Network


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

Hotspot


Dive into the research topics where Ramamoorthy Nagasubramanian is active.

Publication


Featured researches published by Ramamoorthy Nagasubramanian.


Blood | 2011

Genetic predictors for stroke in children with sickle cell anemia.

Jonathan M. Flanagan; Denise M. Frohlich; Thad A. Howard; William H. Schultz; Catherine Driscoll; Ramamoorthy Nagasubramanian; Nicole A. Mortier; Amy C. Kimble; Banu Aygun; Robert J. Adams; Ronald W. Helms; Russell E. Ware

Stroke is a devastating complication of sickle cell anemia (SCA), affecting 5% to 10% of patients before adulthood. Several candidate genetic polymorphisms have been proposed to affect stroke risk, but few have been validated, mainly because previous studies were hampered by relatively small sample sizes and the absence of additional patient cohorts for validation testing. To verify the accuracy of proposed genetic modifiers influencing stroke risk in SCA, we performed genotyping for 38 published single nucleotide polymorphisms (SNPs), as well as α-thalassemia, G6PD A(-) variant deficiency, and β-globin haplotype in 2 cohorts of children with well-defined stroke phenotypes (130 stroke, 103 nonstroke). Five polymorphisms had significant influence (P < .05): SNPs in the ANXA2, TGFBR3, and TEK genes were associated with increased stroke risk, whereas α-thalassemia and a SNP in the ADCY9 gene were linked with decreased stroke risk. Further investigation at these genetic regions may help define mutations that confer stroke risk or protection in children with SCA.


The New England Journal of Medicine | 2018

Efficacy of Larotrectinib in TRK Fusion–Positive Cancers in Adults and Children

Alexander Drilon; Theodore W. Laetsch; Shivaani Kummar; Steven G. DuBois; Ulrik N. Lassen; George D. Demetri; Michael J. Nathenson; Robert C. Doebele; Anna F. Farago; Alberto S. Pappo; Brian Turpin; Afshin Dowlati; Marcia S. Brose; Leo Mascarenhas; Noah Federman; Jordan Berlin; Wafik S. El-Deiry; Christina Baik; John F. Deeken; Valentina Boni; Ramamoorthy Nagasubramanian; Matthew H. Taylor; Erin R. Rudzinski; Funda Meric-Bernstam; Davendra P.S. Sohal; Patrick C. Ma; Luis E. Raez; Jaclyn F. Hechtman; Ryma Benayed; Marc Ladanyi

Background Fusions involving one of three tropomyosin receptor kinases (TRK) occur in diverse cancers in children and adults. We evaluated the efficacy and safety of larotrectinib, a highly selective TRK inhibitor, in adults and children who had tumors with these fusions. Methods We enrolled patients with consecutively and prospectively identified TRK fusion–positive cancers, detected by molecular profiling as routinely performed at each site, into one of three protocols: a phase 1 study involving adults, a phase 1–2 study involving children, or a phase 2 study involving adolescents and adults. The primary end point for the combined analysis was the overall response rate according to independent review. Secondary end points included duration of response, progression‐free survival, and safety. Results A total of 55 patients, ranging in age from 4 months to 76 years, were enrolled and treated. Patients had 17 unique TRK fusion–positive tumor types. The overall response rate was 75% (95% confidence interval [CI], 61 to 85) according to independent review and 80% (95% CI, 67 to 90) according to investigator assessment. At 1 year, 71% of the responses were ongoing and 55% of the patients remained progression‐free. The median duration of response and progression‐free survival had not been reached. At a median follow‐up of 9.4 months, 86% of the patients with a response (38 of 44 patients) were continuing treatment or had undergone surgery that was intended to be curative. Adverse events were predominantly of grade 1, and no adverse event of grade 3 or 4 that was considered by the investigators to be related to larotrectinib occurred in more than 5% of patients. No patient discontinued larotrectinib owing to drug‐related adverse events. Conclusions Larotrectinib had marked and durable antitumor activity in patients with TRK fusion–positive cancer, regardless of the age of the patient or of the tumor type. (Funded by Loxo Oncology and others; ClinicalTrials.gov numbers, NCT02122913, NCT02637687, and NCT02576431.)


Pediatric Blood & Cancer | 2016

Infantile Fibrosarcoma With NTRK3–ETV6 Fusion Successfully Treated With the Tropomyosin-Related Kinase Inhibitor LOXO-101

Ramamoorthy Nagasubramanian; Julie Wei; Paul R. Gordon; Jeff C. Rastatter; Michael C. Cox; Alberto S. Pappo

Infantile fibrosarcoma (IFS) is a rare pediatric cancer typically presenting in the first 2 years of life. Surgical resection is usually curative and chemotherapy is active against gross residual disease. However, when recurrences occur, therapeutic options are limited. We report a case of refractory IFS with constitutive activation of the tropomyosin‐related kinase (TRK) signaling pathway from an ETS variant gene 6–neurotrophin 3 receptor gene (ETV6–NTRK3) gene fusion. The patient enrolled in a pediatric Phase 1 trial of LOXO‐101, an experimental, highly selective inhibitor of TRK. The patient experienced a rapid, radiographic response, demonstrating the potential for LOXO‐101 to provide benefit for IFS harboring NTRK gene fusions.


Cancer Discovery | 2017

A Next-Generation TRK Kinase Inhibitor Overcomes Acquired Resistance to Prior TRK Kinase Inhibition in Patients with TRK Fusion–Positive Solid Tumors

Alexander Drilon; Ramamoorthy Nagasubramanian; James F. Blake; Nora Ku; Brian B. Tuch; Kevin Ebata; Steve Smith; Veronique Lauriault; Gabrielle R. Kolakowski; Barbara J. Brandhuber; Paul D. Larsen; Karyn S. Bouhana; Shannon L. Winski; Robyn Hamor; Wen-I Wu; Andrew Parker; Tony Morales; Francis X. Sullivan; Walter E. DeWolf; Lance Wollenberg; Paul R. Gordon; Dorothea N. Douglas-Lindsay; Maurizio Scaltriti; Ryma Benayed; Sandeep Raj; Bethany Hanusch; Alison M. Schram; Philip Jonsson; Michael F. Berger; Jaclyn F. Hechtman

Larotrectinib, a selective TRK tyrosine kinase inhibitor (TKI), has demonstrated histology-agnostic efficacy in patients with TRK fusion-positive cancers. Although responses to TRK inhibition can be dramatic and durable, duration of response may eventually be limited by acquired resistance. LOXO-195 is a selective TRK TKI designed to overcome acquired resistance mediated by recurrent kinase domain (solvent front and xDFG) mutations identified in multiple patients who have developed resistance to TRK TKIs. Activity against these acquired mutations was confirmed in enzyme and cell-based assays and in vivo tumor models. As clinical proof of concept, the first 2 patients with TRK fusion-positive cancers who developed acquired resistance mutations on larotrectinib were treated with LOXO-195 on a first-in-human basis, utilizing rapid dose titration guided by pharmacokinetic assessments. This approach led to rapid tumor responses and extended the overall duration of disease control achieved with TRK inhibition in both patients.Significance: LOXO-195 abrogated resistance in TRK fusion-positive cancers that acquired kinase domain mutations, a shared liability with all existing TRK TKIs. This establishes a role for sequential treatment by demonstrating continued TRK dependence and validates a paradigm for the accelerated development of next-generation inhibitors against validated oncogenic targets. Cancer Discov; 7(9); 963-72. ©2017 AACR.See related commentary by Parikh and Corcoran, p. 934This article is highlighted in the In This Issue feature, p. 920.


American Journal of Hematology | 2012

Chronic transfusion practices for prevention of primary stroke in children with sickle cell anemia and abnormal TCD velocities

Banu Aygun; Lisa M. Wruck; William H. Schultz; Brigitta U. Mueller; Clark Brown; Lori Luchtman-Jones; Sherron M. Jackson; Rathi V. Iyer; Zora R. Rogers; Sharada A. Sarnaik; Alexis A. Thompson; Cynthia Gauger; Ronald W. Helms; Russell E. Ware; Bogdan R. Dinu; Kusum Viswanathan; Natalie Sommerville-Brooks; Betsy Record; Matthew M. Heeney; Meredith Anderson; Janet L. Kwiatkowski; Jeff Olson; Martha Brown; Lakshmanan Krishnamurti; Regina McCollum; Kamar Godder; Jennifer Newlin; William Owen; Stephen C. Nelson; Katie Bianchi

Chronic transfusions are recommended for children with sickle cell anemia (SCA) and abnormal transcranial Doppler (TCD) velocities ( 200 cm/sec) to help prevent the occurrence of a primary stroke [1]. The goal is usually to maintain the sickle hemoglobin concentration (HbS) <30%; however, this goal is often difficult to achieve in clinical practice. The NHLBI-sponsored trial ‘‘TCD With Transfusions Changing to Hydroxyurea (TWiTCH)’’ will compare standard therapy (transfusions) to alternative therapy (hydroxyurea) for the reduction of primary stroke risk in this patient population. Transfusions will be given according to current transfusion practices at participating sites. To determine current academic community standards for primary stroke prophylaxis in children with SCA, 32 clinical sites collected data on 340 children with abnormal TCD velocities receiving chronic transfusions to help prevent primary stroke. The average (mean ± 1 SD) pretransfusion HbS was 34 ± 11% (institutional average 23–48%); the 75th and 90th percentiles were 41 and 50%, respectively. Lower %HbS was associated with higher pretransfusion Hb values and receiving transfusions on time. These data indicate variable current transfusion practices among academic pediatric institutions and in practice, 30% HbS may not be an easily attainable goal in this cohort of children with SCA and abnormal TCD. Children with sickle cell anemia (SCA) compose a high risk group for the development of stroke. If untreated, 11% will experience a clinical stroke by 20 years of age [2]. Adams et al. have shown that children with SCA who are at risk for primary stroke can be identified by measuring time-averaged mean blood flow velocities in the internal carotid or middle cerebral arteries by TCD [3]. Abnormal TCD velocities ( 200 cm/sec) are associated with high risk for stroke and warrant transfusion therapy to reduce the risk of primary stroke. First stroke can be successfully prevented in 90% of children with SCA and abnormal TCD velocities by the use of chronic transfusion therapy, with a goal of keeping HbS concentrations less than 30% [1]. TCD with Transfusions Changing to Hydroxyurea (TWiTCH) is an NHLBIsponsored, Phase III, multicenter trial comparing standard therapy (monthly transfusions) to alternative therapy (daily hydroxyurea) to reduce the risk of primary stroke in children with SCA and documented abnormal TCD velocities. Since transfusions compose the standard treatment arm, accurate %HbS values achieved in actual clinical practice were needed for protocol development. The majority of our information about transfusing patients with SCA to prevent stroke comes from secondary stroke prevention, i.e., the use of chronic red blood cell transfusions to prevent a second stroke after a first clinical stroke has occurred. Classically, transfusions are administered at 4-week intervals to maintain HbS at less than 30%. After several years of transfusion therapy, a few centers increase transfusion interval to 5–6 weeks and allow HbS to increase toward 50% in selected patients [4,5]. Our previous study in 295 children with SCA who received transfusions for secondary stroke prevention revealed an average pretransfusion HbS of 35 ± 11% with highly variable institutional %HbS levels ranging from 22 to 51% [6] In order to determine the current clinical standard of transfusion therapy for primary stroke prevention for elevated TCD velocities, we performed a larger survey of potential TWiTCH sites. We hypothesized that average pretransfusion HbS values achieved at pediatric academic centers would be higher than 30%. This study defines the current practice at academic medical centers in provision of chronic transfusion therapy to help reduce the risk of primary stroke in children with SCA. A total of 340 children with SCA and history of abnormal TCD velocities receiving chronic PRBC transfusions for primary stroke prophylaxis were identified at 32 institutions (Table I). The number of patients per site ranged from 3 to 33 (median 9 per site). A total of 3,970 transfusions were administered over the 12-month period, with a mean of 11.7 ± 2.8 transfusions per patient. Results were similar when analyzed by each patient contributing equally or each transfusion contributing equally (Table II). The predominant transfusion type by patient was defined as the technique used 6 times over the 12-month period. Most children (79%) received primarily simple transfusions, while 19% had primarily exchange transfusions (11% partial / manual exchange, 8% erythrocytapheresis), and 2% multiple transfusion types. The transfusion goal was <30% at almost all sites (84%), while at five sites, the %HbS was allowed in selected patients to increase to 50% after a period of clinical stability. The majority (95%) of the transfusions were administered within the defined 7-day window. On average, late transfusions were given 1.3 ± 5.5 days after the defined 7-day window. Thirty percent of the patients had at least one late transfusion and 14% had 2 or more late transfusions in the 1-year period. For the 3,653 transfusions with reported %HbS values (representing 92% of the 3,970 transfusions), the mean pretransfusion HbS percentage was 33.2 ± 14.0% (median 32%). The 75th percentile for HbS values was 41%, while the 90th percentile was 51%. There were substantial differences among institutional pretransfusion %HbS values, ranging from 23 ± 14% HbS at one institution where HbS was reported for 103 transfusions given to nine patients during the 12-month period, to 48 ± 15% at another institution where HbS was reported for 95 transfusions administered to nine patients during the same time frame (Table III). The five sites with increased HbS goals to 50% in selected patients did not have higher values than others. For each transfusion, subjects were less likely to have pretransfusion HbS <30% if they were older [OR 0.92 for each year increase in age, 95% CI (0.89, 0.96)] and on transfusions for a longer period of time [OR 0.90 for each year increase in duration, 95% CI (0.86, 0.94)]. Patients with higher pretransfusion Hb levels were more likely to have pretransfusion HbS <30% [OR 1.63 for each g/dL increase in Hb, 95% CI (1.46, 1.83)] and late transfusions were less likely to be associated with a pretransfusion HbS <30% [OR 0.27, 95% CI (0.18, 0.41)]. The Hb result does not appear to be a function of late transfusions since both covariates remained significant when modeled jointly. History of alloor autoantibodies, TCD velocity, and erythrocytapheresis use were not significant predictors of a pretransfusion HbS <30%. During the initial STOP study, transfusions were given to maintain pretransfusion HbS values at less than 30% [3]. However, there were frequent transient rises of HbS above this level [7]. Furthermore, extended follow-up results from the STOP study showed that pretransfusion %HbS values during the post-trial follow-up were higher than those during the STOP study [8]. The average %HbS per patient was 27.5 ± 12.4, still within the desired goal of 30%. However, pretransfusion HbS levels were 30–34.9% in 12%, 35–39.9% in 7%, and greater than 40% in 12% of the transfusions. In the STOP2 study, where children with abnormal TCD velocities whose Doppler readings became normal were randomly assigned to continue or stop transfusions, 24% of the patients had pretransfusion HbS levels greater than 30% [9]. These findings indicate that even in the context of a prospective clinical trial, maintaining HbS <30% was difficult to achieve. With the subsequent recommendation to treat all children with SCA who are at risk for primary stroke with transfusions to maintain HbS <30%, the feasibility of this approach in actual clinical practice is not known. Possible Letters


Lancet Oncology | 2018

Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: phase 1 results from a multicentre, open-label, phase 1/2 study

Theodore W. Laetsch; Steven G. DuBois; Leo Mascarenhas; Brian Turpin; Noah Federman; Catherine M Albert; Ramamoorthy Nagasubramanian; Jessica L Davis; Erin R. Rudzinski; Angela M. Feraco; Brian B. Tuch; Kevin Ebata; Mark Reynolds; Steven M. Smith; Scott Cruickshank; Michael Craig Cox; Alberto S. Pappo; Douglas S. Hawkins

BACKGROUND Gene fusions involving NTRK1, NTRK2, or NTRK3 (TRK fusions) are found in a broad range of paediatric and adult malignancies. Larotrectinib, a highly selective small-molecule inhibitor of the TRK kinases, had shown activity in preclinical models and in adults with tumours harbouring TRK fusions. This study aimed to assess the safety of larotrectinib in paediatric patients. METHODS This multicentre, open-label, phase 1/2 study was done at eight sites in the USA and enrolled infants, children, and adolescents aged 1 month to 21 years with locally advanced or metastatic solid tumours or CNS tumours that had relapsed, progressed, or were non-responsive to available therapies regardless of TRK fusion status; had a Karnofsky (≥16 years of age) or Lansky (<16 years of age) performance status score of 50 or more, adequate organ function, and full recovery from the acute toxic effects of all previous anticancer therapy. Following a protocol amendment on Sept 12, 2016, patients with locally advanced infantile fibrosarcoma who would require disfiguring surgery to achieve a complete surgical resection were also eligible. Patients were enrolled to three dose cohorts according to a rolling six design. Larotrectinib was administered orally (capsule or liquid formulation), twice daily, on a continuous 28-day schedule, in increasing doses adjusted for age and bodyweight. The primary endpoint of the phase 1 dose escalation component was the safety of larotrectinib, including dose-limiting toxicity. All patients who received at least one dose of larotrectinib were included in the safety analyses. Reported here are results of the phase 1 dose escalation cohort. Phase 1 follow-up and phase 2 are ongoing. This trial is registered with ClinicalTrials.gov, number NCT02637687. FINDINGS Between Dec 21, 2015, and April 13, 2017, 24 patients (n=17 with tumours harbouring TRK fusions, n=7 without a documented TRK fusion) with a median age of 4·5 years (IQR 1·3-13·3) were enrolled to three dose cohorts: cohorts 1 and 2 were assigned doses on the basis of both age and bodyweight predicted by use of SimCyp modelling to achieve an area under the curve equivalent to the adult doses of 100 mg twice daily (cohort 1) and 150 mg twice daily (cohort 2); and cohort 3 was assigned to receive a dose of 100 mg/m2 twice daily (maximum 100 mg per dose), regardless of age, equating to a maximum of 173% of the recommended adult phase 2 dose. Among enrolled patients harbouring TRK fusion-positive cancers, eight (47%) had infantile fibrosarcoma, seven (41%) had other soft tissue sarcomas, and two (12%) had papillary thyroid cancer. Adverse events were predominantly grade 1 or 2 (occurring in 21 [88%] of 24 patients); the most common larotrectinib-related adverse events of all grades were increased alanine and aspartate aminotransferase (ten [42%] of 24 each), leucopenia (five [21%] of 24), decreased neutrophil count (five [21%] of 24), and vomiting (five [21%] of 24). Grade 3 alanine aminotransferase elevation was the only dose-limiting toxicity and occurred in one patient without a TRK fusion and with progressive disease. No grade 4 or 5 treatment-related adverse events were observed. Two larotrectinib-related serious adverse events were observed: grade 3 nausea and grade 3 ejection fraction decrease during the 28-day follow-up after discontinuing larotrectinib and while on anthracyclines. The maximum tolerated dose was not reached, and 100 mg/m2 (maximum of 100 mg per dose) was established as the recommended phase 2 dose. 14 (93%) of 15 patients with TRK fusion-positive cancers achieved an objective response as per Response Evaluation Criteria In Solid Tumors version 1.1; the remaining patient had tumour regression that did not meet the criteria for objective response. None of the seven patients with TRK fusion-negative cancers had an objective response. INTERPRETATION The TRK inhibitor larotrectinib was well tolerated in paediatric patients and showed encouraging antitumour activity in all patients with TRK fusion-positive tumours. The recommended phase 2 dose was defined as 100mg/m2 (maximum 100 mg per dose) for infants, children, and adolescents, regardless of age. FUNDING Loxo Oncology Inc.


Pediatric Blood & Cancer | 2018

Targeting NTRK fusions for the treatment of congenital mesoblastic nephroma

Theodore W. Laetsch; Ramamoorthy Nagasubramanian; Michela Casanova

We read with great interest the study by Gooskens et al.1 and would like to comment on a statement in the Discussion section: “Targeted therapy might be an option for cases with progressive disease who do not respond to chemotherapy. In particular, Crizotinib, an ALK inhibitor that proved to be of benefit for acute lymphoblastic leukemia patients harboring the ETV6-NTRK3 transcript, resulting from the same t(12;15)(p13;q25) as described in CMN cases, could be considered as a potential compound in highly selected aggressive cases.” We completely agree that in selected cases of congenital mesoblastic nephroma (CMN), targeted therapy should be considered. NTRK fusions have been shown to be drivers of a broad range of adult and pediatric malignancies, including cellular CMN,2 infantile fibrosarcoma,2,3 sarcoma NOS,3 papillary thyroid cancer,4 diffuse intrinsic pontine gliomas, and other high-grade gliomas.5 It is of note that these fusions have recently been selectively targeted by investigational agents, namely larotrectinib (LOXO-101) and entrectinib. Both are highly potent inhibitors of TRKA, TRKB, and TRKC with IC50s below 10 nM, with entrectinib also inhibiting ROS1, ALK, JAK2, and ACK1 at concentrations below 100 nM.6,7 Conversely, crizotinib is a multikinase inhibitor that inhibits TRK at approximately 100-fold greater concentrations (Table 1).8 Notably, durable responses to larotrectinib and entrectinib have been reported in adultswithmetastatic sarcoma, papillary thyroid cancer, nonsmall cell lung cancer, gastrointestinal stromal tumor, mammary analog secretory carcinoma, and colorectal cancer treated on phase 1 studies.6,9 Nagasubramanian et al. reported the successful treatment of a 16-month-old girl with infantile fibrosarcomaharboring the classic ETV6–NTRK3 fusion with larotrectinib.10 Phase 1 studies of larotrectinib and entrectinib are ongoing in children (NCT02637687, NCT02650401). Recently, the US Food and Drug Administration granted rare pediatric disease designation to larotrectinib for the treatment of infantile fibrosarcoma and breakthrough therapy designation for the treatment of unresectable or metastatic solid tumors harboring NTRK fusions in adult and pediatric patients. It is particularly important to emphasize that no children younger than 1 year were included in the pediatric phase 1 study of crizotinib. In the ongoing pediatric phase 1 study of larotrectinib, infants with an NTRK fusion-positive malignancy as young as age 1 month are eligible. Recruitment of children aged less than 1 year is rare in phase 1 studies, but it is well documented that drug absorption, distribution,


Seminars in Ultrasound Ct and Mri | 2013

The current state of imaging pediatric hemoglobinopathies.

David Dinan; Monica Epelman; Carolina V. Guimaraes; Lane F. Donnelly; Ramamoorthy Nagasubramanian; Nancy A. Chauvin

The hemoglobinopathies are a group of genetic disorders with a broad spectrum of clinical manifestations and radiologic findings. The imaging of pediatric hemoglobinopathies, which is influenced by concomitant hemosiderosis and the sequela of chelation therapy, has evolved over the years along with ever-improving technology. This article reviews and illustrates the most common radiographic and cross-sectional imaging findings of the 2 best known and clinically relevant hemoglobinopathies in pediatric patients, sickle cell disease and β-thalassemia.


Cancer | 2018

The Use of Neoadjuvant Larotrectinib in the Management of Children With Locally Advanced TRK Fusion Sarcomas: Neoadjuvant Larotrectinib in Sarcoma

Steven G. DuBois; Theodore W. Laetsch; Noah Federman; Brian Turpin; Catherine M Albert; Ramamoorthy Nagasubramanian; Megan E. Anderson; Jessica L. Davis; Hope E. Qamoos; Mark Reynolds; Scott Cruickshank; Michael C. Cox; Douglas S. Hawkins; Leo Mascarenhas; Alberto S. Pappo

The highly selective oral tropomyosin receptor kinase (TRK) inhibitor larotrectinib has demonstrated significant activity in adult and pediatric TRK fusion cancers. In the current study, the authors describe the clinical course of children with locally advanced TRK fusion sarcoma who were treated preoperatively with larotrectinib and underwent subsequent surgical resection.


Blood | 2010

Validation of Genetic Predictors for Stroke In Children with Sickle Cell Anemia

Jonathan M. Flanagan; Thad A. Howard; Denise M. Frohlich; William H. Schultz; Catherine Driscoll; Ramamoorthy Nagasubramanian; Nicole A. Mortier; Amy C. Kimble; Banu Aygun; Robert J. Adams; Ronald W. Helms; Russell E. Ware

Collaboration


Dive into the Ramamoorthy Nagasubramanian's collaboration.

Top Co-Authors

Avatar

Alberto S. Pappo

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Theodore W. Laetsch

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Leo Mascarenhas

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Banu Aygun

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Brian Turpin

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michael C. Cox

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Noah Federman

University of California

View shared research outputs
Top Co-Authors

Avatar

Ronald W. Helms

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Russell E. Ware

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge