Lindsay Farrer, PhD Hear my name
Boston University Distinguished Professor of Genetics
Boston University Chobanian & Avedisian School of Medicine
Medicine
Biomedical Genetics

PhD, Indiana University School of Medicine
BA, University of North Carolina at Chapel Hill



Dr. Lindsay Farrer is a medical geneticist at Boston University Schools of Medicine and Public Health where he is the Chobanian and Avedisian School of Medicine Distinguished Professor of Genetics, Chief of Biomedical Genetics, and a Professor of Medicine, Neurology, Ophthalmology, Epidemiology, and Biostatistics. Dr. Farrer is a graduate of the University of North Carolina in Chapel Hill, received his Ph.D. from the Indiana University School of Medicine, and gained additional training in genetic epidemiology at Yale University. He holds adjunct faculty positions at Harvard Medical School, Massachusetts General Hospital, and the Veterans Administration Medical Center in Bedford, Massachusetts. He is a Founding Fellow of the American College of Medical Genetics. Dr. Farrer teaches several courses in human genetics and addiction science at Boston University, directs the BU Transformative Training Program in Addiction Science (TTPAS) that features transdisciplinary training for students enrolled in PhD programs across the Medical and Charles River campuses, directs Boston University’s Molecular Genetics Core Facility which offers DNA genotyping and sequencing services to investigators at Boston University and elsewhere, and provides genetic counseling and testing to patients with a variety of inherited conditions.

Dr. Farrer’s research has lead to more than 450 publications on genetic risk factors for several familial neurodegenerative and other chronic diseases. In collaboration with other laboratories worldwide, his group has localized genes causing a variety of rare and common disorders, most notably Alzheimer disease (AD), substance use disorders (SUDs), age-related macular degeneration (AMD), Wilson disease, Machado-Joseph disease, Waardenburg syndrome, hypertension, sensorineural deafness, and osteoarthritis. His group identified a functional genetic variant in the complement factor H gene which accounts for more than 30% of the attributable risk for AMD, the leading cause of progressive vision loss and blindness in the elderly. In collaboration with other researchers, Dr. Farrer is conducting genome wide association studies (GWAS) and whole genome/exome sequencing studies for several disorders including AD, SUDs (cocaine, opiates, nicotine, alcohol and cannabis), and AMD. Dr. Farrer’s team is also developing methods for locating genes that influence the natural history of complex diseases and pharmacogenetic response.

Under Dr. Farrer’s leadership, the MIRAGE Project, a multi-center study of AD funded since 1991 by the National Institute on Aging, has made several important contributions to our understanding of the interactions between genetic and environmental factors for the disorder. This study has a particular emphasis on the genetics of AD in African Americans. MIRAGE was the first study to demonstrate that genetic factors have a major role in the development of AD and that APOE e4 is more weakly associated with disease in men and persons older than 75 years. Dr. Farrer co-directed the international effort which demonstrated that SORL1 is genetically and functionally associated with AD, thus implicating intracellular protein trafficking as integral pathway in AD. His laboratory conducted genome wide association studies (GWAS) for AD in several populations including African Americans and an inbred Israeli-Arab community, and identified rare AD causal mutations in the AKAP9 gene which are specific to African Americans. Dr. Farrer serves on the Executive Committee of the national Alzheimer Disease Genetics Consortium and co-directs the data analysis effort for this large NIH-funded project. He is also a Principal Investigator of the national Alzheimer Disease Sequencing Project and a study to identify AD risk and protective variants in Koreans. in 2020, Dr. Farrer co-founded the Framingham Heart Study Brain Aging Program (FHS-BAP), an NIH-funded infrastructure program that continues surveillance of FHS participants for cognitive decline and dementia, conducts neuropsychological and brain MRI exams, houses the FHS brain tissue repository, and conducts several projects utilizing genetics, various omics, and wealth of phenotype data on FHS participants to develop predictive models, identify biomarkers and discern vascular and inflammatory processes leading to AD.

Diversity, Equity, Inclusion and Accessibility

My doctoral training in medical genetics introduced me to disorders whose risk and impact on families vary widely according to cultural, social and economic circumstances. This experience also convinced me that the impact of physical and mental disability is greatly influenced by access to treatment and support programs, as well as the family and community environment.

I have designed and led studies, and published many papers about the genetic basis of Alzheimer disease (AD), age-related macular degeneration, hypertension and sickle cell disease in marginalized populations. Many of these papers highlight the population genetic differences related to disease and the scientific and translational medicine benefits of trans-ethnic studies. My lab discovered two rare African-American specific variants in a novel gene that are associated with greatly elevated risk of AD in African Americans. Currently, I am the Principal Investigator (PI) of grants focused on AD genetics in African Americans and Koreans, respectively.

As PI of the Framingham Heart Study Brain Aging Program (FHS-BAP), my research team launched several initiatives to attract and train a diverse group of individuals, particularly those who are not well-represented in biomedical research, at the undergraduate, graduate, post-doctoral and junior faculty levels. International symposia organized by the FHS-BAP emphasize DEIA issues including enrolling marginalized populations in research, the incorporation of social determinants of health in genetic and epidemiological studies, and female and non-White speakers.

As a mentor and Chief of a research section in which White male faculty are the minority, I have sought and promoted the advancement of female and non-White trainees, many of whom have advanced to successful careers in academia and private industry. Since 2007, I have served on Steering Committee of PhenX, a project funded by the National Human Genome Research Institute that is building a set of consensus standards for measures of phenotypes and exposures with particular emphasis on protocols that consider diversity and accessibility.

Section Chief
Boston University Chobanian & Avedisian School of Medicine
Medicine
Biomedical Genetics

Professor
Boston University Chobanian & Avedisian School of Medicine
Medicine
Biomedical Genetics

Professor
Boston University Chobanian & Avedisian School of Medicine
Ophthalmology


Professor
Boston University Chobanian & Avedisian School of Medicine
Neurology


Professor
Boston University School of Public Health
Biostatistics


Professor
Boston University School of Public Health
Epidemiology


Investigator
Framingham Heart Study


Member
Boston University
Evans Center for Interdisciplinary Biomedical Research


Member
Boston University
Genome Science Institute


Member
Boston University
Bioinformatics Graduate Program


Graduate Faculty (Primary Mentor of Grad Students)
Boston University Chobanian & Avedisian School of Medicine, Graduate Medical Sciences




Genetic Studies of Alzheimer's Disease in Jewish and Arab Populations
09/01/2023 - 08/31/2028 (Multi-PI)
PI: Lindsay Farrer, PhD
NIH/National Institute on Aging
1U01AG081230-01

Boston University Alzheimer's Disease Research Center
08/15/2021 - 06/30/2026 (Key Person)
PI: Ann C. McKee, MD
NIH/National Institute on Aging
5P30AG072978-03

Genetic Studies of Alzheimer Disease in Koreans
09/15/2019 - 08/31/2025 (PI)
NIH/National Institute on Aging
5U01AG062602-05

Precision Monitoring and Assessment in the Framingham Study: Cognitive, MRI, Genetic and Biomarker Precursors of AD & Dementia
09/15/2020 - 05/31/2025 (Multi-PI)
PI: Lindsay Farrer, PhD
NIH/National Institute on Aging
5U19AG068753-05

Alzheimer Disease Genetic Architecture in African Americans
05/15/2020 - 04/30/2025 (PI)
NIH/National Institute on Aging
5R01AG048927-10

Metabolic age to define influences of the lipidome on brain aging in Alzheimer's disease
05/15/2023 - 03/31/2025 (Subcontract PI)
Duke University NIH NIA
5R01AG081322-02

Genome Center for Alzheimer's Disease
04/15/2021 - 03/31/2025 (Subcontract PI)
The Trustees of the University of Pennsylvania NIH NIA
5U54AG052427-08

Genome Center for Alzheimer's Disease (GCAD) - Core B
04/15/2021 - 03/31/2025 (Subcontract PI)
The Trustees of the University of Pennsylvania NIH NIA
5U54AG052427-08

Alzheimer Disease Genetics Consortium
04/15/2020 - 03/31/2025 (Subcontract PI)
The Trustees of the University of Pennsylvania NIH NIA
5U01AG032984-14

Metabolomic Signatures for Disease Sub-classification and Target Prioritization in AMP-AD
01/21/2019 - 08/31/2024 (Subcontract PI)
Duke University NIH NIA
5U01AG061359-05

Showing 10 of 43 results. Show All Results


Title


Yr Title Project-Sub Proj Pubs
2024 Core G: Genetics and Molecular Profiling 5P30AG072978-04-9747
2024 Admin Core 5U19AG068753-05-8052
2024 Precision Monitoring and Assessment in the Framingham Study: Cognitive, MRI, Genetic and Biomarker Precursors of AD & Dementia 5U19AG068753-05
2024 Genomic, physiological, and environmental predictors of AD risk, resilience and resistance 5U19AG068753-05-8059
2024 Alzheimer Disease Genetic Architecture in African Americans 5R01AG048927-10
2023 Genetic Studies of Alzheimer's Disease in Jewish and Arab Populations 1U01AG081230-01
2023 Core G: Genetics and Molecular Profiling 5P30AG072978-03-9747
2023 Genomic, physiological, and environmental predictors of AD risk, resilience and resistance 5U19AG068753-04-8059
2023 Precision Monitoring and Assessment in the Framingham Study: Cognitive, MRI, Genetic and Biomarker Precursors of AD & Dementia 5U19AG068753-04
2023 Admin Core 5U19AG068753-04-8052
Showing 10 of 85 results. Show All Results

Publications listed below are automatically derived from MEDLINE/PubMed and other sources, which might result in incorrect or missing publications. Faculty can login to make corrections and additions.

iCite Analysis       Copy PMIDs To Clipboard

  1. Kang M, Li C, Mahajan A, Spat-Lemus J, Durape S, Chen J, Gurnani AS, Devine S, Auerbach SH, Ang TFA, Sherva R, Qiu WQ, Lunetta KL, Au R, Farrer LA, Mez J. Subjective Cognitive Decline Plus and Longitudinal Assessment and Risk for Cognitive Impairment. JAMA Psychiatry. 2024 Jul 03.View Related Profiles. PMID: 38959008; PMCID: PMC11223054; DOI: 10.1001/jamapsychiatry.2024.1678;
     
  2. Ray NR, Kunkle BW, Hamilton-Nelson K, Kurup JT, Rajabli F, Qiao M, Vardarajan BN, Cosacak MI, Kizil C, Jean-Francois M, Cuccaro M, Reyes-Dumeyer D, Cantwell L, Kuzma A, Vance JM, Gao S, Hendrie HC, Baiyewu O, Ogunniyi A, Akinyemi RO, Lee WP, Martin ER, Wang LS, Beecham GW, Bush WS, Xu W, Jin F, Wang L, Farrer LA, Haines JL, Byrd GS, Schellenberg GD, Mayeux R, Pericak-Vance MA, Reitz C. Extended genome-wide association study employing the African genome resources panel identifies novel susceptibility loci for Alzheimer's disease in individuals of African ancestry. Alzheimers Dement. 2024 Aug; 20(8):5247-5261. PMID: 38958117; PMCID: PMC11350055; DOI: 10.1002/alz.13880;
     
  3. Sahelijo N, Rajagopalan P, Qian L, Rahman R, Priyadarshi D, Goldstein D, Thomopoulos SI, Bennett DA, Farrer LA, Stein TD, Shen L, Huang H, Nho K, Andrew SJ, Davatzikos C, Thompson PM, Tcw J, Jun GR. Brain Cell-based Genetic Subtyping and Drug Repositioning for Alzheimer Disease. medRxiv. 2024 Jun 21.View Related Profiles. PMID: 38947056; PMCID: PMC11213108; DOI: 10.1101/2024.06.21.24309255;
     
  4. Ahangaran M, Zhu H, Li R, Yin L, Jang J, Chaudhry AP, Farrer LA, Au R, Kolachalama VB. DREAMER: a computational framework to evaluate readiness of datasets for machine learning. BMC Med Inform Decis Mak. 2024 Jun 04; 24(1):152.View Related Profiles. PMID: 38831432; PMCID: PMC11149315; DOI: 10.1186/s12911-024-02544-w;
     
  5. Nievergelt CM, Maihofer AX, Atkinson EG, Chen CY, Choi KW, Coleman JRI, Daskalakis NP, Duncan LE, Polimanti R, Aaronson C, Amstadter AB, Andersen SB, Andreassen OA, Arbisi PA, Ashley-Koch AE, Austin SB, Avdibegoviç E, Babic D, Bacanu SA, Baker DG, Batzler A, Beckham JC, Belangero S, Benjet C, Bergner C, Bierer LM, Biernacka JM, Bierut LJ, Bisson JI, Boks MP, Bolger EA, Brandolino A, Breen G, Bressan RA, Bryant RA, Bustamante AC, Bybjerg-Grauholm J, Bækvad-Hansen M, Børglum AD, Børte S, Cahn L, Calabrese JR, Caldas-de-Almeida JM, Chatzinakos C, Cheema S, Clouston SAP, Colodro-Conde L, Coombes BJ, Cruz-Fuentes CS, Dale AM, Dalvie S, Davis LK, Deckert J, Delahanty DL, Dennis MF, Desarnaud F, DiPietro CP, Disner SG, Docherty AR, Domschke K, Dyb G, Kulenovic AD, Edenberg HJ, Evans A, Fabbri C, Fani N, Farrer LA, Feder A, Feeny NC, Flory JD, Forbes D, Franz CE, Galea S, Garrett ME, Gelaye B, Gelernter J, Geuze E, Gillespie CF, Goleva SB, Gordon SD, Goçi A, Grasser LR, Guindalini C, Haas M, Hagenaars S, Hauser MA, Heath AC, Hemmings SMJ, Hesselbrock V, Hickie IB, Hogan K, Hougaard DM, Huang H, Huckins LM, Hveem K, Jakovljevic M, Javanbakht A, Jenkins GD, Johnson J, Jones I, Jovanovic T, Karstoft KI, Kaufman ML, Kennedy JL, Kessler RC, Khan A, Kimbrel NA, King AP, Koen N, Kotov R, Kranzler HR, Krebs K, Kremen WS, Kuan PF, Lawford BR, Lebois LAM, Lehto K, Levey DF, Lewis C, Liberzon I, Linnstaedt SD, Logue MW, Lori A, Lu Y, Luft BJ, Lupton MK, Luykx JJ, Makotkine I, Maples-Keller JL, Marchese S, Marmar C, Martin NG, Martínez-Levy GA, McAloney K, McFarlane A, McLaughlin KA, McLean SA, Medland SE, Mehta D, Meyers J, Michopoulos V, Mikita EA, Milani L, Milberg W, Miller MW, Morey RA, Morris CP, Mors O, Mortensen PB, Mufford MS, Nelson EC, Nordentoft M, Norman SB, Nugent NR, O'Donnell M, Orcutt HK, Pan PM, Panizzon MS, Pathak GA, Peters ES, Peterson AL, Peverill M, Pietrzak RH, Polusny MA, Porjesz B, Powers A, Qin XJ, Ratanatharathorn A, Risbrough VB, Roberts AL, Rothbaum AO, Rothbaum BO, Roy-Byrne P, Ruggiero KJ, Rung A, Runz H, Rutten BPF, de Viteri SS, Salum GA, Sampson L, Sanchez SE, Santoro M, Seah C, Seedat S, Seng JS, Shabalin A, Sheerin CM, Silove D, Smith AK, Smoller JW, Sponheim SR, Stein DJ, Stensland S, Stevens JS, Sumner JA, Teicher MH, Thompson WK, Tiwari AK, Trapido E, Uddin M, Ursano RJ, Valdimarsdóttir U, Van Hooff M, Vermetten E, Vinkers CH, Voisey J, Wang Y, Wang Z, Waszczuk M, Weber H, Wendt FR, Werge T, Williams MA, Williamson DE, Winsvold BS, Winternitz S, Wolf C, Wolf EJ, Xia Y, Xiong Y, Yehuda R, Young KA, Young RM, Zai CC, Zai GC, Zervas M, Zhao H, Zoellner LA, Zwart JA, deRoon-Cassini T, van Rooij SJH, van den Heuvel LL, Stein MB, Ressler KJ, Koenen KC. Genome-wide association analyses identify 95 risk loci and provide insights into the neurobiology of post-traumatic stress disorder. Nat Genet. 2024 May; 56(5):792-808.View Related Profiles. PMID: 38637617
     
  6. Wang Y, Huang J, Ang TFA, Zhu Y, Tao Q, Mez J, Alosco M, Denis GV, Belkina A, Gurnani A, Ross M, Gong B, Han J, Lunetta KL, Stein TD, Au R, Farrer LA, Zhang X, Qiu WQ. The association between circulating CD34+CD133+ endothelial progenitor cells and reduced risk of Alzheimer's disease in the Framingham Heart Study. Explor Med. 2024; 5(2):193-214.View Related Profiles. PMID: 38854406; PMCID: PMC11160969; DOI: 10.37349/emed.2024.00216;
     
  7. Zhu C, Tong T, Farrell JJ, Martin ER, Bush WS, Pericak-Vance MA, Wang LS, Schellenberg GD, Haines JL, Lunetta KL, Farrer LA, Zhang X. MitoH3: Mitochondrial Haplogroup and Homoplasmic/Heteroplasmic Variant Calling Pipeline for Alzheimer's Disease Sequencing Project. J Alzheimers Dis Rep. 2024; 8(1):575-587.View Related Profiles. PMID: 38746629; PMCID: PMC11091720; DOI: 10.3233/ADR-230120;
     
  8. Panitch R, Sahelijo N, Hu J, Nho K, Bennett DA, Lunetta KL, Au R, Stein TD, Farrer LA, Jun GR. APOE genotype-specific methylation patterns are linked to Alzheimer disease pathology and estrogen response. Transl Psychiatry. 2024 Feb 29; 14(1):129.View Related Profiles. PMID: 38424036; PMCID: PMC10904829; DOI: 10.1038/s41398-024-02834-x;
     
  9. Malamon JS, Farrell JJ, Xia LC, Dombroski BA, Das RG, Way J, Kuzma AB, Valladares O, Leung YY, Scanlon AJ, Lopez IAB, Brehony J, Worley KC, Zhang NR, Wang LS, Farrer LA, Schellenberg GD, Lee WP, Vardarajan BN. A comparative study of structural variant calling in WGS from Alzheimer's disease families. Life Sci Alliance. 2024 May; 7(5). PMID: 38418088; PMCID: PMC10902710; DOI: 10.26508/lsa.202302181;
     
  10. Leung YY, Naj AC, Chou YF, Valladares O, Schmidt M, Hamilton-Nelson K, Wheeler N, Lin H, Gangadharan P, Qu L, Clark K, Kuzma AB, Lee WP, Cantwell L, Nicaretta H, Haines J, Farrer L, Seshadri S, Brkanac Z, Cruchaga C, Pericak-Vance M, Mayeux RP, Bush WS, Destefano A, Martin E, Schellenberg GD, Wang LS. Human whole-exome genotype data for Alzheimer's disease. Nat Commun. 2024 Jan 23; 15(1):684.View Related Profiles. PMID: 38263370; PMCID: PMC10805795; DOI: 10.1038/s41467-024-44781-7;
     
Showing 10 of 651 results. Show More

This graph shows the total number of publications by year, by first, middle/unknown, or last author.

Bar chart showing 651 publications over 41 distinct years, with a maximum of 40 publications in 2013

YearPublications
19841
19857
19865
198711
19886
198910
19906
19919
199214
199310
199411
199511
199611
19979
199812
19999
200013
20019
20027
200311
20048
200523
200618
200713
200813
200919
201020
201134
201231
201340
201427
201532
201625
201734
201824
201915
202018
202114
202219
202330
202412


Study seeks to explain why Sephardi Jews more likely to get early-onset Alzheimer’

Times of Israel 5/8/2024

Can We Find a Cure for Alzheimer’s Disease?

The Brink 2/21/2024

A new Alzheimer’s monoclonal antibody treatment shows promising results: ‘Hopefully more effective approach’

Boston Herald 10/4/2023

Unique genetic risk factors for Alzheimer’s disease discovered in Ashkenazi Jews

Study Finds 8/4/2023

Novel Alzheimer's Genes Unique to Ashkenazi Jews?

Medscape 6/20/2023

Alzheimer's Risk Factors Unique to Ashkenazi Jews Identified

Technology Networks 6/5/2023

Ashkenazi Jews are more likely to suffer from Alzheimer's, study finds

The Jerusalem Post 6/4/2023

New genetic risk factors for Alzheimer’s disease unique to Ashkenazi Jews identified

News Medical 6/1/2023

Boston University Study Finds Unique Alzheimer's Risks For Ashkenazi Jews

WBZ News Radio 6/1/2023

AMD patients at heightened risk of COVID-19

Optometry Today 2/2/2023

Evidence shows people with eye condition may be more at risk of COVID-19 complications

Diabetes 1/16/2023

Age-Related Eye Disease Tied to Increased COVID-19 Risk

Everyday Health 1/10/2023

Gene associated with vision loss also linked to COVID: Study

WebMD 1/5/2023

Age-related macular degeneration reported to confer higher risk of severe SARS-CoV-2 infection

News Medical 1/3/2023

Why Is Alzheimer’s More Common in Women? New Discovery Brings Scientists Closer to an Answer

SciTech Daily 8/18/2022

The fight against Alzheimer's: Where are we now?

CNN 7/31/2022

Gene discovery may explain why more women get Alzheimer's disease

CNN 6/30/2022

New discovery may explain why women at higher risk for Alzheimer’s disease

The Hill 6/30/2022

Blood Profile at Age 35 Linked to Subsequent Alzheimer's Dementia

Medpage Today 3/25/2022

BU Study Says Blood Sugar, Cholesterol Levels At Age 35 Linked To Later Alzheimer’s Risk

CBS Boston 3/24/2022

Blood Sugar, Cholesterol Issues in 30s Could Raise Alzheimer's Risk

MedicineNet 3/24/2022

Fight Alzheimer's in your mid-30s by tracking these warning signs

CNN 3/23/2022

Having high blood sugar and cholesterol levels in your THIRTIES raise risk of dementia, experts say

Daily Mail 3/23/2022

Alzheimer’s research: Cholesterol and glucose levels at age 35 are linked to future risk of Alzheimer’s, Boston researchers find

Boston Herald 3/23/2022

Ignoring cholesterol and glucose levels at age 35 may impact the chances of getting Alzheimer's later

News Medical 3/23/2022

Blood Sugar, Cholesterol Issues in 30s Could Raise Alzheimer's Risk

Health Day 3/23/2022

High blood sugar, low 'good' cholesterol as young adult raises Alzheimer's risk, study finds

United Press International 3/23/2022

Massachusetts researchers find choices you make in your 30s can affect Alzheimer's risk

WCVB 3/23/2022

Fight Alzheimer’s in your mid-30s by tracking these warning signs

WHDH 3/23/2022

New Approach Needed for AMD Research

Insight 7/31/2019

Rare Genetic Variants Tied To Alzheimer's Disease

Reuters Health 4/10/2019

Research Roundup: Pancreatic Cancer, Dementia Genes, Molecular Surgery and More | BioSpace

Daily News 4/4/2019

Is Klotho the Key to Treating Alzheimer’s and Other Diseases of Aging?

Biospace 4/2/2019

Alzheimer’s Disease: Scientists Discover Two Rare Genes Linked to Condition

Newsweek 3/29/2019

DNA Kits Yield Different Results From Two Genetics Companies

NBC Boston 3/12/2019

DNA Kits Yield Different Results From Two Companies

NECN 3/12/2019

Precision Medicine Researchers Identify New Alzheimer’s Genes

Health IT Analytics 8/17/2018

Dementia breakthrough as scientists discover genes linked to Alzheimer’s disease

Express 8/14/2018

Whole Exome Sequencing Identifies New Alzheimer’s-Related Genes

Genetic Engineering & Biotechnology News 8/14/2018

2019 Boston University School of Medicine: BU Evans Center for Interdisciplinary Biomedical Research Leadership Award
2015 Boston University: Jack Spivack Excellence in Neuroscience Research Award
2015 Boston University School of Medicine: Distinguished Professor of Genetics Award
2011 Boston University School of Medicine: Distinguished Scientist Award
2002 Indiana University School of Medicine: Distinguished Alumnus Award
2000 Case Western University: Joseph M. Foley Lecturer
1999 Who is Who in Science and Engineering (1999 edition)
1998 Who’s Who in the World (1999 edition)
1997 Who’s Who in America (1998 edition)
1997 Who’s Who in Medicine and Health Care (1999-2000 edition)
1991-1993 Alfred P. Sloan Research Fellow
1985-1987 NIH: Postdoctoral Training Fellowship
1984 Indiana University: John H. Edwards Fellowship
In addition to these self-described keywords below, a list of MeSH based concepts is available here.

age-related macular degeneration
Alzheimer disease
gene mapping
genetic epidemiology
illicit drug dependence

Available to Mentor as: (Review Mentor Role Definitions):
  • Advisor
  • Career Mentor
  • Co-Mentor or Peer Mentor
  • Research / Scholarly Mentor
Contact for Mentoring:
  • Email (see 'Contact Info')
  • Assistant
         Name: Hau Nguyen
         Email: haungvn@bu.edu
         Phone: (617) 358-3550

72 E. Concord St Instructional (L)
Boston MA 02118
Google Map


Farrer's Networks
Click the "See All" links for more information and interactive visualizations
Concepts
_
Media Mentions
_
Co-Authors
_
Similar People
_
Same Department