Faculty Fellowship

Our program offers a two-year, non-degree faculty fellowship program for Harvard Medical School junior faculty, which is designed to address the need for additional support to conduct clinical and/or translational research and to free junior faculty from clinical and teaching demands at a key point in their career development. Each faculty fellow will receive $100,000 over a two-year period to support their scholarly efforts. Faculty fellows are required to devote appropriate time toward the development of their academic career, to meet regularly with their mentors, and to present at the annual Minority Health Policy Meeting.

Eligibility

Doctoral degree (e.g. MD, PhD, DO, DMD, DDS). Harvard faculty 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 University. U.S. Citizenship or permanent residency.

How to Apply

Applications are currently being accepted for the Harvard Catalyst Program for Diversity and Inclusion Faculty Fellowship.

DEADLINE: Extended to December 15, 2023

Review the Faculty Fellowship Application Instruction Package.

To apply, submit your application online.

2023-2025 PFDI Faculty Fellows

Diana Lopez, MD, Instructor, Harvard Medical School, Brigham and Women's Hospital

Mentor: Ron Blankstein, MD, FACC, FASNC, MSCCT, FASPC, Associate Director, Cardiovascular Imaging Program, Director, Cardiac Computed Tomography, Co-Director, Cardiovascular Imaging Training Program, Senior Physician, Preventive Cardiology, Brigham and Women’s Hospital; Professor of Medicine and Radiology, Harvard Medical School

Mentor: Marcelo F. Di Carli, MD, Chief, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Executive Director, Cardiovascular Imaging Program, Departments of Radiology and Medicine, Brigham and Women’s Hospital; Seltzer Family Professor of Radiology and Professor of Medicine, Harvard Medical School

Department Chair: John Keaney Jr, MD, Chief, Cardiovascular Medicine, Co-Executive Director, Heart and Vascular Center, Victor J. Dzau Professor of Medicine, Harvard Medical School

Project Title: “The Impact of PCSK9-Inhibition on Myocardial Flow Reserve (EMPOWER Study)

Project Description: Diffuse non-obstructive atherosclerosis and coronary microcirculatory dysfunction (CMD) are prevalent in patients with cardiovascular disease and consistently identify patients at increased risk for adverse outcomes, even in the absence of obstructive CAD. These abnormalities throughout the coronary vasculature are closely inter-related manifestations of atherosclerosis that have been linked to systemic inflammation. Positron Emission Tomography (PET) myocardial flow reserve (MFR) is a robust and reproducible imaging biomarker that integrates the hemodynamic effects of atherosclerosis across the entire coronary circulation and has been shown to predict outcomes, including higher rates of cardiovascular (CV) death. PCSK9 inhibitors are powerful agents that promote epicardial plaque regression and reduce the risk of CV events, but studies are needed to evaluate their effect on the coronary microvasculature and their potential anti-inflammatory effect on atherosclerosis. The investigator-initiated mechanistic clinical trial, “The Impact of PCSK-9 Inhibition on Myocardial Flow Reserve (EMPOWER study)” will enroll 50 participants to assess whether PCSK-9 inhibition for 12 months with Evolocumab improves PET MFR, and if so whether this effect is independent of changes in epicardial plaque and partially mediated by a reduction in inflammation. The findings of this study will provide novel and important insights into the comprehensive effects of Evolocumab on tissue perfusion, endothelial function, and microvascular function in a high-risk population. As such, these data would serve to address a critical gap in the identification of medical therapies that improve microvascular function and can thus target the residual risk of future cardiac events in patients with stable CAD.

Biography: Diana M. Lopez, MD, MSc, is a cardiologist at Brigham and Women’s Hospital (BWH) and an instructor in medicine at Harvard Medical School (HMS). As a clinical general cardiologist, she sees Spanish-speaking patients at BWH and Brigham and Women’s Faulkner Hospital. Additionally, she is the diversity, equity, and inclusion (DEI) officer for the BWH Cardiovascular Medicine Fellowship program. Her research focuses on integrating cardiovascular imaging/testing to better phenotype, risk-stratify, and manage the full spectrum of ischemic heart diseases, with a specific interest in nonobstructive coronary artery disease and coronary microvascular dysfunction. Lopez completed her undergraduate studies at Dartmouth College, medical school training at HMS, and postgraduate internal medicine residency and cardiovascular medicine fellowship at BWH. She subsequently completed a postdoctoral NIH-T32 research fellowship in the BWH Cardiovascular Imaging Research program and received her MS in epidemiology from the Harvard T.H. Chan School of Public Health.

Matthew Yuyun, MD, MPhil, PhD, Assistant Professor, Harvard Medical School, VA Boston Healthcare System

Mentor: Scott Kinlay, MBBS, PhD, Chief of Cardiology, VA Boston Healthcare System; Associate Professor of Medicine, Brigham and Women’s Hospital

Mentor:  Jacob Joseph, MBBS, MD, Chief of Cardiology, VA Providence Healthcare System; Professor, Brown University

Mentor: Jagmeet Singh, MD, PhD, Professor of Medicine, Harvard Medical School

Department Chair: Paul Conlin, MD, Chief, Medical Service, VA Boston Healthcare System; Professor of Medicine, Brigham and Women’s Hospital

Project Title: “Cardiac Arrhythmias in Heart Failure with Preserved Ejection Fraction: The Burden and Impact on Cardiovascular Morbidity and Mortality.”

Project Description: The worldwide burden of heart failure continues to grow with a global prevalence estimated at 64.3 million. Heart failure with preserved ejection fraction (HFpEF) represents approximately 50% of all heart failure patients, with a very poor median survival of 2 years and 5-year mortality of ~75%. The burden and impact of cardiac arrhythmias in HFpEF remains unknown but probably high. It is known that sudden cardiac death (SCD) accounts for 25-30% of total deaths in HFpEF patients, but the mechanism of SCD remains undetermined. Death and hospitalizations resulting from arrhythmias is a promising target for therapeutic interventions once the specific arrhythmic mechanisms are known. The overall objective of the proposed project is to determine the burden and impact of arrhythmias in HFpEF. The specific aims of the study project will be as follows: Aim 1: To determine the burden of arrhythmias in patients with HFpEF in comparison to patients without heart failure. I will compare prevalence and incidence of arrhythmias in veterans with HFpEF with a control group of veterans without heart failure. Aim 2: To determine the impact of cardiac arrhythmias on morbidity and mortality in HFpEF. I will determine the hazard ratios of these outcomes in HFpEF patients with arrhythmias compared to those without. Methods: This study will be conducted using a cohort study design in veterans ≥ 18 years of age with HFpEF (exposed group) and a matched control group without heart failure (unexposed group), derived from national VA databases in the time period 2006-2022.

Biography: Matthew F. Yuyun, MD, M.Phil, PhD, is an assistant professor of medicine at Harvard Medical School (HMS), and an attending cardiac electrophysiologist and general cardiologist at VA Boston Healthcare System, where he is also a member of the IRB committee. Additionally, he is an adjunct assistant professor of medicine at Boston University (BU) Chobanian & Avedisian School of Medicine. Previously, Yuyun’s research has focused on risk factors and biomarkers of cardiovascular diseases. Currently, his research focuses on cardiac arrhythmias in heart failure and cardiac implantable electronic devices, as well as epidemiology of cardiovascular diseases in the developing world. After obtaining an MD degree in Cameroon, he subsequently received the prestigious Commonwealth Scholarship Award to undertake additional training at the University of Cambridge, United Kingdom, where he received both MPhil (biostatistics and epidemiology) and PhD (cardiovascular epidemiology) degrees. He remained in the UK to complete his residency in internal medicine at Cambridge University Hospitals, and a cardiology fellowship training at University Hospitals of Leicester, after which he worked as a consultant general cardiologist at Milton Keynes University Hospital, England. He subsequently completed a clinical cardiac electrophysiology fellowship training at Lahey Hospital & Medical Center/Tufts University School of Medicine, Boston. In 2022, Yuyun received the Excellence in Clinical Teaching Award from medical trainees of HMS and BU.

2022-2024 PFDI Faculty Fellow

Nathaniel Harnett, PhD, Instructor, Harvard Medical School, McLean Hospital

Division Chief and Mentor: Kerry J. Ressler, MD, PhD, Chief, Division of Depression and Anxiety Disorders, Mclean Hospital; Professor of Psychiatry, Harvard Medical School

Project Title:  “The Impacts of Structural Racism on Threat Neurobiology”

Project Description: The present project investigates the neurodevelopmental impacts of structural racism. Structural racism is woven into the fabric of American life and has potentially deleterious consequences for minoritized individuals. However, limited research to date has assessed how disproportionated exposure to racialized stressors contributes to race-related differences in neurobiology relevant to psychiatric disease. Failure to properly consider the moderating role of structural racism and how it may contribute to observed race-related differences in the brain will result in inequitable neurobiological models of psychiatric disease that can contribute to engrained racism in psychiatry. We will use multidimensional assessments of life stressors and multimodal neuroimaging data collected from longitudinal and cohort studies including the Adolescent Brain and Cognitive Development Study and datasets in the Human Connectome Project. Normative age models across brain function and structure metrics will be generated across the datasets after harmonization. Global indices of exposure to structural racism will be derived from participant self-report demographics and neighborhood characteristics. This multidisciplinary project incorporates multiple areas of investigation from neuroscience and psychiatric to development and racism-related stress. The outcome of the project will provide novel insight into the neurobiological consequences of structural racism across the lifespan.

Biography: Nathaniel Harnett, PhD is an assistant neuroscientist at McLean Hospital and instructor in psychiatry at Harvard Medical School. He received his PhD in psychology with a focus on behavioral neuroscience at the University of Alabama at Birmingham under the mentorship of David C. Knight, PhD. Harnett received postdoctoral training in the Neurobiology of Fear Laboratory at McLean Hospital under the mentorship of Kerry J. Ressler, MD, PhD. His research is focused on understanding the neurobiological mechanisms that mediate susceptibility to trauma and stress related disorders. He uses multimodal neuroimaging, psychophysiology, and behavioral assessments to probe cognitive-affective function in individuals exposed to trauma to understand an individual’s potential to later develop posttraumatic stress disorder. In addition, Harnett investigates how structural inequities produce differing neural responses to trauma and how these factors may reinforce racial disparities in mental health. Ultimately, the goal of his research is to develop predictive and preventative neuroscience-based techniques to reduce the prevalence of trauma and stress-related disorder​s. Harnett has received several awards and honors including a Ford Foundation Predoctoral Fellowship and was a DSPAN F99/K00 award. He is a member of professional societies such as the International Society for Traumatic Stress Studies, the Society of Biological Psychiatry, and the Anxiety and Depression Association of America, and his work has been published in journals such as American Journal of Psychiatry, Neuropsychopharmacology, Biological Psychiatry, and NeuroImage.

2022-2023 PFDI Faculty Fellow

Michael Flores, PhD, MPH, Instructor, Harvard Medical School, Cambridge Health Alliance

Department Chair: Phillip Sung-En Wang, Dr.P.H, MD, Head of the Department of Psychiatry, Cambridge Health Alliance; Professor of Practice, Harvard Medical School

Mentor: Benjamin Le Cook, PhD, Associate Professor of Psychiatry, Cambridge Health Alliance

Project Title:  “Examining Racial/Ethnic Disparities in Opioid Treatment”

Project Description: Massachusetts (MA) is one of the top five states with the highest opioid-involved overdose rates. The shift to illicit opioid use has driven opioid-related mortality rates for Black and Latinx individuals to outpace that of White individuals. Despite the high opioid-involved mortality rates, few individuals with opioid use disorder (OUD) engage in evidence-based treatment, such as medication for OUD (MOUD, e.g., methadone, buprenorphine, and naltrexone); in fact, less than 10% receive any substance use treatment in MA. Our long-term goal is to understand how the opioid epidemic has impacted historically marginalized communities. Our overall objective is to examine MOUD access and opioid-related outcomes by race/ethnicity among MA residents using a comprehensive claims-based dataset. Our research objective aligns with the priorities of the MA Department of Public Health, who have partnered with us and granted access to the Public Health Data Warehouse, which we will analyze for this study. We aim to 1) estimate racial/ethnic trends (2011-2019) in OUD, MOUD initiation, MOUD retention, and MOUD availability; and 2) estimate racial/ethnic disparities in fatal/non-fatal opioid-related overdose and time to MOUD prescription. The study is informed by our long-standing collaborations with community partners, substance use specialist, and health services researchers. Our goal is to inform state policymakers, substance use treatment facilities, and clinicians on the extent to which OUD treatment availability and use is equitable across racial/ethnic groups using comprehensive claims-based MA data. Results can inform the allocation of limited resources and help improve opioid-related morbidity and mortality in the state of MA.

Biography: Michael Flores, PhD, MPH, is an instructor in the Department of Psychiatry at Harvard Medical School (HMS) and a research scientist in the Health Equity Research Lab at Cambridge Health Alliance (CHA). His research employs rigorous analytic methods to inform policy development, allocation of limited resources, and to improve the health outcomes and care quality of racial/ethnic minorities with behavioral health disorders. Flores’ current research arc focuses on racial/ethnic minority populations and examines the social determinants of health as they relate to the opioid epidemic, the consequences of opioid use disorder, and ways to improve resource availability to reduce opioid-related morbidity and mortality. He completed a postdoctoral research fellowship at HMS/CHA, where he was awarded the Norman E. Zinberg Fellowship in Addiction Psychiatry Research from HMS. In recognition of his promise as an early-stage investigator, Flores was awarded a New Investigator Award from the National Institute on Drug Abuse and a Diversity Scholar Award from the American Society of Health Economists. He received his PhD in health services research from Brown University.

2021-2023 PFDI Faculty Fellow

Dennis Spencer, MD, PhD, Instructor in Pediatrics, Harvard Medical School, Boston Children’s Hospital

Mentor: Seth Rakoff-Nahoum, MD, PhD, Assistant Professor of Pediatrics, Harvard Medical School; Associate Physician in Pediatrics, Boston Children’s Hospital

Division Chief: Scott B. Snapper, MD PhD, Professor of Medicine, Harvard Medical School; Chief, Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital

Project Title: Modification of Gut Microbiome Antibiotic Resistance through Dietary Glycans

Project Description: The gut microbiome plays an important role in health and disease. One of these important roles is excluding opportunistic enteric pathogens by “colonization resistance.” Bacteroides is the predominant Gram-negative bacteria of the human gut where it competes and survives with a unique capacity to metabolize a diverse range of dietary glycans. Depletion of the Bacteroidetes phylum during antibiotics use is associated with increased risk of enteric infections, including Clostridium difficile. There is a link between diet and the microbiota as well as an important role for metabolism in bacterial antibiotic susceptibility. My preliminary data establishes carbohydrate-specific effects of antibiotic susceptibility in Bacteroides ovatus during cultivation in vancomycin. Further, Transposon Sequencing (TnSeq) studies identified carbohydrate utilization genes associated with antibiotic fitness effects. My project, “Modification of Gut Microbiome Antibiotic Resistance through Dietary Glycans” hypothesizes that specific glycans tune antibiotic susceptibility in Bacteroides and that this may be understood to devise targeted antibiotic-specific prebiotic strategies to promote colonization resistance. To test this, we will: determine which dietary sugars impact antibiotic susceptibility across antibiotic class and Bacteroides strains (Aim 1); use next generation sequencing approaches to elucidate mechanisms by which sugars modulate antibiotic susceptibility (Aim 2); and assemble co-resident Bacteroides communities isolated from pediatric BMT patients’ stool to identify patient-specific optimal sugars that maximize Bacteroides resistance while minimizing opportunistic gut pathogens (Aim 3). This project involves collaboration with BCH Heme/Onc/SCT, utilizing stool from pediatric hematopoietic stem cell transplant patients. The work also benefits from use of the Harvard Biopolymers facility.

Biography: Dennis Spencer, MD, PhD is a pediatric gastroenterologist at Boston Children’s Hospital (BCH) and an instructor in pediatrics at Harvard Medical School (HMS). A physician-scientist, he is an investigator in the Rakoff-Nahoum laboratory (HMS/BCH) with a focus on the impact of diet on the composition and function of the gut microbiome. Spencer is a faculty advisor in the HMS Office of Recruitment and Multicultural Affairs as well as faculty chair of the BCH Graduate Medical Education Committee’s Joint Diversity & Recruitment subcommittee. He was recently awarded the 2021 Harold Amos Faculty Diversity Award from HMS. The National Medical Association has also recognized Spencer as a 2021 Top Physician Under 40 Award recipient. He completed the HMS Fellowship in Pediatric Gastroenterology, Hepatology, and Nutrition at BCH following his residency at Lucile Packard Children’s Hospital/Stanford University. Spencer is a graduate of the Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, obtaining a PhD in Microbial Pathogenesis and Immunology from The Rockefeller University. He is also a proud alumnus of Morehouse College.

Protected Time for Research: Two-year faculty fellowship supports junior faculty at critical juncture
Originally published in HMS News | June 28, 2021

2020-2022 PFDI Faculty Fellows

Hermioni Amonoo, MD, MPP, Assistant Professor of Psychiatry at Brigham and Women’s Hospital

Department Chair: David A. Silbersweig, MD, Stanley Cobb Professor of Psychiatry, Brigham and Women’s Hospital

Mentor: Jeffery C. Huffman, MD, Professor of Psychiatry, Massachusetts General Hospital

Project Title: Development of a positive psychology intervention to improve mood and health related quality of life in patients post hematopoietic stem cell transplantation- Proof of Concept Trial

Project Description: Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative treatment for some hematologic malignancies. Notwithstanding the promising nature, the transplantation process and recovery is intensive and fraught with potential life-threatening complications during recovery. Hence, HSCT recipients have a high burden of distress and quality of life (QOL) deficits. Most efforts to achieve optimal psychological weli-being in this population have targeted the reduction of distress (e.g., depression). However, positive psychological well-being ( e.g., optimism), can buffer against this distress and has been prospectively associated with improved QOL and survival in this population. Positive psychological interventions (PPis), which utilize systematic activities (e.g., recalling positive life events) to promote psychological well-being, have consistently and durably enhanced psychological health and QOL in medical settings, but have never been used in HSCT patients. Given the need for new programs to promote well-being and recovery after HSCT, the proposed project will develop and test a novel PPI in this population to fill this unmet need. I will develop the PATH (Positive psychology for Allogeneic Transplantation of Hematopoietic stem cells) intervention via a review of the literature and application of theoretical frameworks, then test its acceptability (via quantitative participant ratings and qualitative feedback at exit interviews) in a one-arm proof-of-concept trial (N= l0; Aim 1). Next, I will test its feasibility and preliminary efficacy on health outcomes in a pilot randomized controlled trial (N=60; Aim 2). In sum, the HMS-PFDD Award will prepare me to become an independent investigator and leader who develops novel evidence-based supportive oncology interventions.

Biography: Hermioni L. Amonoo, MD, MPP, is an assistant professor at Harvard Medical School (HMS) and a staff physician in the Department of Psychiatry at Brigham and Women’s Hospital (BWH) and the Department of Psychosocial Oncology and Palliative Care at the Dana-Farber Cancer Institute (DFCI). She is also the associate training director of the BWH/HMS Adult Psychiatry Residency Training Program.

Amonoo was born and raised in Accra, Ghana and miraculously moved to Indiana to complete her BSc in biology and chemistry at Purdue University. She then completed her MD and MPP at Harvard Medical School and Harvard Kennedy School of Government. She completed her clinical training at the Massachusetts General Hospital/McLean Hospital Adult Psychiatry Residency Training Program and the BWH/DFCI Consultation Liaison Psychiatry and Psychosocial Oncology Fellowship. Her clinical work in medical psychiatry and psychosocial oncology inspired her research program aimed at understanding distress in vulnerable cancer populations to inform the development of novel supportive oncology interventions that impact health outcomes.

Randy Miles, MD, PhD, Assistant Professor of Radiology at Massachusetts General Hospital

Department Chair and Mentor: Constance D. Lehman, MD, PhD, Professor of Radiology, Massachusetts General Hospital

Project Title: External Validation and Clinical Assessment of a Deep Learning Risk Prediction Model for Mammography Interpretation

Project Description: While recent studies evaluating artificial intelligence (AI) demonstrate promising results, it is imperative that AI tools are thoroughly assessed prior to widespread use to avoid past mistakes with implementation of conventional computer assisted detection (CAD), which led to increased healthcare costs despite only marginal benefit. Current AI studies largely share the following that may limit maximal clinical utility: 1) requirements for large, well-curated datasets to train and validate algorithms, which may not be representative of real-time clinical imaging data 2) validation using internal data sets only, limiting generalizability to diverse patient populations, equipment manufacturers, and/or clinical settings, and 3) lack of evidence on how AI tools can be implemented to maximize clinical impact. We have previously published on our AI model using deep learning techniques, developed from institutional data using 223,109 consecutive screening mammograms performed in 66,661 women from January 1, 2009 to December 31, 2016, that predicts breast cancer risk, based on a woman’s current mammogram. Our model obtained an AUC of 0.82(95%CI:0.80,0.85). Personalized risk prediction scores determined by our model can potentially be used by radiologists to improve radiologist’s interpretative performance, if provided at the time of mammography interpretation. To inform clinical implementation of our model, we aim to 1) validate our AI model utilizing imaging data from a large, diverse external dataset and 2) quantitatively determine the effect of our model on radiologist interpretative performance. Together, this work has the potential to positively impact breast cancer screening by providing a framework for clinical implementation of our model to improve radiologist performance.

Biography: Randy C. Miles MD, MPH, is an assistant professor in the Department of Radiology at Massachusetts General Hospital. He is originally from Green Level, NC., completed his BS in chemistry from Hampton University graduating Summa Cum Laude with honors, and obtained his MD from Mayo Clinic College of Medicine, where he co-led health missions to underserved regions in Haiti and the Dominican Republic. Miles’s international public service led him to obtain a MPH from Harvard T.H. Chan Harvard School of Public Health, where he was awarded the Zuckerman fellowship. During this time, he learned about racial disparities in breast cancer mortality and discovered his passion for improving breast cancer care in traditionally underserved groups. His clinical practice as a board- certified radiologist, specializing in breast imaging, includes image interpretation of digital mammography, digital breast tomosynthesis (“3D mammography”), breast ultrasound (including automated breast ultrasound (ABUS)), and breast MRI. He also performs image- guided procedures including breast biopsies, aspirations, wire/seed localization, and lymphoscintigraphy. His clinical, research, and public health efforts center around improving breast cancer outcomes primarily through 1) identifying barriers to breast cancer screening, 2) creating patient-centered initiatives to improve access to breast imaging services, and 3) examining how to improve delivery of high quality, guideline- concordant breast care for all patients using artificial intelligence. Within the breast imaging division, he has a leadership role in community health, where he has led research efforts focused on reduction of breast cancer screening barriers, appropriate utilization of supplemental screening tools, and assessment of online patient educational materials for factors related to health literacy. In addition, Miles currently works with international hospitals to improve all facets of care in breast imaging related to physician training, workflow optimization, and patient experience.

Read about 2008-2018 Diversity Inclusion Faculty Fellows Alumni.