Network


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

Hotspot


Dive into the research topics where Elena Helman is active.

Publication


Featured researches published by Elena Helman.


Nature | 2013

Mutational heterogeneity in cancer and the search for new cancer-associated genes.

Michael S. Lawrence; Petar Stojanov; Paz Polak; Gregory V. Kryukov; Kristian Cibulskis; Andrey Sivachenko; Scott L. Carter; Chip Stewart; Craig H. Mermel; Steven A. Roberts; Adam Kiezun; Peter S. Hammerman; Aaron McKenna; Yotam Drier; Lihua Zou; Alex H. Ramos; Trevor J. Pugh; Nicolas Stransky; Elena Helman; Jaegil Kim; Carrie Sougnez; Lauren Ambrogio; Elizabeth Nickerson; Erica Shefler; Maria L. Cortes; Daniel Auclair; Gordon Saksena; Douglas Voet; Michael S. Noble; Daniel DiCara

Major international projects are underway that are aimed at creating a comprehensive catalogue of all the genes responsible for the initiation and progression of cancer. These studies involve the sequencing of matched tumour–normal samples followed by mathematical analysis to identify those genes in which mutations occur more frequently than expected by random chance. Here we describe a fundamental problem with cancer genome studies: as the sample size increases, the list of putatively significant genes produced by current analytical methods burgeons into the hundreds. The list includes many implausible genes (such as those encoding olfactory receptors and the muscle protein titin), suggesting extensive false-positive findings that overshadow true driver events. We show that this problem stems largely from mutational heterogeneity and provide a novel analytical methodology, MutSigCV, for resolving the problem. We apply MutSigCV to exome sequences from 3,083 tumour–normal pairs and discover extraordinary variation in mutation frequency and spectrum within cancer types, which sheds light on mutational processes and disease aetiology, and in mutation frequency across the genome, which is strongly correlated with DNA replication timing and also with transcriptional activity. By incorporating mutational heterogeneity into the analyses, MutSigCV is able to eliminate most of the apparent artefactual findings and enable the identification of genes truly associated with cancer.


Nature Biotechnology | 2012

Absolute quantification of somatic DNA alterations in human cancer

Scott L. Carter; Kristian Cibulskis; Elena Helman; Aaron McKenna; Hui Shen; Travis I. Zack; Peter W. Laird; Robert C. Onofrio; Wendy Winckler; Barbara A. Weir; Rameen Beroukhim; David Pellman; Douglas A. Levine; Eric S. Lander; Matthew Meyerson; Gad Getz

We describe a computational method that infers tumor purity and malignant cell ploidy directly from analysis of somatic DNA alterations. The method, named ABSOLUTE, can detect subclonal heterogeneity and somatic homozygosity, and it can calculate statistical sensitivity for detection of specific aberrations. We used ABSOLUTE to analyze exome sequencing data from 214 ovarian carcinoma tumor-normal pairs. This analysis identified both pervasive subclonal somatic point-mutations and a small subset of predominantly clonal and homozygous mutations, which were overrepresented in the tumor suppressor genes TP53 and NF1 and in a candidate tumor suppressor gene CDK12. We also used ABSOLUTE to infer absolute allelic copy-number profiles from 3,155 diverse cancer specimens, revealing that genome-doubling events are common in human cancer, likely occur in cells that are already aneuploid, and influence pathways of tumor progression (for example, with recessive inactivation of NF1 being less common after genome doubling). ABSOLUTE will facilitate the design of clinical sequencing studies and studies of cancer genome evolution and intra-tumor heterogeneity.


Bioinformatics | 2009

A geometric approach for classification and comparison of structural variants

Suzanne S. Sindi; Elena Helman; Ali Bashir; Benjamin J. Raphael

Motivation: Structural variants, including duplications, insertions, deletions and inversions of large blocks of DNA sequence, are an important contributor to human genome variation. Measuring structural variants in a genome sequence is typically more challenging than measuring single nucleotide changes. Current approaches for structural variant identification, including paired-end DNA sequencing/mapping and array comparative genomic hybridization (aCGH), do not identify the boundaries of variants precisely. Consequently, most reported human structural variants are poorly defined and not readily compared across different studies and measurement techniques. Results: We introduce Geometric Analysis of Structural Variants (GASV), a geometric approach for identification, classification and comparison of structural variants. This approach represents the uncertainty in measurement of a structural variant as a polygon in the plane, and identifies measurements supporting the same variant by computing intersections of polygons. We derive a computational geometry algorithm to efficiently identify all such intersections. We apply GASV to sequencing data from nine individual human genomes and several cancer genomes. We obtain better localization of the boundaries of structural variants, distinguish genetic from putative somatic structural variants in cancer genomes, and integrate aCGH and paired-end sequencing measurements of structural variants. This work presents the first general framework for comparing structural variants across multiple samples and measurement techniques, and will be useful for studies of both genetic structural variants and somatic rearrangements in cancer. Availability: http://cs.brown.edu/people/braphael/software.html Contact: [email protected]


Proceedings of the National Academy of Sciences of the United States of America | 2014

Complementary genomic approaches highlight the PI3K/mTOR pathway as a common vulnerability in osteosarcoma

Jennifer A. Perry; Adam Kiezun; Peter Tonzi; Eliezer M. Van Allen; Scott L. Carter; Sylvan C. Baca; Glenn S. Cowley; Ami S. Bhatt; Esther Rheinbay; Chandra Sekhar Pedamallu; Elena Helman; Amaro Taylor-Weiner; Aaron McKenna; David S. DeLuca; Michael S. Lawrence; Lauren Ambrogio; Carrie Sougnez; Andrey Sivachenko; Loren D. Walensky; Nikhil Wagle; Jaume Mora; Carmen Torres; Cinzia Lavarino; Simone dos Santos Aguiar; José Andrés Yunes; Silvia Regina Brandalise; Gabriela Elisa Mercado-Celis; Jorge Melendez-Zajgla; Rocio Cardenas-Cardos; Liliana Velasco-Hidalgo

Significance We present, to our knowledge, the first comprehensive next-generation sequencing of osteosarcoma in combination with a functional genomic screen in a genetically defined mouse model of osteosarcoma. Our data provide a strong rationale for targeting the phosphatidylinositol 3-kinase/mammalian target of rapamycin pathway in osteosarcoma and a foundation for rational clinical trial design. These findings present an immediate clinical opportunity because multiple inhibitors of this pathway are currently in clinical trials. Osteosarcoma is the most common primary bone tumor, yet there have been no substantial advances in treatment or survival in three decades. We examined 59 tumor/normal pairs by whole-exome, whole-genome, and RNA-sequencing. Only the TP53 gene was mutated at significant frequency across all samples. The mean nonsilent somatic mutation rate was 1.2 mutations per megabase, and there was a median of 230 somatic rearrangements per tumor. Complex chains of rearrangements and localized hypermutation were detected in almost all cases. Given the intertumor heterogeneity, the extent of genomic instability, and the difficulty in acquiring a large sample size in a rare tumor, we used several methods to identify genomic events contributing to osteosarcoma survival. Pathway analysis, a heuristic analytic algorithm, a comparative oncology approach, and an shRNA screen converged on the phosphatidylinositol 3-kinase/mammalian target of rapamycin (PI3K/mTOR) pathway as a central vulnerability for therapeutic exploitation in osteosarcoma. Osteosarcoma cell lines are responsive to pharmacologic and genetic inhibition of the PI3K/mTOR pathway both in vitro and in vivo.


Genome Research | 2014

Somatic retrotransposition in human cancer revealed by whole-genome and exome sequencing.

Elena Helman; Michael S. Lawrence; Chip Stewart; Carrie Sougnez; Gad Getz; Matthew Meyerson

Retrotransposons constitute a major source of genetic variation, and somatic retrotransposon insertions have been reported in cancer. Here, we applied TranspoSeq, a computational framework that identifies retrotransposon insertions from sequencing data, to whole genomes from 200 tumor/normal pairs across 11 tumor types as part of The Cancer Genome Atlas (TCGA) Pan-Cancer Project. In addition to novel germline polymorphisms, we find 810 somatic retrotransposon insertions primarily in lung squamous, head and neck, colorectal, and endometrial carcinomas. Many somatic retrotransposon insertions occur in known cancer genes. We find that high somatic retrotransposition rates in tumors are associated with high rates of genomic rearrangement and somatic mutation. Finally, we developed TranspoSeq-Exome to interrogate an additional 767 tumor samples with hybrid-capture exome data and discovered 35 novel somatic retrotransposon insertions into exonic regions, including an insertion into an exon of the PTEN tumor suppressor gene. The results of this large-scale, comprehensive analysis of retrotransposon movement across tumor types suggest that somatic retrotransposon insertions may represent an important class of structural variation in cancer.


Cancer Research | 2012

Abstract 5060: Identification of somatic retrotransposon insertions across cancer types using RetroSeq

Elena Helman; Michael S. Lawrence; Chip Stewart; Gad Getz; Matthew Meyerson

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Retrotransposons comprise over 40% of the human genome and represent an important class of genetic variation. Though most elements are stationary ancient relics, it has recently been shown that some 100 elements remain highly active, copying and pasting themselves throughout the genome with each generation. Retrotransposon insertions can disrupt gene function, modulate gene expression, and lead to genomic rearrangement. These events have previously been implicated in cancer, including an account of insertional mutagenesis in APC as an early event in colon cancer progression. Due to the inherent difficulty in localizing repeat elements, however, an extensive investigation of retrotransposons’ role in cancer has not yet been performed. We developed RetroSeq, a computational framework to identify novel insertions of retrotransposons from paired-end sequencing data. In simulated data, RetroSeq identifies retrotransposon insertions with 99% sensitivity and 99% specificity. We ran RetroSeq on whole-genome sequencing data from a panel of tumors (including 9 colorectal, 20 non-small cell lung, 24 prostate and 22 breast tumors) and matched normal samples to determine the extent of somatic retrotransposon activity. We find a broad range of novel retrotransposon insertion events specific to each tumor, with some insertions falling in exonic as well as intronic and intergenic regions. These events were validated experimentally and integrated with expression and methylation data to provide a global view of retrotransposon activity in these samples. In sum, using RetroSeq we are able to localize novel retrotransposon insertions in paired-end sequencing data and provide evidence for the reactivation of retrotransposons in cancer. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 5060. doi:1538-7445.AM2012-5060


Cancer Research | 2015

Abstract 1457: Genomic characterization of a PDX model of T-DM1-resistant HER2+ invasive ductal carcinoma using augmented exome sequencing

Elena Helman; Michael J. Wick; Michael J. Clark; Lizette Gamez; Sean Boyle; Kyriakos P. Papadopoulos; Shujun Luo; Anthony W. Tolcher; Parin Sripakdeevong; Mirian Karbelashvili; Deanna M. Church; Richard Chen; John West

HER2 amplification/overexpression occurs in 20-30% of breast cancers and is associated with poor prognosis and increased metastatic potential. Patients with HER2+ breast cancers who have progressed on trastuzumab and lapatinib are often prescribed the antibody-drug conjugate, do-trastuzumab emtansine (T-DM1). Results from the TH3RESA and EMILIA studies showed that T-DM1 increased progression-free and overall survival compared to standard therapy and suggested it be considered as standard of care. Despite these favorable efficacy results, most patients treated with T-DM1 eventually progress, but the mechanisms of resistance are not understood. Acquired resistance to T-DM1 has been shown in-vitro, but has not been examined in an in-vivo system. An FNA from a metastatic lung lesion was used to establish a human PDX model from a patient with metastatic HER2+ invasive ductal carcinoma. This model was found resistant to T-DM1 administered weekly at 3 m/k and every three weeks at 10 m/k. We performed whole-exome sequencing on both the metastatic tissue and the PDX model using a content-enhanced exome technology we developed. Our augmented exome is optimized to detect major cancer mutations by enhancing coverage over known sequencing gaps and GC-rich regions across >1300 cancer and 200 miRNA genes. We also performed whole-transcriptome sequencing on the PDX model. All data were analyzed using a cancer bioinformatics pipeline optimized for high-accuracy detection of small variants and indels, somatic copy-number aberrations, gene expression and fusions. We comprehensively profiled the exomes of a metastatic HER2+ ductal carcinoma resistant to T-DM1 treatment and a PDX model derived from it. We found that the PDX model was highly consistent with the neoplastic tissue, with respect to both somatic variants and copy-number alterations, establishing it as an accurate representation of a patient-derived T-DM1-resistant tumor. We verified only 0.27% contamination by mouse DNA in the PDX model. Specifically, we confirmed the continued amplification of HER2 as well as CCNE1 and MYC. Loss-of-heterozygosity of TP53 was coupled with a clonal damaging point mutation. Of note, we found a non-synonymous mutation in HER2, suggesting possible involvement in resistance mechanisms. Transcriptome data confirmed mutation expression in the RNA and gene expression changes of amplified/deleted genes. We present the first preclinical model of a human xenograft derived from a HER2+ metastatic breast cancer with acquired T-DM1 resistance. T-DM1 is effective in treating advanced HER2+ breast cancer in patients who have progressed on standard therapies, but this efficacy is short-lived. Here, we used whole-exome and transcriptome sequencing to characterize the genomic profile of tumors that have become resistant to T-DM1 and present a patient-derived model to reveal insights into acquired T-DM1 resistance mechanisms. Citation Format: Elena Helman, Michael J. Wick, Michael J. Clark, Lizette Gamez, Sean Boyle, Kyriakos P. Papadopoulos, Shujun Luo, Anthony W. Tolcher, Parin Sripakdeevong, Mirian Karbelashvili, Deanna Church, Richard Chen, John West. Genomic characterization of a PDX model of T-DM1-resistant HER2+ invasive ductal carcinoma using augmented exome sequencing. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1457. doi:10.1158/1538-7445.AM2015-1457


Clinical Lung Cancer | 2018

Cell-Free DNA Next-Generation Sequencing Prediction of Response and Resistance to Third-Generation EGFR Inhibitor

Elena Helman; Minh Nguyen; Chris Karlovich; Darrin Despain; A. Karin Choquette; Alexander I. Spira; Helena A. Yu; D. Ross Camidge; Thomas C. Harding; Richard B. Lanman; Andrew Simmons

&NA; We profiled 77 non–small‐cell lung cancer patients with paired baseline and progression blood samples treated with the third‐generation EGFR tyrosine kinase inhibitor (TKI) rociletinib using a broad cell‐free circulating DNA (cfDNA) next‐generation sequencing (NGS) gene panel. We demonstrated a utility of cfDNA NGS to detect EGFR T790M and predict response comparable to tissue‐based tests, even at low allele fractions, and we identified resistance mechanisms. Our findings highlight the genomic heterogeneity observed in disease after progression while receiving therapy with a third‐generation EGFR TKI. Introduction: The genomic alterations driving resistance to third‐generation EGFR tyrosine kinase inhibitors (TKIs) are not well established, and collecting tissue biopsy samples poses potential complications from invasive procedures. Cell‐free circulating DNA (cfDNA) testing provides a noninvasive approach to identify potentially targetable mechanisms of resistance. Here we utilized a 70‐gene cfDNA next‐generation sequencing test to interrogate pretreatment and progression samples from 77 EGFR‐mutated non‐small cell lung cancer (NSCLC) patients treated with a third‐generation EGFR TKI. Patients and Methods: Rociletinib was evaluated in advanced or metastatic (second line or higher) disease with EGFR T790M‐positive NSCLC in the TIGER‐X (NCT01526928) and TIGER‐2 (NCT02147990) studies. Plasma samples were collected at baseline and at the time of systemic progression while receiving rociletinib. The critical exons in 70 genes were sequenced in cfDNA isolated from plasma samples to elucidate a comprehensive genomic profile of alterations for each patient. Results: Plasma‐based cfDNA analysis identified 93% of the initial EGFR activating and 85% of the EGFR T790M resistance mutations in pretreatment samples with detectable tumor DNA. Profiling of progression samples revealed significant heterogeneity, with different variant types (eg, mutations, amplifications, and fusions) detected in multiple genes (EGFR, MET, RB1) that may be driving resistance in patients. Novel alterations not previously described in association with resistance to third‐generation TKIs were also detected, such as an NTRK1 fusion. Conclusion: cfDNA next‐generation sequencing identified initial EGFR activating and secondary T790M resistance mutations in NSCLC patients with high sensitivity, predicted treatment response equivalent to tissue analysis, and identified multiple novel and established resistance alterations.


Cancer Research | 2017

Abstract 1009: Comprehensive ctDNA sequencing reveals mechanisms of resistance to rociletinib in EGFR T790M-mutated NSCLC

Elena Helman; Andrew Simmons; Chris Karlovich; Thomas C. Harding; Mitch Raponi; Darya Chudova; Daniel A. Simon; Richard B. Lanman; AmirAli Talasaz

Background: First and second-generation EGFR tyrosine kinase inhibitors (TKIs) have benefited patients with EGFR-mutated non-small cell lung cancer (NSCLC), but resistance invariably develops after a median of 9-16 months. In ~60% of patients, resistance is mediated by a second mutation in EGFR, namely T790M. Hence, third-generation EGFR TKIs such as osimertinib and rociletinib were developed to target both activating EGFR mutations as well as T790M. Unfortunately, patients also develop resistance to these therapies through mechanisms that have not yet been thoroughly explored. Since repeat tissue biopsies pose potential complications from invasive procedures, circulating tumor DNA (ctDNA) testing is increasingly used in the clinical setting to identify potentially targetable mechanisms of resistance. Methods: Matched pre-treatment and progression plasma from 57 patients with EGFR-mutated NSCLC treated with rociletinib were profiled using a 70-gene ctDNA targeted next-generation sequencing panel (Guardant360) that detects somatic single nucleotide variants, short insertions and deletions, fusions, and copy number variants. Pre-treatment EGFR ctDNA allele fractions were also determined by BEAMing, a technique based on droplet digital PCR followed by flow cytometry. Pre-treatment tumor EGFR status was assessed by the therascreen EGFR test. Results: In all 57 pre-treatment samples profiled, plasma-based ctDNA analysis detected the initial EGFR driver and T790M resistance mutations that were identified in the matched tumor. Interestingly, we found that 12% (7/57) of patients had evidence of compound EGFR driver mutations at baseline, including E709A-L858R, K860I-L858R, and L718V-L858R. EGFR T790M mutations in plasma were observed subclonally (present on average at 40% of the allele fraction of the driver mutation), suggesting tumor heterogeneity at baseline. The correlation coefficients (r) between Guardant360 and BEAMing for EGFR L858R, Exon19Del, and T790M were 0.90, 0.92, 0.95, respectively. Upon progression on rociletinib, 5% of patients (3/57) developed the EGFR C797S resistance mutation, 5% (3/57) developed focal MET amplification, and 2% (1/57) developed a NTRK1 fusion that were not present in the matched baseline plasma. Additionally, 4 deleterious BRCA1/2 alterations (2 germline and 2 somatic) were identified, with the somatic alterations emerging at progression. In 14% (8/57) of the patients, mutations in genes involved in the RAS/RAF signaling pathway, including KRAS Q61H, KRAS K117N and NF1 Q1822*, emerged or increased at progression. Conclusions: Plasma ctDNA revealed heterogeneity and multiple mechanisms of resistance in rociletinib treated patients. Thus comprehensive ctDNA sequencing allows for the identification of potentially actionable alterations and may help inform the choice of next therapy for patients progressing on a third-generation EGFR TKI. Citation Format: Elena Helman, Andrew D. Simmons, Chris A. Karlovich, Thomas C. Harding, Mitch Raponi, Darya I. Chudova, Daniel A. Simon, Richard B. Lanman, AmirAli Talasaz. Comprehensive ctDNA sequencing reveals mechanisms of resistance to rociletinib in EGFR T790M-mutated NSCLC [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1009. doi:10.1158/1538-7445.AM2017-1009


Cancer Research | 2017

Abstract LB-039: Translational research in a phase I proof-of-concept study supports that DCC-2618 is a pan-KIT inhibitor

Filip Janku; Albi Razak; Michael S. Gordon; David C. Brooks; Daniel C. Flynn; Anu Gupta; Michael Kaufman; Cynthia B. Leary; Bryan Smith; Deb Westwood; Neeta Somaiah; Elena Helman; Eric Gerstenberger; Oliver Rosen; Suzanne George

Background: DCC-2618 is a potent switch control inhibitor of KIT and PDGFRα kinases maintains potent inhibition of mutant forms across all exon regions in preclinical models. Gastrointestinal stromal tumor (GIST) is an important disease to achieve a proof-of-concept due to the heterogeneity of resistance mutations in KIT which emerge on treatment with approved KIT inhibitors. In later lines of therapy resistance mechanisms independent of the KIT gene have also been described. Methods: The ongoing phase 1, PK-guided dose escalation study of DCC-2618 given orally BID [28-day cycle] tested doses from 20 mg to 200 mg in patients (pts) with advanced solid tumors including GIST (NCT02571036). We report preliminary longitudinal results of plasma cell free (cf) DNA sequencing by Guardant 360 collected throughout the study and levels of circulating tumor cells (CTCs) based on a viral telomerase promoter-driven GFP expression assay. Results: To date, 24 out of 31 enrolled pts had metastatic KITm GIST refractory to standard therapy. A high, total mean exposure of DCC-2618 and its active metabolite was achieved at 100 and 150 mg BID, affording steady state Cmax >5 µM in Cycle 1. Starting with 50 mg BID dose level, concentrations of total drug exceeding IC90 of the most resistant mutations to DCC-2618 were achieved. Next-generation sequencing of plasma cfDNA revealed a total of 40 KIT mutations in 16 of 18 GIST pts at baseline. DCC-2618 led to rapid decrease and/or clearance of the heterogeneous array of KIT mutations from plasma cfDNA including exons 9, 11, 13, 14, 17, and 18. Independent of suppressed KIT mutation burden, longitudinal monitoring of cfDNA revealed changes in non-KIT oncogenic mutations which may contribute to heterogenous mechanisms of resistance. KIT mutation burden will be correlated with metabolic response assessment by PET scans and exposure to DCC-2618. CTCs have been detected in blood from all GIST patients at baseline using a non-biased assay capable of identifying sarcoma cells. Preliminary result show that CTCs with immunofluorescent detection of KIT or PDGFRα, corresponding to their respective mutational status, show 1 of 3 patterns when compared to radiologic response: most pts show relatively stable low levels at stable disease (SD), a minority of pts with prolonged SD a decline in CTCs and 2 pts with progressive disease had significant increase in KIT positive CTCs. Conclusions: DCC-2618 and its active metabolite achieved high plasma concentrations sufficient to inhibit the most resistant KIT mutations at well-tolerated exposures. Monitoring of cfDNA KIT mutation frequency demonstrates rapid clearance of a broad spectrum of KIT mutations in this heavily pretreated GIST patient population and suggests candidate resistance genes that are independent of KIT. Our data provide a first signal that CTC monitoring might represent a potential marker for tumor control in KIT mutant GIST. Citation Format: Filip Janku, Albi Razak, Michael Gordon, David Brooks, Daniel Flynn, Anu Gupta, Michael Kaufman, Cynthia Leary, Bryan Smith, Deb Westwood, Neeta Somaiah, Elena Helman, Eric Gerstenberger, Oliver Rosen, Suzanne George. Translational research in a phase I proof-of-concept study supports that DCC-2618 is a pan-KIT inhibitor [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr LB-039. doi:10.1158/1538-7445.AM2017-LB-039

Collaboration


Dive into the Elena Helman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Deanna M. Church

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John West

University of Edinburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthony W. Tolcher

University of Texas Health Science Center at San Antonio

View shared research outputs
Researchain Logo
Decentralizing Knowledge