The 2024 application cycle is now open. Deadline: Wednesday, February 7, 2024. 

These awards increase opportunities for underrepresented faculty who are committed to diversity, inclusion and equity, to advance to senior positions in academic medicine and leadership.

The two awards categories are the Physician/Scientist Development Award (PSDA) and the Clinician/Teacher Development Award (CTDA). Each award provides funding over three or four years and is designed for Mass General-appointed faculty pursuing different career goals. 

The Clinician/Teacher Development Award (CTDA) provides funding support for a clinical, educational or community project to a faculty member (junior and mid-career), fellow, or graduating resident pursuing an academic career as a clinician, teacher, administrative or community leader at Mass General. This award offers a total of $120,000 in grant and loan repayment support plus 20% indirect costs to be spent over a four-year period. Up to two awards are given. Click here to download the 2024 CTDA application

The Physician/Scientist Development Award (PSDA) provides transitional grant funding which will aid the applicant in becoming an independent investigator at MGH. $180,000 plus 20% indirect costs will be awarded to be spent over a three or four-year period, with a maximum of  $60,000 plus indirect costs per year for three-year awards and a maximum of $45,000 plus indirect costs per year for four-year awards. Up to six awards are given. Click here to submit a PSDA application.

The CDI Faculty Development Award Consultation Service  is designed to help applicants develop a strong application for the Physician/Scientist and the Clinician-Teacher Development Awards. Through this service, you can speak with a previous award winner for advice on your application, including your research plan, personal statement, and letters. To request a consultation, please complete this survey.

For answers to frequently asked questions, please view PSDA FAQs and CTDA FAQs (PDF).

2023 Clinician-Teacher Development Award Recipients


Linda Herrera Santos, MD, PhD 
Department of Psychiatry  
Director, MGH Hispanic Psychiatry Clinic  
Avery D. Weisman Psychiatry Consultation Service  
Instructor in Psychiatry, Harvard Medical School  

Dr. Linda Herrera Santos serves as the Director of the MGH Hispanic Psychiatry Clinic. She is an Instructor in Psychiatry at Harvard Medical School and an attending psychiatrist in the Psychiatry Consultation Service at MGH. She teaches and supervises MGH/McLean Psychiatry residents and is the co-director of the Sociocultural Didactics Series. Dr. Herrera Santos obtained her medical degree from Texas A&M Health Science Center. She completed the Adult Psychiatry Residency Training at MGH/McLean, followed by the MGH Consultation-Liaison Psychiatry Fellowship. As a trainee, Dr. Herrera Santos was awarded the APA SAMHSA Minority Fellowship, which funded a project addressing mental health in underserved populations. She founded the Hispanic Psychiatry Clinic and secured funding for its pilot through an MGH Collaboration Catalyst Grant and a Research Grant through the MGB Centers of Expertise. Dr. Herrera Santos’ clinical and research interests are focused on delivering mental health services to Hispanic populations. She is committed to expanding initiatives promoting an inclusive healthcare community and delivering excellent, compassionate, and culturally humble care. 

Abstract: Addressing Mental Health Care Disparities Through Resident Training: Evaluation and Expansion of a Hispanic Psychiatry Resident Clinic 

Attaining optimal quality mental health care for Hispanic patients, particularly monolingual Spanish-speaking patients, has been challenging due to the scarcity of bilingual providers with adequate training in culturally-sensitive care. This has led to critical gaps in care and inequitable health outcomes for Hispanic patients. To address this gap, a pilot for a Hispanic Psychiatry Resident Clinic was developed at Mass General to provide Spanish-speaking residents with advanced clinical training in psychiatric care for Hispanic patients. Residents in this pilot provide direct, longitudinal, culturally-sensitive psychopharmacology or psychotherapy services to monolingual Spanish-speaking patients. In addition, residents have access to supervision with bicultural/bilingual faculty to discuss cases. They also attend monthly didactics focused on topics relevant to mental health care delivery in Hispanic communities. This study aims to evaluate and enhance the training mission of the Hispanic Psychiatry Resident Clinic to address the need to develop a workforce of psychiatrists who provide culturally informed care to Spanish-speaking patients. Interviews and impact data collected from residents will guide this work. By the end of this project, we will have a formalized curriculum and clinic structure that could then be easily disseminated for implementation at other institutions.  


Aisha James, MD, MEd 
Internal Medicine and Pediatrics 
Instructor in Medicine, Harvard Medical School 

Originally from Milton, MA, Dr. James earned her undergraduate degree in Psychology and French from Amherst College, her Master’s of Education from Brooklyn College, and her MD from Mount Sinai School of Medicine. She worked as a special education teacher in NYC prior to medical school. She completed a combined residency in Internal Medicine & Pediatrics at Massachusetts General Hospital, and served as a Chief Resident. Dr James now works as a PCP at MGH Everett Family Care and the MGH Sickle Cell Disease Center. She also works to promote equity at MGH as the director of Racial Justice for the Department of Medicine, and Associate Director for Diversity and Equity for the Department of Pediatrics. Outside of MGH, she engages in legislative advocacy as an active member of the Massachusetts’ American College of Physicians Health and Public Policy Committee and the Massachusetts’ American Academy of Pediatrics Legislative Committee. Dr. James’ current CTDA project is focused on infusing graduated medical education with an anti-racist and equity lens through a coaching program.   

Abstract: Utilizing the Department of Medicine’s Anti-Racism and Equity Educational Initiative to Improve Equity in Graduate Medical Education

Provider-held bias and systemic inequities contribute to racial and ethnic health disparities. Graduate medical education (GME) curricula have been expanded to include stand-alone health equity series. However, GME curricula may still, inadvertently, reinforce stereotypes and further health disparities. To support the integration of anti-racism and equity across GME curricular content we developed and implemented the Department of Medicine’s Anti-Racism and Equity Educational Initiative (DARE). DARE trained volunteer coaches to support educators using a checklist of best practices to bring an anti-racism and equity lens to their lecture content. Evaluation of the DARE pilot program demonstrated improvement in the anti-racism and equity content of existing internal medicine residency educational conferences. The current project aims to further expand and evaluate DARE across different departments and specialties.  


See list of prior CTDA recipients >

2023 Physician/Scientist Development Award Recipients


Christiano R. Alves, P.h.D. 
Department of Neurology 
Center for Genomic Medicine 
Instructor in Neurology, Harvard Medical School 

Dr. Alves is a junior faculty (Instructor) in the Department of Neurology and in the Center for Genomic Medicine at Massachusetts General Hospital (MGH) and Harvard Medical School. He is interested in the convergence between genome editing and applied physiology to make an impact on human health. His current research efforts focus on the development of genome editing technologies to treat neuromuscular diseases. In parallel, he is dedicating research to identifying novel biomarkers to track muscular and neuromuscular diseases. Dr. Alves holds a Ph.D. in muscle physiology from the University of Sao Paulo located in his hometown Sao Paulo, Brazil. Before joining the Kleinstiver lab at MGH, he was a postdoctoral research fellow at Joslin Diabetes Center, Harvard Medical School (2017-2018), and at the Center for Genomic Medicine, MGH (2018-2021). 

Abstract: Development of in vivo base editing as a genetic treatment for spinal muscular atrophy

Spinal muscular atrophy (SMA) is a devastating neuromuscular disease and a leading genetic cause of infantile death worldwide. Despite exciting progress in the neuromuscular field that has resulted in novel therapies, there remains no permanent cure for SMA. Therefore, developing a permanent treatment that treats the underlying genetic perturbation and all systemic manifestations of this disease would transform patients’ life. SMA is caused by mutations in the Survival Motor Neuron 1 (SMN1) gene. An important modifier of SMA severity is the number of copies of a paralogous gene SMN2. The sequence of SMN2 mainly differs from SMN1 by a C•G-to-T•A transition in exon 7 (“C6T”), which causes the skipping of exon 7 in most SMN2 transcripts due to alternative splicing. While previous studies have demonstrated that antisense oligonucleotides (ASOs) or small molecules targeted to SMN2 gene can transiently increase full-length SMN protein expression, this approach requires repeated dosing and has incomplete penetrance that poses challenges for patients. Therefore, we are developing optimized base editors (BEs) that enable precise genetic alterations within defined regions of the SMN2 gene. In this proposal, we plan to extend this work by optimizing the most optimal genome editing approaches and investigating the use of adeno-associated viruses (AAVs) to deliver these novel editing technologies in SMA mouse models. In principle, this will create a lasting single-dose permanent treatment for SMA with several advantages compared to targeting mutations in the SMN1 gene or exogenous gene replacement. The success of this proposal will provide solutions to questions at the intersection of the fields of genome editing and neurogenetics, with broad implications for the entire neuromuscular scientific community. 


Pierre Ankomah, MD PhD 
Department of Medicine, Division of Infectious Diseases 
Instructor, Harvard Medical School 

Dr. Ankomah’s research investigates microbial and host immune response dynamics in infectious syndromes. His current primary focus is using transcriptional analysis of immune cells at single-cell resolution to study the immunological heterogeneity underlying sepsis. Dr. Ankomah received his MD and PhD degrees from Emory University. He completed Internal Medicine residency at MGH and the combined Infectious Diseases fellowship training program at MGH/BWH. His work is also supported by a Harvard KL2/Catalyst Medical Research Investigator Training (CMeRIT) award. Dr. Ankomah is a recipient of the American Society for Clinical Investigation’s Emerging Generation Award in recognition of his potential as an academic physician-scientist.  

Abstract: Single-cell transcriptional analysis of immune cells to investigate patient heterogeneity in sepsis

Sepsis is a common and deadly disease that causes about 11 million deaths worldwide annually. It is characterized by organ failure from dysfunctional immune responses to infection; yet the specifics of immune dysfunction are poorly understood and vary widely among different patients. Current management strategies for sepsis ignore this heterogeneity, instead relying on a “one-size-fits-all” approach to diagnostics and treatment that has been a major reason for limited progress in improving sepsis outcomes for decades.  

Transcriptional analysis of cells at single-cell resolution (single-cell sequencing) is a transformative new tool to probe disease pathogenesis. Recent studies in our laboratory using this methodology have made novel findings, including identification of a new immune cell subtype enriched in sepsis. In this project, we will use single-cell sequencing to investigate the immune cell heterogeneity within a cohort of sepsis patients. These studies will evaluate immune cell abundance and transcriptional changes over time, allowing us to discover the response of each individual immune cell type to infection. Ultimately, we anticipate that this project will expand our understanding of disease mechanisms in sepsis, allow precise distinction between sepsis and its mimics, and characterize patient subtypes to identify those that may respond favorably to certain therapies. 


Adjoa Anyane-Yeboa, MD, MPH 
Division of Gastroenterology, Department of Medicine 
Instructor of Investigation, Mongan Institute 
Assistant Professor of Medicine, Harvard Medical School 

Dr. Anyane-Yeboa is a gastroenterologist at Massachusetts General Hospital and an Assistant Professor of Medicine at Harvard Medical School. She received her medical degree from the University of Cincinnati, completed her internal medicine residency and a 4th year chief residency at the University of Illinois at Chicago, and completed her gastroenterology fellowship at the University of Chicago Medicine. After completing her gastroenterology training, she went on to pursue additional training to further her drive to achieve equity in health outcomes for patients from vulnerable communities. She completed the Commonwealth Fund Fellowship in Minority Health Policy through Harvard Medical School where she also received her Master’s in Public Health with a focus in Health Policy from the Harvard T.H. Chan School of Public Health.  

Dr. Anyane-Yeboa’s research focuses on health equity in gastroenterology, with a specific focus on understanding IBD in minority populations and increasing colorectal cancer (CRC) screening in community health centers. Her long-term goal is to reduce inequities in CRC screening in marginalized groups using equity-driven implementation science methods. She has previously received funding from a National Cancer Institute diversity supplement, American Cancer Society, Trefler foundation and Pfizer Medical Grants Program to support her work. She is actively involved in her national gastroenterology societies and serves on the Diversity Committees for the American College of Gastroenterology and the American Gastroenterological Association. She is a prior recipient of the Rising Star Award from the University of Cincinnati Alumni Association and the University of Chicago Division of Gastroenterology Walter L Palmer Junior Alumni award.

Abstract:  

Colorectal cancer (CRC) is the third leading cause of cancer death in Black men and women, with mortality rates that are 44% and 31% higher in Black men and women compared to White men and women. Approximately 40% of the difference in incidence and 20% of the difference in mortality are due to disparities in screening. Community health centers (CHCs) in urban areas serve a large share of racial and ethnic minority patients and have lower screening rates (41.9%) compared to the general population (69.7%). Low screening rates, and rising incidence and mortality from CRC in adults at younger ages also suggests that focusing on increasing screening uptake from 45 to 54 is a critical target. The objective of this study is to use equity-driven implementation strategies to initiate CRC screening in unscreened individuals aged 45 to 54 at Codman Square Health Center (CSHC) using principles of patient activation. This award leverages an existing partnership with CSHC, a predominantly Black CHC in inner-city Boston. This proposal has three scientific aims: (1) identify system-level and patient-level determinants of CRC screening in individuals 45 to 54 using mixed methods; (2) identify equity-driven implementation strategies to initiate CRC screening matched to determinants; and (3) conduct a randomized pilot evaluation to determine impact of implementation strategies on CRC screening. 

Study Design: The proposed project utilizes mixed methods and equity-driven implementation science methods to increase CRC screening in a predominantly Black CHC. To achieve this goal we will (1) identify patient and system-level determinants of CRC screening through an organizational survey and focus groups with patients and staff guided by an equity-driven CFIR framework; (2) develop implementation strategies matched to determinants using implementation roadmaps; and (3) perform a rapid cycle pilot evaluation to test implementation strategies at the patient- and system-level using an equity-driven RE-AIM framework. The combination of formal training and mentored research outlined in this application is designed to ensure that Dr. Anyane-Yeboa will emerge as a nationally recognized independent investigator with the skills and experience to reduce CRC inequities in marginalized patient populations. 


Maria Agustina Battistone, P.h.D.
Department of Medicine, Nephrology Division, MGH Program in Membrane Biology
Assistant Professor of Medicine, Harvard Medical School

Dr. Battistone have been an Assistant Professor of Medicine at Harvard Medical School (HMS) and Massachusetts General Hospital (MGH) since April 2022. Her laboratory explores the molecular basis of mucosal immunity in the urogenital tract and applies the knowledge acquired through basic research to identify new diagnostic/therapeutic targets for kidney injury and male infertility.

She obtained her bachelor’s degree in Biochemistry in 2008 at the University of Buenos Aires, Argentina. She conducted her Ph.D. studies at the same university, focused on sperm physiology. From June 2015 to July 2019, she performed her Postdoctoral training in Dr. Breton’s laboratory at MGH, and was awarded the Lalor Fellowship to study molecular mechanisms involved in luminal acidification in the urogenital tract. While examining the role of proton-secreting cells in epithelial dynamics, they uncovered a completely novel role for these cells in immune regulation she  started her research group in April 2022, when her first NIH R01 was awarded. Her scientific contributions were recognized by different awards, including the DOM MGH Sanchez and Ferguson Research Faculty Award, and the MGH Claflin Distinguished Scholar. In addition, she have been involved in the training of postdoctoral fellows, international Ph.D. students, and pre-medical school students.

Abstract: Immune tolerance disruption induces kidney injury

Acute kidney injury (AKI) is an important clinical disorder affecting many patients worldwide. Given its high prevalence and mortality, AKI is currently diagnosed only after renal injury has occurred, and there is no specific cure. Therefore, there is a clear need for the development of early markers of AKI and an associated therapy to reverse/prevent this complication. The current proposal addresses this clinical gap by leveraging novel mechanisms by which epithelial proton-secreting intercalated cells (ICs) communicate damage to immunocytes in the kidney. Uncontrolled inflammation is a leading cause of AKI. Our hypothesis is that the ICs are strategically positioned to interact with immunocytes, the so-called mononuclear phagocytes (MPs), to survey the renal epithelial barrier and regulate the balance between inflammation and tolerance. In the first aim, we will elucidate the major roles of ICs together with MPs after the ablation of immunotolerance. In the second aim, we will evaluate IC - MP crosstalk to mount an anti-inflammatory response during AKI, performing renal ischemia followed by reperfusion injury. The research proposed will use an innovative multidisciplinary approach to describe how specialized epithelial cells and immunocytes communicate in the kidney. Ultimately, such knowledge will fill gaps to understand the pathogenesis of AKI.


Margarete Diaz Cuadros, PhD 
Department of Molecular Biology 
Instructor in Investigation 
Lecturer in Genetics, Harvard Medical School 

Dr. Diaz Cuadros is an Instructor in Investigation in the Department of Molecular Biology at Massachusetts General Hospital. She earned her Bachelor’s degree from Columbia University (2014), where she worked on neural development in the nematode Caenorhabditis elegans with Dr. Martin Chalfie. She later completed her PhD under the supervision of Dr. Olivier Pourquie at Harvard Medical School (2022), where she established in vitro models of the mammalian segmentation clock based on pluripotent stem cells. Through the Junior Fellows Program hosted by the Department of Molecular Biology, Margarete was able to launch her independent research career directly after graduate school. Her research focuses on understanding the mechanisms that allow some mammalian species to complete embryonic development faster than others. For instance, a mouse embryo is formed in 15 days, but a human embryo requires 56 days to complete the same process. In the long term, the goal of this research program is to provide a basis for the acceleration of human pluripotent stem cell differentiation for disease modeling and cell-based therapies. 

Abstract: Role of NADH Shuttles in Regulating Species-Specific Developmental Rates

All mammalian embryos undergo a highly conserved sequence of developmental events, but different species complete this sequence at significantly different speeds. For instance, a mouse embryo is formed in 15 days, but a human embryo requires 56 days to complete the same process. How species-specific developmental rates are set represents a major unanswered question in developmental biology. We recently established an in vitro system that recapitulates the twofold difference in developmental rate between mouse and human embryos. This system provides a quantitative measure of developmental speed as revealed by the period of the segmentation clock, a molecular oscillator associated with the rhythmic production of vertebral precursors. Using this system, we uncovered that a higher cytosolic NAD+/NADH ratio in mouse cells compared to human cells gives rise to faster protein production, which ultimately allows mouse cells to proliferate and differentiate faster [5]. In the PSDA-funded project, we seek to elucidate the basis of cytosolic NAD+/NADH differences between mouse and human cells by investigating the activity of biochemical shuttles that transport reducing equivalents from the cytosol to mitochondria. Specifically, we will test the hypothesis that mouse cells feature increased activity of the glycerol phosphate shuttle compared to human cells, thereby underlying differences in cytosolic NAD+/NADH and developmental rate. We will then leverage this new understanding to develop approaches for the modulation of developmental rate in vitro by manipulating glycerol phosphate shuttle activity. Acceleration of developmental rate would allow faster production of mature cell types from human induced pluripotent stem cells (iPSCs) for cell-based therapies and disease modeling; whereas its deceleration would provide new avenues to halt tumor growth and to slow down the aging process. 


Juan D. Matute, MD
Department of Pediatrics, Division of Newborn Medicine
Director of Equity, Newborn Medicine
Instructor of Pediatrics, Harvard Medical School

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 and Boston Medical Center. He further specialized in Perinatal and Neonatal Medicine through the Harvard Combined Program. Following his clinical training, Dr. Matute joined MGH and Harvard Medical School HMS as an instructor. He 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. Dr. Matute has conducted research on the host-microbiota interface starting in early life. He has focused his research efforts on how the intestinal microbiota and the host interact in 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.

Abstract: The role of Intelectin-1 in obesity

Genetic and environmental factors interact in the development of obesity from early life. However, research is still needed to understand how specific genes and environmental factors promote obesity development. The collection of microorganisms that live in our intestine is called the microbiota and is acquired after birth. Multiple lines of evidence have shown that the microbiota influences obesity development. In this project, we will define mechanistically how a genetic risk factor for obesity named Intelectin-1 regulates host metabolism potentially through the microbiota. Intelectin-1 is secreted to our gastrointestinal tract and interacts only with sugars found in the wall of microbes. We will study how Intelectin-1 in the gastrointestinal tract influences obesity using new genetically modified mice. This research project will enhance our understanding of Intelectin-1 on metabolic syndrome and will trailblaze the path to study the implications of Intelectin-1 interactions with the microbiota in humans with obesity. The proposed studies are relevant as they are expected to identify new insights into factors at the host and intestinal microbiome interface associated with metabolic syndrome, elucidate potential mechanisms, and, thus, novel therapeutic strategies for preventing obesity starting during the first years of life.


See list of prior PSDA recipients >