Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Aleksandra Pavlovic is active.

Publication


Featured researches published by Aleksandra Pavlovic.


The Lancet | 2010

Clinical assessment incorporating a personal genome

Euan A. Ashley; Atul J. Butte; Matthew T. Wheeler; Rong Chen; Teri E. Klein; Frederick E. Dewey; Joel T. Dudley; Kelly E. Ormond; Aleksandra Pavlovic; Alexander A. Morgan; Dmitry Pushkarev; Norma F. Neff; Louanne Hudgins; Li Gong; Laura M. Hodges; Dorit S. Berlin; Caroline F. Thorn; Joan M. Hebert; Mark Woon; Hersh Sagreiya; Ryan Whaley; Joshua W. Knowles; Michael F. Chou; Joseph V. Thakuria; Abraham M. Rosenbaum; Alexander Wait Zaranek; George M. Church; Henry T. Greely; Stephen R. Quake; Russ B. Altman

BACKGROUND The cost of genomic information has fallen steeply, but the clinical translation of genetic risk estimates remains unclear. We aimed to undertake an integrated analysis of a complete human genome in a clinical context. METHODS We assessed a patient with a family history of vascular disease and early sudden death. Clinical assessment included analysis of this patients full genome sequence, risk prediction for coronary artery disease, screening for causes of sudden cardiac death, and genetic counselling. Genetic analysis included the development of novel methods for the integration of whole genome and clinical risk. Disease and risk analysis focused on prediction of genetic risk of variants associated with mendelian disease, recognised drug responses, and pathogenicity for novel variants. We queried disease-specific mutation databases and pharmacogenomics databases to identify genes and mutations with known associations with disease and drug response. We estimated post-test probabilities of disease by applying likelihood ratios derived from integration of multiple common variants to age-appropriate and sex-appropriate pre-test probabilities. We also accounted for gene-environment interactions and conditionally dependent risks. FINDINGS Analysis of 2.6 million single nucleotide polymorphisms and 752 copy number variations showed increased genetic risk for myocardial infarction, type 2 diabetes, and some cancers. We discovered rare variants in three genes that are clinically associated with sudden cardiac death-TMEM43, DSP, and MYBPC3. A variant in LPA was consistent with a family history of coronary artery disease. The patient had a heterozygous null mutation in CYP2C19 suggesting probable clopidogrel resistance, several variants associated with a positive response to lipid-lowering therapy, and variants in CYP4F2 and VKORC1 that suggest he might have a low initial dosing requirement for warfarin. Many variants of uncertain importance were reported. INTERPRETATION Although challenges remain, our results suggest that whole-genome sequencing can yield useful and clinically relevant information for individual patients. FUNDING National Institute of General Medical Sciences; National Heart, Lung And Blood Institute; National Human Genome Research Institute; Howard Hughes Medical Institute; National Library of Medicine, Lucile Packard Foundation for Childrens Health; Hewlett Packard Foundation; Breetwor Family Foundation.


Science Translational Medicine | 2012

Patient-Specific Induced Pluripotent Stem Cells as a Model for Familial Dilated Cardiomyopathy

Ning Sun; Masayuki Yazawa; Jianwei Liu; Leng Han; Veronica Sanchez-Freire; Oscar J. Abilez; Enrique G. Navarrete; Shijun Hu; Wang L; Andrew Lee; Aleksandra Pavlovic; Shin Lin; Rui Chen; Roger J. Hajjar; Michael Snyder; Ricardo E. Dolmetsch; Manish J. Butte; Euan A. Ashley; Michael T. Longaker; Robert C. Robbins; Joseph C. Wu

Human induced pluripotent stem cells generated from patients with familial dilated cardiomyopathy model cardiovascular disease in these patients. iPSCs Make the Heart Beat Faster Mutations in genes expressed in the heart can cause dilated cardiomyopathy (DCM), a form of heart disease in which a weakened and enlarged heart is unable to pump sufficient blood for the body’s needs. DCM can lead to progressive heart failure that eventually requires heart transplantation. This disease has been challenging to study because cardiomyocytes from the hearts of DCM patients are difficult to obtain and do not survive long. Mouse models of DCM are established and have provided important clues about the disease mechanisms for DCM. However, the mouse heart is very different in physiology compared to the human heart; for example, the mouse heart rate is 10 times faster than that of human. In a new study, Sun et al. generated induced pluripotent stem cells (iPSCs) from skin cells of patients in a family with inherited DCM. This family carries a deleterious mutation in TNNT2, a gene that is expressed specifically in the heart and regulates cardiomyocyte contraction. Using iPSCs, the authors generated a large number of individual-specific cardiomyocytes carrying the specific TNNT2 mutation and analyzed their functional properties. Compared to cardiomyocytes derived from iPSCs of healthy controls in the same family, cardiomyocytes derived from iPSCs of DCM patients exhibited an increased heterogeneous myofilament organization, susceptibility to stress, compromised ability to regulate calcium flux, and decreased contraction force. These results suggest that the mutation in TNNT2 causes abnormalities in the cardiomyocytes and contributes to the development of DCM disease. Using these DCM iPSC–derived cardiomyocytes, the researchers also showed that several current treatments that clinically benefit DCM disease improved DCM cardiomyocyte function in culture. The current study shows that human iPSC-derived cardiomyocytes could provide an important platform to investigate the specific disease mechanisms of DCM as well as other inherited cardiovascular disorders and for screening new drugs for cardiovascular disease. Characterized by ventricular dilatation, systolic dysfunction, and progressive heart failure, dilated cardiomyopathy (DCM) is the most common form of cardiomyopathy in patients. DCM is the most common diagnosis leading to heart transplantation and places a significant burden on healthcare worldwide. The advent of induced pluripotent stem cells (iPSCs) offers an exceptional opportunity for creating disease-specific cellular models, investigating underlying mechanisms, and optimizing therapy. Here, we generated cardiomyocytes from iPSCs derived from patients in a DCM family carrying a point mutation (R173W) in the gene encoding sarcomeric protein cardiac troponin T. Compared to control healthy individuals in the same family cohort, cardiomyocytes derived from iPSCs from DCM patients exhibited altered regulation of calcium ion (Ca2+), decreased contractility, and abnormal distribution of sarcomeric α-actinin. When stimulated with a β-adrenergic agonist, DCM iPSC–derived cardiomyocytes showed characteristics of cellular stress such as reduced beating rates, compromised contraction, and a greater number of cells with abnormal sarcomeric α-actinin distribution. Treatment with β-adrenergic blockers or overexpression of sarcoplasmic reticulum Ca2+ adenosine triphosphatase (Serca2a) improved the function of iPSC-derived cardiomyocytes from DCM patients. Thus, iPSC-derived cardiomyocytes from DCM patients recapitulate to some extent the morphological and functional phenotypes of DCM and may serve as a useful platform for exploring disease mechanisms and for drug screening.


PLOS Genetics | 2011

Phased Whole-Genome Genetic Risk in a Family Quartet Using a Major Allele Reference Sequence

Frederick E. Dewey; Rong Chen; Sergio Cordero; Kelly E. Ormond; Colleen Caleshu; Konrad J. Karczewski; Michelle Whirl-Carrillo; Matthew T. Wheeler; Joel T. Dudley; Jake K. Byrnes; Omar E. Cornejo; Joshua W. Knowles; Mark Woon; Li Gong; Caroline F. Thorn; Joan M. Hebert; Emidio Capriotti; Sean P. David; Aleksandra Pavlovic; Anne West; Joseph V. Thakuria; Madeleine Ball; Alexander Wait Zaranek; Heidi L. Rehm; George M. Church; John West; Carlos Bustamante; Michael Snyder; Russ B. Altman; Teri E. Klein

Whole-genome sequencing harbors unprecedented potential for characterization of individual and family genetic variation. Here, we develop a novel synthetic human reference sequence that is ethnically concordant and use it for the analysis of genomes from a nuclear family with history of familial thrombophilia. We demonstrate that the use of the major allele reference sequence results in improved genotype accuracy for disease-associated variant loci. We infer recombination sites to the lowest median resolution demonstrated to date (<1,000 base pairs). We use family inheritance state analysis to control sequencing error and inform family-wide haplotype phasing, allowing quantification of genome-wide compound heterozygosity. We develop a sequence-based methodology for Human Leukocyte Antigen typing that contributes to disease risk prediction. Finally, we advance methods for analysis of disease and pharmacogenomic risk across the coding and non-coding genome that incorporate phased variant data. We show these methods are capable of identifying multigenic risk for inherited thrombophilia and informing the appropriate pharmacological therapy. These ethnicity-specific, family-based approaches to interpretation of genetic variation are emblematic of the next generation of genetic risk assessment using whole-genome sequencing.


JAMA Cardiology | 2017

Feasibility of Obtaining Measures of Lifestyle From a Smartphone App: The MyHeart Counts Cardiovascular Health Study

Michael V. McConnell; Anna Shcherbina; Aleksandra Pavlovic; Julian R. Homburger; Rachel L. Goldfeder; Daryl Waggot; Mildred K. Cho; Mary Rosenberger; William L. Haskell; Jonathan Myers; Mary Ann Champagne; Emmanuel Mignot; M Landray; Lionel Tarassenko; Robert A. Harrington; Alan C. Yeung; Euan A. Ashley

Importance Studies have established the importance of physical activity and fitness, yet limited data exist on the associations between objective, real-world physical activity patterns, fitness, sleep, and cardiovascular health. Objectives To assess the feasibility of obtaining measures of physical activity, fitness, and sleep from smartphones and to gain insights into activity patterns associated with life satisfaction and self-reported disease. Design, Setting, and Participants The MyHeart Counts smartphone app was made available in March 2015, and prospective participants downloaded the free app between March and October 2015. In this smartphone-based study of cardiovascular health, participants recorded physical activity, filled out health questionnaires, and completed a 6-minute walk test. The app was available to download within the United States. Main Outcomes and Measures The feasibility of consent and data collection entirely on a smartphone, the use of machine learning to cluster participants, and the associations between activity patterns, life satisfaction, and self-reported disease. Results From the launch to the time of the data freeze for this study (March to October 2015), the number of individuals (self-selected) who consented to participate was 48 968, representing all 50 states and the District of Columbia. Their median age was 36 years (interquartile range, 27-50 years), and 82.2% (30 338 male, 6556 female, 10 other, and 3115 unknown) were male. In total, 40 017 (81.7% of those who consented) uploaded data. Among those who consented, 20 345 individuals (41.5%) completed 4 of the 7 days of motion data collection, and 4552 individuals (9.3%) completed all 7 days. Among those who consented, 40 017 (81.7%) filled out some portion of the questionnaires, and 4990 (10.2%) completed the 6-minute walk test, made available only at the end of 7 days. The Heart Age Questionnaire, also available after 7 days, required entering lipid values and age 40 to 79 years (among 17 245 individuals, 43.1% of participants). Consequently, 1334 (2.7%) of those who consented completed all fields needed to compute heart age and a 10-year risk score. Physical activity was detected for a mean (SD) of 14.5% (8.0%) of individuals’ total recorded time. Physical activity patterns were identified by cluster analysis. A pattern of lower overall activity but more frequent transitions between active and inactive states was associated with equivalent self-reported cardiovascular disease as a pattern of higher overall activity with fewer transitions. Individuals’ perception of their activity and risk bore little relation to sensor-estimated activity or calculated cardiovascular risk. Conclusions and Relevance A smartphone-based study of cardiovascular health is feasible, and improvements in participant diversity and engagement will maximize yield from consented participants. Large-scale, real-world assessment of physical activity, fitness, and sleep using mobile devices may be a useful addition to future population health studies.


Circulation-cardiovascular Genetics | 2012

Randomized Trial of Personal Genomics for Preventive Cardiology Design and Challenges

Joshua W. Knowles; Themistocles L. Assimes; Michaela Kiernan; Aleksandra Pavlovic; Benjamin A. Goldstein; Veronica Yank; Michael V. McConnell; Devin Absher; Carlos Bustamante; Euan A. Ashley; John P. A. Ioannidis

Background Genome-wide association studies (GWAS) have identified more than 1500 disease-associated single nucleotide polymorphisms (SNPs), including many related to atherosclerotic cardiovascular disease (CVD). Associations have been found for most traditional risk factors (TRFs), including lipids,1,2 blood pressure/hypertension,3,4 weight/body mass index,5,6 smoking behavior,7 and diabetes.8–13 GWAS have also identified susceptibility variants for coronary heart disease (CHD). The first and, so far, strongest of these signals was found in the 9p21.3 locus, where common variants in this region increase the relative risk of CVD by 15% to 30% per risk allele in most race/ethnic groups.13–20 Subsequent largescale GWAS meta-analyses and replication studies in largely white/European populations have led to the reliable identification of an additional 26 loci conferring susceptibility to CHD,2,20–23 all with substantially lower effects sizes compared with the 9p21 locus. Many of these CVD susceptibility loci appear to be conferring risk independent of TRFs and thus cannot currently be assessed by surrogate clinical measures (Table 1). Among the 27 independent loci identified in the most recent large meta-analyses of CVD, 21 were reported not to be associated with any of the TRFs.20,21 Several studies have explored whether initial CVD-related genetic markers can improve risk prediction over standard models restricted to TRFs using a genetic risk score (GRS) constructed on the basis of the number of risk alleles inherited.24–26 Results to date have been mixed. Although all have shown that a GRS is strongly associated with the outcome of interest independent of TRFs, none were able to demonstrate a significant improvement in the c-statistic. Two of the 3 studies showed some modest improvement in newly defined discrimination indices, including the integrated discrimination index, the net reclassification index, and the clinical net reclassification index (net reclassification index in the intermediate-risk subjects). Thus, the use of these markers has not yet been shown to convincingly outperform models that include TRFs and family history alone. One important reason for the failure of these markers to demonstrate clinically meaningful improvement of risk prediction relates to the small proportion of the genetic variance explained by these markers, a phenomenon commonly referred to as the heritability gap. The basis for this heritability gap is the focus of intense investigation. Despite this gap, it is still possible that knowledge of genetic risk may improve patient outcomes through means other than enhanced risk reclassification. For instance, genetic testing may improve patient adherence and CVD risk factor reduction for Mendelian disorders related to CHD, such as familial hypercholesterolemia.27 This effect may be owing to an increase in patient motivation (eg, people who recognize and accept their high risk are more encouraged to reduce it); however, no clinical trial to date has demonstrated that newly discovered genetic markers improve risk factor profiles by improving adherence to prescribed therapy for complex (garden variety) CVD. Here, we describe the design of an ongoing randomized trial to investigate whether CVD risk factor profiles can be improved by providing participants with knowledge related to their inherited risk of CVD in addition to information on their risk related to measured TRFs. We also discuss some of the challenges that arise in the design and conduct of such a trial and how they were addressed.


Journal of Cardiology | 2015

Long-term outcomes of septal reduction for obstructive hypertrophic cardiomyopathy.

Daniel Sedehi; Gherardo Finocchiaro; Yen Tibayan; Jeffrey Chi; Aleksandra Pavlovic; Young Min Kim; Frederick A. Tibayan; Bruce A. Reitz; Robert C. Robbins; Joseph Woo; Richard Ha; David P. Lee; Euan A. Ashley

BACKGROUND Surgical myectomy and alcohol septal ablation (ASA) aim to decrease left ventricular outflow tract (LVOT) gradient in hypertrophic cardiomyopathy (HCM). Outcome of myectomy beyond 10 years has rarely been described. We describe 20 years of follow-up of surgical myectomy and 5 years of follow-up for ASA performed for obstructive HCM. METHODS We studied 171 patients who underwent myectomy for symptomatic LVOT obstruction between 1972 and 2006. In addition, we studied 52 patients who underwent ASA for the same indication and who declined surgery. Follow-up of New York Heart Association (NYHA) functional class, echocardiographic data, and vital status were obtained from patient records. Mortality rates were compared with expected mortality rates of age- and sex-matched populations. RESULTS Surgical myectomy improved NYHA class (2.74±0.65 to 1.54±0.74, p<0.001), reduced resting gradient (67.4±43.4mmHg to 11.2±16.4mmHg, p<0.001), and inducible LVOT gradient (98.1±34.7mmHg to 33.6±34.9mmHg, p<0.001). Similarly, ASA improved functional class (2.99±0.35 to 1.5±0.74, p<0.001), resting gradient (67.1±26.9mmHg to 23.9±29.4mmHg, p<0.001) and provoked gradient (104.4±34.9mmHg to 35.5±38.6mmHg, p<0.001). Survival after myectomy at 5, 10, 15, and 20 years of follow-up was 92.9%, 81.1%, 68.9%, and 47.5%, respectively. Of note, long-term survival after myectomy was lower than for the general population [standardized mortality ratio (SMR)=1.40, p<0.005], but still compared favorably with historical data from non-operated HCM patients. Survival after ASA at 2 and 5 years was 97.8% and 94.7%, respectively. Short-term (5 year) survival after ASA (SMR=0.61, p=0.48) was comparable to that of the general population. CONCLUSION Long-term follow-up of septal reduction strategies in obstructive HCM reveals that surgical myectomy and ASA are effective for symptom relief and LVOT gradient reduction and are associated with favorable survival. While overall prognosis for the community HCM population is similar to the general population, the need for surgical myectomy may identify a sub-group with poorer long-term prognosis. We await long-term outcomes of more extensive myectomy approaches adopted in the past 10 years at major institutions.


Journal of the American College of Cardiology | 2015

Systems Genomics Identifies a Key Role for Hypocretin/Orexin Receptor-2 in Human Heart Failure.

Marco V Perez; Aleksandra Pavlovic; Ching Shang; Matthew T. Wheeler; Clint L. Miller; Jing Liu; Frederick E. Dewey; Stephen Pan; Porama Thanaporn; Devin Absher; Jeffrey Brandimarto; Heidi Salisbury; Khin Chan; Rupak Mukherjee; Roda P. Konadhode; Richard M. Myers; Daniel Sedehi; Thomas E. Scammell; Thomas Quertermous; Thomas P. Cappola; Euan A. Ashley

BACKGROUND The genetic determinants of heart failure (HF) and response to medical therapy remain unknown. We hypothesized that identifying genetic variants of HF that associate with response to medical therapy would elucidate the genetic basis of cardiac function. OBJECTIVES This study sought to identify genetic variations associated with response to HF therapy. METHODS This study compared extremes of response to medical therapy in 866 HF patients using a genome-wide approach that informed the systems-based design of a customized single nucleotide variant array. The effect of genotype on gene expression was measured using allele-specific luciferase reporter assays. Candidate gene transcription-deficient mice underwent echocardiography and treadmill exercise. The ability of the target gene agonist to rescue mice from chemically-induced HF was assessed with echocardiography. RESULTS Of 866 HF patients, 136 had an ejection fraction improvement of 20% attributed to resynchronization (n = 83), revascularization (n = 7), tachycardia resolution (n = 2), alcohol cessation (n = 1), or medications (n = 43). Those with the minor allele for rs7767652, upstream of hypocretin (orexin) receptor-2 (HCRTR2), were less likely to have improved left ventricular function (odds ratio: 0.40 per minor allele; p = 3.29 × 10(-5)). In a replication cohort of 798 patients, those with a minor allele for rs7767652 had a lower prevalence of ejection fraction >35% (odds ratio: 0.769 per minor allele; p = 0.021). In an HF model, HCRTR2-deficient mice exhibited poorer cardiac function, worse treadmill exercise capacity, and greater myocardial scarring. Orexin, an HCRTR2 agonist, rescued function in this HF mouse model. CONCLUSIONS A systems approach identified a novel genetic contribution to human HF and a promising therapeutic agent efficacious in an HF model.


Journal of Cardiac Failure | 2014

Latent Obstruction and Left Atrial Size Are Predictors of Clinical Deterioration Leading to Septal Reduction in Hypertrophic Cardiomyopathy

Gherardo Finocchiaro; Francois Haddad; Aleksandra Pavlovic; Gianfranco Sinagra; Ingela Schnittger; Joshua W. Knowles; Marco V Perez; Emma Magavern; Jonathan Myers; Euan A. Ashley

BACKGROUND Exercise echocardiography is a reliable tool to assess left ventricular (LV) dynamic obstruction in hypertrophic cardiomyopathy (HCM). The aim of this study was to determine the role of exercise echocardiography in the evaluation of latent obstruction and in predicting clinical deterioration in HCM patients. METHODS AND RESULTS We considered 283 HCM patients studied with exercise echocardiography. The end point was clinical deterioration leading to septal reduction (myectomy or alcohol septal ablation). LV latent obstruction was present at enrollment in 67 patients (24%). During a mean follow-up of 42 ± 31 months, 42 patients had clinical deterioration leading to septal reduction therapy: in 12/67 (22%) patients with a latent obstruction at enrollment, in 28/84 (33%) patients with obstruction at rest, and in 2/132 (1.5%) with obstruction neither at rest or during stress. Multivariate analysis identified the following variables as independently associated with the end point: LV gradient >30 mm Hg at rest (hazard ratio [HR] 2.56, 95% CI 1.27-5.14; P = .009), LV gradient >30 mm Hg during stress (HR 4.96, 95% CI 1.81-13.61; P = .002), and indexed left atrial volume (LAVi ) >40 mL/m(2) (HR 2.86, 95% CI 1.47-5.55; P = .002). In patients with a latent obstruction, the strongest independent predictor of outcome was LAVi >40 mL/m(2) (HR 3.75, 95% CI 1.12-12.51; P = .032). CONCLUSIONS Assessment of LV gradient during stress with exercise echocardiography is an important tool for the evaluation of latent obstruction in HCM and may have a role in risk stratification of these patients.


Frontiers in Cardiovascular Medicine | 2017

Impact of a Genetic Risk Score for Coronary Artery Disease on Reducing Cardiovascular Risk: A Pilot Randomized Controlled Study

Joshua W. Knowles; Shirin Zarafshar; Aleksandra Pavlovic; Benjamin A. Goldstein; Sandra A. Tsai; Jin Li; Michael V. McConnell; Devin Absher; Euan A. Ashley; Michaela Kiernan; John P. A. Ioannidis; Themistocles L. Assimes

Purpose We tested whether providing a genetic risk score (GRS) for coronary artery disease (CAD) would serve as a motivator to improve adherence to risk-reducing strategies. Methods We randomized 94 participants with at least moderate risk of CAD to receive standard-of-care with (N = 49) or without (N = 45) their GRS at a subsequent 3-month follow-up visit. Our primary outcome was change in low density lipoprotein cholesterol (LDL-C) between the 3- and 6-month follow-up visits (ΔLDL-C). Secondary outcomes included other CAD risk factors, weight loss, diet, physical activity, risk perceptions, and psychological outcomes. In pre-specified analyses, we examined whether there was a greater motivational effect in participants with a higher GRS. Results Sixty-five participants completed the protocol including 30 participants in the GRS arm. We found no change in the primary outcome between participants receiving their GRS and standard-of-care participants (ΔLDL-C: −13 vs. −9 mg/dl). Among participants with a higher GRS, we observed modest effects on weight loss and physical activity. All other secondary outcomes were not significantly different, including anxiety and worry. Conclusion Adding GRS to standard-of-care did not change lipids, adherence, or psychological outcomes. Potential modest benefits in weight loss and physical activity for participants with high GRS need to be validated in larger trials.


Journal of Cardiovascular Translational Research | 2008

Genetics of Arrhythmia: Disease Pathways Beyond Ion Channels

Marco V Perez; Matthew T. Wheeler; Michael Ho; Aleksandra Pavlovic; Paul J. Wang; Euan A. Ashley

Diseases of the electrical conduction system that lead to irregularities in cardiac rhythm can have morbid and often lethal clinical outcomes. Linkage analysis has been the principal tool used to discover the genetic mutations responsible for Mendelian arrhythmic disease. Although the majority of arrhythmias can be accounted for by mutations in genes encoding ion channels, linkage analysis has also uncovered the role of other gene families such as those encoding members of the desmosome. With a list of candidates in mind, mutational analysis has helped confirm the suspicion that proteins found in caveolae or gap junctions also play a role in arrhythmogenesis. Atrial fibrillation and sudden cardiac death are relatively common arrhythmias that may be caused by multiple factors including common genetic variants. Genome-wide association studies are already revealing the important and poorly understood role of intergenic regions in atrial fibrillation. Despite the great advancements that have been made in our understanding of the genetics of these diseases, we are still far from able to routinely use genomic data to make clinical management decisions. There remain several hurdles in the study of genetics of arrhythmia, including the costs of genotyping, the need to find large affected families for linkage analysis, or to recruit large numbers of patients for genome-wide studies. Novel techniques that incorporate epigenetic information, such as known gene–gene interactions, biologic pathways, and experimental gene expression, will need to be developed to better interpret the large amount of genetic data that can now be generated. The study of arrhythmia genetics will continue to elucidate the pathophysiology of disease, help identify novel therapies, and ultimately allow us to deliver the individualized medical therapy that has long been anticipated.

Collaboration


Dive into the Aleksandra Pavlovic's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge