Network


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

Hotspot


Dive into the research topics where Hailun Wang is active.

Publication


Featured researches published by Hailun Wang.


Annals of Oncology | 2014

Prognostic value of CpG island methylator phenotype among colorectal cancer patients: a systematic review and meta-analysis

Y. Y. Juo; Fabian M. Johnston; D. Y. Zhang; H. H. Juo; Hailun Wang; Emmanouil Pappou; T. Yu; Hariharan Easwaran; Stephen B. Baylin; M. van Engeland; Nita Ahuja

BACKGROUND Divergent findings regarding the prognostic value of CpG island methylator phenotype (CIMP) in colorectal cancer (CRC) patients exist in current literature. We aim to review data from published studies in order to examine the association between CIMP and CRC prognosis. MATERIALS AND METHODS A comprehensive search for studies reporting disease-free survival (DFS), overall survival (OS), or cancer-specific mortality of CRC patients stratified by CIMP is carried out. Study findings are summarized descriptively and quantitatively, using adjusted hazard ratios (HRs) as summary statistics. RESULTS Thirty-three studies reporting survival in 10 635 patients are included for review. Nineteen studies provide data suitable for meta-analysis. The definition of CIMP regarding gene panel, marker threshold, and laboratory method varies across studies. Pooled analysis shows that CIMP is significantly associated with shorter DFS (pooled HR estimate 1.45; 95% confidence interval (CI) 1.07-1.97, Q = 3.95, I(2) = 0%) and OS (pooled HR estimate 1.43; 95% CI 1.18-1.73, Q = 4.03, I(2) = 0%) among CRC patients irrespective of microsatellite instability (MSI) status. Subgroup analysis of microsatellite stable (MSS) CRC patients also shows significant association between shorter OS (pooled HR estimate 1.37; 95% CI 1.12-1.68, Q = 4.45, I(2) = 33%) and CIMP. Seven studies have explored CIMPs value as a predictive factor on stage II and III CRC patients DFS after receiving adjuvant 5-fluorouracil (5-FU) therapy: of these, four studies showed that adjuvant chemotherapy conferred a DFS benefit among CIMP(+) patients, one concluded to the contrary, and two found no significant correlation. Insufficient data was present for statistical synthesis of CIMPs predictive value among CRC patients receiving adjuvant 5-FU therapy. CONCLUSION CIMP is independently associated with significantly worse prognosis in CRC patients. However, CIMPs value as a predictive factor in assessing whether adjuvant 5-FU therapy will confer additional survival benefit to CRC patients remained to be determined through future prospective randomized studies.


Neoplasia | 2015

Structure-Function Studies of the bHLH Phosphorylation Domain of TWIST1 in Prostate Cancer Cells

Rajendra P. Gajula; Sivarajan T. Chettiar; Russell Williams; Katriana Nugent; Yoshinori Kato; Hailun Wang; Reem Malek; Kekoa Taparra; Jessica Cades; Anvesh Annadanam; A-Rum Yoon; Elana Fertig; Beth A. Firulli; Lucia Mazzacurati; Timothy F. Burns; Anthony B. Firulli; Steven S. An; Phuoc T. Tran

The TWIST1 gene has diverse roles in development and pathologic diseases such as cancer. TWIST1 is a dimeric basic helix-loop-helix (bHLH) transcription factor existing as TWIST1-TWIST1 or TWIST1-E12/47. TWIST1 partner choice and DNA binding can be influenced during development by phosphorylation of Thr125 and Ser127 of the Thr-Gln-Ser (TQS) motif within the bHLH of TWIST1. The significance of these TWIST1 phosphorylation sites for metastasis is unknown. We created stable isogenic prostate cancer cell lines overexpressing TWIST1 wild-type, phospho-mutants, and tethered versions. We assessed these isogenic lines using assays that mimic stages of cancer metastasis. In vitro assays suggested the phospho-mimetic Twist1-DQD mutation could confer cellular properties associated with pro-metastatic behavior. The hypo-phosphorylation mimic Twist1-AQA mutation displayed reduced pro-metastatic activity compared to wild-type TWIST1 in vitro, suggesting that phosphorylation of the TWIST1 TQS motif was necessary for pro-metastatic functions. In vivo analysis demonstrates that the Twist1-AQA mutation exhibits reduced capacity to contribute to metastasis, whereas the expression of the Twist1-DQD mutation exhibits proficient metastatic potential. Tethered TWIST1-E12 heterodimers phenocopied the Twist1-DQD mutation for many in vitro assays, suggesting that TWIST1 phosphorylation may result in heterodimerization in prostate cancer cells. Lastly, the dual phosphatidylinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) inhibitor BEZ235 strongly attenuated TWIST1-induced migration that was dependent on the TQS motif. TWIST1 TQS phosphorylation state determines the intensity of TWIST1-induced pro-metastatic ability in prostate cancer cells, which may be partly explained mechanistically by TWIST1 dimeric partner choice.


Cancer Research | 2017

TWIST1-WDR5-Hottip regulates Hoxa9 chromatin to facilitate prostate cancer metastasis.

Reem Malek; Rajendra P. Gajula; Russell Williams; Belinda Nghiem; Brian W. Simons; Katriana Nugent; Hailun Wang; Kekoa Taparra; Ghali Lemtiri-Chlieh; A-Rum Yoon; Lawrence D. True; Steven S. An; Theodore L. DeWeese; Ashley E. Ross; Edward M. Schaeffer; Kenneth J. Pienta; Paula J. Hurley; Colm Morrissey; Phuoc T. Tran

TWIST1 is a transcription factor critical for development that can promote prostate cancer metastasis. During embryonic development, TWIST1 and HOXA9 are coexpressed in mouse prostate and then silenced postnatally. Here we report that TWIST1 and HOXA9 coexpression are reactivated in mouse and human primary prostate tumors and are further enriched in human metastases, correlating with survival. TWIST1 formed a complex with WDR5 and the lncRNA Hottip/HOTTIP, members of the MLL/COMPASS-like H3K4 methylases, which regulate chromatin in the Hox/HOX cluster during development. TWIST1 overexpression led to coenrichment of TWIST1 and WDR5 as well as increased H3K4me3 chromatin at the Hoxa9/HOXA9 promoter, which was dependent on WDR5. Expression of WDR5 and Hottip/HOTTIP was also required for TWIST1-induced upregulation of HOXA9 and aggressive cellular phenotypes such as invasion and migration. Pharmacologic inhibition of HOXA9 prevented TWIST1-induced aggressive prostate cancer cellular phenotypes in vitro and metastasis in vivo This study demonstrates a novel mechanism by which TWIST1 regulates chromatin and gene expression by cooperating with the COMPASS-like complex to increase H3K4 trimethylation at target gene promoters. Our findings highlight a TWIST1-HOXA9 embryonic prostate developmental program that is reactivated during prostate cancer metastasis and is therapeutically targetable. Cancer Res; 77(12); 3181-93. ©2017 AACR.


Cells Tissues Organs | 2017

Therapeutic Targeting of Epithelial Plasticity Programs: Focus on the Epithelial-Mesenchymal Transition

Reem Malek; Hailun Wang; Kekoa Taparra; Phuoc T. Tran

Mounting data points to epithelial plasticity programs such as the epithelial-mesenchymal transition (EMT) as clinically relevant therapeutic targets for the treatment of malignant tumors. In addition to the widely realized role of EMT in increasing cancer cell invasiveness during cancer metastasis, the EMT has also been implicated in allowing cancer cells to avoid tumor suppressor pathways during early tumorigenesis. In addition, data linking EMT to innate and acquired treatment resistance further points towards the desire to develop pharmacological therapies to target epithelial plasticity in cancer. In this review we organized our discussion on pathways and agents that can be used to target the EMT in cancer into 3 groups: (1) extracellular inducers of EMT, (2) the transcription factors that orchestrate the EMT transcriptome, and (3) the downstream effectors of EMT. We highlight only briefly specific canonical pathways known to be involved in EMT, such as the signal transduction pathways TGFβ, EFGR, and Axl-Gas6. We emphasize in more detail pathways that we believe are emerging novel pathways and therapeutic targets such as epigenetic therapies, glycosylation pathways, and immunotherapy. The heterogeneity of tumors and the dynamic nature of epithelial plasticity in cancer cells make it likely that targeting only 1 EMT-related process will be unsuccessful or only transiently successful. We suggest that with greater understanding of epithelial plasticity regulation, such as with the EMT, a more systematic targeting of multiple EMT regulatory networks will be the best path forward to improve cancer outcomes.


Molecular Cancer Research | 2017

A First-in-Class TWIST1 Inhibitor with Activity in Oncogene-driven Lung Cancer

Zachary A. Yochum; Jessica Cades; Lucia Mazzacurati; Neil M. Neumann; Susheel Khetarpal; Suman Chatterjee; Hailun Wang; Myriam Attar; Eric H.-B. Huang; Sarah N. Chatley; Katriana Nugent; Ashwin Somasundaram; Johnathan A. Engh; Andrew J. Ewald; Yoon Jae Cho; Charles M. Rudin; Phuoc T. Tran; Timothy F. Burns

TWIST1, an epithelial–mesenchymal transition (EMT) transcription factor, is critical for oncogene-driven non–small cell lung cancer (NSCLC) tumorigenesis. Given the potential of TWIST1 as a therapeutic target, a chemical–bioinformatic approach using connectivity mapping (CMAP) analysis was used to identify TWIST1 inhibitors. Characterization of the top ranked candidates from the unbiased screen revealed that harmine, a harmala alkaloid, inhibited multiple TWIST1 functions, including single-cell dissemination, suppression of normal branching in 3D epithelial culture, and proliferation of oncogene driver-defined NSCLC cells. Harmine treatment phenocopied genetic loss of TWIST1 by inducing oncogene-induced senescence or apoptosis. Mechanistic investigation revealed that harmine targeted the TWIST1 pathway through its promotion of TWIST1 protein degradation. As dimerization is critical for TWIST1 function and stability, the effect of harmine on specific TWIST1 dimers was examined. TWIST1 and its dimer partners, the E2A proteins, which were found to be required for TWIST1-mediated functions, regulated the stability of the other heterodimeric partner posttranslationally. Harmine preferentially promoted degradation of the TWIST1-E2A heterodimer compared with the TWIST-TWIST1 homodimer, and targeting the TWIST1-E2A heterodimer was required for harmine cytotoxicity. Finally, harmine had activity in both transgenic and patient-derived xenograft mouse models of KRAS-mutant NSCLC. These studies identified harmine as a first-in-class TWIST1 inhibitor with marked anti-tumor activity in oncogene-driven NSCLC including EGFR mutant, KRAS mutant and MET altered NSCLC. Implications: TWIST1 is required for oncogene-driven NSCLC tumorigenesis and EMT; thus, harmine and its analogues/derivatives represent a novel therapeutic strategy to treat oncogene-driven NSCLC as well as other solid tumor malignancies. Mol Cancer Res; 15(12); 1764–76. ©2017 AACR.


Oncogene | 2018

Targeting the EMT transcription factor TWIST1 overcomes resistance to EGFR inhibitors in EGFR- mutant non-small-cell lung cancer

Zachary A. Yochum; Jessica Cades; Hailun Wang; Suman Chatterjee; Brian W. Simons; James P. O’Brien; Susheel Khetarpal; Ghali Lemtiri-Chlieh; Kayla V. Myers; Eric H.-B. Huang; Charles M. Rudin; Phuoc T. Tran; Timothy F. Burns

Patients with EGFR-mutant non-small-cell lung cancer (NSCLC) have significantly benefited from the use of EGFR tyrosine kinase inhibitors (TKIs). However, long-term efficacy of these therapies is limited due to de novo resistance (~30%) as well as acquired resistance. Epithelial–mesenchymal transition transcription factors (EMT-TFs), have been identified as drivers of EMT-mediated resistance to EGFR TKIs, however, strategies to target EMT-TFs are lacking. As the third generation EGFR TKI, osimertinib, has now been adopted in the first-line setting, the frequency of T790M mutations will significantly decrease in the acquired resistance setting. Previously less common mechanisms of acquired resistance to first generation EGFR TKIs including EMT are now being observed at an increased frequency after osimertinib. Importantly, there are no other FDA approved targeted therapies after progression on osimertinib. Here, we investigated a novel strategy to overcome EGFR TKI resistance through targeting the EMT-TF, TWIST1, in EGFR-mutant NSCLC. We demonstrated that genetic silencing of TWIST1 or treatment with the TWIST1 inhibitor, harmine, resulted in growth inhibition and apoptosis in EGFR-mutant NSCLC. TWIST1 overexpression resulted in erlotinib and osimertinib resistance in EGFR-mutant NSCLC cells. Conversely, genetic and pharmacological inhibition of TWIST1 in EGFR TKI-resistant EGFR-mutant cells increased sensitivity to EGFR TKIs. TWIST1-mediated EGFR TKI resistance was due in part to TWIST1 suppression of transcription of the pro-apoptotic BH3-only gene, BCL2L11 (BIM), by directly binding to BCL2L11 intronic regions and promoter. As such, pan-BCL2 inhibitor treatment overcame TWIST1-mediated EGFR TKI resistance and were more effective in the setting of TWIST1 overexpression. Finally, in a mouse model of autochthonous EGFR-mutant lung cancer, Twist1 overexpression resulted in erlotinib resistance and suppression of erlotinib-induced apoptosis. These studies establish TWIST1 as a driver of resistance to EGFR TKIs and provide rationale for use of TWIST1 inhibitors or BCL2 inhibitors as means to overcome EMT-mediated resistance to EGFR TKIs.


Journal of Clinical Investigation | 2018

O-GlcNAcylation is required for mutant KRAS -induced lung tumorigenesis

Kekoa Taparra; Hailun Wang; Reem Malek; Audrey Lafargue; Mustafa A. Barbhuiya; Xing Wang; Brian W. Simons; Matthew Ballew; Katriana Nugent; Jennifer Groves; Russell Williams; Takumi Shiraishi; James E. Verdone; Gokben Yildirir; Roger Henry; Bin Zhang; John Wong; Ken Kang-Hsin Wang; Barry D. Nelkin; Kenneth J. Pienta; Dean W. Felsher; Natasha E. Zachara; Phuoc T. Tran

Mutant KRAS drives glycolytic flux in lung cancer, potentially impacting aberrant protein glycosylation. Recent evidence suggests aberrant KRAS drives flux of glucose into the hexosamine biosynthetic pathway (HBP). HBP is required for various glycosylation processes, such as protein N- or O-glycosylation and glycolipid synthesis. However, its function during tumorigenesis is poorly understood. One contributor and proposed target of KRAS-driven cancers is a developmentally conserved epithelial plasticity program called epithelial-mesenchymal transition (EMT). Here we showed in novel autochthonous mouse models that EMT accelerated KrasG12D lung tumorigenesis by upregulating expression of key enzymes of the HBP pathway. We demonstrated that HBP was required for suppressing KrasG12D-induced senescence, and targeting HBP significantly delayed KrasG12D lung tumorigenesis. To explore the mechanism, we investigated protein glycosylation downstream of HBP and found elevated levels of O-linked &bgr;-N-acetylglucosamine (O-GlcNAcylation) posttranslational modification on intracellular proteins. O-GlcNAcylation suppressed KrasG12D oncogene-induced senescence (OIS) and accelerated lung tumorigenesis. Conversely, loss of O-GlcNAcylation delayed lung tumorigenesis. O-GlcNAcylation of proteins SNAI1 and c-MYC correlated with the EMT-HBP axis and accelerated lung tumorigenesis. Our results demonstrated that O-GlcNAcylation was sufficient and required to accelerate KrasG12D lung tumorigenesis in vivo, which was reinforced by epithelial plasticity programs.


Clinical Cancer Research | 2018

Abstract B12: Centrosome clustering inhibition as a novel strategy to sensitize non-small cell lung cancer to radiation treatment and immunotherapy

Hailun Wang; Katriana Nugent; Matthew Ballew; Ghali Lemtiri-Chlieh; Natasha Raman; Michelle S. Levine; Andrew J. Holland; Phuoc T. Tran

The centrosome is a microtubule-organizing center that plays an important role during M-phase and cell division where it organizes the two poles of the bipolar microtubule spindle apparatus from which chromosomes are segregated. In contrast to most normal cells, the vast majority of lung cancers contain extra copies of centrosomes, and a large body of circumstantial evidence links extra centrosomes to the development of cancer. Cells with supernumerary centrosomes form multipolar mitotic spindles, which, if not corrected, lead to lethal multipolar divisions or “mitotic catastrophe.” To overcome this, lung cancer cells cluster their centrosomes into two spindle poles, enabling tumor cells to survive. Thus, inhibition of centrosome clustering (CCi) can force cancer cells into multipolar divisions, lead to chromosomal instability, make cancer cells more sensitive to radiation treatment, and eventually result in the selective killing of cancer cells. At the same time, this approach should spare healthy tissues with normal centrosome numbers, resulting in a favorable therapeutic index. To examine the relationship between CCi and radiation-treatment sensitivity, we first treated three NSCLC cell lines (A549, H460 and H358) and one immortalized primary human bronchial epithelial cell (HBEC) with griseofulvin—a centrosomal clustering inhibitor. Immunofluorescent staining for pericentrin and alpha-tubulin, we observed a significant increase in the number of cells with multipolar spindles in NSCLC cells compared to HBECs (40-80% vs. 15%). Next, we examined the ability of griseofulvin to radiosensitize these cells to radiation treatment. By using clonogenic assays, we saw significantly less clonogenic potential of NSCLC cells following radiation treatment after exposure to 30 uM of griseofulvin, but griseofulvin did not radiosensitize HBECs. To further confirm our results, we genetically targeted KIFC1, a member of kinesin-14 family of motor proteins that have been shown to be essential for clustering centrosomes in cancer cells, but are not required for division in normal cells. In our experiments, knockdown of KIFC1 expression with siRNA also significantly sensitized NSCLC cells to radiation treatment. Finally, when we treated the NSCLC cells with griseofulvin, we saw a significant increase in the formation of micronuclei in cancer cells. Recently, it has been shown that micronuclei are important sources of immunostimulatory DNA via activation of the cGAS-STING pathway. Therefore, inhibition of CC could potentially augment immunogenicity and increase efficiency of immunotherapy (IMT). In conclusion, inhibition of CC resulted in the formation of multipolar spindle in NSCLCs and further sensitized NSCLCs to radiation treatment. CC is not typically required for normal cells, and thus CCi may be a more specific and perhaps less toxic therapy for NSCLCs. The potential of CCi to enhance IMT is under investigation. Citation Format: Hailun Wang, Katriana Nugent, Matthew Ballew, Ghali Lemtiri-Chlieh, Natasha Raman, Michelle Levine, Andrew Holland, Phuoc Tran. Centrosome clustering inhibition as a novel strategy to sensitize non-small cell lung cancer to radiation treatment and immunotherapy [abstract]. In: Proceedings of the Fifth AACR-IASLC International Joint Conference: Lung Cancer Translational Science from the Bench to the Clinic; Jan 8-11, 2018; San Diego, CA. Philadelphia (PA): AACR; Clin Cancer Res 2018;24(17_Suppl):Abstract nr B12.


Cancer Research | 2018

Abstract B47: Modeling epithelial plasticity-induced erlotinib resistance in non-small cell lung cancer

Hailun Wang; Jessica Cades; Zachary A. Yochum; Katriana Nugent; Charles M. Rudin; Timothy F. Burns; Phuoc T. Tran

Background: Advanced non-small lung cancer (NSCLC) patients with EGFR mutations initially respond to treatment with the EGFR-targeted tyrosine kinase inhibitors (TKIs) such as erlotinib, but will invariably acquire resistance with progression of disease within 10–16 months. Mechanisms of EGFR TKIs resistance include second-site EGFR mutations (>50%), MET amplfication (5–10%), and mutations in PIK3CA ( Methods: To examine the relationship between epithelial plasticity and erlotinib resistance in EGFR mutant lung cancers, we created an inducible CCSP-rtTA/tetO-EgfrL858R/Twist1 (CET) transgenic mice (Twist1 is a key regulator of EMT). We utilized the tetracycline-inducible gene expression system to control EgfrL858R/Twist1 gene expression in the lung by providing or withdrawing doxycycline to the mice. The mice were treated for 3 weeks with erlotinib and scanned by CT each week, and survival of the mice was also recorded. The tumor tissues were collected 1 week and 3 weeks after the start of treatment and used for immunohistochemical staining for HE 2017 Sep 24-27; Orlando, Florida. Philadelphia (PA): AACR; Cancer Res 2018;78(10 Suppl):Abstract nr B47.


Cancer Research | 2017

Abstract 4118: The EMT transcription factor TWIST1 mediates resistance to EGFR inhibitors inEGFR-mutant non-small cell lung cancer

Zachary A. Yochum; Hailun Wang; Jessica Cades; Susheel Khetarpal; Eric H.-B. Huang; Phouc T. Tran; Timothy F. Burns

Recent advances in the treatment of non-small cell lung cancer (NSCLC) stem from the paradigm shift of classifying patients into subtypes based upon the presence of distinct molecular drivers. Subsets of patients, such as those with EGFR mutations and ALK translocations, have dramatic responses in their tumors to tyrosine kinase inhibitors (TKIs) that specifically inhibit these oncogenic drivers. While many patients initially response to TKIs, therapeutic resistance is inevitable. For EGFR-mutant NSCLC, there are multiple described mechanisms of resistance to EGFR TKIs, including epithelial-mesenchymal transition (EMT). Previous studies have implicated the AXL kinase and ZEB1, an EMT transcription factor (EMT-TF), in EMT-mediated EGFR TKI resistance. We have previously demonstrated that the EMT-TF, TWIST1, is required for oncogene-driven NSCLC tumorigenesis, including those tumors with EGFR mutations. In this study, we investigated the role of TWIST1 in EMT-mediated resistance to EGFR TKIs. We have demonstrated that genetic or pharmacologic inhibition of TWIST1 resulted in growth inhibition in a panel of EGFR-mutant NSCLC cell lines and apoptosis in a subset of these lines. Interestingly, TWIST1 overexpression in EGFR-mutant NSCLC cell lines led to EGFR TKI resistance. Conversely, knockdown of TWIST1 in an erlotinib resistant EGFR-mutant NSCLC cell line restored erlotinib sensitivity. We found that TWIST1 mediates resistance to EGFR TKIs through suppression of apoptosis possibly through decreasing the expression of the pro-apoptotic Bcl-2 member, BCL2L11 (BIM). We observed that TWIST1 knockdown increased BIM levels, while TWIST1 overexpression decreased BIM expression. Furthermore, TWIST1-mediated resistance was overcome by treatment with the BCL-2/BCL-XL inhibitor, ABT-737. Knockdown of BIM recapitulated the resistance seen following TWIST1 overexpression, suggesting that TWIST1 suppression of BIM is a mechanism through which TWIST1 leads to EGFR TKI resistance. To explore the role of TWIST1 in modulating EGFR inhibitor sensitivity in vivo, we used an inducible EGFR-mutant transgenic mouse model, CCSP-rtTA/tetO-EGFRL858R (CE), which expresses EGFRL858R in the lung and a EGFR-mutant/Twist1 transgenic model, CCSP-rtTA/tetO-EGFRL858R/ Twist1- tetO7-luc (CET), which expresses both Twist1 and EGFRL858R in the lung. CET mice had a significantly increased tumor burden, decreased apoptosis and a decreased overall survival compared to CE mice following erlotinib treatment. In summary, we found that TWIST1 overexpression leads to EGFR TKI resistance by suppressing EGFR TKI-induced apoptosis through suppressing BIM expression. Future studies aim to establish the mechanisms of TWIST1 suppression of BIM expression and determine if our TWIST1 inhibitor, harmine, is effective in overcoming EMT-mediated resistance. Citation Format: Zachary A. Yochum, Hailun Wang, Jessica A. Cades, Susheel Khetarpal, Eric H. Huang, Phouc T. Tran, Timothy F. Burns. The EMT transcription factor TWIST1 mediates resistance to EGFR inhibitors in EGFR-mutant non-small cell lung cancer [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 4118. doi:10.1158/1538-7445.AM2017-4118

Collaboration


Dive into the Hailun Wang's collaboration.

Top Co-Authors

Avatar

Phuoc T. Tran

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kekoa Taparra

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Reem Malek

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Jessica Cades

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian W. Simons

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge