drug recommendations based on individual genotype and not a delivery of genetic test results. This presentation will discuss the delivery of a user friendly report that for the first time holds the promise of taking pharmacogenomics from bench to bedside in a large scale.
Speaker: Dietrich Stephan, Silicon Valley Biosystems
All human disease has a genetic component. SV Bio enables physician access to genetics in a simple, accurate, and comprehensive manner so as to improve patient outcomes. The presentation will describe SV Bio's unique platform which promises to democratize the practice of genetic testing so it is a pervasive part of healthcare. Easing access to, and interpretation of, the genome for physicians is the key to empowering them to use the information. That being said, the internal complexities of generating clinical grade NGS data, managing massive amounts of data using commercial grade IT systems, and interpreting genomes in silico and at scale will be described as some of the unique differentiating features of SV Bio.
PanGenX has developed a Business-to-Business (B2B) knowledgebase and data analytics solution for pharmaceutical companies, hospitals, national health systems, and health plans to more meaningfully analyze and apply the vastly increasing array of pharmacogenetic data.
NeuroGeneration is a biotechnology company designing stem cell therapies and biological modifiers for neurodegenerative disorders. Their mission is to provide individualized biotherapeutics and be at the forefront of central nervous system repair and restoration.
Saladax Biomedical develops and commercializes novel diagnostic assays for the practical delivery of personalized medicine. Saladax’s initial focus is oncology, with a portfolio of 13 chemotherapy drug dose management assays in various stages of development.
Assay Depot has created a web-based application called a Scientific Research Exchange or Marketplace that enables scientists from all over the world to access experts and purchase thousands of research services online.
Boston Heart Diagnostics is a heart health management company providing integrated diagnostic and patient management solutions that are advancing cardiovascular disease risk assessment, monitoring and treatment. The Company’s goal is to predict, prevent, manage and reverse cardiovascular disease by improving patient assessment and management.
telmedx enables doctors and clinicians to remotely see patients via live medical-grade videoTM from patient cell phones. Their HIPAA-compliant telemedicine platforms reduce costs and create revenue opportunities by improving clinician triage, remote diagnosis, follow-up care, drug and protocol compliance, and health status monitoring without a patient clinic visit.
Omixon helps clinical and diagnostic labs to adopt next generation sequencing by developing precise and validated NGS data analysis software for the benefit of patients.
Bell Biosystems provides new methods for testing efficacy of cancer drugs and for evaluating cell therapies in animal models. Their tools offer researchers total control of injected cells via imaging, homing, and kill-switch functionalities. The heart of our innovation is the inheritability of these functionalities as cells divide.
Goalpost helps prevent and manage chronic disease by changing behavior through social influence, behavioral analytics and medical advice.
uBiome seeks to spark the era of personalized medicine by providing the public with easily accessible information about their own bodies using the latest in high-throughput DNA sequencing technology. In addition, uBiome compares participants’ microbiomes with numerous past studies on the role of the microbiome in health, diet and lifestyle.
Portable Genomics™ develops mobile software for comprehensive visualization and use of personal genomic data for informed healthcare planning and lifestyle adjustments in connection with healthcare professionals.
Talon Biometrics develops a technology for molecular diagnostics, the J-Probe™, offering advantages in sensitivity, selectivity and multiplicity over alternatives. The J-Probe can distinguish and quantify tens of thousands of genetic sequences in a simple single-tube reaction.
SomaLogic uses its proprietary proteomic technology to develop enhanced protein analysis tools and reagents for the life sciences community, to facilitate biomarker discovery and validation for diagnostic and therapeutic applications, and to develop and commercialize clinical diagnostic products that will improve the delivery of healthcare by offering diagnostic information to physicians and patients.
PLC diagnostics is developing the next generation handheld point of care platform. Based on proprietary waveguide technology, this platform is capable of multiplexing protein and nucleic acid assays. No wash requirements and short turnaround time enable POC monitoring of biomarker panels required for different aspects of personalized medicine.
TOMA Biosciences uses the latest genomic sequencing technology and a patented approach to reveal tumor specific information that physicians can use to improve cancer treatment for patients.
Stacy Feld, J.D., is a Partner at Physic Ventures. Stacy focuses on personalized health solutions to improve and transform care for the health consumer. Specifically, Stacy is interested in life science opportunities offering more precise prevention, diagnosis, and treatment of chronic disease. She is on the board of T2 Biosystems and an observer on the boards of On-Q-ity and Own.
Careticker helps patients better prepare before surgery and provide personalized support after surgery for a safe road to recovery. Their applications are procedure-specific, allowing patients to plan, connect and monitor their own needs throughout a healthcare event.
Speaker: Joseph Victor, President and CEO, DVS Sciences
Privately held DVS Sciences Inc. manufactures and globally distributes a revolutionary platform technology using mass spectrometry for multi-parameter cell analysis, or “Mass Cytometry”. Mass Cytometry is being adopted globally at an unprecedented pace driven by significant advantages over conventional cell analysis technology (like flow cytometry) primarily derived by eliminating the inherent spectral overlap from fluorescent probes, thereby enabling quantitative, highly multi-parametric analysis that can simultaneously identify up to 100 biomarkers with high resolution and wide dynamic range. Key commercial products include the CyTOF®, a patented multi-parameter mass cytometer instrument for quantitative individual cell analysis, & proprietary reagents.
ReSurfX uses novel data generation and utilization technologies to enable better decisions from large volumes of data. They leverage patented, integrative solutions and products to increase Pharma/healthcare productivity as well as to improve sensitivity and accuracy of measurement technologies.
Station X develops software products for scientists and clinicians who work with large-scale human genome information in either a research or clinical setting, enabling them to extract maximal biological meaning from complex genomic data.
Syapse is a biomedical data platform helping researchers, providers, and payers use omics to improve medical outcomes.
Astrid’s focus is data management and analysis for genomics research - utilizing core competences in mathematics and information technology. Clients include academic research institutes, pharmaceutical and biotech companies, who are seeking comprehensive IT solutions for diagnostic marker and therapeutic target identification.