Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lauren Peter is active.

Publication


Featured researches published by Lauren Peter.


Nature | 2014

Rapid seeding of the viral reservoir prior to SIV viraemia in rhesus monkeys

James B. Whitney; Alison L. Hill; Srisowmya Sanisetty; Pablo Penaloza-MacMaster; Jinyan Liu; Mayuri Shetty; Lily Parenteau; Crystal Cabral; Jennifer Shields; Stephen Blackmore; Jeffrey Y. Smith; Amanda L. Brinkman; Lauren Peter; Sheeba Mathew; Kaitlin M. Smith; Erica N. Borducchi; Daniel I. S. Rosenbloom; Mark G. Lewis; Jillian Hattersley; Bei Li; Joseph Hesselgesser; Romas Geleziunas; Merlin L. Robb; Jerome H. Kim; Nelson L. Michael; Dan H. Barouch

The viral reservoir represents a critical challenge for human immunodeficiency virus type 1 (HIV-1) eradication strategies. However, it remains unclear when and where the viral reservoir is seeded during acute infection and the extent to which it is susceptible to early antiretroviral therapy (ART). Here we show that the viral reservoir is seeded rapidly after mucosal simian immunodeficiency virus (SIV) infection of rhesus monkeys and before systemic viraemia. We initiated suppressive ART in groups of monkeys on days 3, 7, 10 and 14 after intrarectal SIVMAC251 infection. Treatment with ART on day 3 blocked the emergence of viral RNA and proviral DNA in peripheral blood and also substantially reduced levels of proviral DNA in lymph nodes and gastrointestinal mucosa as compared with treatment at later time points. In addition, treatment on day 3 abrogated the induction of SIV-specific humoral and cellular immune responses. Nevertheless, after discontinuation of ART following 24 weeks of fully suppressive therapy, virus rebounded in all animals, although the monkeys that were treated on day 3 exhibited a delayed viral rebound as compared with those treated on days 7, 10 and 14. The time to viral rebound correlated with total viraemia during acute infection and with proviral DNA at the time of ART discontinuation. These data demonstrate that the viral reservoir is seeded rapidly after intrarectal SIV infection of rhesus monkeys, during the ‘eclipse’ phase, and before detectable viraemia. This strikingly early seeding of the refractory viral reservoir raises important new challenges for HIV-1 eradication strategies.


Nature | 2017

An immunogenic personal neoantigen vaccine for patients with melanoma

Patrick A. Ott; Zhuting Hu; Derin B. Keskin; Sachet A. Shukla; Jing Sun; David J. Bozym; Wandi Zhang; Adrienne M. Luoma; Anita Giobbie-Hurder; Lauren Peter; Christina Chen; Oriol Olive; Todd A. Carter; Shuqiang Li; David J. Lieb; Thomas Eisenhaure; Evisa Gjini; Jonathan Stevens; William J. Lane; Indu Javeri; Kaliappanadar Nellaiappan; Andres M. Salazar; Heather Daley; Michael S. Seaman; Elizabeth I. Buchbinder; Charles H. Yoon; Maegan Harden; Niall J. Lennon; Stacey Gabriel; Scott J. Rodig

Effective anti-tumour immunity in humans has been associated with the presence of T cells directed at cancer neoantigens, a class of HLA-bound peptides that arise from tumour-specific mutations. They are highly immunogenic because they are not present in normal tissues and hence bypass central thymic tolerance. Although neoantigens were long-envisioned as optimal targets for an anti-tumour immune response, their systematic discovery and evaluation only became feasible with the recent availability of massively parallel sequencing for detection of all coding mutations within tumours, and of machine learning approaches to reliably predict those mutated peptides with high-affinity binding of autologous human leukocyte antigen (HLA) molecules. We hypothesized that vaccination with neoantigens can both expand pre-existing neoantigen-specific T-cell populations and induce a broader repertoire of new T-cell specificities in cancer patients, tipping the intra-tumoural balance in favour of enhanced tumour control. Here we demonstrate the feasibility, safety, and immunogenicity of a vaccine that targets up to 20 predicted personal tumour neoantigens. Vaccine-induced polyfunctional CD4+ and CD8+ T cells targeted 58 (60%) and 15 (16%) of the 97 unique neoantigens used across patients, respectively. These T cells discriminated mutated from wild-type antigens, and in some cases directly recognized autologous tumour. Of six vaccinated patients, four had no recurrence at 25 months after vaccination, while two with recurrent disease were subsequently treated with anti-PD-1 (anti-programmed cell death-1) therapy and experienced complete tumour regression, with expansion of the repertoire of neoantigen-specific T cells. These data provide a strong rationale for further development of this approach, alone and in combination with checkpoint blockade or other immunotherapies.


The Journal of Infectious Diseases | 2013

First-in-human evaluation of the safety and immunogenicity of a recombinant adenovirus serotype 26 HIV-1 Env vaccine (IPCAVD 001).

Lindsey R. Baden; Stephen R. Walsh; Michael S. Seaman; Robert P. Tucker; Kathleen H. Krause; Alka Patel; Jennifer A. Johnson; Jane A. Kleinjan; Katherine E. Yanosick; James R. Perry; Elise Zablowsky; Peter Abbink; Lauren Peter; M. Justin Iampietro; Ann Cheung; Maria G. Pau; Mo Weijtens; Jaap Goudsmit; Edith Swann; Mark Wolff; Hayley Loblein; Raphael Dolin; Dan H. Barouch

BACKGROUND We report the first-in-human safety and immunogenicity assessment of a prototype Ad26 vector-based human immunodeficiency virus (HIV) vaccine in humans. METHODS Sixty Ad26-seronegative, healthy, HIV-uninfected subjects were enrolled in a randomized, double-blinded, placebo-controlled, dose-escalation phase 1 study. Five groups of 12 subjects received 10(9)-10(11) vp of the Ad26-EnvA vaccine (N = 10/group) or placebo (N = 2/group) at weeks 0 and 24 or weeks 0, 4, and 24. Safety and immunogenicity were assessed. RESULTS Self-limited reactogenicity was observed after the initial immunization at the highest (10(11) vp) dose. No product-related SAEs were observed. All subjects who received the Ad26-EnvA vaccine developed Ad26 NAb titers, EnvA-specific enzyme-linked immunosorbent assays (ELISA) titers, and EnvA-specific enzyme-linked immunospot assays (ELISPOT) responses. These responses persisted at week 52. At week 28 in the 10(9), 10(10), 10(11) vp 3-dose and the 10(10) and 5 × 10(10) vp 2-dose groups, geometric mean EnvA ELISA titers were 6113, 12 470, 8545, 3470, and 9655 and mean EnvA ELISPOT responses were 397, 178, 736, 196, and 1311 SFC/10(6) peripheral blood mononuclear cells, respectively. CONCLUSION This Ad26 vectored vaccine was generally safe and immunogenic at all doses tested. Reactogenicity was minimal with doses of 5 × 10(10) vp or less. Ad26 is a promising new vaccine vector for HIV-1. CLINICAL TRIALS REGISTRATION NCT00618605.


The Journal of Infectious Diseases | 2013

Characterization of Humoral and Cellular Immune Responses Elicited by a Recombinant Adenovirus Serotype 26 HIV-1 Env Vaccine in Healthy Adults (IPCAVD 001)

Dan H. Barouch; Jinyan Liu; Lauren Peter; Peter Abbink; M. Justin Iampietro; Ann Cheung; Galit Alter; Amy W. Chung; Anne Sophie Dugast; Nicole Frahm; M. Juliana McElrath; Holger Wenschuh; Ulf Reimer; Michael S. Seaman; Maria G. Pau; Mo Weijtens; Jaap Goudsmit; Stephen R. Walsh; Raphael Dolin; Lindsey R. Baden

BACKGROUND Adenovirus serotype 26 (Ad26) has been developed as a novel candidate vaccine vector for human immunodeficiency virus type 1 (HIV-1) and other pathogens. The primary safety and immunogenicity data from the Integrated Preclinical/Clinical AIDS Vaccine Development Program (IPCAVD) 001 trial, the first-in-human evaluation of a prototype Ad26 vector-based vaccine expressing clade A HIV-1 Env (Ad26.ENVA.01), are reported concurrently with this article. Here, we characterize in greater detail the humoral and cellular immune responses elicited by Ad26.ENVA.01 in humans. METHODS Samples from the IPCAVD 001 trial were used for humoral and cellular immunogenicity assays. RESULTS We observed a dose-dependent expansion of the magnitude, breadth, and epitopic diversity of Env-specific binding antibody responses elicited by this vaccine. Antibody-dependent cell-mediated phagocytosis, virus inhibition, and degranulation functional activity were also observed. Env-specific cellular immune responses induced by the vaccine included multiple CD8(+) and CD4(+) T-lymphocyte memory subpopulations and cytokine secretion phenotypes, although cellular immune breadth was limited. Baseline vector-specific T-lymphocyte responses were common but did not impair Env-specific immune responses in this study. CONCLUSION Ad26.ENVA.01 elicited a broad diversity of humoral and cellular immune responses in humans. These data support the further clinical development of Ad26 as a candidate vaccine vector. CLINICAL TRIALS REGISTRATION NCT00618605.


Science | 2016

Antibody-mediated protection against SHIV challenge includes systemic clearance of distal virus

Jinyan Liu; Khader Ghneim; Devin Sok; William J. Bosche; Yuan Li; Elizabeth Chipriano; Brian Berkemeier; Kelli Oswald; Erica N. Borducchi; Crystal Cabral; Lauren Peter; Amanda L. Brinkman; Mayuri Shetty; Jessica Jimenez; Jade Mondesir; Benjamin C. Lee; Patricia B. Giglio; Abishek Chandrashekar; Peter Abbink; Arnaud D. Colantonio; Courtney Gittens; Chantelle Baker; Wendeline Wagner; Mark G. Lewis; Wenjun Li; Rafick-Pierre Sekaly; Jeffrey D. Lifson; Dennis R. Burton; Dan H. Barouch

HIV-1–specific broadly neutralizing antibodies (bNAbs) can protect rhesus monkeys against simian-human immunodeficiency virus (SHIV) challenge. However, the site of antibody interception of virus and the mechanism of antibody-mediated protection remain unclear. We administered a fully protective dose of the bNAb PGT121 to rhesus monkeys and challenged them intravaginally with SHIV-SF162P3. In PGT121-treated animals, we detected low levels of viral RNA and viral DNA in distal tissues for seven days following challenge. Viral RNA–positive tissues showed transcriptomic changes indicative of innate immune activation, and cells from these tissues initiated infection after adoptive transfer into naïve hosts. These data demonstrate that bNAb-mediated protection against a mucosal virus challenge can involve clearance of infectious virus in distal tissues.


Nature | 2016

Ad26/MVA therapeutic vaccination with TLR7 stimulation in SIV-infected rhesus monkeys

Erica N. Borducchi; Crystal Cabral; Kathryn E. Stephenson; Jinyan Liu; Peter Abbink; David Ng’ang’a; Joseph P. Nkolola; Amanda L. Brinkman; Lauren Peter; Benjamin C. Lee; Jessica Jimenez; David Jetton; Jade Mondesir; Shanell Mojta; Abishek Chandrashekar; Katherine Molloy; Galit Alter; Jeffrey M. Gerold; Alison L. Hill; Mark G. Lewis; Maria G. Pau; Hanneke Schuitemaker; Joseph Hesselgesser; Romas Geleziunas; Jerome H. Kim; Merlin L. Robb; Nelson L. Michael; Dan H. Barouch

The development of immunologic interventions that can target the viral reservoir in HIV-1-infected individuals is a major goal of HIV-1 research. However, little evidence exists that the viral reservoir can be sufficiently targeted to improve virologic control following discontinuation of antiretroviral therapy. Here we show that therapeutic vaccination with Ad26/MVA (recombinant adenovirus serotype 26 (Ad26) prime, modified vaccinia Ankara (MVA) boost) and stimulation of TLR7 (Toll-like receptor 7) improves virologic control and delays viral rebound following discontinuation of antiretroviral therapy in SIV-infected rhesus monkeys that began antiretroviral therapy during acute infection. Therapeutic vaccination with Ad26/MVA resulted in a marked increase in the magnitude and breadth of SIV-specific cellular immune responses in virologically suppressed, SIV-infected monkeys. TLR7 agonist administration led to innate immune stimulation and cellular immune activation. The combination of Ad26/MVA vaccination and TLR7 stimulation resulted in decreased levels of viral DNA in lymph nodes and peripheral blood, and improved virologic control and delayed viral rebound following discontinuation of antiretroviral therapy. The breadth of cellular immune responses correlated inversely with set point viral loads and correlated directly with time to viral rebound. These data demonstrate the potential of therapeutic vaccination combined with innate immune stimulation as a strategy aimed at a functional cure for HIV-1 infection.


The Journal of Infectious Diseases | 2015

Induction of HIV-1–Specific Mucosal Immune Responses Following Intramuscular Recombinant Adenovirus Serotype 26 HIV-1 Vaccination of Humans

Lindsey R. Baden; Jinyan Liu; Hualin Li; Jennifer A. Johnson; Stephen R. Walsh; Jane A. Kleinjan; Brian A. Engelson; Lauren Peter; Peter Abbink; Danny A. Milner; Kevin L. Golden; Kyle L. Viani; Matthew D. Stachler; Benjamin J. Chen; Maria G. Pau; Mo Weijtens; Brittany R. Carey; Caroline A. Miller; Edith Swann; Mark Wolff; Hayley Loblein; Michael S. Seaman; Raphael Dolin; Dan H. Barouch

BACKGROUND Defining mucosal immune responses and inflammation to candidate human immunodeficiency virus type 1 (HIV-1) vaccines represents a current research priority for the HIV-1 vaccine field. In particular, it is unclear whether intramuscular immunization can elicit immune responses at mucosal surfaces in humans. METHODS In this double-blind, randomized, placebo-controlled clinical trial, we evaluated systemic and mucosal immune responses to a candidate adenovirus serotype 26 (Ad26) vectored HIV-1 envelop (Env) vaccine in baseline Ad26-seronegative and Ad26-seropositive healthy volunteers. Systematic mucosal sampling with rectal Weck-Cel sponges and rectal biopsies were performed. RESULTS Intramuscular immunization elicited both systemic and mucosal Env-specific humoral and cellular immune responses in the majority of subjects. Individuals with preexisting Ad26-specific neutralizing antibodies had vaccine-elicited immune responses comparable to those of subjects who were Ad26 seronegative. We also observed no increase in activated total or vector-specific mucosal CD4+ T lymphocytes following vaccination by either histopathology or flow cytometry. CONCLUSIONS These data demonstrate that a single intramuscular administration of this Ad26-vectored HIV-1 Env vaccine elicited both systemic and mucosal immune responses in humans. Induction of antigen-specific humoral and cellular mucosal immunity was not accompanied by a detectable increase in mucosal inflammation. CLINICAL TRIALS REGISTRATION NCT01103687.


The Journal of Infectious Diseases | 2014

First-in-human evaluation of a hexon chimeric adenovirus vector expressing HIV-1 Env (IPCAVD 002).

Lindsey R. Baden; Stephen R. Walsh; Michael S. Seaman; Jennifer A. Johnson; Robert P. Tucker; Jane A. Kleinjan; Jon A. Gothing; Brian A. Engelson; Brittany R. Carey; Avinash Oza; Shringkhala Bajimaya; Lauren Peter; Chelsea Bleckwehl; Peter Abbink; Maria G. Pau; Mo Weijtens; Meghan Kunchai; Edith Swann; Mark Wolff; Raphael Dolin; Dan H. Barouch

BACKGROUND We report the first-in-human safety and immunogenicity assessment of a prototype hexon chimeric adenovirus (Ad) serotype 5 (Ad5) vector containing the hexon hypervariable regions of Ad serotype 48 (Ad48) and expressing human immunodeficiency virus (HIV) type 1 EnvA. METHODS Forty-eight Ad5 and Ad48 seronegative, HIV-uninfected subjects were enrolled in a randomized, double-blind, placebo-controlled, dose escalation phase 1 study. Four groups of 12 subjects received 10(9) to 10(11) viral particles (vp) of the Ad5HVR48.EnvA.01 vaccine (n = 10 per group) or placebo (n = 2 per group) at week 0 or weeks 0, 4, and 24. Safety and immunogenicity were assessed. RESULTS Self-limited reactogenicity was observed after the initial immunization in the highest (10(11) vp) dose group. Responses in vaccinees included Ad48 neutralizing antibody (nAb) titers higher than Ad5 nAb titers, EnvA-specific enzyme-linked immunosorbent assay titers, and EnvA-specific enzyme-linked immunospot assay responses, and these responses generally persisted at week 52. At week 28 in the 10(9), 10(10), and 10(11) vp 3-dose groups, geometric mean EnvA enzyme-linked immunosorbent assay titers were 5721, 10 929, and 3420, respectively, and Ad48 nAb titers were a median of 1.7-fold higher than for Ad5. CONCLUSIONS Ad5HVR48.ENVA.01 was safe, well tolerated, and immunogenic at all doses tested. Vector-elicited nAb responses were greater for Ad48 than Ad5, confirming that Ad-specific nAbs in humans are primarily, but not exclusively, directed against the hexon hypervariable regions. Clinical Trials Registration. NCT00695877.


Journal of Acquired Immune Deficiency Syndromes | 2016

Immunization of HIV-1-Infected Persons With Autologous Dendritic Cells Transfected With mRNA Encoding HIV-1 Gag and Nef: Results of a Randomized, Placebo-Controlled Clinical Trial

Rajesh T. Gandhi; Douglas S. Kwon; Eric A. Macklin; Janet Shopis; Anna P. McLean; Nicole McBrine; Theresa Flynn; Lauren Peter; Amy Sbrolla; Daniel E. Kaufmann; Filippos Porichis; Bruce D. Walker; Nina Bhardwaj; Dan H. Barouch; Daniel G. Kavanagh

Background:HIV-1 eradication may require reactivation of latent virus along with stimulation of HIV-1-specific immune responses to clear infected cells. Immunization with autologous dendritic cells (DCs) transfected with viral mRNA is a promising strategy for eliciting HIV-1-specific immune responses. We performed a randomized controlled clinical trial to evaluate the immunogenicity of this approach in HIV-1-infected persons on antiretroviral therapy. Methods:Fifteen participants were randomized 2:1 to receive intradermal immunization with HIV-1 Gag- and Nef-transfected DCs (vaccine) or mock-transfected DCs (placebo) at weeks 0, 2, 6, and 10. All participants also received DCs pulsed with keyhole limpet hemocyanin (KLH) to assess whether responses to a neo-antigen could be induced. Results:After immunization, there were no differences in interferon-gamma enzyme-linked immunospot responses to HIV-1 Gag or Nef in the vaccine or placebo group. CD4 proliferative responses to KLH increased 2.4-fold (P = 0.026) and CD8 proliferative responses to KLH increased 2.5-fold (P = 0.053) after vaccination. There were increases in CD4 proliferative responses to HIV-1 Gag (2.5-fold vs. baseline, 3.4-fold vs. placebo, P = 0.054) and HIV-1 Nef (2.3-fold vs. baseline, 6.3-fold vs. placebo, P = 0.009) among vaccine recipients, but these responses were short-lived. Conclusion:Immunization with DCs transfected with mRNA encoding HIV-1 Gag and Nef did not induce significant interferon-gamma enzyme-linked immunospot responses. There were increases in proliferative responses to HIV-1 antigens and to a neo-antigen, KLH, but the effects were transient. Dendritic cell vaccination should be optimized to elicit stronger and long-lasting immune responses for this strategy to be effective as an HIV-1 therapeutic vaccine.


The Lancet | 2018

Evaluation of a mosaic HIV-1 vaccine in a multicentre, randomised, double-blind, placebo-controlled, phase 1/2a clinical trial (APPROACH) and in rhesus monkeys (NHP 13-19)

Dan H. Barouch; Frank Tomaka; Frank Wegmann; Daniel J. Stieh; Galit Alter; Merlin L. Robb; Nelson L. Michael; Lauren Peter; Joseph P. Nkolola; Erica N. Borducchi; Abishek Chandrashekar; David Jetton; Kathryn E. Stephenson; Wenjun Li; Bette T. Korber; Georgia D. Tomaras; David C. Montefiori; Glenda Gray; Nicole Frahm; M. Juliana McElrath; Lindsey R. Baden; Jennifer A. Johnson; Julia Hutter; Edith Swann; Etienne Karita; Hannah Kibuuka; Juliet Mpendo; Nigel Garrett; Kathy Mngadi; Kundai Chinyenze

BACKGROUND More than 1·8 million new cases of HIV-1 infection were diagnosed worldwide in 2016. No licensed prophylactic HIV-1 vaccine exists. A major limitation to date has been the lack of direct comparability between clinical trials and preclinical studies. We aimed to evaluate mosaic adenovirus serotype 26 (Ad26)-based HIV-1 vaccine candidates in parallel studies in humans and rhesus monkeys to define the optimal vaccine regimen to advance into clinical efficacy trials. METHODS We conducted a multicentre, randomised, double-blind, placebo-controlled phase 1/2a trial (APPROACH). Participants were recruited from 12 clinics in east Africa, South Africa, Thailand, and the USA. We included healthy, HIV-1-uninfected participants (aged 18-50 years) who were considered at low risk for HIV-1 infection. We randomly assigned participants to one of eight study groups, stratified by region. Participants and investigators were blinded to the treatment allocation throughout the study. We primed participants at weeks 0 and 12 with Ad26.Mos.HIV (5 × 1010 viral particles per 0·5 mL) expressing mosaic HIV-1 envelope (Env)/Gag/Pol antigens and gave boosters at weeks 24 and 48 with Ad26.Mos.HIV or modified vaccinia Ankara (MVA; 108 plaque-forming units per 0·5 mL) vectors with or without high-dose (250 μg) or low-dose (50 μg) aluminium adjuvanted clade C Env gp140 protein. Those in the control group received 0·9% saline. All study interventions were administered intramuscularly. Primary endpoints were safety and tolerability of the vaccine regimens and Env-specific binding antibody responses at week 28. Safety and immunogenicity were also assessed at week 52. All participants who received at least one vaccine dose or placebo were included in the safety analysis; immunogenicity was analysed using the per-protocol population. We also did a parallel study in rhesus monkeys (NHP 13-19) to assess the immunogenicity and protective efficacy of these vaccine regimens against a series of six repetitive, heterologous, intrarectal challenges with a rhesus peripheral blood mononuclear cell-derived challenge stock of simian-human immunodeficiency virus (SHIV-SF162P3). The APPROACH trial is registered with ClinicalTrials.gov, number NCT02315703. FINDINGS Between Feb 24, 2015, and Oct 16, 2015, we randomly assigned 393 participants to receive at least one dose of study vaccine or placebo in the APPROACH trial. All vaccine regimens demonstrated favourable safety and tolerability. The most commonly reported solicited local adverse event was mild-to-moderate pain at the injection site (varying from 69% to 88% between the different active groups vs 49% in the placebo group). Five (1%) of 393 participants reported at least one grade 3 adverse event considered related to the vaccines: abdominal pain and diarrhoea (in the same participant), increased aspartate aminotransferase, postural dizziness, back pain, and malaise. The mosaic Ad26/Ad26 plus high-dose gp140 boost vaccine was the most immunogenic in humans; it elicited Env-specific binding antibody responses (100%) and antibody-dependent cellular phagocytosis responses (80%) at week 52, and T-cell responses at week 50 (83%). We also randomly assigned 72 rhesus monkeys to receive one of five different vaccine regimens or placebo in the NHP 13-19 study. Ad26/Ad26 plus gp140 boost induced similar magnitude, durability, and phenotype of immune responses in rhesus monkeys as compared with humans and afforded 67% protection against acquisition of SHIV-SF162P3 infection (two-sided Fishers exact test p=0·007). Env-specific ELISA and enzyme-linked immunospot assay responses were the principal immune correlates of protection against SHIV challenge in monkeys. INTERPRETATION The mosaic Ad26/Ad26 plus gp140 HIV-1 vaccine induced comparable and robust immune responses in humans and rhesus monkeys, and it provided significant protection against repetitive heterologous SHIV challenges in rhesus monkeys. This vaccine concept is currently being evaluated in a phase 2b clinical efficacy study in sub-Saharan Africa (NCT03060629). FUNDING Janssen Vaccines & Prevention BV, National Institutes of Health, Ragon Institute of MGH, MIT and Harvard, Henry M Jackson Foundation for the Advancement of Military Medicine, US Department of Defense, and International AIDS Vaccine Initiative.

Collaboration


Dive into the Lauren Peter's collaboration.

Top Co-Authors

Avatar

Dan H. Barouch

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michael S. Seaman

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lindsey R. Baden

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Peter Abbink

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Erica N. Borducchi

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jinyan Liu

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Stephen R. Walsh

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Edith Swann

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Raphael Dolin

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge