Department of Pharmacy and Pharmaceutical Sciences | 2025 Annual Report

Page 1


2025 ANNUAL REPORT

DEPARTMENT OF

PHARMACY AND PHARMACEUTICAL SCIENCES

FACULTY

FULL MEMBERS

William E. Evans, PharmD (Emeritus)

James M. Hoffman, PharmD, MS

Mark Leggas, PhD

Mary V. Relling, PharmD (Emerita)

P. David Rogers, PharmD, PhD (Chair)

John D. Schuetz, PhD

Clinton F. Stewart, PharmD

Jun J. Yang, PhD (Division Director, Pharmaceutical Sciences)

ASSOCIATE MEMBERS

Brooke Bernhardt, PharmD (Division Director, Pharmaceutical Services)

Kelly Caudle, PharmD, PhD

Cyrine Haidar, PharmD

Daniel Savic, PhD

ASSISTANT MEMBERS

Samuel Brady, PhD

Peijun Ma, PhD

Jeffrey Rybak, PharmD, PhD

Liqin Zhu, PhD

DEPARTMENT

LEADERSHIP TEAM

David Aguero, PharmD, MSTL

Carla Cook, MS

Kristine R. Crews, MS, PharmD

William Humphrey, MS, MBA, BS Pharm

John McCormick, PharmD

Alejandro Molinelli, PhD

Andrew Pappas

Steve Pate, DPh

Jennifer Robertson, PharmD

PHARMACEUTICAL SCIENCES STAFF

Mohammad Aslam Khan, PhD

Kathy Barker, PhD

Kelly Barnett, PhD

Tony Brown, PhD

LaChelle Buckley

Kami Chauncy

Divyabharathi Chepyala, PhD

Landon Choi

Erin DeWitt

Guoqing Du, PhD

Dawn Fisher

Amanda Gilbert

Wenbo Ge

Sadakatali Gori, PhD

Tomoka Gose, PhD

Murad Hasan

Jamie Horn, PhD

Yu-Chih Hsiao, MS

Sarah Jones

Julia Kincaide, MS

Elaine King

Brandon Klee, PharmD

Zhenhua Li, PhD

Xiaoli Liu, PhD

John Lynch, PhD

Andrey Matlin

Pamela McGill

Ben McKinley

Claire Mills

Robert Mobley, PhD

Sreenath Nair, PhD

Carl Panetta, PhD

Tracy Peters, MS

Nick Selvo

Hanfu Shi, MS

Ana Oliveira Souza, PhD

Abi Stolarski

Megan Walker, PhD

Yao Wang, MD

Kazuto Yasuda, PhD

Qing Zhang

Wenjian Yang, PhD

PHARMACEUTICAL SERVICES STAFF

Tekoa Akins

Delia Allen, PharmD

Eric Andrews, MS

Chris Askins, PharmD

Dina Bachor

PJ Barker, PharmD

Amy Bass, PharmD

Alysa Baumann, PharmD, MBA

Debra Black

Hui Black, PhD

Ellie Boldreghini, RN

Kymira Boone

Melissa Bourque, PharmD

Toya Bowman

Becky Boyd

Allison Bragg, PharmD

Cindy Brasher, PharmD, MS

Sharon Brittman

Felicia Brooks

Mike Brown

Thomas Bryant

Derrick Calhoun

Liz Carlton

Susan Carr, PharmD

Andy Christensen, PharmD

Misti Clark, PharmD

Rochelle Connors

Madison Cole, PharmD

Jennifer Cooper

Nicole Cox

Shane Cross, PharmD

Maria Darby

Paula Davies

LaChaka Davis

Regenia Davis

Nousheen DeRenzo, PharmD

Roseann Donnelly, PharmD

Katie Duncan

Ryan Duncan, PharmD

Nicole Edwards-Durden, PharmD

Krystal Effinger

Maureen Esposito, PharmD

Debra Ethridge, PharmD

Joseph Evans, PharmD

Tamra Filkins

Liz Gallimore, PharmD

Emma Gant, PharmD

Randy Garrett

Amanda Gillispie, PharmD

Bruce Girdler

Elbony Gray

Catina Greer

Danaka Hancock, PharmD

Torrery Hardin

Charlisa Harris

Nekia Harris

Jaclyn Hopp, PharmD

Timothy Howze, PharmD

Carlton Hughes

Kristen Hughes, PharmD

Kipp Hunter

Pat Hunter

Million Irvin

Servin Izadian, PharmD

Nevonda Jackson, PharmD

Tim Jacobs, PharmD

Sherree Johns

Clara Johnson, MS

Jennifer Kemper, PharmD

Melissa Key

Jenny Knych, PharmD

Kristen Kuehn

Terri Kuehner

Trevor Lannom, PharmD

Subrena Lesure-Johnson

David Lewis

Nichole Lewis, RN

Lie Li, PhD

Alvertis London Sr.

Maira Lopez

Natiya Lundy

William Mabry

Bertha Macklin

Shane Marshall, PharmD

Ashley Martin

Elvis Martinez

Astrid Mathis, MS

Kim McClelland, MBA

Anne McCormick, BS Pharm

Kristy McRae

Shevette Milan

Camille Miller

Thomas Mills, PharmD

Chris Moon

Ben Moore, PharmD

Heather Morgan, PharmD

Ted Morton, PharmD

Heather Mullins, PharmD

Tiffany Nason, PharmD

Geannie Navarro

Tam Nguyen

Christina Owens

Monica Patel, PharmD

Jennifer Pauley, PharmD

Trina Peery, PharmD

Jackie Quackenbush, BS Pharm

Julie Richardson, PharmD

Sheri Ring, RN

Jennifer Robertson, PharmD

Charles Rose

Margaret Russell

Robert Rutschman, PharmD

Kaley Salamon

Courtney Sartain

Linda Schiff, PharmD

Joe Sciasci, PharmD

Chris Scobey, PharmD

Nicole Sims

Camille Smith, PharmD

Kelsey Finnell Smith, PharmD

Kevin Smith, PharmD, HMBA

Terrilyn Smith

Hope Swanson, PharmD

Kelly Tartera, PharmD

Paula Taylor

Willie Taylor III

Nick Thomas, MS

Laura Todd

Tabetha Todd, PharmD

Deni Trone, PharmD

Dagny Ulrich, PharmD, PhD

Liz Velazquez

Huy Vi, PharmD

Madison Wade

Lynlie Wallach, RN

Deborah Ward, PharmD

Angela Watson

Yvette White

Courtney Wilburn Lee

Cheri Wilkerson, PharmD

Barry Williams, BS Pharm

Diana Wu, PharmD

Paula Yates

Curtis Yeh, PharmD

Keith Young, BS Pharm

POSTDOCTORAL FELLOWS

Srijan Acharya, PhD

Stefanie Baril, PhD

Akash Bhaskar, PhD

Kashi Raj Bhattarai, PhD

DEPARTMENT STAFF LISTING

Laura Doorley, PhD

Carolin Escherich, PhD

Meghan Garg, PhD

Luisa Gomez Londono, PhD

Keito Hoshitsuki, PharmD

Ruida Hou, MD, PhD

Priyanka Karmokar, PhD

Ke Liu, PhD

Qian Liu, PhD

Maud Maillard, PhD

Sabina Ranjit, PhD

Darian Santana, PhD

Gustavo Santiago-Collazo, PhD

Cheng Tian, PhD

Bailey Tibben, PhD

Rajesh Yetirajam, PhD

Satoshi Yoshimura, MD, PhD

Huanbin Zhao, PhD

PHARMACY RESIDENTS

Ashton Bellamy, PharmD

Lily Higgins, PharmD

Meghan McNulty, PharmD

Gina Peng, PharmD

Sophie Tilley, PharmD

GRADUATE STUDENTS

Farrah Altamimi

Joanne Canedo

Ashley Gray

Jeremy Hunt

Joey Miller

Kumar Niloy

Faten Usrof

Garrett Weeks

Jingwen Zhu, PhD

PHARMACY INTERNS

Jylexus Burks

Jennifer Hampton

Cameron Harrison

Monica Lewis

May Lin

Diana Makram

Mary Marcos

Madeline Matheson

Will Rhinehardt

Braxton Snuffer

Simone Yates

VISITING STUDENT

Linus Folkesson

The mission of the Department of Pharmacy & Pharmaceutical Sciences is to generate new knowledge to advance pharmacotherapy and provide the best patient care for children with catastrophic diseases. With a focus on patient care, discovery and implementation, and training and advocacy, the department comprises the Division of Pharmaceutical Sciences and the Division of Pharmaceutical Services. Both research and treatment are highly integrated at St. Jude Children’s Research Hospital, and many of our departmental faculty and staff members are extensively involved in both domains. Indeed, the synergies and efficiencies of having the research and patient care components in a single academic department have been hallmarks of St. Jude since its establishment in 1962, and they facilitate the success of our institution.

Our vision is to transform pharmacotherapy for patients with catastrophic childhood diseases. Survival rates for children with cancer, rare neurological diseases, hematologic disorders and infectious diseases continue to increase, largely through the improved use of drug therapy. Failure of current therapies and unacceptable adverse effects are partly due to suboptimal use of medications. We aim to elucidate the biological basis of inter-individual differences in pharmacologic response, translate our findings into more rational therapeutics and improve patient care.

Heterogeneity in many drugs’ metabolism, transport, elimination, targets and receptors and consequent variability in therapeutic or adverse effects may result from germline genetic differences or genetic alterations in malignant cells. Nongenetic factors, such as drug interactions, host organ function and maturity, disease severity, and adherence to therapy also influence drug response. Genetic variability in the infecting pathogen can further impact anti-infective therapy, resulting in anti-infective tolerance or resistance.

Many aspects comprise the Department of Pharmacy and Pharmaceutical Sciences vision.

Patient Care

■ Translate research findings into better treatments

■ Optimize treatments for individual patients

■ Improve overall care

Discovery and Implementation

■ Clarify biological basis of drug response differences

■ Identify genetic determinants of drug effects

■ Develop preclinical models and integrate them into translational clinical studies

Training and Advocacy

■ Faculty-led interdisciplinary St. Jude programs

■ Lead and participate in national and international research collaborations and consortia

■ Regular workshops, journal clubs and seminars

We develop preclinical models to systematically characterize the determinants of human variation in drug response and integrate our work into translational clinical studies. Laboratory work informs clinical studies, and clinical challenges drive much of our laboratory work.

Faculty members lead and participate in interdisciplinary St. Jude programs and national cooperative research collaborations. Our research integrates genome-wide analyses, molecular analyses, functional genomics, and pharmacokinetics and pharmacodynamics to identify genetic determinants of drug effects, with the long-term goal of optimizing therapy for individual patients. The Department comprises faculty members, post-doctoral fellows,

residents, undergraduate and graduate students, over 25 board-certified pharmacists, and full-time staff members working as computing experts, research nurses, technical, laboratory, administrative and clinical staff.

The department is led by David Rogers, PharmD, PhD, who serves as chair, and Jun J. Yang, PhD, and Brooke Bernhardt, PharmD, MS, vice-chairs of the Divisions of Pharmaceutical Sciences and Pharmaceutical Services, respectively.

The department is supported in part by grants from industry, foundations, and the National Institutes of Health. The research in the department includes clinical and fundamental pharmacology, pharmacokinetics, pharmacodynamics and pharmacogenomics. The Division of Pharmaceutical Sciences occupies over 15,000 square feet of contiguous, state-of-the-art laboratory and office space, and the Division of Pharmaceutical Services occupies over 18,000 square feet of space in the clinical areas of St. Jude. The department hosts weekly research workshops and journal clubs, as well as monthly seminars that are open to the entire institution. Additionally, we host multiple laboratory- and services–specific meetings, webinars with national and international colleagues, and regular pharmacogenomics meetings.

Chair: David Rogers, PharmD, PhD

Research and clinical implementation often inform each other in a continuous cycle.

Basic Research Integrated Models in Translational Studies

Clinical Questions or Problems

Learn more about our department and the work we do:

Clinical Research

Division Director of Pharmaceutical Sciences: Jun J. Yang, PhD

Division Director of Pharmaceutical Services: Brooke Bernhardt, PharmD, MS

About the Chair

David Rogers, PharmD,PhD

Member and Chair

St. Jude Endowed Chair in Pharmaceutical Sciences

Scan the QR code to view of Dr. Rogers’ lab website

David Rogers, PharmD, PhD, serves as Chair of the Department of Pharmacy and Pharmaceutical Sciences at St. Jude and holds the St. Jude Endowed Chair in Pharmaceutical Sciences. He received his Doctor of Pharmacy degree from the University of Tennessee and his Master of Science and Doctor of Philosophy in Microbiology and Immunology from the University of Mississippi. Dr. Rogers completed his residency in pharmacy practice (PGY1) at the Regional Medical Center, Memphis, and residency (PGY2) and clinical fellowship in infectious diseases pharmacy practice at the University of Mississippi Medical Center.

Before joining the faculty at St. Jude, he was a Professor of Clinical Pharmacy and Translational Science at the University of Tennessee College of Pharmacy. There, he also held the First Tennessee Endowed Chair of Excellence in Clinical Pharmacy, served at various times as Vice Chair of the Department of Clinical Pharmacy and Translational Science, Associate Dean for Clinical and Translational Research, and Director of the UTHSC Center of Excellence for Pediatric Experimental Therapeutics. He is an elected Fellow of the American College of Clinical Pharmacy (ACCP) and the American Academy of Microbiology. He has served on the Board of Trustees for the ACCP Research Institute and on the Board of Directors for the Society of Infectious Diseases Pharmacists. In 2021, he received both the ACCP Russell R. Miller Award and the ACCP Therapeutic Frontiers Lecture Award. He recently completed a six-year term as a member of the NIH Drug Discovery and Mechanisms of Antimicrobial Resistance (DDR)

Study Section, and he currently serves on the Board of Trustees of the ACCP Foundation and has been appointed as an Ex Officio Member of the National Academies Forum on Antimicrobial Threats. Dr. Rogers’ research program is focused on improving antifungal pharmacotherapy by studying how pathogenic fungi develop resistance to antifungal agents. He has authored over 300 publications and scientific abstracts, and his laboratory has been continuously funded by the NIH for over two decades.

Division of Pharmaceutical Sciences

The core mission of the Division of Pharmaceutical Sciences is to generate new and fundamental knowledge about how to individualize drug therapy for children with catastrophic diseases. Basic science has always been represented in our research portfolio, with faculty exploring fundamental biology of drug transport, metabolism, toxicity and resistance. We continue to build on our groundbreaking efforts in pharmacogenomics and lead the world in the discovery and clinical implementation of pharmacogenomics-guided precision medicine. Our faculty study genomic and epigenomic architecture of drug phenotypes, develop more accurate models of pediatric cancers for preclinical evaluation, and elucidate the molecular basis of resistance to anticancer and antiinfective agents. The Division integrates expertise in pharmacokinetics, pharmacodynamics, and pharmacogenomics with state-of-the-art technologies to drive discovery research in therapeutic science across disease specialties. Importantly, our unique strength also lies in the ability to translate our laboratory discoveries into patient care, in some cases shifting the paradigm of how pharmacotherapy is practiced. Housed on the 5th floor of the Chili’s Care Center, the laboratories of the division faculty provide superb infrastructure for translational research and an outstanding training environment for graduate students, postdoctoral fellows, and clinician-scientist trainees.

Jun J. Yang, PhD

Member, Division Director

Division of Pharmaceutical Sciences

St. Jude Endowed Chair in Pharmacogenomics

Dr. Jun J. Yang is a Member and holds the St. Jude Endowed Chair in Pharmacogenomics and serves as the Director for the Division of Pharmaceutical Sciences. His research focuses on pharmacogenomics of anticancer drugs in children with a particular interest in childhood acute lymphoblastic leukemia (ALL). The goal is to ultimately develop pharmacogenomics-guided treatment strategies to personalize therapy for children with catastrophic diseases.

Toward this effort, in 2024, Dr. Yang and his team published work that analyzed the single-cell transcriptomes of human B-cells, leading to the discovery that the developmental state of leukemia cells correlates with asparaginase sensitivity and the identification of a potential target to enhance aspariginase efficacy pharmaceutically. The work emphasized the potential of single-cell systems pharmacology to predict effective combination therapies based on the developmental heterogeneity within leukemia.

The Yang lab continued exploring how genetic variants affect thiopurine toxicity and tolerability in children with ALL. With findings regarding lower dose tolerations in patients of Hispanic heritage with TPMT mutations and NUDT15 genetic variants, the work highlights the importance of genetic testing, particularly in Hispanic populations, for TPMT and NUDT15 variants to guide thiopurine dosing in pediatric ALL patients to reduce toxicity and improve therapeutic outcomes.

In an additional effort, the team published an investigation into how age influences drug sensitivity and treatment responses in B-cell ALL (B-ALL). Specifically, they showed that children exhibited greater sensitivity to mercaptopurine, independent of leukemia subtype, stressing the need for age-specific treatment considerations.

Dr. Yang continues research on drug-induced liver toxicity through an innovative collaborative project that has challenged our scientists’ ingenuity and maximized our shared resources. The team developed an ultrahigh parameter flow cytometry assay using 48 distinct fluorophores to precisely track cellular changes in the liver following drug treatment. By integrating these findings with single-cell sequencing, spatial-omics and data from a rich clinical cohort, they are uncovering the fundamental mechanisms of drug-induced liver toxicity while generating promising avenues for potential interventions.

Scan the QR code to view of Dr. Yang’s lab website

Samuel Brady, PhD

Scan the QR code to view all of Dr. Brady’s lab website

Samuel Brady, PhD, joined the Department of Pharmacy and Pharmaceutical Sciences in 2023. Since joining, he has established his lab and hired staff to support their research endeavors.

The overarching objective within the Brady lab is to improve cancer treatment and reduce toxicity using genomics as a starting point. Genomic alterations in a tumor can predict response or resistance to various therapeutics; the Brady lab aims to understand why and how this occurs so that therapies can be tailored appropriately.

The team also uses mutational signatures to understand how existing therapies may induce secondary cancers in survivors later in life. Many pediatric cancer survivors will undergo treatment and be cured of their initial cancer but then develop a second cancer due to their prior therapy. The Brady lab uses mutational signatures and other genomic techniques to understand how therapy can induce specific mutations that initiate second cancers. The results of this work could help inform which therapies should be reduced to a lower dosage and which should be avoided entirely in certain patient populations. Therapy-induced mutational signatures can also fuel tumor heterogeneity and drug resistance. Thus, they may help identify approaches to sequence therapies so that highly mutagenic therapies are avoided during certain phases of therapy when drug resistance is more likely to develop.

Finally, the Brady lab aims to tackle new therapeutic targets using existing or new therapies. Many current cancer therapies are toxic, which is a significant

problem for pediatric patients because their bodies are still developing. In order to develop treatments that will have the maximum impact on cancer cells with minimal impact on healthy tissue, the team is working to identify the genes that are unnecessary for human survival but vital to a cancer cell’s survival. This research will allow the identification of existing drugs that can target the function of these genes or the development of new therapies to target specific proteins and minimize treatment toxicity.

William E. Evans, PharmD

Scan the QR code to view all of Dr. Evans’ lab website

The Evans lab examined the pharmacogenomics of anticancer agents, specializing in childhood acute lymphoblastic leukemia (ALL). Evans and his team aimed to elucidate genomic determinants of toxicity and efficacy of anticancer agents and translate this knowledge into new diagnostics and treatment strategies to optimize ALL therapy. They achieved their aims by evaluating patient cohorts’ leukemia cells for drug sensitivity and clinical response in prospective clinical trials. They used comprehensive DNA and RNA sequencing to identify genes and genome variations that are important determinants of antileukemic agents’ disposition and effects.

The Evans lab closed in 2021 and no longer accepts students or post-doctoral trainees. As an emeritus faculty member, he and his collaborators continue to analyze and report data generated over the last several years to improve the safety and efficacy of medications used to treat children with cancer.

Mark Leggas, PhD

for Translational Pharmacology

Scan

the QR code to view all of Dr. Leggas’ lab website

Dr. Mark Leggas is a Member of the Department of Pharmacy and Pharmaceutical Sciences and the Director of the Center for Translational Pharmacology. The Leggas Lab has a vested interest in translational pharmacology to optimize the clinical use of existing drugs or new drug candidates. The lab has a longstanding project to develop a new targeted therapy for Ewing sarcoma and is concluding a project assessing the utility of a non-opioid medication to treat neonates physically dependent on opioids. A major new effort aims to optimize lymphodepletion and use patient metabolic phenotyping to improve outcomes in patients treated with adoptive cell therapies such as CAR T–cell products.

The Ewing sarcoma project focuses on semisynthetically modified mithramycin analogs, which have shown the ability to interfere with the oncogenic transcription factor responsible for Ewing sarcoma growth. This is significant because this oncogenic transcription factor (i.e., EWS-FLI1) is recognized as the Achilles’ heel of Ewing sarcoma yet attempts to develop treatments that disrupt its function have failed. Currently, several analogs are advancing through preclinical evaluation and, if successful, will move to the NCI Experimental Therapeutics program to complete all the preclinical assessments needed for entry into the clinic.

The neonate project is centered around a randomized controlled clinical trial that evaluated the use of clonidine, an alpha-2 adrenoceptor agonist, compared to morphine for treating neonates born physically

dependent on opioids due to prenatal exposure. In 2024, the Leggas Lab further advanced the study of neonatal opioid exposure by publishing clinical trial results and a pharmacometric model to optimize clonidine dosing regimens. These efforts have positively contributed to the treatment of neonatal opioid withdrawal syndrome (NOWS).

The CAR T–cell project aims to optimize lymphodepletion conditioning therapy for all CAR T–cell products under development at St. Jude. This is essential because the effects of lymphodepleting agents on CAR T–cell kinetics and pharmacodynamics have not been systematically studied. Additionally, the lab is developing advanced analytical methods and informatics platforms that will enable comprehensive metabolic phenotyping of patients receiving CAR T–cell treatment. This is important because host-related small molecules derived from physiological processes, nutritional status, or environmental sources may significantly influence CAR T–cell signaling. Ultimately, biomarkers of CAR T–cell disposition (similar to drug metabolism) and phenotype in relation to efficacy and toxicity (pharmacodynamics) will be developed, along with semi-mechanistic models that establish exposure (by phenotype) – response relationships.

Peijun Ma, PhD

Scan the QR code to view all of Dr. Ma’s lab website

Dr. Peijun Ma joined the Department of Pharmacy and Pharmaceutical Sciences as an Assistant Member in 2023. Her previous research focused on developing bacterial single-cell RNA sequencing technology and understanding the evolution of antibiotic resistance in gram-negative bacterial pathogens. At St. Jude, Dr. Ma’s research group aims to investigate bacterial heterogeneity and its impact on the evolution of antibiotic resistance and persistence using bacterial single-cell transcriptomics.

The emergence of antibiotic resistance and persistence can directly lead to treatment failure, posing a significant threat to human health. A major issue in our current efforts to combat antibiotic resistance and persistence is the predominant characterization of bacteria as whole populations. However, resistant and persistent clones typically arise from single-cell events. The heterogeneity in bacterial populations has not been well-characterized, and the mechanisms leading to this heterogeneity remain unclear. To address this problem, the Ma Lab will apply innovative bacterial single-cell transcriptomics to characterize bacterial drug-induced heterogeneity at the single-cell level. Additionally, Ma’s team will characterize the microbiome using bacterial single-cell transcriptomics to understand the evolution of antibiotic resistance and persistence in the context of the host microenvironment, particularly in pediatric patients with recurrent bacterial infections. This work will provide novel and exciting insight into bacterial heterogeneity and lead to the identification of drug targets for treating bacterial infections associated with antibiotic resistance and persistence.

Mary V. Relling, PharmD

Scan the QR code to view all of Dr. Relling’s lab website

Mary Relling, PharmD, became a faculty member in the Department of Pharmaceutical Sciences at St. Jude in 1988 and served as department chair from 2003 to 2020. Her primary research efforts focused on translational research in childhood acute lymphoblastic leukemia (ALL), aiming to identify the host- and treatment-related risk factors for adverse treatment outcomes. The Relling lab’s use of precision medicine approaches to tailor therapeutic dosages enabled them to incorporate pharmacogenetically-based dosage adjustments in ALL trials and for all other St. Jude patients via the clinical protocol PG4KDS.

Relling co-founded the Clinical Pharmacogenetics Implementation Consortium (CPIC), an NIH-supported genomics resource focused on facilitating the clinical implementation of germline pharmacogenetic testing. After stepping down as department chair and moving to part-time status in 2021, Relling retired in June of 2023, but her collaborators continue to build on her tireless work.

DIVISION

Jeffrey M. Rybak, PharmD, PhD

Scan the QR code to view of Dr. Rybaks’s lab website

Jeffrey M. Rybak, PharmD, PhD, joined the Department of Pharmacy and Pharmaceutical Sciences faculty at St. Jude Children’s Research Hospital in September 2020. Previously, his research focused on discovering the molecular mechanisms underpinning antifungal resistance among challenging fungal pathogens such as Aspergillus fumigatus and Candidozyma (Candida) auris. Employing whole genome and transcriptome sequencing, in vitro evolution studies and targeted allelic replacement, Rybak and his colleagues revealed mutations in the A. fumigatus HMG-CoA reductase gene, hmg1, and the C. auris zinc-cluster transcription factor (ZCF) gene, TAC1B, as widespread genetic determinants of clinical triazole antifungal resistance.

Rybak’s current research focuses on advancing the treatment of invasive fungal infections by developing new therapeutic strategies to overcome difficult-totreat fungal pathogens. In pursuit of this objective, the Rybak lab has developed the C. auris-optimized Episomal Plasmid Induced Cas9 (EPIC) system, capable of single nucleotide-editing, and used this system to interrogate the role of the fungal-specific zinc-cluster transcription factors (ZCF) in regulating the response to antifungal-induced stress in C. auris. They have demonstrated that targeted disruption of one of these ZCF genes, UPC2, results in ablation of the normal C. auris transcriptional response to triazole treatment, reduced production of the key fungal sterol ergosterol, and diminished resistance to triazole antifungals. Furthermore, the Rybak lab has observed that while triazoles are conventionally limited to fungistatic activity against species of Candida, disruption of C. auris UPC2 conferred rapid fungicidal activity to the triazole

agents at pharmacologically achievable concentrations both in vitro and in vivo. Their ongoing studies seek to characterize the C. auris UPC2 regulatory network and to reveal molecular weak points that can be targeted to restore and enhance the activity of the triazoles and amphotericin B.

The Rybak lab also seeks to advance the treatment of invasive mold infections. Recently, they demonstrated that a specific group of commonly observed mutations in the cyp51A gene, which alter the glycine-54 residue (G54), confer increased resistance to long-lipophilic triazoles such as posaconazole while creating a previously unknown collateral sensitivity to compact triazoles. They then demonstrated that this vulnerability could be therapeutically exploited to treat even pantriazole-resistant A. fumigatus clinical isolates with G54 altering mutations by utilizing targeted combinations of clinically available triazoles in a murine model of pulmonary aspergillosis.

In addition to his benchtop research, Rybak is a member of the Antimicrobial Utilization and Improvement Committee (AUIC) and co-authored the updated St. Jude Antifungal Utilization Guidelines.

Daniel Savic, PhD

Dr. Daniel Savic leads a research program focused on understanding how the noncoding genome impacts chemotherapeutic drug resistance and treatment outcomes in childhood acute lymphoblastic leukemia (ALL). His laboratory employs advanced genomic techniques, including chromatin accessibility profiling (ATAC-seq), high-throughput chromosome conformation capture (Hi-C), massively parallel reporter assays and epigenomic CRISPR-based screening, to identify gene regulatory disruptions impacting the pharmacogenomics of childhood ALL treatment. The research program encompasses three main areas. First, the laboratory investigates cis-regulatory element activity and gene regulatory network structure in ALL. This work led to the creation of the largest chromatin accessibility dataset in ALL to date, analyzing primary cells from over 150 patients across distinct molecular subtypes. Complementary to ALL chromatin accessibility data, capture Hi-C data was collected for select ALL cell samples to guide association of cisregulatory elements with genes to better define gene regulatory networks.

Second, the Savic lab maps cis-regulatory elements and gene regulatory responses to antileukemic agents that impact drug resistance. This research has revealed important insights into glucocorticoid drug resistance mechanisms, including the discovery of genomic interactions between glucocorticoid and canonical Wnt signaling pathways that affect ALL cell survival. This finding suggests that Wnt signaling antagonists could be beneficial for patients with glucocorticoid-resistant disease.

Third, the Savic lab examines how noncoding genetic variants influence drug resistance, treatment response and relapse in ALL patients. This work culminated in the most extensive functional investigation to date of the impact of inherited noncoding genetic variants on ALL pharmacogenomics using massively parallel reporter assays.

The ultimate objective of Dr. Savic’s research is to advance precision medicine in childhood ALL by developing a deeper understanding of genome function and the gene regulatory factors that lead to chemotherapy failure. Beyond his primary research role in the Department of Pharmacy and Pharmaceutical Sciences, Dr. Savic maintains important affiliations with the Hematological Malignancies Program at St. Jude, the Pharmacogenomics Global Research Network, and the Children’s Oncology Group, enabling broader collaboration and impact in the field.

Scan the QR code to view of Dr. Savic’s lab website

John D. Schuetz, PhD

Member

Scan the QR code to view all of Dr. Schuetz’s lab website

Investigations in the John Schuetz lab determine how transporters and metabolic pathways impact disease, cancer and therapeutics.

Dyschromatosis universalis hereditaria (DUH) is a rare skin pigmentation disorder linked to the protein ABCB6 — a member of the ABC family of proteins responsible for transmembrane transport with a responsibility in cellular toxin removal. Relatively little is understood about the long list of other DUH symptoms, which range from hearing deficits to learning difficulties. Prior to our genetic findings, a link to hearing impairments was unknown. By complementary biochemical and biophysical approaches to study ABCB6 mutations identified in DUH patients, the Schuetz lab found an unexpected link between ABCB6 and hearing deficits, implying that certain cases of hearing deficits may be directly attributable to defects in the protein.

We also recently identified a medulloblastoma tumorderived somatic mutation (Q393K) that occurred in a highly conserved amino acid across mammalian species. This mutation did not affect expression or localization but nonetheless impaired function. Structural modeling and molecular dynamics simulations based on ABCG2 cryo-EM structures suggested that the Q393K interacts with E446 to create a strong salt bridge that stabilized the transporter in anon-functional, inward-facing conformation. This finding has implications in the communication needed between substrate binding and transport domains.

The findings of the Schuetz Lab reveal insights into key transporters and pathways that can help guide the development of novel therapeutic approaches in the future. These efforts and discoveries led to Dr. Schuetz’s election as Chair of AAAS section S (Pharmaceutical Sciences) and his nomination as Chair of the 2027 Gordon Research Conference (GRC) on multidrug efflux pairs with duties in 2025 as Vice Chair. Schuetz was also selected as the inaugural editor of ASPET Discovery, a new journal ASPET (American Society for Pharmacology and Experimental Therapeutics), which launched in January 2025.

Clinton F. Stewart, PharmD

Scan the QR code to view all of Dr. Stewart’s lab website

Dr. Clinton Stewart joined St. Jude in 1991, and during his tenure his work has focused on translational research of anti-cancer drugs in children with solid tumors and brain tumors. The Stewart Lab has addressed clinically relevant problems encountered in the therapy of children with brain tumors, primarily the central nervous system (CNS) penetration of novel compounds used to treat pediatric brain tumors. The process used includes tumor subgroup-specific models of pediatric CNS tumors grown in mice, cerebral microdialysis sampling of tumor extracellular fluid and ventricular cerebrospinal fluid, drug measurement using mass spectrometry, and pharmacokinetic modeling and simulation of the derived data to directly assess the unbound partition coefficient for the drug under study.

As little is known about the disposition of most anticancer agents in children with cancer, the Stewart Lab has collaborated with clinical investigators to conduct comprehensive pharmacology studies in clinical trials. These studies include pharmacokinetic, pharmacogenetic, and pharmacodynamic studies of novel drugs to gain a better understanding of the variability in drug disposition in children with cancer. The lab has contributed to studies that advance the understanding of pharmacologic therapies across diseases states such as medulloblastoma, glioma, AT/RT, neuroblastoma, retinoblastoma and sarcoma.

Beyond these contributions, the Stewart Lab has translated findings from the lab to clinic for several drugs. For example, we found that mice treated with

topotecan had better antitumor responses. This led to a series of studies where Bayesian analysis of pharmacokinetic data determined a patient’s system clearance and allowed for individualized topotecan dosage. According to our most recent research, this can be done with a simple single-sample approach.

Further delving into clinical dosing, sorafenib improves outcomes in adult hepatocellular carcinoma, but not without toxicities. Both efficacy and toxicity are related to sorafenib systemic exposure, which is highly variable in children based upon studies conducted at St. Jude. Thus, as part of an ongoing clinical trial our lab conducted a study of pharmacokinetically-guided sorafenib dosing. A primary objective of the trial was to determine if sorafenib systemic exposure can be targeted to a systemic exposure by day 21 of course 1 in 60% of patients when given in combination. The safety run-in phase met the objective of successfully targeting at least 60% of the individuals. The median targeted sorafenib dosage was 41% lower than the conventional dose of 90 mg/m2 and we have not observed any grade 3 or higher sorfenib-related toxicities.

Dr. Liqin Zhu, Assistant Member in the Department of Pharmacy and Pharmaceutical Sciences, studies the molecular and cellular mechanisms driving the metastasis and drug resistance of pediatric and adult liver cancer. Her research aims to determine the role of the host liver microenvironment in liver cancer metastasis and investigate the newly acquired vulnerability of liver tumor cells induced by standard treatment to identify new drug targets for adjuvant therapies.

Using multiple metastatic models of pediatric and adult liver cancer, the Zhu Lab found that there is a developmentally prometastatic niche to pediatric hepatoblastoma in the neonatal liver mediated by the Cxcl1/Cxcr2 axis. Further analysis showed it as an ongoing competitive process between the attempt of the activated myofibroblasts as part of the host liver defense to block local growth of the tumor, and that of tumor cells to break through the suppression and metastasize in a Vcam1-dependent manner. In work that advances therapeutic knowledge, Zhu demonstrated there is a dynamic, reversible switching between the two ribonucleotide reductase M2 subunits, RRM2 and RRM2B, that supports the transition of hepatoblastoma cells between a growing state and a surviving state under standard chemotherapy. In 2023, Dr. Zhu also published work showcasing differences in hepatocellular iron metabolism between male and females with hepatocyte ferroptosis, accounting for sexual dimorphism in liver diseases.

Through these studies, it has become evident that the molecular and cellular networks supporting liver tumor metastasis and drug response are highly plastic and dependent on changes in both intra- and extra-tumoral microenvironment.

Center for Translational Pharmacology

The Center for Translational Pharmacology advances the implementation of pharmacokinetic and pharmacodynamic translational research at St. Jude. The mission of the Center is to centralize high-quality, competitively funded, peer-reviewed clinical pharmacology research in the context of clinical trials.

Director

Mark Leggas, PhD

Associate Director

Research Operations

Kristine Crews, MS, PharmD

Associate Director

Bioanalytical Operations

Clinton Stewart, PharmD

Equipped with state-of-the-art equipment and experienced personnel, the Center is positioned to increase the impact and value of clinical trials at St. Jude by providing expertise in pharmacokinetic and pharmacodynamic study design, developing and applying novel mathematical models, and providing high-quality data. The staff assists with building electronic sample orders, laboratory test procedures, and educating clinical staff on protocol-specific procedures. Dedicated research nurses facilitate proper sample collection and logistical support to ensure efficient sample handling, processing, storage, and distribution. The Center offers bioanalytical assay development and validation under FDA guidelines and sample analysis for accurate measurement of drug and metabolite concentrations in plasma, blood, and other relevant biological matrices. The Center also offers state-of-the-art mathematical modeling and simulation, providing quantitative input toward rational study design and interpretation. Services are available to all faculty who initiate St. Jude trials as well as to those participating in externally collaborative projects, industry-sponsored studies, and consortium-led clinical initiatives.

The Pharmacotyping Resource

Over the past 40 years, pharmacogenomics has been a central research endeavor for the Department of Pharmacy and Pharmaceutical Sciences at St. Jude. Our discoveries of the genetic determinants of drug toxicity and response have fundamentally improved pediatric cancer therapy and made St. Jude a leader in pharmacogenomics-driven precision medicine. Pharmacotyping — defining interpatient variability in drug sensitivity — is the starting point for correlating drug phenotype to patient genomic profiles.

Director

Jun J. Yang, PhD

Associate Director

Kristine Crews, MS, PharmD

The Pharmacotyping Resource, located within the Division of Pharmaceutical Sciences, supports the opportunity to “embrace the challenge to create a new tomorrow.” The mission of the Pharmacotyping Resource is to leverage drug screening technology to enable collaborative pharmacogenomics research that improves cancer treatment outcomes. The Resource has supported research leading to discoveries that have uncovered novel mechanisms of drug resistance and have pointed to strategies for overcoming them.

The Resource offers a state-of-the-art imaging platform to test drug sensitivity profiles (pharmacotyping) of primary patient tumor cells as a functional precision medicine method. A team of research technologists, led by Jun J. Yang, PhD and Kristine Crews, PharmD, operates the Resource with expertise in high-content imaging, deep machine learning, drug assay development, preclinical drug evaluation and research informatics. Building upon our strength in this field, we use a modern pharmacotyping platform to discover new therapeutic opportunities for leukemia.

To this end, the Pharmacotyping Resource is equipped with a PerkinElmer Operetta CLS high-content analysis system for imaging-based drug sensitivity testing. The Operetta CLS is an automated confocal spinning disk fluorescence microscope. It is capable of imaging in 96 or 384 well plate formats. This imager is combined with the PerkinElmer Harmony data analysis software that allows for quantitative image analysis with image analysis capabilities powered by machine learning.

The team offers a pharmacotyping assay validated in tumor cells collected from patients treated for leukemia in clinical trials run by the Hematological Malignancies Program. Leukemia cells obtained from patients are tested for ex vivo sensitivity to 40 anti-leukemia drugs, and genomic profiling is done in parallel by whole genome sequencing and other assays. Our published data shows wide variability among patients in drug sensitivity and an association between sensitivity profiles and event free survival. These findings point to therapeutic implications of genomic alterations in acute lymphoblastic leukemia (ALL). The integration of tumor cell sensitivity measures with genomic data advances pharmacogenomics discovery research at St. Jude and guides the next generation of pediatric cancer therapy.

Division of Pharmaceutical Services

Pharmaceutical Services is led by Brooke Bernhardt, PharmD and is staffed by pharmacists, pharmacy technicians, research and administrative staff and faculty, all dedicated to providing patients the best pharmaceutical care possible at St. Jude while supporting a collective research endeavor. Working with other clinicians in a cutting-edge and highly collaborative environment, our personnel ensure the best possible drug therapy outcomes. Nearly 180 pharmacy staff are involved in these care efforts. Their work emphasizes integrating optimal conduct of St. Jude’s medication-related clinical research, helping to fulfill our organizational mission to “advance cures and means of prevention for pediatric catastrophic diseases through research and treatment.” Over 30 pharmacists are certified as specialists by the Board of Pharmacy Specialties, and four other pharmacists carry at least one credential from other certifying organizations, testifying to the staff’s commitment to a deep understanding and high level of clinical practice of pharmacotherapy and research.

Pharmaceutical Services had several significant achievements in 2024. Specialty Pharmacy Services successfully completed its reaccreditation with ACHC, securing dual accreditation for another three years. Home Infusion Pharmacy Services had two significant achievements in 2024. They were one of six early adopters of Epic’s home infusion software and went live in December 2024; this was a considerable team achievement that included both our Home Infusion team and our Pharmacy Informatics team. The Home Infusion Pharmacy also completed its reaccreditation survey with Accreditation Commission for Health Care, Inc. (ACHC), which provides them with another three-year accreditation term. Pharmaceutical Services won the Tennessee Pharmacist Association Excellence in Innovation award for the group’s work on St. Jude’s first domicile-based medication dispensing kiosk. On the pharmacy education front, the St. Jude Continuing Pharmacy Education (CPE) program has expanded its reach by identifying and working with internal and external strategic partners. These partnerships include St. Jude Grand Rounds, the Children’s Oncology Group (COG) and the Midsouth chapter of ACCP (MSCCP), to name a few. Doing so facilitated over 20 CE programs during the 2024 calendar year that awarded nearly 1000 continuing pharmacy education hours. Lastly, the Midsouth Pharmacy Residents Conference continues to grow in its 16th year, expecting more than 150 residents from the Midsouth to present their research and collaborate.

Brooke Bernhardt, PharmD, MS

Associate Member, Division Director

Division of Pharmaceutical Services

Chief Pharmacy Officer

Scan the QR code to view all of Dr. Bernhardt’s lab website

Brooke Bernhardt, PharmD, MS, joined St. Jude in 2023 as Associate Member, Division Director, and Chief Pharmacy Officer for the Division of Pharmaceutical Services. As a board-certified pediatric and oncology pharmacist, Dr. Bernhardt brings a wealth of experience to her leadership roles within the department.

Her interests encompass pharmacogenomic factors that influence drug disposition, metabolomics, toxicity, and survival in children with cancer. She works in close collaboration with other department faculty as they research the role of pharmacogenomics and examine disparate outcomes across different populations of children with cancer.

Dr. Bernhardt has also served as a pharmacist leader in cooperative group clinical trials as an NCI-funded pharmacist for the Children’s Oncology Group (COG) and as the Vice-Chair of the Pharmacy Discipline within COG. In this capacity, she has also contributed to over 50 cooperative group clinical trials through COG and the NCI’s Pediatric Early Phase Clinical Trials Network (PEP-CTN). She continues to lead in this space and is actively designing new studies leveraging COG data, including data obtained through the NCI’s Molecular Characterization Initiative, through a collaboration between the COG Pharmacy Discipline and COG Epidemiology Domain Committee.

As the senior leader of the Division of Pharmaceutical Services, Bernhardt strives to ensure that the Division provides the highest level of pharmaceutical care to the patients of St. Jude. To achieve this, the Division collaborates closely with the Division of Pharmaceutical Sciences to optimize opportunities for translational care. The Division also collaborates with other clinical departments, including Oncology, Hematology, and Epidemiology and Cancer Control. The Division of Pharmaceutical Services is also committed to training the next generation of pharmacists, with a significant focus on training pharmacy students and pharmacy residents. Bernhardt has created a new Clinical Pharmacy Fellowship in Pharmacoepidemiology and Pharmacogenomics to allow for additional subspecialty research training and has recruited the first fellow to start in FY25. Finally, Bernhardt is leading the launch of two new initiatives for the Division, specifically clinical and research programs in Clinical Pharmaceutics and Pharmacoeconomics.

Kelly Caudle, PharmD, PhD

Scan the QR code to view all of Dr. Caudle’s lab website

Dr. Kelly Caudle is an Associate Member within the Department of Pharmacy and Pharmaceutical Sciences. Through global collaboration, Dr. Caudle’s lab mission is to curate and synthesize knowledge using rigorous scientific standards to support informed prescribing decisions and enhance medication-related health outcomes. With a vision for widespread integration of pharmacogenomics and other medication safety measures into standard health care practice worldwide, Dr. Caudle is dedicated to ensuring that every patient receives personalized, effective and safe medication — ultimately improving outcomes and reducing health care costs.

As the Principal Investigator and Director of the Clinical Pharmacogenetics Implementation Consortium (CPIC)—a National Institutes of Health (NIH)-funded consortium with over 700 members globally—Dr. Caudle leads efforts to develop and disseminate freely available, peer-reviewed and updatable gene-drug clinical practice guidelines. In addition to pharmacogenomic guideline development, her research focuses on implementation science, addressing the challenges clinicians face in adopting pharmacogenomic testing and other medication safety initiatives. As a clinical pharmacist and scientist, she is uniquely positioned to identify these barriers and drive solutions. Her work also extends to the standardization of pharmacogenomics, ensuring its seamless integration into clinical practice. She actively collaborates with the Pharmacogenomics Program to implement these guidelines and incorporate pharmacogenomic testing into patient care at St. Jude.

Cyrine-Eliana Haidar, PharmD

Scan the QR code to view all of Dr. Haidar’s lab website

Cyrine-Eliana Haidar, PharmD, joined St. Jude Children’s Research Hospital in 2004 and was promoted to an associate faculty member in January 2024. Her work within the Department of Pharmacy and Pharmaceutical Sciences implements preemptive pharmacogenomic testing into clinical practice, which helps clinicians tailor the pharmacotherapy of patients based on their pharmacogenomic profile. This involves developing ways to best integrate pharmacogenomics testing into the electronic health record. For clinicians, this creates easy access and utilization of the pharmacogenomic test results during their day-to-day patient treatment. In addition, patients are taught about the lifelong implications of their pharmacogenomic test results, which hopefully empowers them to share these results with their non-St. Jude health care providers when they return home.

In 2023, Haidar focused on starting the Pharmacogenomics Clinic at St. Jude. This clinic saw its first patient in November of 2023 and focuses on the return of pharmacogenomic test results to patients and their families. Additionally, she has worked closely with Epic in establishing and coleading the Pharmacogenomics Brain Trust, Epic’s first genomic-based brain trust. Drawing from the St. Jude pharmacogenomic implementation expertise and Epic’s experience, the Brain Trust’s goal is to create robust and standardized support for pharmacogenomics within Epic and across organizations. This would enable a more seamless integration of pharmacogenomics within the electronic health record frameworks.

Haidar’s clinical research explores the impact of patients’ genetics combined with certain medications

or dosage levels on treatment outcomes. Notably, her collaboration with Jun J. Yang, “Clinical actionability of the NUDT15*4 (p.R139H) allele and its association with Hispanic ethnicity,” was published in August 2024 in the Clinical Pharmacology and Therapeutics and investigates the relationship between the rare NUDT15*4 variant and tolerance to thiopurine medicines.

James M. Hoffman, PharmD, MS

Scan the QR code to view all of Dr. Hoffman’s lab website

James Hoffman, PharmD, MS, joined St. Jude and the Department of Pharmaceutical Sciences in 2004. In 2015, he took on the hospital leadership role of Chief Patient Safety Officer, and in 2022, he took on additional responsibilities as Senior Vice President –Quality and Safety.

Hoffman has focused on identifying and evaluating risk for patient harm and improving patient safety. He concentrates on refining existing tools and identifying new means of patient safety event detection, improving patient safety culture, and developing and improving informatics and clinical decision support at St. Jude. Hoffman continues to lead the St. Jude transition to DNV as the institution’s accreditation partner, which helps further develop the St. Jude quality system. The DNV process started in 2022, with annual surveys beginning in 2023; the current focus is to progress toward the IS0 9001 quality standard.

Hoffman builds the reputation of St. Jude as an academic leader in quality, patient safety and improvement science for children with catastrophic diseases. In 2024, he was the senior author on a publication in Hospital Pediatrics on the St. Jude effort to optimize the use of dose error reduction software on intravenous infusion pumps. This paper builds on the commitment of St. Jude to deploy the latest medication-use technology and verify it is used to its full potential to guide safe care. Hoffman is deeply engaged with the Institute for Healthcare Improvement (IHI). He was a co-author on proceedings from the international meeting’s science stream published in BMJ Leader. At the IHI annual forum, he presented multiple times, including on the role of the Chief Quality

Officer and the potential to use artificial intelligence in improvement science.

Hoffman works to implement pharmacogenomics as a patient safety strategy. These efforts include PG4KDS: Clinical Implementation of Pharmacogenetics at St. Jude and internationally through the NIH–funded Clinical Pharmacogenetic Implementation Consortium (CPIC).

Hoffman remains a leader in various topics relevant to pharmaceutical policy and pharmacy practice. He serves as Deputy Editor for the ASHP Pharmacy Forecast, an annual report he has contributed to for over a decade that provides strategic advice for health system pharmacy leaders. He is also an author of an annual paper forecasting drug expenditures, and he serves as the contributing editor for content strategy for the American Journal of Health-System Pharmacy.

The Clinical Pharmacokinetics (CPK) Laboratory

The Clinical Pharmacokinetics (CPK) Laboratory, located in the Division of Pharmaceutical Sciences, supports the St. Jude mission by providing state-of-the-art therapeutic drug monitoring and pharmacogenetic testing that clinical pharmacists interpret to ensure optimal drug dosing. It is directed by Alejandro Molinelli, PhD, with translational support from Kristine Crews, PharmD.

Director Alejandro Molinelli, PhD

The CPK lab is certified as a high-complexity laboratory by CLIA (Clinical Laboratory Improvement Amendments) and is accredited by the College of American Pathologists. Our staff consists of licensed medical laboratory scientists. Every year, the laboratory processes and analyzes approximately 8000 clinical specimens and sends out another 550 to reference laboratories. The laboratory’s in-house test menu includes multiple high-complexity assays ranging from therapeutic drug determinations (e.g., immunosuppressant, antifungal drugs) to glomerular filtration rate estimation using 99mTc-DTPA. Some of our resources include random access immunochemistry analyzers (e.g., Roche Cobas c303) and analytical instrumentation (e.g., LC-MS/MS, HPLC).

Most of our instruments have bidirectional interfaces with the Epic electronic health record. The laboratory also handles pharmacogenetic testing for the hospital, offering genotyping results that accompany consults prepared by clinical pharmacists or pharmacy specialty residents.

The laboratory staff and pharmacists at St. Jude work closely to provide prompt results. Once a test result is obtained, the laboratory scientists alert the pharmacist, who then prepares a clinical consult. This close integration of care ensures that our patients receive the best treatment while minimizing adverse drug effects. The laboratory staff is also involved in clinical translational science projects, for which tests developed in the research laboratories are validated and incorporated into the CPK lab test menu to bring cutting-edge care to our patients.

In addition to the samples for clinical testing, the CPK laboratory staff members also process thousands of patient research specimens yearly in support of St. Jude clinical trials for the Center for Translational Pharmacology.

Clinical Pharmacogenomics Program

The Clinical Pharmacogenomics Program aims to integrate preemptive pharmacogenomic testing into patient care to improve the safety and efficacy of medication use.

A pharmacist-led Pharmacogenomics Clinic, one of the first nationwide, provides in-person counseling to patients and families about their individualized pharmacogenomic test results and emphasizes the importance of sharing these results with healthcare providers. Integration with clinical informatics remains critical to preemptive pharmacogenomic testing.

We recently published an analysis of 4,783 clinical decision support alerts guided by pharmacogenomics presented to St. Jude clinicians for 12 genes and 60 drugs. We found clinicians adhered to the therapeutic recommendations provided in 4392 alerts (92%). The Clinical Pharmacogenomics Program advances the ability for other organizations to build similar pharmacogenomics capabilities in their electronic health record. Notably, Dr. Haidar serves as one of the founding Chairs of the Epic Pharmacogenomics Brain Trust. She was also elected to the Epic Genetics Specialty Steering Board to represent the pharmacogenomics perspective of genomic implementation.

To facilitate evidence-based clinical pharmacogenomic testing worldwide, the Clinical Pharmacogenetics Implementation Consortium (CPIC®) was created in 2011. Kelly Caudle, PharmD, PhD, leads CPIC as co-PI and Director with James Hoffman, PharmD, as co- leader of CPIC informatics. The Department is well represented in CPIC with additional involvement as co-authors and other roles from Cyrine Haidar, PharmD; Kristine Crews, PharmD; Alejandro Molinelli, PhD; Jun Yang, PhD; and others. CPIC membership has grown to over 700 members of international clinicians and scientists and continues to provide education and valuable resources to guide the implementation of pharmacogenomics into clinical practice. A significant 2023 accomplishment was successful renewal of NIH funding in collaboration with colleagues at Stanford University and the University of Pennsylvania and continued planning for how CPIC will connect with other established genomic resources. St. Jude is also involved in the NIH-funded Clinical Genome Resource (ClinGen), which was recently outlined in Clinical Chemistry. CPIC guidelines are indexed in PubMed as clinical guidelines, endorsed by ASHP and ASCPT and referenced in ClinGen and PharmGKB.

The goal at St. Jude is to implement all genes and drugs encompassed by CPIC guidelines. As of 2025, the Clinical Pharmacogenomics Program has implemented 16 genes and 75 drugs at St. Jude. Over 7,700 patients have consented for pre-emptive pharmacogenomic testing through the multidisciplinary PG4KDS protocol.

To continually advance clinical implementation, the Clinical Pharmacogenomics Program personnel train the next generation of pharmacogenomics professionals through the PGY2 residency in Clinical Pharmacogenomics and educate health care professionals and patients. Through the Department’s Clinical Pharmacogenomics Program, pharmacogenomic testing benefits St. Jude patients and resources to advance pharmacogenomics implementation.

Kelly Caudle, PharmD Kristine Crews, MS, PharmD Cyrine Haidar, PharmD James Hoffman, PharmD, MS Jun J. Yang, PhD

Clinical Pharmacy Services

By providing clinical pharmacy services to the institution, Clinical Pharmacy Specialists extend the reach of Pharmaceutical Services to direct patient care. Our clinicians provide support to the Leukemia/Lymphoma, Solid Tumor, Neuro-Oncology, Bone Marrow Transplant & Cellular Therapy, Intensive Care, Surgery, (non-malignant) Hematology and HIV care teams. As we continue to grow, we have developed a clinical/academic clinical pharmacy role that is embedded in the St. Jude Life/After Completion of Therapy (ACT) clinics. We are also expanding our services to provide assistance to our colleagues in the Quality-of-Life program. Our board-certified Clinical Pharmacy Specialists are credentialed members of the medical staff with collaborative authority to perform direct patient assessment and prescribe medication therapy and associated laboratory tests.

These pharmacists provide pharmacokinetic assessments, which include individualized targeted therapy for antineoplastics and other medications. Further individualization incorporates the use of clinical pharmacogenomics in medication selection and dosing approaches. Our pharmacists work closely with clinicians to provide oversight and development of care plans (non-protocol treatment plan) for patients who are not managed on an approved research protocol. We also provide departmental and institutional support for research protocol development and implementation.

Our pharmacists offer expertise for services including intra- and inter-departmental education as well as post graduate training in pediatric hematology/oncology medication management and disease states. Team members continue involvement in multiple collaborative efforts. We provide leadership and direction for the antimicrobial stewardship program which helps maintain the institution’s status as an Antimicrobial Stewardship Center of Excellence as designated by the Infectious Diseases Society of America. This is an ongoing collaboration between our department and the Department of Infectious Diseases. We continue to assist with the implementation of the new electronic health record (EPIC). Expanding our reach beyond St. Jude and Memphis, our clinical pharmacists support patient care needs in collaboration with our colleagues at St. Jude affiliate sites and provide educational assistance in support of our Global Pediatric Medicine program.

Pharmaceutical Services Medication Safety and Policy Team

The Pharmaceutical Services Medication Safety and Policy team, led by Jennifer Robertson, PharmD, ensures safe and standardized medication systems throughout St. Jude. The team has five main functions: guideline and policy development, formulary management, medication safety improvements, infusion pump library management, and quality improvement initiatives.

The team works with the institution’s Pharmacy and Therapeutics (P&T) Committee to set and revise policies related to medication use within the institution; establish and maintain high-quality drug information resources and drug therapy guidelines; maintain the drug formulary and review nonformulary drug requests; and implement cost savings efforts, when relevant. Delia Allen, PharmD, the Medication Use Policy Manager, serves as Vice-Chair of the P&T Committee and leads these efforts. In partnership with Pharmacy Supply Chain Services, the department has saved over $8 million with formulary improvements since 2020. Madison Cole, PharmD, the newest member of the Medication Safety and Policy team, helps with these efforts.

The Medication Use Safety Team (MUST), chaired by Dr. Robertson, leads routine and ongoing efforts to improve the safety of medication use systems, based on both reactive and proactive review of events and risk. Specifically, medication events are investigated, and actions are taken to improve medication systems; special emphasis is placed on improving the electronic health record (EHR) to prevent future safety events. Cause analyses, such as root cause analysis, are conducted as appropriate. The team works closely with the Office of Quality and Patient Safety to coordinate actions. Proactive efforts include review and implementation of the Institute for Safe Medication Practices’ (ISMP) recommendations as well as benchmarking and discussion with other institutions. A Failure Modes and Effects Analysis (FMEA), a systematic method used to identify, analyze, and prioritize potential failure modes within a product, process, or system, may also be conducted before innovative technologies or other major changes are implemented.

Infusion pumps are a joint management effort between the Division of Pharmaceutical Services, Informatics, Nursing and Biomedical Engineering. The Medication Infusion Technology Task Force was formed in 2024 to lead efforts related to the improvement of infusion pumps at St. Jude, including implementation of innovative technology, maintenance of drug libraries, and review of drug library compliance. Kristen Hughes, PharmD, from the Medication Use and Policy group, co-leads this task force. Under her leadership, smart pump library compliance has increased from 50% to 90%.

Also led by Dr. Robertson, a division quality improvement committee regularly reviews ongoing improvement projects and provides direction as needed. A quality improvement related to the reduction in nuisance alerts with dose warnings in the EHR is underway. The smart pump library improvements were a part of a quality improvement project, and results were published in Hospital Pediatrics.

Finally, the group provides didactic lectures to the University of Tennessee College of Pharmacy, as well as precepts students on Medication Safety rotations. The team is recruiting its eleventh Medication-Use Safety and Policy resident and precepts residents in the Oncology and Informatics pharmacy residency programs.

Pharmacy Informatics

The Pharmacy Informatics Community is primarily composed of the Pharmacy Automation Team, Epic – Willow Team, and Pharmacy Analytics Services. Together they implement, maintain and optimize best-in-class medication technology solutions and analytics to support exceptional pharmaceutical care. By collaborating with clinical staff, Quality and Patient Care, and Information Services, Pharmacy Informatics works to develop practical solutions that enhance patient outcomes, improve workflow efficiency, and promote resource stewardship within the medication use process at St. Jude.

The informatics ecosystem at St. Jude is highly complex, and this group plays a critical role in ensuring its functionality to support excellent patient care. The Pharmacy Automation Team manages a wide range of solutions, including inventory control tools, infusion pumps, compounding devices and other essential pharmacy automation technologies. The Epic – Willow Team is responsible for building and configuring all medication management workflows within the Epic electronic health record (EHR), while Clinical Informatics Pharmacists bridge the gap between operational practice and technical functionality.

Medication management data is essential for guiding departmental operations and serves as a benchmark for pharmaceutical data stewardship both within St. Jude and externally. As a federated (credentialed) analytics team, Pharmacy Analytics Services leverages this data to develop and maintain a comprehensive business intelligence portfolio as part of the department’s data strategy, empowering data-driven decision-making across the organization.

Pharmacy Supply Chain Services

Pharmacy Supply Chain Services (PSCS) is an operational coalition overseeing a service line focused on two key areas:

Strategic Sourcing - Manages pharmaceutical vendor relationships, accounts payable, contracts, and drug shortage monitoring. The team collaborates with our Group Purchasing Organization (Vizient) and leverages our strategic partnership with McKesson to maintain a reliable supply while ensuring responsible stewardship of St. Jude resources

Pharmacy Supply Chain Operations - Oversees inventory movement, ensures consistent customer service, supervises vendor purchasing decisions, optimizes inventory, aligns inventory systems with compliance requirements and facilitates communication and coordination during drug shortages

This team plays a vital role in maintaining a stable and efficient pharmaceutical supply chain to support patient care at St. Jude.

Acute Care Pharmacy Operations

The Acute Care Pharmacy team is led by William Humphrey, DPh, MS, MBA. Two pharmacy managers support Humphrey: Alysa Baumann, PharmD, MBA, BCPS, DPLA and Cindy Brasher, PharmD, MS, BCSCP. Acute Care operations comprehensively address the needs of St. Jude patients across the inpatient and ambulatory infusion spaces.

Director

William Humphrey, DPh, MS, MBA

A team of investigational drug services pharmacists and certified pharmacy technicians helps execute many drugrelated clinical trials conducted through St. Jude, supported by manager Chris Scobey, PharmD, HMBA, CCRP. This team assists with proper protocol development, handling investigational drugs and developing information for all pharmacy staff related to each protocol. Each pharmacist is dedicated to the research mission, assessing and ensuring proper drug therapy compliance with the definitions of each unique protocol to which individual patients consent.

The Inpatient Pharmacy team is responsible for verifying medication orders and coordinating the preparation and dispensing of all inpatient doses of medications. The inpatient team is also responsible for all sterile compounding for hazardous and nonhazardous intravenous medications, including chemotherapy, intravenous fluids and anti-infectives. An extension of this service includes the Infusion Center Pharmacy, which provides sterile products for patients treated in the clinics, procedure areas, diagnostic areas and the infusion center. A second extension of this service is staffing the Bone Marrow & Cellular Therapies pharmacy satellite, likewise with order verification and sterile compounding for nonhazardous and hazardous medications. The inpatient pharmacists also respond to Harvey Team (code) events and provide the needed drug support to the responding team. Decentralized clinical staff pharmacists also support the Inpatient Chemo Service and the Intensive Care Unit. The acute care pharmacy team comprises clinical staff pharmacists and certified pharmacy technicians.

Ambulatory Pharmacy Operations

The Ambulatory Pharmacy team is led by Steve Pate, DPh, supported by pharmacy managers Cheri Wilkerson, PharmD; Tiffany Nason, PharmD; and Ryan Duncan, PharmD. Ambulatory Pharmacy Operations comprehensively addresses the needs of St. Jude patients across the outpatient continuum of care.

Because medications are shipped nationwide, the team carefully complies with the unique regulatory requirements of each state. Similarly, the broad geographic dispersion of patients necessitates interaction with many state and commercial insurance programs, requiring attention to reimbursement and payer network requirements. Specialty pharmacy services are dually accredited by URAC and ACHC. This helps ensure optimal use of these high-cost, high-risk medications, facilitates reimbursement, and allows patients to receive more of their medications at home.

The Outpatient Pharmacy team includes the ACU Pharmacy, specialty pharmacy, prescription mail service and the home infusion pharmacy. These pharmacies are accredited by outside accreditation organizations to ensure highquality patient care, regardless of location. The outpatient team continually seeks opportunities to bring care closer to patients. The ACU Pharmacy recently installed the first medication dispensing kiosk at St. Jude, allowing patients to pick up their medications when most convenient. When patients from outside Memphis return home, pharmacists ensure continuity of care through mail order or local contracted or affiliate pharmacies.

Acute Care and Ambulatory Pharmacy Operations Services extend beyond the typical services, as St. Jude provides home infusion therapy to patients requiring medications at home or in a local domicile. This service supplies medications to support therapy in the local delivery area and collaborates with outside agencies to deliver care to patients outside that region. The Home Infusion Pharmacy is accredited by ACHC to ensure patients receive the highest levels of care.

Clinical staff pharmacists and certified pharmacy technicians deliver optimal inpatient and outpatient care consistent with the defined research protocol or non-protocol care plan. Technical staff prepare and provide medications, while pharmacists manage medication therapy to align with treatment plans and prevent adverse effects. These pharmacists serve as patient and family educators and resources when drug-related questions arise. Clinical staff pharmacists collaborate with clinical pharmacy specialists to ensure proper conduct of medications reconciliation as patients move from inpatient to outpatient care and back. Finally, the department collaborates directly with the eight St. Jude Affiliates to help ensure that patients managed at these locations throughout the United States have all their medication-related needs addressed.

PGY2 Residencies

The Department of Pharmacy and Pharmaceutical Sciences hosts PGY2 Residencies in Oncology Pharmacy, Medication Use Safety and Policy, Clinical Pharmacogenomics, Infectious Diseases, and Informatics. All programs are accredited by the American Society of Health System Pharmacists (ASHP). Trainees at St. Jude are supported by an institutional Clinical Education and Training Office, whose goal is to assist our investigators and professional staff to improve the quality of experiences, training, benefits, and support for our undergraduate, graduate, professional, and postdoctoral trainees. Over 300 postdoctoral trainees (post-PhD, MD, and PharmD) are at St. Jude.

Ashton Bellamy, PharmD Pharmacy Infectious Diseases

Meghan McNulty, PharmD Pharmacogenomics

Sophie Tilley, PharmD Pharmacy Oncology

Lily Higgins, PharmD Pharmacy Oncology

Gina Peng, PharmD Pharmacy Informatics

Administrative Team

Director Carla Cook, MS

The Department of Pharmacy & Pharmaceutical Sciences Administrative Team, under the leadership of Carla Cook, MS, plays an integral role in advancing both the department’s mission and the lifesaving work of St. Jude Children’s Research Hospital. This dedicated team provides critical support across various operational and strategic areas, including large-scale event coordination, executive calendar management, recruitment initiatives, guest relations, pre- and post-award grant administration and budget oversight, among numerous other responsibilities. Their efforts ensure the seamless functioning of departmental activities and contribute meaningfully to the hospital’s broader objectives.

As of 2025, the team comprises nine administrative professionals with a combined total of more than 43 years of experience at St. Jude. Four team members — Carla Cook (Administrative Director), Dawn Fisher (Senior Administrative Coordinator), April Blankenship (Executive Coordinator) and LaChelle Buckley (Senior Administrative Assistant) — are alumni of the St. Jude Leadership Academy, reflecting their commitment to professional development and leadership excellence, and in 2025 Dawn Fisher was recognized as a St. Jude Leading Lady at the “Unstoppable: Celebrating Women’s Journeys” event. The team’s expertise and reputation for high-quality support have led to frequent collaborations with other departments, where they serve as trainers and mentors for administrative staff across the hospital. Their contributions extend beyond routine responsibilities, as they actively participate in community outreach initiatives, on-campus events, patient activities and various St. Jude Employee Resource Groups, exemplifying their dedication as engaged and compassionate St. Jude citizens.

In 2024 and 2025, the team welcomed several new members: Jasmine Williams (Senior Administrative Specialist, Division of Pharmaceutical Services), Andrea Jones (Senior Administrative Specialist, Division of Pharmaceutical Sciences, Jane Holton (Senior Administrative Specialist, Division of Pharmaceutical Sciences) and Liz Edmundson (Senior Administrative Specialist, Division of Pharmaceutical Services). Additionally, Dina Bachor, (Division of Pharmaceutical Services) was promoted to Administrative Coordinator. These additions and promotions further strengthen the team’s capacity to support the department and its vital work.

Community Culture Committee

The Department of Pharmacy and Pharmaceutical Sciences at St. Jude Children’s Research Hospital established a Community Culture Committee in January 2023 and continues through current state with a mix of colleagues that includes all pharmacy areas, shifts, and roles from the department. The vision of the committee is to model St. Jude Values and create spaces for connection that champion a safe, respectful, supportive working environment and encourage joy in our daily patient care.

Embracing key principles from a leadership book entitled, The Culture Code, by Daniel Coyle, the committee has incorporated various culture building concepts to strengthen the pharmacy team to prioritize felt safety and belonging, shared vulnerability and unified purpose. The shared purpose comes naturally as we strive towards the institution’s mission to advance cures, and means of prevention, for pediatric catastrophic diseases through research and treatment. The committee has created many opportunities for building relationships and promoting resilience both inside and out of the normal work environment, including holiday celebrations, Joy Kit for resets at workplace, friendly competitions, shared mealtimes, patient service events (Bingo games), as well as periodic treats and staff recognition. All of these have fostered stronger relationships within the department and have impacted morale at both an individual and collective level.

Honors & Awards

Dr. Brooke Bernhardt, Director, Division of Pharmaceutical Services, was honored in the 2024 St. Jude Values Book for Value 7, “Make the most of St. Jude resources and be mindful of those who provided them.”

Dr. Ted Morton, Clinical Pharmacy Specialist - Antimicrobial Stewardship, was honored in the 2024 St. Jude Values Book for Value 2, “Do what is right; take ownership of what you do.”

Dr. Jun Yang, Director, Division of Pharmaceutical Sciences, was appointed to the NIH NCI Board of Scientific Counselors and will serve for 4 years.

Dr. Dave Rogers, Chair, Department of Pharmacy and Pharmaceutical Sciences, was honored with an invitation to deliver the 59th annual Gordon A. Bergy Lecture, Alpha Mu Chapter of the Rho Chi Society, at West Virgina University Robert C. Byrd Health Science Center.

Dr. Steve Pate, Sr. Director, Pharmacy, Deputy Chief Pharmacy Officer, was honored by Upsher-Smith Laboratories, LLC and the National Alliance of State Pharmacy Association for, “Celebrating Pharmacy Trailblazers: 2024 Excellence in Innovation Award.” This award honors innovative patient care solutions.

Dr. David Aguero, Director, Medication Systems, and Informatics, was elected a Fellow of American Society of Health-System Pharmacists and American Medical Informatics Association.

Dr. William L. Greene, Member and Chief Pharmaceutical Officer (ret.) received the American Society of Health-System Pharmacists Distinguished Leadership Award.

Dr. Shane Cross, Clinical Pharmacy Specialist-Antimicrobial Stewardship and Dr. Ted Morton, Clinical Pharmacy SpecialistAntimicrobial Stewardship, received renewal for our Antimicrobial Stewardship Program as an Infectious Diseases Society of America Antimicrobial Stewardship Center of Excellence.

Dr. Ted Morton, Sr. Clinical Pharmacy Specialist – Antimicrobial Stewardship was promoted to Professor at University Tennessee Health Science Center.

Dr. John Schuetz, Member, was elected chair of American Association for the Advancement of Science, Pharmaceutical Sciences –Section 5 and was named Editor in Chief, American Society for Pharmacology and Experimental Therapeutics, Discovery journal.

Dr. Kelly Caudle, Associate Member, and Mary Relling, (Emerita Faculty), were St. Jude scientists listed on the 2024 Highly Cited Researchers list in the field of Pharmacology and Toxicology.

Meghan McNulty, PGY2 in Pharmacogenomics, received the Abstract of the Year Award from the Midsouth Pharmacy Residency Conference

Lily Higgins, PGY2 in Oncology, received second place poster at the 13th Annual Clinical Fellow Research Symposium at St Jude

Sharon Brittmon was awarded the Shining Star Compounding Pharmacy Technician by the Tennessee Pharmacists Association

David Lewis was awarded Health-System Pharmacy Technicians of the Year by the Tennessee Pharmacists Association

Ted Morton, and Shane Cross were awarded the Health-System Residency Program of the Year by the Tennessee Pharmacists Association

Dr. Cyrine Haidar, and Dawn Fisher, Sr. Administrative Coordinator, were recognized as St. Jude Leading Ladies at the “Unstoppable: Celebrating Women’s Journeys” event.

Departmental Extramural Funding

NEW AWARDS FOR FY24

Kelly Caudle

NHGRI U24 – $1,380,782

September 2023 to June 2026

Clinical Implementation Resources for Pharmacogenomics (CIRP)

Peijun Ma

CIDC – $50,000

July 2023 to June 2024

Carbapenem-induced heterogeneous expression of penicillin-binding proteins and antibiotic persistence

Peijun Ma

CIDR, SJCRH - $49,000

July 2023 to June 2024

Systematic Characterization of Bacterial Isolates

Causing Antibiotic Treatment Failure in Immunocompromised Children

Ted Morton

SIDP – $75,000

July 2023 to July 2024

SIDP Pharmacotherapy Residency Award 2023-2024

Jeffrey Rybak

NIAID R03 – $186,500

July 2023 to June 2025

Development of M-Drive: A recyclable Mucor-optimized CAS9 gene-drive system cable of multi-target gene editing

Liqin Zhu

ACS – $300,000

July 2023 to June 2025

Neonatal hypoxia on hepatoblastoma metastasis

NEW AWARDS FOR FY25

Peijun Ma

NIGMS R35 - $2,312,500

January 2025 to November 2029

Deciphering the genetic regulation of stress-response mediated antibiotic persistence using bacterial single-cell transcriptomics

Milre Matherne

HOPA - $50,000

December 2024 to June 2026

Pilot study of inotuzumab ozogamicin pharmacokinetics in newly diagnosed pediatric acute lymphoblastic leukemia

Daniel Savic

NHGRI R01 - $3,264,297

April 2024 to March 2029

Deciphering the underlying structure of the glucocorticoid gene regulatory network

Liqin Zhu

ACS – $300,000

July 2024 to March 2025

Neonatal hypoxia on hepatoblastoma metastasis

OTHER ACTIVE FUNDING FY2025

Mark Leggas

NIH P30 – $277,500

March 2025 to February 2026

CCSG - Pharmacokinetics Shared Resources

P. David Rogers

NIAID R01 – $1,206,879

March 2025 to February 2026

Mapping the genomic and molecular mechanisms of antifungal resistance in the emerging fungal pathogen

Candida auris

P. David Rogers

NIAID R01 – $21,366

April 2025 to March 2026

Antifungal Antagonism as a Cause of Treatment Failure for Invasive Mycoses

Jeffrey Rybak

NIAID R03 – $186,500

July 2025 to June 2026

Development of M-Drive: A recyclable Mucor-optimized CAS9 gene-drive system cable of multi-target gene editing

Daniel Savic

NIH R01 – $68,929

August 2023 to July 2024

Admixture analysis of acute lymphoblastic leukemia in African American children: the ADMIRAL Study

John Schuetz

NIH R01 – $39,569

June 2025 to March 2026

Inhibition of Apical cAMP/cGMP transporter (MRP4) in Gut Induces Diarrhea

John Schuetz

NCI R01 – $406,501

April 2024 to March 2025

Transporters and Medulloblastoma

Clinton Stewart

Pfizer ONITT – $18,316

February 2025 to February 2026

A Randomized Phase I/II Study of Onivyde in Combination with Talazoparib or Temozolomide in Children and Young Adults With Recurrent Solid Malignancies and Ewing Sarcoma

Jun J. Yang

NCI R01 – $569,213

July 2024 to June 2025

Biomarkers of Dasatinib Response and Resistance in T-cell Acute Lymphoblastic Leukemia

Jun J. Yang

LLS TRP – $263,400

July 2024 to June 2025

LCK-targeted Therapy in T cell Acute Lymphoblastic Leukemia

Jun J. Yang

NCI R01 – $267,455

August 2024 to July 2025

Molecular Epidemiology of ALL in Children with Down Syndrome

Jun J. Yang

NCI U01 – $223,792

September 2024 to August 2025

Clonal Therapy for Pediatric T-cell Acute Lymphoblastic Leukemia

Jun J. Yang

NIGMS R35 – $516,960

February 2025 to January 2026

Pharmacogenomics of Nucleobase and Nucleoside

Analog Drugs

Jun J. Yang

NIH R01 – $35,102

March 2025 to February 2026

Predictors of Systemic Exposure to Oral 6MP During Maintenance in Adolescents and Young Adults with Acute Lymphoblastic Leukemia (wolfson)

Jun J. Yang

NCI R01 – $29,249

May 2025 to April 2026

Understanding the Increased Risk of Childhood Acute Lymphoblastic Leukemia in Latinos

Highlighted Publications

Nature Communications

The role of ATP-binding cassette subfamily B member 6 in the inner ear

Baril SA, Wilson KA, Shaik MM, Fukuda Y, Umans RA, Barbieri A, Lynch J, Gose T, Myasnikov A, Oldham ML, Wang Y, Zhu J, Fang J, Zuo J, Kalathur RC, Ford RC, Coffin A, Taylor MR, O’Mara ML, Schuetz JD. Nat Commun 15:9885-,2024. PMCID: PMC11574312 PUBMEDID: 39557842

Antimicrobial Agents and Chemotherapy

Analysis of clinical Candida parapsilosis isolates reveals copy number variation in key fluconazole resistance genes

Bergin S, Doorley LA, Rybak JM, Wolfe KH, Butler G, Cuomo CA, Rogers PD. Antimicrob Agents Chemother 68:e0161923,2024. PMCID: PMC11620501 PUBMEDID: 38712935

Nature Communications

Investigation of inherited noncoding genetic variation impacting the pharmacogenomics of childhood acute lymphoblastic leukemia treatment

Clinical Pharmacology & Therapeutics

Clinical Pharmacogenetics Implementation Consortium guideline (CPIC) for CYP2D6, ADRB1, ADRB2, ADRA2C, GRK4, and GRK5 genotypes and beta-blocker therapy

Bhattarai KR, Mobley RJ, Barnett KR, Ferguson DC, Hansen BS, Diedrich JD, Bergeron BP, Yoshimura S, Yang W, Crews KR, Manring CS, Jabbour E, Paietta E, Litzow MR, Kornblau SM, Stock W, Inaba H, Jeha S, Pui CH, Cheng C, Pruett-Miller SM, Relling MV, Yang JJ, Evans WE, Savic D. Nat Commun. 2024 May 1;15(1):3681. doi: 10.1038/s41467-024-48124-4.PMID: 38693155

Duarte JD, Thomas CD, Lee CR, Huddart R, Agundez JAG, Baye JF, Gaedigk A, Klein TE, Lanfear DE, Monte AA, Nagy M, Schwab M, Stein CM, Uppugunduri CRS, van Schaik RHN, Donnelly RS, Caudle KE, Luzum JA. Clin Pharmacol Ther 116:939-947,2024. PMCID: PMC11502236 PUBMEDID: 38951961

Blood

Germ line genetic NBN variation and predisposition to B-cell acute lymphoblastic leukemia in children

Escherich CS, Chen W, Li Y, Yang W, Nishii R, Li Z, Raetz EA, Devidas M, Wu G, Nichols KE, Inaba H, Pui CH, Jeha S, Camitta BM, Larsen E, Hunger SP, Loh ML, Yang JJ. Blood 143:22702283,2024. PMCID: PMC11443573 PUBMEDID: 38446568

Hepatology

HKDC1 promotes liver cancer stemness under hypoxia via stabilizing β-catenin

Fan L, Tian C, Yang W, Liu X, Dhungana Y, Yang W, Tan H, Glazer ES, Yu J, Peng J, Ma L, Ni M, Zhu L. Hepatology :-,2024. PMCID: PUBMEDID: 39250463

Nature Communications

The net electrostatic potential and hydration of ABCG2 affect substrate transport

Cancer Cell

Single-cell systems pharmacology identifies development-driven drug response and combination therapy in B cell acute lymphoblastic leukemia

Gose T, Aitken HM, Wang Y, Lynch J, Rampersaud E, Fukuda Y, Wills M, Baril SA, Ford RC, Shelat A, O’Mara ML, Schuetz JD. Nat Commun 15:8956-,2024. PMCID: PMC11486965

PUBMEDID: 39420170

Hospital Pediatrics

Optimizing the Use of Dose Error Reduction Software on Intravenous Infusion Pumps

The Pediatric Infectious Disease Journal

Evaluation of continuous infusion vancomycin in a pediatric hematology/oncology population

Huang X, Li Y, Zhang J, Yan L, Zhao H, Ding L, Bhatara S, Yang X, Yoshimura S, Yang W, Karol SE, Inaba H, Mullighan C, Litzow M, Zhu X, Zhang Y, Stock W, Jain N, Jabbour E, Kornblau SM, Konopleva M, Pui CH, Paietta E, Evans W, Yu J, Yang JJ. Cancer Cell 42:552-567.e6,2024. PMCID: PMC11008188 PUBMEDID: 38593781 NIHMSID: NIHMS1978911

Pediatric Blood & Cancer

Blinatumomab infusion interruptions in pediatric patients rarely lead to readmission

Hughes K, Cole M, Tims D, Wallach T, Spencer C, Page V, Robertson J, Hoffman JM. Hosp Pediatr. 2024 Jun 1;14(6):448-454. doi: 10.1542/hpeds.2023-007385.PMID: 38716570

Pharmaceutics

Population pharmacokinetic and pharmacodynamic study of palbociclib in children and young adults with recurrent, progressive, or refractory brain tumors

King MA, Cross SJ, Morton TH, Hijano DR, Greene WL, Sun Y, Tang L, Pauley JL, Bourque MS, Christensen AM. Pediatr Infect Dis J 43:520-524,2024. PMCID: PMC11098708 PUBMEDID: 38359358 NIHMSID: NIHMS1962322

Moody J, Barker PJ, Sciasci J, Pauley JL, Bragg A, McMillan C, Triplett BM, Swanson HD. Pediatr Blood Cancer 71:e31223,2024. PUBMEDID: 39054702

Panetta JC, Selvo NS, Mater DV, Stewart CF. Pharmaceutics 16:1528-,2024. PMCID: PMC11676693 PUBMEDID: 39771507

Nature Communications

A secondary mechanism of action for triazole antifungals in Aspergillus fumigatus mediated by hmg1

Journal of the National Cancer Institute

Optimizing early phase clinical trial washout periods: A report from the Therapeutic Advances in Childhood Leukemia and Lymphoma consortium

Rybak JM, Xie J, Martin-Vicente A, Guruceaga X, Thorn HI, Nywening AV, Ge W, Souza ACO, Shetty AC, McCracken C, Bruno VM, Parker JE, Kelly SL, Snell HM, Cuomo CA, Rogers PD, Fortwendel JR. Nat Commun 15:3642-,2024. PMCID: PMC11059170 PUBMEDID: 38684680

Schafer ES, Rushing T, Crews KR, Annesley C, Colace SI, Kaiser N, Pommert L, Ramsey LB, Sabnis HS, Wong K, Chang BH, Cooper TM, Shah NN, Rheingold SR, Place AE, Chi YY, Bhojwani D, Wayne AS, Bernhardt MB. J Natl Cancer Inst 116:1721-1729,2024. PMCID: PMC11542989 PUBMEDID: 38964343

The Pediatric Infectious Disease Journal

Sulfamethoxazole-trimethoprim prophylaxis in pediatric oncology patients with glucose-6phosphate dehydrogenase deficiency

Stone RM, Haidar CE, Kornegay NM, Barker PJ, Karol SE, Wolf J, Hankins JS, Relling MV, Crews KR. Pediatr Infect Dis J :-,2024. PUBMEDID: 39163536

Journal of Clinical Pharmacology

Pharmacokinetic modeling and model-based hypothesis generation for dose optimization of clonidine in neonates with neonatal opioid withdrawal syndrome

Cancer Research

The imipridone ONC213 targets a-ketoglutarate dehydrogenase to induce mitochondrial stress and suppress oxidative phosphorylation in acute myeloid leukemia

Tang F, Ng CM, Horn J, Bada HS, Leggas M. Clin Pharmacol Ther :-,2024. PUBMEDID: 39575611

Journal of Clinical Oncology

Impact of age on pharmacogenomics and treatment outcomes of B-cell acute lymphoblastic leukemia

Yongwei Su, Jenna L. Carter, Xinyu Li, Yu Fukuda, Ashley Gray, John Lynch, Holly Edwards, Jun Ma, Patrick Schreiner, Lisa Polin, Juiwanna Jushner, Sijana H. Dzinc, Steven A . Buck, Shondra M Pruett-Miller, Katie Hege Hurrish, Camenzind Robinson, Xinan Qiao, Shuang Liu, Shuangshuang Wu, Guan Wang, Jing Li, Joshua E. Allen, Varun V. Prabhu, Aaron D. Schimmer, Dhananjay Joshi, Shiva Kalhor0Monfared, Iain D. G. Watson, Richard Marcellus, Methvin B. Isaac, Rima Al-Awar, Jeffrey W. Taub, Hai Lin, John D. Schuetz, Yubin Ge. Cancer Res. 2024 PMID: 38266099

Yoshimura S, Li Z, Gocho Y, Yang W, Crews KR, Lee SHR, Roberts KG, Mullighan CG, Relling MV, Yu J, Yeoh AEJ, Loh ML, Saygin C, Litzow MR, Jeha S, Karol SE, Inaba H, Pui CH, Konopleva M, Jain N, Stock W, Paietta E, Jabbour E, Kornblau SM, Evans WE, Yang JJ. J Clin Oncol 42:3478-3490,2024. PMCID: PMC11458355 PUBMEDID: 39102629 NIHMSID: NIHMS1996246COIS-

Publications

Aguero D, Nelson SD. The potential application of large language models in pharmaceutical supply chain management. J Pediatr Pharmacol Ther 29:200-205,2024. PMCID: PMC11001215 PUBMEDID: 38596417

Aguero D, Vest MH, Tryon J. The role of the chief pharmacy officer in leading analytics strategy to support the enterprise. Am J Health Syst Pharm 81:e648-e652,2024. PMCID: PUBMEDID: 38469877

Aguero D, Allen D. Weathering the storm: Commentary on the Hurricane Helene IV fluid shortage. J Pediatr Pharmacol Ther 29:667-669,2024. PMCID: PMC11627566 PUBMEDID: 39659851

Ansari SS, Dillard ME, Zhang Y, Austria MA, Boatwright N, Shelton EL, Stewart DP, Johnson A, Wang CE, Young BM, Rankovic Z, Hansen BS, Pruett-Miller SM, Carisey AF, Schuetz JD, Robinson CG. Sonic Hedgehog activates prostaglandin signaling to stabilize primary cilium length. J Cell Biol 223:-,2024. PMCID: PMC11166601 PUBMEDID: 38856684

Bada HS, Westgate PM, Sithisarn T, Yolton K, Charnigo R, Pourcyrous M, Tang F, Gibson J, Shearer-Miller J, Giannone P, Leggas M. Clonidine as monotherapy for neonatal opioid withdrawal syndrome: A randomized trial. Pediatrics 154:e2023065610-,2024. PMCID: PMC11524040 PUBMEDID: 39403061

Baril SA, Wilson KA, Shaik MM, Fukuda Y, Umans RA, Barbieri A, Lynch J, Gose T, Myasnikov A, Oldham ML, Wang Y, Zhu J, Fang J, Zuo J, Kalathur RC, Ford RC, Coffin A, Taylor MR, O’Mara ML, Schuetz JD. The role of ATP-binding cassette subfamily B member 6 in the inner ear. Nat Commun 15:9885-,2024. PMCID: PMC11574312 PUBMEDID: 39557842

Barker P, Carias DC, Jacobs T, Carpiniello P, Takemoto C, Lieberman J, Adderson E. Promoting penicillin allergy de-labeling for children attending a hematology clinic. Pediatr Blood Cancer 71:e31034-,2024. PMCID: PUBMEDID: 38679842

Bayoumy AB, Mulder CJJ, De Boer NKH, Derijks LJJ, Yang JJ, Schmiegelow K. Comment on: A soaring price, a silent fight: Thioguanine portends new access barriers to life-saving treatments for children with cancer: The ugly duck or the chicken with the golden eggs: The crisis in thioguanine accessibility. Pediatr Blood Cancer :e31464-,2024. PMCID: PUBMEDID: 39616415

Bergin S, Doorley LA, Rybak JM, Wolfe KH, Butler G, Cuomo CA, Rogers PD. Analysis of clinical Candida parapsilosis isolates reveals copy number variation in key fluconazole resistance genes. Antimicrob Agents Chemother 68:e0161923,2024. PMCID: PMC11620501 PUBMEDID: 38712935

Bhattarai KR, Mobley RJ, Barnett KR, Ferguson DC, Hansen BS, Diedrich JD, Bergeron BP, Yoshimura S, Yang W, Crews KR, Manring CS, Jabbour E, Paietta E, Litzow MR, Kornblau SM, Stock W, Inaba H, Jeha S, Pui CH, Cheng C, PruettMiller SM, Relling MV, Yang JJ, Evans WE, Savic D. Investigation of inherited noncoding genetic variation impacting the pharmacogenomics of childhood acute lymphoblastic leukemia treatment. Nat Commun 15:3681-,2024. PMCID: PMC11063049 PUBMEDID: 38693155

Campagne O, Huang J, Lin T, Reddick WE, Selvo NS, Onar-Thomas A, Ward D, Robinson G, Gajjar A, Stewart CF. Population pharmacokinetics of methotrexate and 7-hydroxymethotrexate and delayed excretion in infants and young children with brain tumors. Eur J Pharm Sci 193:106669-,2024. PMCID: PMC10843628 PUBMEDID: 38070781

Carolus H, Sofras D, Boccarella G, Sephton-Clark P, Biriukov V, Cauldron NC, Lobo Romero C, Vergauwen R, Yazdani S, Pierson S, Jacobs S, Vandecruys P, Wijnants S, Meis JF, Gabaldón T, van den Berg P, Rybak JM, Cuomo CA, Van Dijck P. Acquired amphotericin B resistance leads to fitness trade-offs that can be mitigated by compensatory evolution in Candida auris. Nat Microbiol 9:3304-3320,2024. PMCID: PUBMEDID: 39567662

Chauhan S, Lian E, Habib I, Liu Q, Anders NM, Bugg MM, Federman NC, Reid JM, Stewart CF, Cates T, Michalek JE, Keller C. Entinostat as a combinatorial therapeutic for rhabdomyosarcoma. Sci Rep 14:18936-,2024. PMCID: PMC11327338

PUBMEDID: 39147820

Chavana AN, Taylor ZL, DeGroote N, Lindsay HB, Sauer HE, Mason EJ, Schafer ES, Miller TP, Castellino SM, Pommert L, O’Brien MM, Ramsey LB, Bernhardt MB, Brown AL. Toxicity profile of high-dose methotrexate in young children with central nervous system tumors. Pediatr Blood Cancer 71:e31213-,2024. PMCID: PUBMEDID: 39039774

de Smith AJ, Wahlster L, Jeon S, Kachuri L, Black S, Langie J, Cato LD, Nakatsuka N, Chan TF, Xia G, Mazumder S, Yang W, Gazal S, Eng C, Hu D, Burchard EG, Ziv E, Metayer C, Mancuso N, Yang JJ, Ma X, Wiemels JL, Yu F, Chiang CWK, Sankaran VG. A noncoding regulatory variant in IKZF1 increases acute lymphoblastic leukemia risk in Hispanic/Latino children. Cell Genom 4:100526-,2024. PMCID: PMC11019360 PUBMEDID: 38537633

Diedrich JD, Dong Q, Ferguson DC, Bergeron BP, Autry RJ, Qian M, Yang W, Smith C, Papizan JB, Connelly JP, Hagiwara K, Crews KR, Pruett-Miller SM, Pui CH, Yang JJ, Relling MV, Evans WE, Savic D, Correction: Profiling chromatin accessibility in pediatric acute lymphoblastic leukemia identifies subtype-specific chromatin landscapes and gene regulatory networks. Leukemia 38:1867-,2024. PMCID: PUBMEDID: 38961139

Donnelly RS, Whirl-Carrillo M, Klein TE, Caudle KE. The value of clinical pharmacogenomic guidelines that recommend standard of care over genotype-based prescribing. Clin Pharmacol Ther 116:897-898,2024. PMCID: PMC11452270 PUBMEDID: 39115927

Duarte JD, Thomas CD, Lee CR, Huddart R, Agundez JAG, Baye JF, Gaedigk A, Klein TE, Lanfear DE, Monte AA, Nagy M, Schwab M, Stein CM, Uppugunduri CRS, van Schaik RHN, Donnelly RS, Caudle KE, Luzum JA. Clinical Pharmacogenetics Implementation Consortium guideline (CPIC) for CYP2D6, ADRB1, ADRB2, ADRA2C, GRK4, and GRK5 genotypes and betablocker therapy. Clin Pharmacol Ther 116:939-947,2024. PMCID: PMC11502236 PUBMEDID: 38951961

Escherich CS, Chen W, Li Y, Yang W, Nishii R, Li Z, Raetz EA, Devidas M, Wu G, Nichols KE, Inaba H, Pui CH, Jeha S, Camitta BM, Larsen E, Hunger SP, Loh ML, Yang JJ. Germ line genetic NBN variation and predisposition to B-cell acute lymphoblastic leukemia in children. Blood 143:2270-2283,2024. PMCID: PMC11443573 PUBMEDID: 38446568

Escherich CS, Moriyama T, Li Z, Hsiao Y, Yang W, Li Y, Reyes N, Walker M, Budhraja A, Bhatara S, Diaz-Flores E, Stock W, Paietta E, Konopleva MY, Kornblau SM, Litzow MR, Inaba H, Pui CH, Opferman JT, Loh ML, Yu J, O’Brien M, Evans WE, Yang JJ. Dntt-mediated DNA damage response drives inotuzumab ozogamicin resistance in B-cell acute lymphoblastic leukemia. Blood :-,2024. PMCID: PUBMEDID: 39791601

Fan L, Tian C, Yang W, Liu X, Dhungana Y, Yang W, Tan H, Glazer ES, Yu J, Peng J, Ma L, Ni M, Zhu L. HKDC1 promotes liver cancer stemness under hypoxia via stabilizing β-catenin. Hepatology :-,2024. PMCID: PUBMEDID: 39250463

Friedman JM, Bombard Y, Carleton B, Issa AM, Knoppers B, Plon SE, Rahimzadeh V, Relling MV, Williams MS, van Karnebeek C, Vears D, Cornel MC, Global Alliance for Genomics and Health Regulatory and Ethics Workstream. Should secondary pharmacogenomic variants be actively screened and reported when diagnostic genome-wide sequencing is performed in a child? Genet Med 26:101033-,2024. PMCID: PUBMEDID: 38007624

Gaedigk A, Turner AJ, Haidar CE, Empey PE, Offer SM. Response to “DPYD genotyping panels: Impact of population diversity”. Clin Transl Sci 17:e13806-,2024. PMCID: PMC11026573 PUBMEDID: 38637962

Gaietto A, Panetta JC, Pauley JL, Relling MV, Ribeiro R, Ehrhardt MJ, Pui CH, Inaba H, Swanson HD. Ommaya reservoir use in pediatric ALL and NHL: A review at St. Jude Children’s Research Hospital. Cancer Chemother Pharmacol 93:617-625,2024. PMCID: PMC11376002 PUBMEDID: 38416167

Geller JI, Renfro LA, Grundy PE, Perlman EJ, Kalapurakal JA, Ehrlich PF, Biegel J, Huff V, Warwick AB, Paulino A, Mullen EA, Daw NC, Hoffer FA, Tochner Z, Gow K, Gratias E, Ward DA, Anderson JR, Fernandez CV, Dome JS. Rhabdoid tumor of the kidney and soft tissues: Results from National Wilms Tumor Study-5 and Children’s Oncology Group Study AREN0321. Pediatr Blood Cancer :e31490-,2024. PMCID: PUBMEDID: 39702900

Gose T, Rasouli A, Dehghani-Ghahnaviyeh S, Wen PC, Wang Y, Lynch J, Fukuda Y, Shafi T, Ford RC, Tajkhorshid E, Schuetz JD. Tumor-acquired somatic mutation affects conformation to abolish ABCG2-mediated drug resistance. Drug Resist Updat 73:101066-,2024. PMCID: PMC11137617 PUBMEDID: 38387283

Gose T, Aitken HM, Wang Y, Lynch J, Rampersaud E, Fukuda Y, Wills M, Baril SA, Ford RC, Shelat A, O’Mara ML, Schuetz JD. Publisher Correction: The net electrostatic potential and hydration of ABCG2 affect substrate transport. Nat Commun 15:8956-,2024. PMCID: PMC11486965 PUBMEDID: 39420170

Harris RD, Taylor OA, Gramatges MM, Hughes AE, Zobeck M, Pruitt S, Bernhardt MB, Chavana A, Huynh V, Ludwig K, Klesse L, Heym K, Griffin T, Erana R, Bernini JC, Choi A, Ohno Y, Richard MA, Morrison AC, Chen H, Yu B, Lupo PJ, Rabin K, Scheurer ME, Brown AL. Evaluation of methotrexate Pharmacogenomic variation to predict acute neurotoxicity in children with acute lymphoblastic leukemia. Pharmacotherapy :-,2024. PMCID:11806518 PUBMEDID:39734275

Huang X, Li Y, Zhang J, Yan L, Zhao H, Ding L, Bhatara S, Yang X, Yoshimura S, Yang W, Karol SE, Inaba H, Mullighan C, Litzow M, Zhu X, Zhang Y, Stock W, Jain N, Jabbour E, Kornblau SM, Konopleva M, Pui CH, Paietta E, Evans W, Yu J, Yang JJ. Single-cell systems pharmacology identifies development-driven drug response and combination therapy in B cell acute lymphoblastic leukemia. Cancer Cell 42:552-567.e6,2024. PMCID: PMC11008188 PUBMEDID: 38593781

Huemmer SE, Patnaik JL, Ybarra S, Congdon N, Cherwek DH, Wilson MW. Teleophthalmology through online mentorship over a 20-year period: education and capacity building. JCO Glob Oncol 10:e2400297-,2024. PMCID: PUBMEDID: 39541557

Hughes K, Cole M, Tims D, Wallach T, Spencer C, Page V, Robertson J, Hoffman JM. Optimizing the use of dose error reduction software on intravenous infusion pumps. Hosp Pediatr 14:448-454,2024. PMCID: PUBMEDID: 38716570

Hurley C, McArthur J, Gossett JM, Hall EA, Barker PJ, Hijano DR, Hines MR, Kang G, Rains J, Srinivasan S, Suliman A, Qudeimat A, Ghafoor S. Intrapulmonary administration of recombinant activated factor VII in pediatric, adolescent, and young adult oncology and hematopoietic cell transplant patients with pulmonary hemorrhage. Front Oncol 14:1375697-,2024. PMCID: PMC11055461 PUBMEDID: 38680864

Jarusiewicz JA, Yoshimura S, Actis M, Li Y, Fu X, Yang L, Narina S, Pruett-Miller SM, Zhou S, Wang X, High AA, Nishiguchi G, Yang JJ, Rankovic Z. Development of an orally bioavailable LCK PROTAC degrader as a potential therapeutic approach to T-cell acute lymphoblastic leukemia. J Med Chem 67:11868-11884,2024. PMCID: PUBMEDID: 38973320

Junco JJ, Rochette M, Alozie M, Rashid R, Terrell M, Zorman B, Sumazin P, Rowland L, Dettorre G, Powell RT, Stephan CC, Davies PJ, Martinez-Moczygemba M, Yang JJ, Rabin KR. A transgenic mouse model of Down syndrome acute lymphoblastic leukemia identifies targetable vulnerabilities. Haematologica :-,2024. PMCID: PMC11609804 PUBMEDID: 39049603

Kager L, Evans WE. Pharmacogenomics in pediatric oncology research and treatment. J Pediatr Pharmacol Ther 29:554557,2024. PMCID: PMC11472404 PUBMEDID: 39411408

Kannan S, Li Y, Baran N, Yang X, Ghotbaldini S, Tatarata QZ, Yoshimura S, Li Z, Hsiao YC, Balachander SB, Andersen CL, Cidado J, Yu J, Jain N, Yang JJ, Konopleva MY. Anti-leukemia efficacy of the dual BCL2/BCL-XL inhibitor AZD0466 in acute lymphoblastic leukemia preclinical models. Blood Adv :-,2024. PMCID: PUBMEDID: 39561378

Kimura S, Park CS, Montefiori LE, Iacobucci I, Pölönen P, Gao Q, Arnold ED, Attarbaschi A, Brown A, Buldini B, Caldwell KJ, Chang Y, Chen C, Cheng C, Cheng Z, Choi J, Conter V, Crews KR, de Groot-Kruseman HA, Deguchi T, Eguchi M, Muhle HE, Elitzur S, Escherich G, Freeman BB 3rd, Gu Z, Han K, Horibe K, Imamura T, Jeha S, Kato M, Chiew KH, Khan T, Kicinski M, Köhrer S, Kornblau SM, Kotecha RS, Li CK, Liu YC, Locatelli F, Luger SM, Paietta EM, Manabe A, Marquart HV, Masetti R, Maybury M, Mazilier P, Meijerink JPP, Mitchell S, Miyamura T, Moore AS, Oshima K, Pawinska-Wasikowska K, Pieters R, Prater MS, Pruett-Miller SM, Pui CH, Qu C, Reiterova M, Reyes N, Roberts KG, Rowe JM, Sato A, Schmiegelow K, Schrappe M, Shen S, Skoczeń S, Spinelli O, Stary J, Svaton M, Takagi M, Takita J, Tang Y, Teachey DT, Thomas PG, Tomizawa D, Trka J, Varotto E, Vincent TL, Yang JJ, Yeoh AEJ, Zhou Y, Zimmermann M, Inaba H, Mullighan CG. Biologic and clinical analysis of childhood gamma delta T-ALL identifies LMO2/STAG2 rearrangements as extremely high risk. Cancer Discov 14:18381859,2024. PMCID: PMC11452281 PUBMEDID: 38916500

King MA, Cross SJ, Morton TH, Hijano DR, Greene WL, Sun Y, Tang L, Pauley JL, Bourque MS, Christensen AM. Evaluation of continuous infusion vancomycin in a pediatric hematology/oncology population. Pediatr Infect Dis J 43:520-524,2024. PMCID: PMC11098708 PUBMEDID: 38359358

Li L, Atkinson N, Crews KR, Molinelli AR. Quantification of thiopurine metabolites in human erythrocytes by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Methods Mol Biol 2737:443-452,2024. PMCID: PUBMEDID: 38036845

Li L, Atkinson N, Crews KR, Molinelli AR. Determination of thiopurine S-methyltransferase (TPMT) activity in human erythrocytes by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Methods Mol Biol 2737:465-472,2024. PMCID: PUBMEDID: 38036847

Li L, Sun J, Fu Y, Changrob S, McGrath JJC, Wilson PC. A hybrid demultiplexing strategy that improves performance and robustness of cell hashing. Brief Bioinform 25:bbae254-,2024. PMCID: PMC11145454 PUBMEDID: 38828640

Li Z, Yeoh AEJ. SEQ-ing the genetic constellation of acute lymphoblastic leukemia. Haematologica 109:1640-1642,2024. PMCID: PMC11141640 PUBMEDID: 38124627

Li H, Chen Y, Ding M, Liu J, Sun H, Fang H, Brady SW, Xu Y, Glaser F, Ma X, Tang Y, Du L, Wu X, Wang S, Zhu L, Li B, Shen S, Zhang J, Zheng L, Yu J, Assaraf YG, Zhou BS. Folylpolyglutamate synthetase inactivation in relapsed ALL induces a druggable folate metabolic vulnerability. Drug Resist Updat 77:101141-,2024. PMCID: PUBMEDID: 39181011

Leung KT, Cai J, Liu Y, Chan KYY, Shao J, Yang H, Hu Q, Xue Y, Wu X, Guo X, Zhai X, Wang N, Li X, Tian X, Li Z, Xue N, Guo Y, Wang L, Zou Y, Xiao P, He Y, Jin R, Tang J, Yang JJ, Shen S, Pui CH, Li CK. Prognostic implications of CD9 in childhood acute lymphoblastic leukemia: Insights from a nationwide multicenter study in China. Leukemia 38:250-257,2024. PMCID: PMC10844073 PUBMEDID: 38001171

Maillard M, Nishii R, Yang W, Hoshitsuki K, Chepyala D, Lee SHR, Nguyen JQ, Relling MV, Crews KR, Leggas M, Singh M, Suang JLY, Yeoh AEJ, Jeha S, Inaba H, Pui CH, Karol SE, Trehan A, Bhatia P, Antillon Klussmann FG, Bhojwani D, Haidar CE, Yang JJ. Additive effects of TPMT and NUDT15 on thiopurine toxicity in children with acute lymphoblastic leukemia across multiethnic populations. J Natl Cancer Inst 116:702-710,2024. PMCID: PMC11077315 PUBMEDID: 38230823

Maillard M, Nguyen JQ, Yang W, Hoshitsuki K, Relling MV, Caudle KE, Crews KR, Jeha S, Inaba H, Pui CH, Bhatia S, Karol SE, Antillon-Klussmann FG, Haidar CE, Bhojwani D, Yang JJ. Clinical actionability of the NUDT15 *4 (p.R139H) allele and its association with Hispanic ethnicity. Clin Pharmacol Ther :-,2024. PMCID: PUBMEDID: 39688234

Martin-Vicente A, Souza ACO, Guruceaga X, Thorn HI, Xie J, Nywening AV, Ge W, Fortwendel JR. A conserved fungal morphogenetic kinase regulates pathogenic growth in response to carbon source diversity. Nat Commun 15:8945-,2024. PMCID: PMC11484838 PUBMEDID: 39414804

Miao DNR, Wilke MAP, Pham J, Ladha F, Singh M, Arsenio J, Luca E, Dabdoub A, Yang W, Yang JJ, Drögemöller BI. Leveraging large-scale datasets and single cell omics data to develop a polygenic score for cisplatin-induced ototoxicity. Hum Genomics 18:112-,2024. PMCID: PMC11463131 PUBMEDID: 39380081

Miller JS, Bada HS, Westgate PM, Sithisarn T, Leggas M. Neonatal abstinence signs during treatment: Trajectory, resurgence, and heterogeneity. Children (Basel) 11:203-,2024. PMCID: PMC10887053 PUBMEDID: 38397314

Mittal P, Myers JA, Carter RD, Radko-Juettner S, Malone HA, Rosikiewicz W, Robertson AN, Zhu Z, Narayanan IV, Hansen BS, Parrish M, Bhanu NV, Mobley RJ, Rehg JE, Xu B, Drosos Y, Pruett-Miller SM, Ljungman M, Garcia BA, Wu G, Partridge JF, Roberts CWM. PHF6 cooperates with SWI/SNF complexes to facilitate transcriptional progression. Nat Commun 15:7303,2024. PMCID: PMC11344777 PUBMEDID: 39181868

Moody J, Barker PJ, Sciasci J, Pauley JL, Bragg A, McMillan C, Triplett BM, Swanson HD. Blinatumomab infusion interruptions in pediatric patients rarely lead to readmission. Pediatr Blood Cancer 71:e31223-,2024. PMCID: PUBMEDID: 39054702

Moore JN, Campagne O, Bhojwani D, Crowe D, Stewart CF. Developing therapy for every kid With cancer - Summary of a scientific session at the 2023 ASCPT Meeting. Clin Pharmacol Ther 115:185-187,2024. PMCID: PUBMEDID: 37997700

Nair S, Selvo NS, Stolarski A, Klee B, Federico SM, Stewart CF. Quantitative determination of liposomal irinotecan and SN38 concentrations in plasma samples from children with solid tumors: Use of a cryoprotectant solution to enhance liposome stability. J Chromatogr B Analyt Technol Biomed Life Sci 1245:124273-,2024. PMCID: PMC11416028 PUBMEDID: 39146822

Panetta JC, Selvo NS, Mater DV, Stewart CF. Population pharmacokinetic and pharmacodynamic study of palbociclib in children and young adults with recurrent, progressive, or refractory brain tumors. Pharmaceutics 16:1528-,2024. PMCID: PMC11676693 PUBMEDID: 39771507

Rabin KR, Devidas M, Chen Z, Ji L, Kairalla J, Hitzler JK, Yang JJ, Carroll AJ, Heerema NA, Borowitz MJ, Wood BL, Roberts KG, Mullighan CG, Harvey RC, Chen IM, Willman CL, Reshmi SC, Gastier-Foster JM, Bhojwani D, Rheingold SR, Maloney KW, Mattano LA, Larsen EC, Schore RJ, Burke MJ, Salzer WL, Winick NJ, Carroll WL, Raetz EA, Loh ML, Hunger SP, Angiolillo AL. Outcomes in children, adolescents, and young adults with Down syndrome and ALL: A report from the Children’s Oncology Group. J Clin Oncol 42:218-227,2024. PMCID: PMC10824380 PUBMEDID: 37890117

Pereira L, Haidar CE, Haga SB, Cisler AG, Hall A, Shukla SK, Hebbring SJ, Leary EJW. Assessment of the current status of real-world pharmacogenomic testing: Informed consent, patient education, and related practices. Front Pharmacol 15:1355412-,2024. PMCID: PMC10895424 PUBMEDID: 38410134

Pölönen P, Di Giacomo D, Seffernick AE, Elsayed A, Kimura S, Benini F, Montefiori LE, Wood BL, Xu J, Chen C, Cheng Z, Newman H, Myers J, Iacobucci I, Li E, Sussman J, Hedges D, Hui Y, Diorio C, Uppuluri L, Frank D, Fan Y, Chang Y, Meshinchi S, Ries R, Shraim R, Li A, Bernt KM, Devidas M, Winter SS, Dunsmore KP, Inaba H, Carroll WL, Ramirez NC, Phillips AH, Kriwacki RW, Yang JJ, Vincent TL, Zhao Y, Ghate PS, Wang J, Reilly C, Zhou X, Sanders MA, Takita J, Kato M, Takasugi N, Chang BH, Press RD, Loh M, Rampersaud E, Raetz E, Hunger SP, Tan K, Chang TC, Wu G, Pounds SB, Mullighan CG, Teachey DT. The genomic basis of childhood T-lineage acute lymphoblastic leukaemia. Nature 632:1082-1091,2024. PMCID: PMC11611067 PUBMEDID: 39143224

Radko-Juettner S, Yue H, Myers JA, Carter RD, Robertson AN, Mittal P, Zhu Z, Hansen BS, Donovan KA, Hunkeler M, Rosikiewicz W, Wu Z, McReynolds MG, Roy Burman SS, Schmoker AM, Mageed N, Brown SA, Mobley RJ, Partridge JF, Stewart EA, Pruett-Miller SM, Nabet B, Peng J, Gray NS, Fischer ES, Roberts CWM. Targeting DCAF5 suppresses SMARCB1-mutant cancer by stabilizing SWI/SNF. Nature 628:442-449,2024. PMCID: PMC11184678 PUBMEDID: 38538798

Radko-Juettner S, Yue H, Myers JA, Carter RD, Robertson AN, Mittal P, Zhu Z, Hansen BS, Donovan KA, Hunkeler M, Rosikiewicz W, Wu Z, McReynolds MG, Roy Burman SS, Schmoker AM, Mageed N, Brown SA, Mobley RJ, Partridge JF, Stewart EA, Pruett-Miller SM, Nabet B, Peng J, Gray NS, Fischer ES, Roberts CWM. Author Correction: Targeting DCAF5 suppresses SMARCB1-mutant cancer by stabilizing SWI/SNF. Nature 629:E12-,2024. PMCID: PUBMEDID: 38684813

Ramirez-Zavala B, Kruger I, Schwanfelder S, Barker KS, Rogers PD, Morschhauser J. The zinc cluster transcription factor Znc1 regulates Rta3-dependent miltefosine resistance in Candida albicans. mSphere 9:e0027024-,2024. PMCID: PMC11288014 PUBMEDID: 38860767

Ramírez-Zavala B, Hoffmann A, Krüger I, Schwanfelder S, Barker KS, Rogers PD, Morschhäuser J. Probing gene function in Candida albicans wild-type strains by Cas9-facilitated one-step integration of two dominant selection markers: A systematic analysis of recombination events at the target locus. mSphere 9:e0038824-,2024. PMCID: PMC11288041 PUBMEDID: 38940507

Robinson KM, Eum S, Desta Z, Tyndale RF, Gaedigk A, Crist RC, Haidar CE, Myers AL, Samer CF, Somogyi AA, Zubiaur P, Iwuchukwu OF, Whirl-Carrillo M, Klein TE, Caudle KE, Donnelly RS, Kharasch ED. Clinical Pharmacogenetics Implementation Consortium guideline for CYP2B6 genotype and methadone therapy. Clin Pharmacol Ther 116:932-938,2024. PMCID: PMC11587193 PUBMEDID: 38863207

Rybak JM, Xie J, Martin-Vicente A, Guruceaga X, Thorn HI, Nywening AV, Ge W, Souza ACO, Shetty AC, McCracken C, Bruno VM, Parker JE, Kelly SL, Snell HM, Cuomo CA, Rogers PD, Fortwendel JR. A secondary mechanism of action for triazole antifungals in Aspergillus fumigatus mediated by hmg1. Nat Commun 15:3642-,2024. PMCID: PMC11059170 PUBMEDID: 38684680

Schafer ES, Rushing T, Crews KR, Annesley C, Colace SI, Kaiser N, Pommert L, Ramsey LB, Sabnis HS, Wong K, Chang BH, Cooper TM, Shah NN, Rheingold SR, Place AE, Chi YY, Bhojwani D, Wayne AS, Bernhardt MB. Optimizing early phase clinical trial washout periods: A report from the Therapeutic Advances in Childhood Leukemia and Lymphoma consortium. J Natl Cancer Inst 116:1721-1729,2024. PMCID: PMC11542989 PUBMEDID: 38964343

Shah A, Hoffman JM, Twum-Danso N, Burlison J, Barker P. Current state and future directions for improvement science: Reflections from the 2024 International Forum on Quality and Safety in Healthcare. BMJ Lead :-,2024. PMCID: PUBMEDID: 39653394

Shen CJ, Kry SF, Buchsbaum JC, Milano MT, Inskip PD, Ulin K, Francis JH, Wilson MW, Whelan KF, Mayo CS, Olch AJ, Constine LS, Terezakis SA, Vogelius IR. Retinopathy, optic neuropathy, and cataract in childhood cancer survivors treated with radiation therapy: A PENTEC comprehensive review. Int J Radiat Oncol Biol Phys 119:431-445,2024. PMCID: PUBMEDID: 37565958

Short NJ, Jabbour E, Jain N, Senapati J, Nasr L, Haddad FG, Li Z, Hsiao YC, Yang JJ, Pemmaraju N, Ohanian M, Wierda WG, Montalban-Bravo G, Borthakur G, Han L, Xiao L, Huang X, Abramova R, Zhao M, Garris R, Konopleva M, Ravandi F, Kantarjian H. A phase 1/2 study of mini-hyper-CVD plus venetoclax in patients with relapsed/refractory acute lymphoblastic leukemia. Blood Adv 8:909-915,2024. PMCID: PMC10875259 PUBMEDID: 38207208

Stone RM, Haidar CE, Kornegay NM, Barker PJ, Karol SE, Wolf J, Hankins JS, Relling MV, Crews KR. Sulfamethoxazoletrimethoprim prophylaxis in pediatric oncology patients with glucose-6-phosphate dehydrogenase deficiency. Pediatr Infect Dis J :-,2024. PMCID: PUBMEDID: 39163536

Su Y, Carter JL, Li X, Fukuda Y, Gray A, Lynch J, Edwards H, Ma J, Schreiner P, Polin L, Kushner J, Dzinic SH, Buck SA, Pruett-Miller SM, Hege-Hurrish K, Robinson C, Qiao X, Liu S, Wu S, Wang G, Li J, Allen JE, Prabhu VV, Schimmer AD, Joshi D, Kalhor-Monfared S, Watson IDG, Marcellus R, Isaac MB, Al-Awar R, Taub JW, Lin H, Schuetz JD, Ge Y. The imipridone ONC213 targets α-ketoglutarate dehydrogenase to induce mitochondrial stress and suppress oxidative phosphorylation in acute myeloid leukemia. Cancer Res 84:1084-1100,2024. PMCID: PMC11380567 PUBMEDID: 38266099

Tang F, Ng CM, Horn J, Bada HS, Leggas M. Pharmacokinetic modeling and model-based hypothesis generation for dose optimization of clonidine in neonates with neonatal opioid withdrawal syndrome. Clin Pharmacol Ther :-,2024. PMCID: PUBMEDID: 39575611

Thorn HI, Guruceaga X, Martin-Vicente A, Nywening AV, Xie J, Ge W, Fortwendel JR. MOB-mediated regulation of septation initiation network (SIN) signaling is required for echinocandin-induced hyperseptation in Aspergillus fumigatus. mSphere 9:e0069523-,2024. PMCID: PMC10964416 PUBMEDID: 38349166

Turner AJ, Haidar CE, Yang W, Boone EC, Offer SM, Empey PE, Haddad A, Tahir S, Scharer G, Broeckel U, Gaedigk A. Updated DPYD HapB3 haplotype structure and implications for pharmacogenomic testing. Clin Transl Sci 17:e13699-,2024. PMCID: PMC10777430 PUBMEDID: 38129972

Umeda M, Ma J, Westover T, Ni Y, Song G, Maciaszek JL, Rusch M, Rahbarinia D, Foy S, Huang BJ, Walsh MP, Kumar P, Liu Y, Yang W, Fan Y, Wu G, Baker SD, Ma X, Wang L, Alonzo TA, Rubnitz JE, Pounds S, Klco JM. A new genomic framework to categorize pediatric acute myeloid leukemia. Nat Genet 56:281-293,2024. PMCID: PMC10864188 PUBMEDID: 38212634

Vermeire S, Dubinsky MC, Rabizadeh S, Panetta JC, Spencer EA, Dreesen E, D’Haens G, Dervieux T, Laharie D. Forecasted infliximab concentrations during induction predict time to remission and sustained disease control of inflammatory bowel disease. Clin Res Hepatol Gastroenterol 48:102374-,2024. PMCID: PUBMEDID: 38750934

Whitlock BD, Ma Y, Conseil G, O’Brien AR, Banerjee M, Swanlund DP, Lin ZP, Wang Y, Le XC, Schuetz JD, Cole SPC, Leslie EM. Differential selectivity of human and mouse ABCC4/Abcc4 for arsenic metabolites. Drug Metab Dispos 52:14171428,2024. PMCID: PMC11585317 PUBMEDID: 39313329

Wright EK, Chaparro M, Gionchetti P, Hamilton AL, Schulberg J, Gisbert JP, Chiara Valerii M, Rizzello F, De Cruz P, Panetta JC, Everts-van der Wind A, Kamm MA, Dervieux T. Adalimumab clearance, rather than trough level, may have greatest relevance to Crohn’s disease therapeutic outcomes assessed clinically and endoscopically. J Crohns Colitis 18:212-222,2024. PMCID: PUBMEDID: 37594369

Wu X, Quan M, Hadisurya M, Hu J, Liu YK, Zhuang Y, Li L, Iliuk AB, Yang JJ, Kuang S, Tao WA. Monitoring drug metabolic pathways through extracellular vesicles in mouse plasma. PNAS Nexus 3:pgae023-,2024. PMCID: PMC10833468 PUBMEDID: 38312223

Xu J, Chen C, Sussman JH, Yoshimura S, Vincent T, Pölönen P, Hu J, Bandyopadhyay S, Elghawy O, Yu W, Tumulty J, Chen CH, Li EY, Diorio C, Shraim R, Newman H, Uppuluri L, Li A, Chen GM, Wu DW, Ding YY, Xu JA, Karanfilovski D, Lim T, Hsu M, Thadi A, Ahn KJ, Wu CY, Peng J, Sun Y, Wang A, Mehta R, Frank D, Meyer L, Loh ML, Raetz EA, Chen Z, Wood BL, Devidas M, Dunsmore KP, Winter SS, Chang TC, Wu G, Pounds SB, Zhang NR, Carroll W, Hunger SP, Bernt K, Yang JJ, Mullighan CG, Tan K, Teachey DT. A multiomic atlas identifies a treatment-resistant, bone marrow progenitor-like cell population in T cell acute lymphoblastic leukemia. Nat Cancer :-,2024. PMCID: PUBMEDID: 39587259

Yoshimura S, Li Z, Gocho Y, Yang W, Crews KR, Lee SHR, Roberts KG, Mullighan CG, Relling MV, Yu J, Yeoh AEJ, Loh ML, Saygin C, Litzow MR, Jeha S, Karol SE, Inaba H, Pui CH, Konopleva M, Jain N, Stock W, Paietta E, Jabbour E, Kornblau SM, Evans WE, Yang JJ. Impact of age on pharmacogenomics and treatment outcomes of B-cell acute lymphoblastic leukemia. J Clin Oncol 42:3478-3490,2024. PMCID: PMC11458355 PUBMEDID: 39102629

Zhang C, Chan KYY, Ng WH, Cheung JTK, Sun Q, Wang H, Chung PY, Cheng FWT, Leung AWK, Zhang XB, Lee PY, Fok SP, Lin G, Poon ENY, Feng JH, Tang YL, Luo XQ, Huang LB, Kang W, Tang PMK, Huang J, Chen C, Dong J, Mejstrikova E, Cai J, Liu Y, Shen S, Yang JJ, Yuen PMP, Li CK, Leung KT. CD9 shapes glucocorticoid sensitivity in pediatric B-cell precursor acute lymphoblastic leukemia. Haematologica 109:2833-2845,2024. PMCID: PMC11367191 PUBMEDID: 38572553

Zhang X, Wang Y, Tian X, Sun L, Jiang H, Chu J, Zhou F, Shen S, Hu S, Fang Y, Lai C, Ju X, Xu X, Zhai X, Jiang H, Yang M, Leung AWK, Xue N, Zhang Y, Yang J, Pui CH, Yu J, Gao J, Hu Q, Zhu X. Delineation of features, responses, outcomes, and prognostic factors in pediatric PDGFRB-positive acute lymphoblastic leukemia patients: A retrospective multicenter study. Cancer 130:3902-3912,2024. PMCID: PUBMEDID: 39136180

Zhang W, Cai J, Wang X, Ma Y, Zhu X, Yu J, Xiao P, Gao J, Fang Y, Liang C, Li X, Zhou F, Zhai X, Xu X, Tian X, Liu A, Wang N, Zhu J, Wang L, Cheng FWT, Yang L, Zhang G, Cheng C, Yang JJ, Shen S, Li CK, Li B, Jiang H, Pui CH. Prognostic and therapeutic implications of measurable residual disease levels during remission induction of childhood ALL. Blood :-,2024. PMCID: PUBMEDID: 39786460

Zhou Y, Phelps GA, Mangrum MM, McLeish J, Phillips EK, Lou J, Ancajas CF, Rybak JM, Oelkers PM, Lee RE, Best MD, Reynolds TB. The small molecule CBR-5884 inhibits the Candida albicans phosphatidylserine synthase. MBio 15:e0063324,2024. PMCID: PMC11077991 PUBMEDID: 38587428

Zhu L. Reply: HKDC1 promotes liver cancer stemness under hypoxia via stabilizing β-catenin. Hepatology :-,2024. PMCID: PUBMEDID: 39523584

Howze, T. J., Nason, T. N., Carr, S. D., Pate, S. M., Roe, A. L., & Patel, N. D. (2025). Improving medication adherence and viral load suppression in pediatric, adolescent, and young adult patients living with HIV using a specialty pharmacy. HIV Research & Clinical Practice, 26(1). https://doi.org/10.1080/25787489.2025.2513847

Howze, T. J., Nason, T. N., Carr, S. D., Pate, S. M., Roe, A. L., & Patel, N. D. (2025). Improving medication adherence and viral load suppression in pediatric, adolescent, and young adult patients living with HIV using a specialty pharmacy. HIV Research & Clinical Practice, 26(1). https://doi.org/10.1080/25787489.2025.2513847

Howze, T. J., Nason, T. N., Carr, S. D., Pate, S. M., Roe, A. L., & Patel, N. D. (2025). Improving medication adherence and viral load suppression in pediatric, adolescent, and young adult patients living with HIV using a specialty pharmacy. HIV Research & Clinical Practice, 26(1). https://doi.org/10.1080/25787489.2025.2513847

Turn static files into dynamic content formats.

Create a flipbook
Issuu converts static files into: digital portfolios, online yearbooks, online catalogs, digital photo albums and more. Sign up and create your flipbook.