Faculty Fellowships

Office for Diversity Inclusion and Community Partnership (DICP) Faculty Fellowship

A two-year, non-degree Faculty Fellowship Program for Harvard Medical School (HMS) junior faculty that enables fellows to pursue activities that enhance their development as researchers and clinicians/teachers, leads to their advancement within the Harvard system, and promotes diversity within the HMS community.  The Program will provide two years of fellowship support in the amount of $50,000 per year int ended to provide release time from clinical work to conduct an individual, mentored research project, participate in Fellowship-related activities, meet regularly with mentors, and present research findings at the annual Minority Health Policy Meeting.


Eligibility

  • Doctoral degree (e.g. MD, PhD, DO, DMD, DDS)
  • Harvard Medical School appointment at the level of Instructor or Assistant Professor
  • Applications will also be considered from clinical or research fellows who are in the process of appointment/promotion to instructor and/or assistant professor at Harvard Medical School


DICP Faculty Fellows

2023-2025 DICP Faculty Fellowship Recipients

Jessica Addison, MD, MPH
Instructor, Boston Children’s Hospital

Mentor: Sabra Katz-Wise, PhD, Associate Professor of Pediatrics, Senior Faculty Advisor, Office of Health Equity and Inclusion, Boston Children's Hospital; Co-Director, Harvard SOGIE Health Equity Research Collaborative; Associate Professor, Harvard T.H Chan School of Public Health

Department Chair: Amy Divasta, MD, MMSc, Chief, Division of Adolescent and Young Adult Medicine, Co-Director, Reproductive Endocrinology and PCOS Program, Co-Director, Adolescent Long Acting Reversible Contraception (LARC) Program, Boston Children’s Hospital; Associate Professor of Pediatrics, Harvard Medical School

Project Title: “Retention of Care in Adolescents and Young Adults Living HIV Who Are Engaged in a Multidisciplinary Healthcare Program”

Project Description: Adolescents and young adults (AYA) living with HIV experience significant challenges to engagement in healthcare. This population is known to have lower rates of retention in care compared to their adult counterparts, resulting in poor adherence to antiretroviral therapy, and ultimately, higher viral load and lower CD4 counts that negatively impact their health. Understanding constraints and barriers to care through the lens of AYA living with HIV is vital to improving access to care and can inform future interventions to address barriers to adherence.

To reduce barriers to care, the Boston HAPPENS |LS|Human immunodeficiency virus (HIV) Adolescent Provider and Peer Education Network for Services|RS| offers an open access, multi-disciplinary approach to caring for AYA living with HIV.

The purpose of this study is to determine if this multi-disciplinary care approach keeps adolescents and young adults (AYA) living with HIV engaged in care. Our specific aims are to:

1) Understand experiences of healthcare engagement and retention, including facilitators and barriers, from the perspectives of AYA living with HIV, and  2) Determine if a patient’s health status improves when engaged with a multi-disciplinary care team.

To address Aim 1, we will conduct semi-structured qualitative interviews with AYA living with HIV who are engaged in care with the Boston HAPPENS program.  To address Aim 2, we will analyze objective measures to determine whether a patient’s health status has improved since engaging in care with the Boston HAPPENS program. A chart review will be performed on all Boston HAPPENS patients from 2017-2022.

Biography: As and Adolescent Medicine Fellow at Children’s National Health Center in Washington DC I honed my skills to become an effective medical educator, researcher, and clinician. It was during this time I started my work in advocating and providing care to youth living with HIV and transgendered youth through my work in the Burgess Clinic and the Pride Clinic respectively. I am currently a faculty member in the Division of Adolescent/Young Adult Medicine and Instructor of Pediatrics and Harvard Medical School. In this role,  provide primary and consultative care to adolescents/young adults (AYA). One specific clinical interest and passion of mine that I have been able to continue in this role is providing care to youth living with HIV and youth who engage in sexually behaviors that may increase their risk of HIV. I am Co-Medical Director of Boston HAPPENS, a multidisciplinary program that provides medical, mental health, and nutritional services to AYA living with HIV. In addition to providing medical services for youth  at risk for the HIV and other sexually transmitted infection’s (STI’s) and victims of sexual assault. I also prescribe and counsel patients about PrEP. I have presented at the National Association of Pediatric and Adolescent Gynecology conference in addition to numerous local presentations about PrEP.  I have also collaborated with other members of the Boston HAPPENS team on many grants and received funding for the following projects: Ryan White part A (Health Education Risk Reduction and Medical Nutrition Services), Ending the Epidemic funding, and Testing Together (support services that encouraged sexual partners to get STI testing together. I was also involved with developing a clinical pathway to assist medical providers in navigating counseling and prescribing or Pre-Exposure Prophylaxis. I have also worked collaboratively with colleagues to develop a non-occupational post exposure prophylaxis (nPEP) guide for patients after sexual assault.  The proposed project aligns perfectly within my clinical and research priorities of engaging, educating, and advocating for PrEP uptake in AYA. My work and experience have prepared me to successfully contribute to the proposed project. My responsibilities for this project will be recruitment at the Martha Eliot Health Center, a community health center where I provide medical care to AYA; in addition to engaging facilitating discussions with clinicians around PrEP.

Medina Jackson-Browne, PhD, MPhil, MS
Member of the Faculty of Pediatrics, Boston Children’s Hospital

Mentor: Wanda Phipitanakul, MD, Attending Physician, Division of Immunology, Director, Research Center, Division of Immunology, Boston Children’s Hospital; S. Jean Emans Professor of Pediatrics, Harvard Medical School.

Department Chair: Christopher Landrigan, MD, MPH, Chief, Division of General Pediatrics, Boston Children’s Hospital; Director, Sleep and Patient Safety Program, Brigham and Women’s Hospital; William Berenberg Professor of Pediatrics, Harvard Medical School

Project Title: “Associations between Perfluoroalkyl Substances (PFAS) Exposure and Aeroallergen Sensitization among Infants at High-Risk for Developing Asthma”

Project Description: We propose evaluating the extent to which PFAS exposure plays a role in aeroallergen sensitization, a known precursor to asthma development. The proposed study population will be approximately 200 children aged 24, and up to 47 months of age who are at high risk for asthma enrolled in the on-going NIH-funded pediatric clinical trial (PARK). Perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) are PFAS almost universally detected in the serum of pregnant women and neonates worldwide. Serum PFOA and PFOS concentrations will be quantified from children serum samples and aeroallergen sensitization status will be assessed by allergen skin testing at the PARK clinical centers. Additionally, the PARK clinical trial will have biomarkers of immune function and other subclinical outcomes measures from urine and serum samples, medical history from questionnaires, and other extensive clinical and medical data including anthropometrics and medical/healthcare utilization measures. Using the baseline data, we propose to:

1. Evaluate the cross-sectional associations of PFAS with aeroallergen sensitization and other asthma development risk factors in high-risk infants. We will explore whether PFAS exposure are associated with allergen skin prick test results and other secondary subclinical outcomes (e.g., biomarkers of immune function, hypersensitivity biomarkers). We will adjust for sociodemographic and allergic disease risk factors.

2. We will also evaluate effect modification and potential mediators of the associations in Aim 1. Priority modifiers and mediators will include sex, race/ethnicity, home environment, and other chemical exposures.

Biography: My long-term career goal is to develop an independent career of high-quality research in children's environmental health. I study early life environmental chemical exposures, hormonal/molecular pathways that are sensitive to environmental chemical exposures, and health disparities in urban children. Specifically, I have used prospective cohort studies to quantify the association of early life exposure to phenols and perfluoroalkyl substances, with children’s neurodevelopment and immune and respiratory system function. Through this DICP Faculty Fellowship proposal entitled “Associations between Perfluoroalkyl Substances (PFAS) Exposure and Aeroallergen Sensitization among Infants at High-Risk for Developing Asthma”, we propose to evaluate the cross-sectional between PFAS serum levels and allergic sensitization (IgE antibody production) in a cohort of infants from age 24-47 months with a highrisk of developing asthma enrolled in the “Controlling and Preventing Asthma Progression and Severity in Kids” short title PARK (Preventing Asthma in High-Risk Kids), U01 AI 126614, PI: Wanda Phipatanakul, MD) clinical trial. This fellowship focuses on enhancing my clinical research capabilities and community-based pediatric cohort building skills. I am privileged to have an outstanding research mentor, Dr. Wanda Phipatanakul, MD, who has devoted considerable time and effort to my research and career development since August 2019. With Dr. Phipatanakul’s guidance, I recently joined the division of pediatrics at Boston Children's Hospital as a full-time research scientist, leaving a tenure-track position at the University of Delaware. This transition gave me the opportunity to focus on research full-time at one of the top clinical pediatric institutions in the country. Under Dr. Phipatanakul I have also successfully submitted four NIH proposals of which three have been awarded: 1) NIH Research Supplement to Promote Diversity in Health-Related Research, 2) NIH Pediatric Loan Repayment Renewal award, and 3) 2 separate NIH Human Health Exposure Analysis Resource (HHEAR) awards. Additionally, I have published 3 first author manuscripts during this time as well. My contributions to Dr. Phipatanakul’s research team will, enrich the overall goal of the parent study by undertaking paralleling and complimentary specific aims to deepen the understanding of early- life PFAS exposures and the immune and respiratory systems in children at high risk for developing asthma. Longer term, this clinical trial will also allow us to measure the effects of these and other chemical exposures on the efficacy of the intervention on the primary outcome (asthma development). Additionally, I plan to extend my training by learning how to successfully recruit and develop a pediatric clinical trial and to learn cutting-edge methodologies that will allow me to begin to disentangle the complex relationship between environmental exposures and early-life allergic disease development. This award will contribute to the fulfillment of my career goal to develop into an independent interdisciplinary researcher in pediatric environmental health research by giving me the tools to identify risk factors for allergic disease development and morbidity in children.

I was born and raised in Harlem, NYC and all my primary family member still reside there and all work in healthcare. Living alone in a new city, starting a new job away from my family during the COVID-19 pandemic heavily impacted my research progression during my time at UD, specifically, manuscript publication. However, I recently joined the research faculty in the Division of General Pediatrics at BCH as a Senior Research Scientist in September 2022. Although it was difficult to leave a hard-money, tenure track faculty position, I believed the great working relationship with Dr. Phipatanakul and the resources available to support my career development at BCH and Harvard Medical School were unmatched.

Malika Boudries, PhD
Assistant Professor of Medicine, Beth Israel Deaconess Medical Center/ Center for Virology and Vaccine Research

Mentor: Omar K. Siddiqi, MD, MPH, Assistant Professor of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School; Visiting Lecturer, University of Zambia School of Medicine

Division Chief: Dan H. Barouch, MD, PhD, Director, Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center; Member, Ragon Institute of MGH, MIT, and Harvard; Williams Bosworth Castle Professor of Medicine, Professor of Microbiology and Immunology, Harvard Medical School;

Project Title: “HIVEpi-Lat: Longitudinal Profiling of the Genomics and Epigenetics Features of SIV Integration Site in ART suppressed rhesus macaques at the single cell level.”

Project Description: In this project, we will implement a new computational tool for multi-level profiling of SIV integration and host interaction. We will employ a multi-omics TEA-Seq approach, combined with integration site profiling of SIV latent cells in SIV-infected-ART suppressed macaques at the single cell level. Longitudinal samples from SIVmac251-infected, ART-suppressed macaques were obtained from an archived study at baseline, week 0, weeks 4, 72, and 136 on ART suppression. PBMCs, LNMCs, and plasma samples were collected from all animals at the specified time points. We plan to implement a mathematical model to establish a multi-level association network between integration sites, proviral sequences, and the epigenetic and plasma features associated with transcriptionally active or latent infected cells. By capturing the complex relationships among different layers of the generated data, these models will not only reveal the factors governing proviral integration but also help to elucidate the role of various genomic and epigenetic features in the persistence and survival of SIV-infected cells during ART suppression. By leveraging the power of multi-omics profiling, proviral integration, and advanced analytical techniques, this proposal aims to address the critical questions surrounding HIV latency and integration and pave the way for groundbreaking advancements in HIV research and therapy. If validated in humans, our work will provide invaluable insights into the complex dynamics of HIV latency and pave the way for novel therapeutic strategies.

Biography: I am an Assistant Professor of Medicine specializing in Computational and Systems Biology and Bioinformatics at the Center for Virology and Vaccine Research (CVVR). My Primary research focuses on developing and implementing machine learning-based computational approaches for gene signature associate with disease pathogenesis, innate correlate, and biomarkers of vaccine response efficacy. I am also involved in studies identifying transcriptomics and proteomics correlates of vaccine protection durability. These methods were successfully applied to the recent Ad26 COVID-19 vaccine using transcriptomics, and proteomics data sets from individuals, rhesus macaques, and hamsters vaccinated with Ad26.COV2.S vaccine. At CVVR, my work bridges multiple disciplines, including immunology, virology, computer science, statistics, mathematical modeling, and machine learning to analyze and interpret biological omics data sets that include but are not limited to bulk RNA-Seq, single RNA-Seq, Path-Seq, ATAC-Seq, Metabolome, Proteome, System serology, and Flow cytometry.

George Molina, MD, MPH
Assistant Professor, Brigham and Women’s Hospital

Mentor: Quoc-Dien Trinh, MD, Director, Ambulatory Clinical Operations, Division of Urology Surgery, Department of Surgery, Brigham And Women’s Hospital; Associate Professor of Surgery, Harvard Medical School

Department Chair: Chandrajit P. Raut, MD, MSc, Chief, Division of Surgical Oncology, BWH Distinguished Chair for Cancer Care, Brigham and Women’s Hospital; Surgery Director, Center for Sarcoma and Bone Oncology, Dana Farber Cancer Institute; Professor of Surgery, Harvard Medical School

Project Title: “Improving Survival among Patients with Colorectal Liver Metastasis by Using a Regional and Hospital System-level Approach to Improve Equitable access to Liver Surgery.”

Project Description: The overall objective of the project is to determine modifiable regional and hospital system-level drivers of variation in access to liver surgery for colorectal liver metastasis (CRLM) by characterizing these drivers for a group of complex gastrointestinal cancers (CRLM, pancreatic cancer, and gastric cancer). The central hypothesis is that a regional and hospital system-level understanding is needed to address variation in resources and systems and thus improve survival for patients with CRLM. This project addresses variation in undergoing a liver surgical resection for CRLM, despite significant survival benefit in undergoing a liver metastasectomy. Additionally, variation in access to a liver surgical resection for CRLM is a significant problem because it disproportionately impacts patients who have been historically underserved. I will address the following specific aims: (1) Characterize the source and uncover the pattern of variation in surgical treatment for this group of cancers across health service areas in the US; (2) Design a comparative case study to describe predictive regional factors that are common among this group of cancers and are associated with access to surgery. The findings from this study could improve survival for all patients with CRLM, regardless of race or place of residence, by improving access to and reducing regional disparities in surgery for CRLM.

Biography: I am a practicing Surgical Oncologist specializing in hepatopancreatobiliary cancers in the Division of Surgical Oncology at Brigham and Women's Hospital (BWH) and the Gastrointestinal Cancer Treatment Center at Dana-Farber Cancer Institute (DFCI), and an Assistant Professor of Surgery at Harvard Medical School. I obtained an MPH in Quantitative Methods from the Harvard T.H. Chan School of Public Health (HSPH) and I have skills in using SAS and Stata programming. As part of my training in general surgery at Massachusetts General Hospital (MGH), I completed a two-year post-graduate research fellowship at Ariadne Labs, a joint health systems innovation center of BWH and HSPH. During this professional development time, I was involved in or led projects on health systems innovation and research, global surgery modeling, clinical studies in surgical oncology, surgical safety culture in inpatient and ambulatory settings, and impact of a surgical safety checklist program. As a fellow in complex general surgical oncology at DFCI, I have collaborated in projects using the National Cancer Database evaluating the association between neoadjuvant therapy and overall survival for pancreatic cancer. As an early-career academic surgeon, I have led work evaluating the association between volume of major liver surgery and receiving care at a Commission on Cancer-accredited hospital and undergoing a liver metastasectomy for colorectal liver metastasis.

Azeesat Babajide, MD, MBA
Instructor, Cambridge Health Alliance

Mentor: Nicholas Carson, MD, Chief, Division of Child and Adolescent Psychiatry, Director, Child and Adolescent Outpatient Psychiatry, Clinical Research Associate, Health Equity Research Lab, Cambridge Health Alliance; Assistant Professor of Psychiatry, Harvard Medical School

Mentor: Benjamin Le Cook, PhD, MPH, Director of the Health Equity Research Lab, Director ofResearch, Department of Psychiatry, Cambridge Health Alliance; Associate Professor, Harvard Medical School

Department Chair: Carl Fulwiler, MD, PhD, Chair and Chief of Psychiatry (Interim), Cambridge Health Alliance; Associate Professor in Psychiatry, Harvard Medical School

Project Title: "Identifying Barriers and Facilitators to Mental Health Care Transitions for Young Adults in a Safety Net System"

Project Description: Young adults (18-25 years) often fall out of mental health (MH) care as they transition into adulthood and are less likely to engage in MH care compared to other age groups despite higher rates of mental illness. This treatment gap is worse for racial, ethnic, and linguistic (REL) minorities. Few services exist to support youth with these MH care transitions, and even fewer focus on the needs of REL minority youth in public healthcare settings. Clinical entities serving minority youth, particularly community health systems, require strategies for using electronic medical record (EMR) data to provide insight into MH care use and to inform interventions that target those at highest risk of failure to transition. This mixed methods research project seeks to 1) use EMR data to assess MH care use and transition rates as patients enter adulthood, 2) examine predictors of MH care follow-up and transition to adult services among REL minority young adults compared to White young adults, and 3) understand barriers and facilitators to MH care engagement and transition using semi-structured qualitative interviews. The outcomes of this research will expand understanding of MH care transitions for REL youth in a community health system and lay the groundwork for the translation of these methods for use in other health systems. All three aims will provide insight into barriers to transition and support the development of an intervention targeting youth transition.

Biography: Currently, I am an Instructor in psychiatry at Harvard Medical School and a staff psychiatrist at Cambridge Health Alliance (CHA). Throughout my career, I have maintained interests in systems of care, including considerations around the utility of integrated care models and challenges around transitions of care for young adults, particularly thoughts from underserved backgrounds. I have many years of clinical experience working with youth in underserved communities, which has led to a deep appreciation for the challenges they face when transitioning out of pediatric mental health services. I have presented work in this area at the American Psychiatric Association’s Annual Conference andhave served on the American Psychiatric Association's Council for Children Adolescents and their Families. As part of my work with the Council, I helped write a position statement advocating for increased attention to the needs of these transition-aged youth. I have published a review article with first authorship focusing on the developmental needs for transition-aged youth with depression and anxiety disorders and the challenges they face during the transition to adulthood. This review explored the possibility of integrated care models as a way to address transition needs for these patients. At CHA, I am one of a few faculty selected for the Learning Health Scholars Program, a program thatinvolves seminars on research design and methods and affords me 4 hours of protected time a week for research. In my time as a Learning Health Scholar, I have conducted preliminary work that provided critical information regarding the mental health transition landscape for youth transitioning to adult services from the provider perspective. As part of that work, I conducted five focus groups with CHA psychotherapists, psychiatrists, and care partners to identify themes around provider experiences in working with and engaging young adults. I also administered and analyzed survey data from focus group participants with a 40% response rate. The focus groups and survey data provided important information about potential barriers to transition for youth at CHA. I intend to build upon this work with my current project, which seeks to quantify rates of mental health care utilization among youth as they transition into adulthood, identify predictors for engagement in services in adulthood, and explore barriers and facilitators to successful transition to adult services with a particular focus on racial, ethnic, and linguistic minoritized youth. The time afforded by the Learning Health Scholar’s program has allowed me to familiarize myself with the EMR variables available through the Health Equity Research Lab’s data warehouse. This provides foundational understanding that will be imperative for the quantitative analyses that I will conduct as part of this project

Juan Matute, MD
Instructor, Massachusetts General Hospital

Mentor: Richard Blumberg, MD, Jerry S. Trier Distinguished Chair in Gastroenterology, Hepatology and Nutrition, Brigham and Women’s Hospital; Vice Chair, Department of Medicine, Professor of Medicine, Harvard Medical School

Mentor: Alessio Fasano, MD, W. Allen Walker Chair in Pediatric Gastroenterology and Nutrition, Division Chief, Pediatrics Gastroenterology, and Nutrition, Massachusetts General Hospital;Professor of Pediatrics, Harvard Medical School

Division Chief: Paul Lerou, MD, Chief, Division of Neonatology and Newborn Medicine, Massachusetts General Hospital; Assistant Professor of Pediatrics, Harvard Medical School

Project Title: “Gut Intelectin-1 protects from Obesity.”

Project Description: Obesity is a complex condition influenced by both genetic factors and the environment, starting early in life. However, we still have limited understanding of how specific genetic variations contribute to obesity and how they can be targeted for therapeutic interventions. Our project focuses on the role of a genetic factor called Intelectin-1, which has been linked to obesity and type 2 diabetes.

Intelectin-1 is a protein produced by cells in the intestines and has been found to bind to certain molecules present in bacteria. Interestingly, we have observed that levels of Intelectin-1 are reduced in individuals with obesity and diabetes. In mouse studies, we have also discovered that Intelectin-1 in the gut can protect against obesity.

While previous research on Intelectin-1 in the pathophysiology of obesity has primarily focused on the direct effects of Intelectin-1 on the hypothalamus and visceral fat, we aim to investigate its protective role in obesity through the gastrointestinal tract. By using transgenic mice and studying human samples, we seek to unravel how Intelectin-1 in the intestines can influence obesity susceptibility.

Understanding how Intelectin-1 acts in the gut may provide valuable insights for developing novel strategies to prevent and treat obesity. Our research has the potential to contribute to the development of innovative approaches to modulate obesity risk factors. By bridging the gap between genetic associations and therapeutic interventions, we hope to make a meaningful impact on the fight against obesity.

Biography: Dr. Matute is a neonatologist and researcher in mucosal immunology at Massachusetts General Hospital (MGH). He obtained his Medical Degree from La Universidad de Antioquia (Colombia). Prior to his pediatric internship, he engaged in basic immunology research at Indiana University. Subsequently, he completed his pediatric residency through the Boston Combined Residency Program, receiving training at Boston Children’s Hospital (Harvard Medical School) and Boston Medical Center (Boston University School of Medicine). He further specialized in Perinatal and Neonatal Medicine through the Harvard Combined Program, which included training at MGH, Boston Children’s Hospital, Brigham and Women’s Hospital, and Beth Israel Deaconess Medical Center. Following his clinical training, Dr. Matute joined MGH and Harvard Medical School HMS as an instructor. He currently dedicates his time to mucosal immunology research and clinical care in the MGH Neonatal Intensive Care Unit. Additionally, he leads initiatives focused on optimizing neonatal gastrointestinal health and serves as the Director of Equity in the Division of Newborn Medicine at MGH. Under the guidance of Dr. Richard Blumberg and Dr. Fasano, Dr. Matute has conducted extensive research on the host-microbiota interface starting in early life, with a specific focus on its implications for homeostasis, the pathogenesis of intestinal inflammation and obesity. His work has received support from the American Heart Association, the Pediatric Scientist Development Program, the Crohn’s and Colitis Foundation, and the Harvard Digestive Disease Center.

2022 DICP Faculty Fellowship Recipients

Oren Ganor, MD

Oren Ganor, MD
Assistant Professor of Surgery, Harvard Medical School, Boston Children’s Hospital

Division Chief and Mentor: John G. Meara, MD, DMD, MBA, Kletjian Professor of Global Surgery, Professor of Surgery in the Field of Pediatrics Plastic Surgery, Harvard Medical School; Plastic Surgeon-in-Chief, Boston Children’s Hospital 

Project Title: “Effects of Gender-Affirming Hormones on Facial Anthropometrics in Transgender Youth”

Project Description: Gender-affirming, or "cross-sex", hormone therapy, is one of the most common interventions prescribed to transgender individuals with the intention of improving congruence between their sense of self and their external characteristics. While many changes resulting from these hormones have been documented, little is known about the impact of gender-affirming hormones on facial sexual dimorphism and what relationship these changes may have on psychosocial wellbeing. The proposed research addresses this dearth by examining the effects of gender-affirming hormones on facial anthropometrics and psychosocial wellbeing in a prospective cohort of 50 trans-masculine individuals starting gender-affirming testosterone at Boston Children's Hospital. In the context of increasing interest in gender-affirming surgeries, it becomes increasingly important for surgeons and patients alike to have quality data to guide decision-making. The proposed study is the first of its kind to prospectively measure the effects of gender-affirming hormones on facial anthropometrics and to study the relationship between facial anthropometric changes and gender congruence. These data have the potential to shape guidelines for decision-making around facial masculinization by providing estimates of the extent and speed of facial changes expected to occur on testosterone and may improve the ability of providers to counsel transgender patients on expected changes from gender-affirming hormones and how these hormones may affect the ideal timing of any desired facial surgical procedures.

Biography: Dr. Oren Ganor, Assistant Professor at Harvard Medical School, is a plastic and reconstructive surgeon who specializes in complex reconstruction, microsurgery, and gender-affirming surgeries. He is a co-director of the Center for Gender Surgery at Boston Children’s Hospital, which is proud to be the first transgender surgical center in a pediatric hospital in North America. Dr. Ganor received his education and training in Israel. He later completed two clinical fellowships in Microsurgery at BIDMC, and Craniofacial & Pediatric Plastic Surgery at BCH, prior to joining as faculty at Boston Children’s Hospital. As part of his clinical work offering chest, genital, and facial surgeries, he is also deeply invested in clinical research and is currently working on several projects designed to improve surgical outcomes and quality of life of gender non-conforming individuals. Through his work, Dr. Ganor aspires to use research to enhance the field of gender-affirmation surgery, provide the highest quality of care to patients, and educate the next generation of healthcare providers in highest quality gender care.

Bon Trinh, PhD

Bon Trinh, PhD
Instructor, Harvard Medical School,  Beth Israel Deaconess Medical Center

Mentor: Daniel G. Tenen, MD, Professor of Medicine, Harvard Medical School; Professor of Hematology and Oncology, Beth Israel Deaconess Medical Center

Division Chief: David E. Avigan, MD, Chief of Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center; Professor of Medicine, Harvard Medical School

Project Title:  " Preclinical Development of RNA-based Therapeutics for Acute Myeloid Leukemia"

Project Description: Survival rate of patients with Acute Myeloid Leukemia (AML) remain at less than 30% and have not been significantly improved over the last decade. Innovative approaches for AML therapeutics are therefore needed. The PU.1 transcription factor is a critical regulator of normal hematopoiesis and functions as a key tumor suppressor in AML. However, there are no available therapies directly focus on this pivotal molecule. In general, therapeutically activating tumor suppressor transcription factors remains a technical challenge. Recently, we discovered a long noncoding RNA that functions as an RNA inducer of PU.1. We further demonstrated that the RNA inhibits AML cell growth and promote cell differentiation, indicating that it exhibits AML inhibitory functions. This proposal aims to utilize the RNA in developing RNA-based therapeutic approaches for AML. We propose to boost this RNA in AML cell lines and in mice using two FDA-approved delivery systems for clinical use, namely the Adeno-associated virus-mediated gene delivery and the poly(DL-lactide-co-glycolide)-based nanoparticle (PLGA-NP) RNA delivery. We will validate the effects of the RNA on growth and differentiation of AML cell lines. Furthermore, we will determine whether the RNA reduces leukemia features and prolongs survival in AML mouse models. The success of this preclinical study will provide a proof-of-concept for approaches that utilize tumor-suppressing noncoding RNAs in AML therapeutics. Ultimately, this could result in further clinical studies leading to development of effective RNA-based drugs for blood malignancies and cancer.

Biography: Bon Trinh, PhD is an Instructor of Medicine at Harvard Medical School and a Staff Scientist at Beth Israel Deaconess Medical Center. He received his PhD degree from the University of Texas Graduate School of Biomedical Sciences. Throughout his research training at UT MD Anderson Cancer Center and Harvard, he has been investigating protein- and RNA-mediated gene regulations in diverse biological systems including normal blood development, cancer cell proliferation, drug resistance, tumor angiogenesis, and metastasis. Dr. Trinh’s current work focuses on understanding the role of proteins and RNAs, via modulation of chromatin architecture, in normal immune cell development and abnormalities in this molecular interplay in leukemia progression as well as developing therapeutic strategies targeting chromatin structure. His research has been supported by an K01 training grant from the National Cancer Institute. His work was selected for featuring at an early career highlight seminar for instructors and assistant professors of Beth Israel Medical Center, and was awarded an Abstract Achievement Award from the American Society of Hematology.

Ana Paula Abreu, MD, MPH

Ana Paula Abreu, MD, MPH
Assistant Professor of Medicine, Harvard Medical School,  Brigham and Women’s Hospital

Mentor: Ursula Kaiser, MD, Chief, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital; Professor of Medicine, Harvard Medical School

Department Chair: Joseph Loscalzo, MD, PhD, Hersey Professor of the Theory and Practice of Physics, Harvard Medical School; Head of the Department of Medicine, Brigham and Women’s Hospital

Project Title:  “Genomic and Clinicopathologic characterization of aggressive pituitary adenoma”

Project Description: Pituitary corticotroph adenomas (CA) are a pituitary tumor subtype with positive immunohistochemistry for ACTH. The pathogenesis of CA is poorly understood. These tumors can secrete bioactive ACTH resulting in excess of cortisol levels and Cushing disease (CD) or be extremely aggressive without secretion of bioactive ACTH, these are called Silent corticotroph adenomas (SCA). CA have high morbidity and mortality. CD is associated with obesity, metabolic syndrome, anxiety and several other symptoms. Adenomas causing CD are usually small and difficult to identify in brain MRI, therefore full surgical removal is challenging. Medical treatment is usually not efficacious to treat the adenoma and drugs targeting the over secretion of cortisol are usually used with several side effects. The identification of somatic mutations in USP8 in CD provided exciting advances in this field identifying the molecular pathway driving these tumors with potential for target therapies. SCA are usually large adenomas presenting with compressive symptoms with aggressive markers in the pathology report. They are difficult to remove surgically due to invasiveness. Very little is known about the molecular pathways of these tumors. I have identified mutations in TP53, ATRX and DAXX in one third of these tumors. This proposal will explore target treatments for CD with USP8 mutations and the molecular pathways by which the loss of TP53, ATRX and DAXX result in SCA. I will also test target therapies for SCA. Dataobtained in this proposal will expand the knowledge on SCA generate potential tools for the treatment of CA.

Biography:  Ana Paula Abreu Metzger, M.D., Ph.D., is an Assistant Professor of Medicine at Harvard Medical School (HMS), an Associate Physician in Endocrinology at Brigham and Women's Hospital (BWH), Division of Endocrinology, Diabetes and Hypertension, and a member of the faculty at Harvard-MIT Health Science & Technology. obtained her M.D. at the Faculty of Medicine of Universidade Federal de Minas Gerais in Brazil. She later obtained her Ph.D. degree at the Universidade de São Paulo. During her Ph.D. training, she worked to identify genes associated with Hypogonadotropic Hypogonadism and Central Precocious Puberty and performed in-vitro experiments to understand the molecular mechanisms by which these genes act centrally to cause these disorders. Thereafter, she was awarded an NIH F05 International Neuroscience Fellowship to do her Postdoctorate Fellowship at BWH in the Division of Endocrinology, Diabetes, and Hypertension. During this time, she initiated a collaborative study to perform whole-exome sequencing analysis of families with central precocious puberty.This study culminated in identifying the most common genetic cause of central precocious puberty, loss-of-function mutations in MKRN3. More recently, she was the recipient of an NIH K99/R00, "Pathway to Independence Award," to perform studies on the molecular mechanisms of action of MKRN3. Her work has been internationally recognized and she is the recipient of several awards, including the "Neena B. Schwartz Award" for best basic abstract by the Women in Endocrinology and the "Young Scientist Award" from the American Society for Clinical Investigation. She received the "2018 Department of Medicine Innovation Evergreen Fund Award" for studying genetic drivers of ACTH positive pituitary adenomas, leading her to start a new innovative translational research area to investigate molecular mechanisms driving the formation of corticotroph pituitary lesions and their behavior. Clinically she is interested in and takes care of patients with neuroendocrine, reproductive, and endocrine genetic disorders. She is one of the founders and now co-director of the Brigham Center for Endocrine Genetics, which seeks to expand the access to genetic counseling and testing in the Division of Endocrinology as well as to provide better training to trainees (or fellows) and provide personalized medicine to patients.

Ayobami Akenroye, MBChB, MPH

Ayobami Akenroye, MBChB, MPH
Member of Faculty, Harvard Medical School, Brigham and Women’s Hospital

Mentor: Scott Weiss, MD, Professor of Medicine, Harvard Medical School, Brigham and Women’s Hospital

Department Chair: Joshua Boyce, MD, Albert L. Sheffer Professor of Medicine in the field of Allergic Disease, Harvard Medical School; Chief, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital

Project Title:  “Integrative Profiling of Response to Monoclonal Antibodies in Asthma"

Project Description: Multiple monoclonal antibodies are currently approved for the treatment of severe asthma with many individuals with severe asthma meeting eligibility criteria for two or more of these agents, i.e., “multiply eligible”. However, the monoclonal antibody that would provide the greatest clinical benefits in these multiply eligible patients is usually unknown. To date, models for prediction of response to these therapies based on clinical biomarkers such as peripheral blood eosinophil counts have demonstrated low accuracy. In this study, we seek to improve the predictive accuracy of these models by integrating sociodemographic and clinical features with immunologic profiling. We will apply supervised machine learning methods to develop models predictive of response which incorporate 1.) Sociodemographic and clinical features; 2.) Immunologic and metabolomic signatures. Subsequently, we will integrate all available features in a multidimensional model to identify novel clusters which will facilitate our understanding of treatment response to monoclonal antibodies in asthma.

Biography:  Ayobami Akenroye is an Associate Physician in the Division of Allergy and Clinical Immunology, and Associate Epidemiologist in the Channing Division of Network Medicine at the Brigham and Women’s Hospital (BWH). Dr. Akenroye graduated top of her medical school class in Ile-Ife, Nigeria. Subsequently. she arrived in the US for the MPH program at Harvard School of Public Health. She completed her residency in internal medicine at the Albert Einstein College of Medicine, NY, and clinical and research fellowship in Allergy/Immunology at Johns Hopkins University. Her research interests are in asthma and pharmacoepidemiology. Specifically, her work focuses on the comparative effectiveness of monoclonal antibodies approved for the treatment of asthma, and the identification of predictors of response. She is the recipient of the BWH Minority Faculty Career Development Award and the NIH K99/R00 MOSAIC award to support her work using real-world data to generate evidence on the comparative effectiveness of therapies for the treatment of asthma. During the period of the Diversity Inclusion and Community Partnership Faculty Fellowship, Dr. Akenroye will expand her skills to the use of pharmaco-omics approaches in predicting treatment response.

Asishana Osho, MD, MPH

Asishana Osho, MD, MPH
Member of the Faculty of Surgery, Harvard Medical School, Massachusetts General Hospital

Mentor: Oluwaseun Johnson-Akeju, MD, Henry Isaiah Dorr Associate Professor of Research and Teaching in Anaesthetics and Anaesthesia, Harvard Medical School; Head of the Department of Anaesthesia, Massachusetts General Hospital

Department Chair: Keith Lillemoe, MD, W. Gerald Austen Professor of Surgery, Harvard Medical School; Head of the Department of Surgery, Massachusetts General Hospital

Project Title:  "A Randomized Controlled Trial of Low-dose Amiodarone for Preventing Atrial Fibrillation following Major non-coronary Cardiac Surgery"

Project Description: The incidence of atrial fibrillation following major cardiac valve surgery is unacceptably high. 30-50% of these patients will suffer post-operative atrial fibrillation with associated increases in the risk of developing further complications including stroke, heart failure and death. Despite this high rate of occurrence, the literature on postoperative atrial fibrillation is insufficient, with gaps in the knowledge about optimal prevention, duration of disease and patterns of presentation. Amiodarone, a medication that modulates the transmission of electrical signals in the heart, has been shown to significantly reduce the incidence of postoperative atrial fibrillation in patients undergoing coronary surgery. However, this medication is not routinely used in cardiac surgery. Additionally, dedicated studies have not been performed in patients undergoing non-coronary cardiac operations (Including major valvular surgery and aortic procedures). We have designed a randomized controlled trial to evaluate the efficacy and safety of low-dose prophylactic amiodarone for the prevention of post-operative atrial fibrillation following major, non-coronary cardiac surgery. Integration of an analysis using wearable biosensors placed on the wrists and/or chests of study participants at the time of discharge will delineate the duration and characteristics of atrial fibrillation in discharged patients. Overall, this project innovates clinically and technologically to advance our understanding of a major cause of morbidity and mortality in cardiac surgery.

Biography:  Dr. Asishana Osho is a surgeon in the Corrigan Minehan Heart Center at Massachusetts General Hospital who specializes in adult cardiac surgery, heart failure and thoracic transplantation. He earned a BA with high honors from Oberlin College, an MPH from the Yale University School of Public Health, and an MD from the Duke University School of Medicine. He completed residencies in general and cardiothoracic surgery at Massachusetts General Hospital/Harvard Medical School. Dr. Osho conducts clinical, translational and health services research to understand and improve health outcomes in patients undergoing cardiothoracic surgery. He also has significant experience designing and implementing clinical trials in cardiothoracic surgery. Dr. Osho’s research projects have been supported by the American Heart Association, the Thoracic Surgery Foundation, and the Bollinger research program at Duke University.

Luis Gonzalez Castro, MD, PhD

Luis Gonzalez Castro, MD, PhD
Instructor of Neurology, Harvard Medical School, Brigham and Women’s Hospital/ Dana Farber Cancer Institute

Mentor: Mario Suva, MD, PhD, Associate Professor of Pathology, Harvard Medical School and Brigham and Women’s Hospital

Department Chair: Tracy T. Batchelor, MD, Miriam Sydney Joseph Professor of Neurology, Harvard Medical School; Neurologist in Chief, Brigham and Women’s Hospital

Project Title:  "Modulating the Disease Activity of Glioblastoma Via Cellular Reprogramming"

Project Description:  Glioblastoma, the most common malignant brain tumor, has a median overall survival of only 16 months. The cellular heterogeneity of glioblastoma is the main barrier to the effective treatment of this disease. Our work profiling gene expression in glioblastoma at single-cell resolution has demonstrated that the cellular heterogeneity of glioblastoma can be explained by the combination of four transcriptional programs - neural-progenitor-cell-like (NPC-like), oligodendrocyte-progenitor-cell-like (OPC-like), astrocytic-cell-like (AC-like), and mesenchymal-cell-like (MES-like). However, the gene regulation determinants of the transcriptional programs in each cellular state remain unknown. Knowledge of these regulatory elements will enable cellular reprogramming to decrease the tumor’s heterogeneity, making it less aggressive and more responsive to therapy. To establish the regulatory elements that underlie each gene expression program, I propose to profile available chromatin (as characterized by the assay of transposase accessible chromatin, ATAC) and gene expression at single-cell resolution in fresh tumor specimens from glioblastoma patients at the time of surgical resection. I will then perform motif enrichment analyses to determine which DNA-binding transcription factors control the transcription of the genes that characterize each transcriptional program. Subsequently, these findings will be validated through transcription factor overexpression experiments using patient-derived gliomaspheres, to demonstrate reprogramming of cellular states. Once the transcription factors that underlie cellular reprogramming are validated, I will perform a small molecule screen to identify candidates to disrupt transcription factor activity. These compounds will enable preclinical testing and eventual evaluation in clinical trials for the treatment of glioblastoma.

Biography:  L. Nicolas Gonzalez Castro, MD, PhD is a neurologist and neuro-oncologist at the Department of Neurology, Brigham and Women’s Hospital, and the Center for Neuro-Oncology, Dana-Farber Cancer Institute. He trained in systems neuroscience and computational biology as an MD-PhD student at the Harvard-MIT Division of Health Sciences and Technology, and completed his neurology residency at the Harvard Neurology Residency Program (Massachusetts General Hospital / Brigham and Woman’s Hospital). He did his neuro-oncology training at the Massachusetts General Hospital Cancer Center / Brigham and Woman’s Hospital - Dana Farber Cancer Center Fellowship Program. His current research focus is in understanding the cellular programs that underlie differentiation, survival and resistance to treatment in primary brain tumors.

View Fellowship Alumni