Preview Mode Links will not work in preview mode

Getting Personal: Omics of the Heart


Apr 20, 2018

Jane Ferguson:                 Hello, welcome to Getting Personal: Omics of the Heart. This is podcast Episode 15 from April 2018. I'm Jane Ferguson, an Assistant Professor of Medicine at Vanderbilt University Medical Center, and this podcast is brought to you by Circulation Genomic and Precision Medicine and the AHA Council on Genomic and Precision Medicine.

                                                As usual, we have a great lineup of papers in Circ Genomic and Precision Medicine this month. The first is actually the subject of our interview this month. Sony Tuteja talked to Craig Lee from the University of North Carolina about his manuscript entitled, "Clinical Outcomes and Sustainability of Using CYP2C19 Genotype Guided Antiplatelet Therapy After Percutaneous Coronary Intervention." This manuscript investigated the use of pharmacogenomics to improve treatment after PCI, and you can hear a lot more about it directly from the first author later in the podcast.

                                                Our next manuscript also used pharmacogenomics approaches to look for snips associated with plasma renin activity and to assess the effect of top snips with blood pressure response to atenolol and hydrochlorothiazide. The first and last authors are Caitrin McDonough and Julie Johnson from the University of Florida. And their manuscript is entitled, "Genetic Variants Influencing Plasma Renin Activity in Hypertensive Patients from the Pharmacogenomic Evaluation of Antihypertensive Response," or PEAR study. They find that snips in the SNNTXNDC11 gene region associate with higher baseline plasma renin activity in their sample of over 700 subjects and with a smaller systolic blood pressure reduction to hydrochlorothiazide. Variation in the region may act through modulation of TXNDC11 gene expression. They also identified several other candidate genes of interest. These new candidates may allow for precision medicine approach to selection of hypertensive treatment and further study the mechanisms may reveal novel biology on blood pressure response to pharmacological treatment.

                                                Next up is a manuscript by Deirdre Tobias and colleagues entitled, "Circulating Branch Chain Amino Acids and Incident Cardiovascular Disease in a Prospective Cohort of U.S. Women." I actually had the chance to talk to Deirdre about her research last month. So check out the March podcast, Episode 14, to hear more from Dr. Tobias about this study.

                                                A study of hypertrophic cardiomyopathy from Hannah [inaudible 00:02:36] and Michelle Michels and colleagues from the Erasmus Medical Center in the Netherlands assessed the effects of genetic screening in family members of patients with a known hypertrophic cardiomyopathy mutation. In their manuscript entitled, "Outcomes of Contemporary Family Screening and Hypertrophic Cardiomyopathy," they described their study which assessed cascade screening in 777 relatives of 209 probans between 1985 and 2016. Genetic and clinical screening resulted in a diagnosis of HCM in 30% of family members at the time of testing. An additional 16% of family members developed HCM over seven years of follow up. Of the 43% of family members who were genotype positive, 37% were ultimately diagnosed with HCM. There was no difference in survival between genotype positive and genotype negative family members or with relatives who did not undergo genetic testing.

                                                There are genetic considerations that are unique to the ancestral composition of the Netherlands with a high proportion of individuals with a founder mutation, so the proportion of probans with identified mutations is higher than in other reported studies. This paper demonstrates the potential benefit of genetic screening in family members, which can identify individuals who should undergo intensive screening, and at the same time reduce concerns for family members who are genotype negative. However, the classification of the pathogenicity of variants and understanding variable penetrance remains a challenge.

                                                A manuscript entitled, "Exome Sequencing in Children with Pulmonary Arterial Hypertension Demonstrates Differences Compared to Adults." From Na Zhu, Claudia Gonzaga-Jauregui, Carrie Welch, Wendy Chung, and colleagues from Columbia University, ask the question whether there were differences in the genetic mutations responsible for early onset pulmonary arterial hypertension, or PAH, in a pediatric sample compared with adult onset disease. While some mutations, particularly in BMPR2 appear to be similar in the pediatric and adult samples there were significantly more mutations in TBX4 in the children compared with adults.

                                                Further, children were more likely to have de novo mutations identified through exam sequencing that were predicted missense variants. Given the additional complications associated with pediatric onset of PAH, understanding the genetic differences in this population is an important step towards identifying novel genes and mechanisms which could guide future therapeutic development.

                                                Our next manuscript authored by Iisan Kadhen, Carolyn Macdonald, Mark Lindsay, and colleagues from Harvard Medical School is entitled, "Prospective Cardiovascular Genetics Evaluation in Spontaneous Coronary Artery Dissection," or SCAD. They genotyped individuals with SCAD to find out the genetic contribution to the disease. Of the patients for whom genetic testing was performed, six of them were 8.2%. Identifiable mutations in genes known to be involved in vascular disease, including COL3A1, LMX1B, PKD1, and SMAD3. These individuals were significantly younger at the time of their first SCAD event compared to patients with no identifiable mutation. Given the relatively higher rate of mutations identified in this sample, there may be a rationale to conduct genetic testing in all individuals presenting with SCAD, particularly in younger individuals.

                                                Shiu Lun Au Yeung, Maria-Carolina Borges, and Debbie Lawlor, from the University of Hong Kong and the University of Bristol, set out to find out if reduced lung function is causal in coronary artery disease. As reported in their manuscript, entitled "The Association of Genetic Instrumental Variables for Lung Function on Coronary Artery Disease Risk, A 2-Sample Mendelian Randomization Study," they used a Mendelian Randomization approach to assess causal relationships between two measures of lung function. Forced expiratory volume in one second, and forced vital capacity on CAD. Genetic predictors of increased forced expiratory volume were associated with lower risk of CAD. While there was a similar association with forced vital capacity, this was attenuated in sensitivity analyses. Overall, the data suggests that higher forced expiratory volume may independently protect against CAD. However, the mechanisms remain unclear.

                                                Finally, the April issue also contains a white paper from Kiran Musunuru, Xiao-zhong Luo, and colleagues entitled, "Functional Assays to Screen and Dissect Genomic Hits, Doubling Down on the National Investment in Genomic Research." This paper lays out strategies to followup on findings from high-throughput genomic analyses, including the use of novel technologies, assays, and model systems that can help to effectively translate big data findings and capitalize on previous investment in genomic discovery.

                                                To see the latest issue of Circulation Genomic and Precision Medicine, and to access all the papers we talked about and to browse previous issues, go to "circgenetics.ahajournals.org."

Sony Tuteja:                       Hello, my name is Sony Tuteja, I'm an assistant Professor of Medicine at the University of Pennsylvania in Philadelphia, I'm also an early career member of the American Heart Association Council on Genomic and Precision Medicine. Today I'm joined by Dr. Craig Lee, an associate Professor of Pharmacy at the University of North Carolina School of Pharmacy. Dr. Lee is a first author of an article published in April 2018 issue of Circulation Genomic and Precision Medicine entitled, "Clinical Outcomes and Sustainability of Using CYP2C19 Genotype Guided Anti-Platelet Therapy After Percutaneous Coronary Intervention." Welcome Dr. Lee, and thank you for joining me today.

Craig Lee:                            Thanks for having me.

Sony Tuteja:                       First let me just say congratulations on spearheading such impactful work on the implementation of CYP2C19 pharmacogenetic testing.

Craig Lee:                            Thanks, this has been a very complicated project, but a lot of fun.

Sony Tuteja:                       Great. So I think some of our listeners may have not had time to read your paper yet so I was wondering if you could provide a brief overview of the paper and what the study was about.

Craig Lee:                            Sure. Although it's been widely described that loss of function polymorphisms in the drug metabolizing enzyme, CYP2C19, which is responsible for the bio-activation of the antiplatelet drug clopidogrel, impairs its effectiveness, there remains considerable debate and uncertainties surrounding whether CYP2C19 genetic testing should be used clinically for guiding antiplatelet therapy in percutaneous coronary intervention, or PCI patients. As the evidence base is expanded, an increasing number of institutions are seeking to implement CYP2C19 genetic testing despite limited data on the use and impact of using this genetic testing to guide antiplatelet therapy selection following PCI in real world clinical settings.

                                                UNC was an early adopter for CYP2C19 genotype-guided antiplatelet therapy in high-risk PCI patients. Our algorithm recommends that patients carrying one or two loss of function alleles in CYP2C19 be prescribed an alternative antiplatelet therapy such as prasugrel or ticagrelor. Our algorithm was implemented back in the summer of 2012, under our then-director of the Catheterization Laboratory, and now Chief of Cardiology, Dr. Rick Stouffer. We conducted the study to better understand the feasibility, sustainability, and clinical impact of using CYP2C19 genetic testing to optimize antiplatelet therapy selection in PCI patients in real-world clinical practice.

                                                Basically what we did was following the implementation of our algorithm in the summer of 2012, we've been retrospectively collecting data from all patients that come through our Cath lab that undergo a PCI. We collect information on their clinical characteristics, whether or not they underwent CYP2C19 genetic testing, what antiplatelet therapy they were prescribed when they were in the hospital at discharge and over the course of followup, and more recently we've been assessing clinical outcomes, both ischemic outcomes and bleeding outcomes. The data presented in our paper described the algorithm's use at our institution over the first two years following its implementation from 2012 to 2014 with one year of followup data. Since we do about 600 PCI procedures per year on our Cath lab, the study population is just under 1200 patients.

                                                Our main findings were that CYP2C19 genotypes were frequently ordered, efficiently returned, and routinely used to guide antiplatelet therapy selection after PCI over this two year period. However, we also observed that the frequency of genotype testing and frequency of using alternative therapy such as prasugrel or ticagrelor in the patients that carried CYP2C19 loss of function alleles fluctuated over time. We also observed that use of clopidogrel in patients that were tested, but carried either one or two copies of a CYP2C19 loss of function allele was associated with a significantly higher risk of experiencing a major ischemic cardiovascular event compared to use of alternative therapy. These risks were particularly evident in the highest risk patients, and largely driven by patients who carry only one copy of the loss of function allele, the so-called intermediate metabolizers.

                                                Our primary takeaway from this analysis is that implementing a genotype-guided antiplatelet therapy algorithm is feasible, sustainable, and associated with better clinical outcomes in a real-world clinical settling, but challenging to maintain at a consistently high level over time.

Sony Tuteja:                       Great. I know it's always challenging to implement new work flow and new testing into the clinical setting. Can you describe how the algorithm was incorporated in the cardiologist workflow to minimize disruption?

Craig Lee:                            Absolutely. This algorithm was spearheaded by our interventional cardiologists with the support of our clinical pharmacy specialists and pathology laboratory. They key element to our success is that we have the capacity to do the genotype testing in our molecular pathology lab on site. Dr. Karen Weck is the director of that laboratory and is a coauthor on our paper. Since the prescribing decision for antiplatelet occurs in a highly specialized clinical setting, we have all the pieces in place to do this in-house at UNC, which seems to make things very efficient.

                                                There really wasn't very much disruption in the workflow given that the testing is done on-site and the test seems to be treated like another laboratory test that's done, which is really the ultimate goal of pharmacogenomics. We don't currently actually have clinical decision support built into our electronic health record, so the reason we could actually get this off the ground was because of the substantial collaboration between our physicians, pharmacists, and pathology lab.

                                                But one of the things we learned through this experience, which is described in the paper, is that there are fluctuations in the use of the genetic testing to guide prescribing over time that we believe could be remedied by developing more automated clinical decision support, to help make things a little bit more efficient for the clinicians. But at the start of it, it was really just a will to do it, which was really exciting to observe.

Sony Tuteja:                       Absolutely. That's exciting that everybody was on board with this project. What do you think were the most challenging aspects of the implementation?

Craig Lee:                            That's a great question, and one that often comes up. I think that the education on the front end is really, really important. It needs to recur as the implementation spans over a period of time. For example, there's turnover in the interventional cardiology fellows every summer as well as occasional turnover of attending physicians and clinical pharmacy specialists. As individuals come and go into the clinical environment, it is important that they understand how the algorithm works, and how it can be applied in practice. And this is accomplished by recurring education and communication.

                                                The other thing that's been a challenge is turnaround time. Even though our molecular pathology lab typically turns tests around within one day of a PCI procedure, if the test result isn't available or the antiplatelet therapy isn't changed in response to the genetic test before the patient is discharged from the hospital, we found that it can be challenging to followup on the result before the next encounter. Typically, if a change in medication needs to occur after discharge and prior to the first followup clinic visit, the communication piece has proven to be very important. It's not an insurmountable barrier, but one we observe that created one additional challenge. Other institutions around the country that are doing this have expressed similar things.

Sony Tuteja:                       You showed in your study that during the middle of the implementation there was a decline in testing. What do you think were the major reasons that led to decrease in testing?

Craig Lee:                            Yeah, that's a great question. We're not sure. We didn't collect information prospectively, and more specifically, we did not survey the physicians in terms of why they ordered the test. But we believe, just based on anecdotal experience and talking about this with everyone, there was this big surge of momentum, with the initial implementation, and as the practice evolved there was just sort-of a settling of individuals in terms of, I think, the practice patterns.

                                                Overall, the test was ordered and over 70% of PCI patients, an alternative therapy was prescribed and approximately 70% a loss of function allele carriers. These numbers exceeded 80% early on which was higher than we expected. They dropped down to about 60%, which is still a pretty high utilization rate when you compare to other institutions that have implemented. After some educational efforts, the testing rates and use of alternative therapy and loss of function allele carriers began to increase again over the last six months.

Sony Tuteja:                       Yeah I was just gonna ask, since the study is completed, have you taken any further steps to maintain the frequency of the testing at the high level that you initially started with?

Craig Lee:                            Yeah, so again recurring education has been really important particularly with interventional cardiology fellows, since they're the ones that really execute this in terms of ordering the tests and working with the clinical pharmacy specialists. And as I mentioned, we're in the process of developing clinical decision support to help make this a little bit easier on the prescribers. When a test result is available, we believe this will make it a little easier for the result to be more readily available for the clinical decision.

Sony Tuteja:                       Yeah I think the CDS tools will be key to have more compliance with the results in adherence to the test results. I'm just curious, who pays for the genotype tests at your center and are you billing for these tests?

Craig Lee:                            Yes. We're billing for these tests as part of routine clinical care.

Sony Tuteja:                       Great, and you've had good success with reimbursement?

Craig Lee:                            As far as we can tell, yes.

Sony Tuteja:                       That's great to hear. I think that will really incentivize other centers to pursue similar lines of testing. So what do you think are the broader implications for implementing genetically guided care for other drugs?

Craig Lee:                            Yeah, I think that it's interdisciplinary collaboration. Communication is really important among physicians, clinical pathologists, and clinical pharmacists. We found that this has been essential to success of the program here at UNC with this one gene drug pair. And again, this is fueled by a spirit of collaboration and will for our clinicians to work together to optimize patient care. And really, I think clinical pharmacists are uniquely positioned to help make this happen. Clinical pharmacists are uniquely positioned to interpret pharmacogenomic test results, provide medication recommendations, as well as counsel patients on how to interpret the tests and why the prescribing decision is being made. Our clinical pharmacists at UNC are fantastic and have really embraced this. They've shown that pharmacogenomics can be an important part of medication therapy management.

                                                Although implementation of pharmacogenomics testing is clearly a challenge, it is now part of the routine in our Cath lab and in our cardiology services. And again, that's been really exciting to observe. I also think this experience provides a foundation in an example for other pharmacogenomic implementations to occur at our institution.

Sony Tuteja:                       That's great, it's so nice to hear about the team working together to get this accomplished. What has been the patient response to the testing? How have they responded to receiving genetic test results?

Craig Lee:                            We think it has been overall positive. And again, it's now part of the workup in terms of providing the best possible care for the patient given the evidence that we have. And so again, since it's part of the clinical work flow, there's not a separate research consent that's done. The testing is part of the consent to the procedure.

Sony Tuteja:                       Well great, that's all the questions I have for you today. Do you have any final thoughts you wanna share with our listeners?

Craig Lee:                            No, other than just a thank you again for having me in for talking about our paper. And I guess, I would just urge those that are out there that are either planning to do this or doing this, to collect data. It's really important to evaluate the practice, evaluate the frequency of testing, the frequency of prescribing decisions being altered by the testing, and trying to understand what the barriers are. And if possible, evaluate clinical outcomes.

                                                You know, we started this study under the umbrella of continuous quality improvement and it really has taught us a lot. I think it has helped optimize how the algorithm is used, and as other centers around the country have been doing this, it provided a basis to collaborate and really evaluate the impact on clinical outcomes, which is really the question that is on everybody's minds. And as the evidence base expands, I think there will be a lot more comfort with doing these things, but we should always strive to generate the evidence we need to assure that we're making the right decisions in practice.

Sony Tuteja:                       Absolutely, I think that outcomes piece will be critical to getting this in the mainstream. Well I'd like to thank you for your time today, it was a pleasure speaking to you and once again, congratulations.

Craig Lee:                            Thanks.

Jane Ferguson:                 That's all for this month. As a reminder you can follow us on Twitter, @Circ_Gen or connect with us on Facebook. Thanks for listening, and I look forward to bringing you more on genomics and precision medicine of the heart next month.