Network


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

Hotspot


Dive into the research topics where Danny Simpson is active.

Publication


Featured researches published by Danny Simpson.


Toxins | 2016

From Mollusks to Medicine: A Venomics Approach for the Discovery and Characterization of Therapeutics from Terebridae Peptide Toxins

Aida Verdes; Prachi Anand; Juliette Gorson; Stephen A. Jannetti; Patrick J. Kelly; Abba Leffler; Danny Simpson; Girish Ramrattan; Mandë Holford

Animal venoms comprise a diversity of peptide toxins that manipulate molecular targets such as ion channels and receptors, making venom peptides attractive candidates for the development of therapeutics to benefit human health. However, identifying bioactive venom peptides remains a significant challenge. In this review we describe our particular venomics strategy for the discovery, characterization, and optimization of Terebridae venom peptides, teretoxins. Our strategy reflects the scientific path from mollusks to medicine in an integrative sequential approach with the following steps: (1) delimitation of venomous Terebridae lineages through taxonomic and phylogenetic analyses; (2) identification and classification of putative teretoxins through omics methodologies, including genomics, transcriptomics, and proteomics; (3) chemical and recombinant synthesis of promising peptide toxins; (4) structural characterization through experimental and computational methods; (5) determination of teretoxin bioactivity and molecular function through biological assays and computational modeling; (6) optimization of peptide toxin affinity and selectivity to molecular target; and (7) development of strategies for effective delivery of venom peptide therapeutics. While our research focuses on terebrids, the venomics approach outlined here can be applied to the discovery and characterization of peptide toxins from any venomous taxa.


Genome Biology and Evolution | 2018

Are Fireworms Venomous? Evidence for the Convergent Evolution of Toxin Homologs in Three Species of Fireworms (Annelida, Amphinomidae)

Aida Verdes; Danny Simpson; Mandë Holford

Abstract Amphinomids, more commonly known as fireworms, are a basal lineage of marine annelids characterized by the presence of defensive dorsal calcareous chaetae, which break off upon contact. It has long been hypothesized that amphinomids are venomous and use the chaetae to inject a toxic substance. However, studies investigating fireworm venom from a morphological or molecular perspective are scarce and no venom gland has been identified to date, nor any toxin characterized at the molecular level. To investigate this question, we analyzed the transcriptomes of three species of fireworms—Eurythoe complanata, Hermodice carunculata, and Paramphinome jeffreysii—following a venomics approach to identify putative venom compounds. Our venomics pipeline involved de novo transcriptome assembly, open reading frame, and signal sequence prediction, followed by three different homology search strategies: BLAST, HMMER sequence, and HMMER domain. Following this pipeline, we identified 34 clusters of orthologous genes, representing 13 known toxin classes that have been repeatedly recruited into animal venoms. Specifically, the three species share a similar toxin profile with C-type lectins, peptidases, metalloproteinases, spider toxins, and CAP proteins found among the most highly expressed toxin homologs. Despite their great diversity, the putative toxins identified are predominantly involved in three major biological processes: hemostasis, inflammatory response, and allergic reactions, all of which are commonly disrupted after fireworm stings. Although the putative fireworm toxins identified here need to be further validated, our results strongly suggest that fireworms are venomous animals that use a complex mixture of toxins for defense against predators.


Journal of Translational Medicine | 2018

Baseline antibody profiles predict toxicity in melanoma patients treated with immune checkpoint inhibitors

Michael Gowen; Keith M. Giles; Danny Simpson; Jeremy Tchack; Hua Zhou; Una Moran; Zarmeena Dawood; Anna C. Pavlick; Shaohui Hu; Melissa Wilson; Hua Zhong; Michelle Krogsgaard; Tomas Kirchhoff; Iman Osman

BackgroundImmune checkpoint inhibitors (anti-CTLA-4, anti-PD-1, or the combination) enhance anti-tumor immune responses, yielding durable clinical benefit in several cancer types, including melanoma. However, a subset of patients experience immune-related adverse events (irAEs), which can be severe and result in treatment termination. To date, no biomarker exists that can predict development of irAEs.MethodsWe hypothesized that pre-treatment antibody profiles identify a subset of patients who possess a sub-clinical autoimmune phenotype that predisposes them to develop severe irAEs following immune system disinhibition. Using a HuProt human proteome array, we profiled baseline antibody levels in sera from melanoma patients treated with anti-CTLA-4, anti-PD-1, or the combination, and used support vector machine models to identify pre-treatment antibody signatures that predict irAE development.ResultsWe identified distinct pre-treatment serum antibody profiles associated with severe irAEs for each therapy group. Support vector machine classifier models identified antibody signatures that could effectively discriminate between toxicity groups with > 90% accuracy, sensitivity, and specificity. Pathway analyses revealed significant enrichment of antibody targets associated with immunity/autoimmunity, including TNFα signaling, toll-like receptor signaling and microRNA biogenesis.ConclusionsOur results provide the first evidence supporting a predisposition to develop severe irAEs upon immune system disinhibition, which requires further independent validation in a clinical trial setting.


PLOS ONE | 2017

Apolipoprotein L1 risk variants associate with prevalent atherosclerotic disease in African American systemic lupus erythematosus patients

Ashira D. Blazer; Binhuan Wang; Danny Simpson; Tomas Kirchhoff; Sean Heffron; Robert R. Clancy; Adriana Heguy; Karina Ray; Matija Snuderl; Jill P. Buyon

Objective Atherosclerosis is exaggerated in African American (AA) systemic lupus erythematosus (SLE) patients, with doubled cardiovascular disease (CVD) risk compared to White patients. The extent to which common Apolipoprotein L1 (APOL1) risk alleles (RA) contribute to this trend is unknown. This retrospective cohort study assessed prevalent atherosclerotic disease across APOL1 genotypes in AA SLE patients. Methods One hundred thirteen AA SLE subjects were APOL1-genotyped and stratified as having: zero risk alleles, one risk allele, or two risk alleles. Chart review assessed CVD manifestations including abdominal aortic aneurysm, angina, carotid artery disease, coronary artery disease, myocardial infarction, peripheral vascular disease, stroke, and vascular calcifications. Associations between the genotypes and a composite endpoint defined as one or more CVD manifestations were calculated using logistic regression. Symptomatic atherosclerotic disease, excluding incidental vascular calcifications, was also assessed. Results The 0-risk-allele, 1-risk-allele and 2-risk-allele groups, respectively, comprised 34%, 53%, and 13% of the cohort. Respectively, 13.2%, 41.7%, and 60.0% of the 0-risk allele, 1-risk-allele, and 2-risk-allele groups met the composite endpoint of atherosclerotic CVD (p = 0.001). Adjusting for risk factors–including smoking, ESRD, BMI >25 and hypertension–we observed an association between carrying one or more RA and atherosclerotic CVD (OR = 7.1; p = 0.002). For symptomatic disease, the OR was 3.5 (p = 0.02). In a time-to-event analysis, the proportion of subjects free from the composite primary endpoint, symptomatic atherosclerotic CVD, was higher in the 0-risk-allele group compared to the 1-risk-allele and 2-risk-allele groups (χ2 = 6.5; p = 0.04). Conclusions Taken together, the APOL1 RAs associate with prevalent atherosclerotic CVD in this cohort of AA SLE patients, perhaps reflecting a potentiating effect of SLE on APOL1-related cardiovascular phenotypes.


Journal of the National Cancer Institute | 2018

Primary Melanoma Histologic Subtype: Impact on Survival and Response to Therapy

Michael Lattanzi; Yesung Lee; Danny Simpson; Una Moran; Farbod Darvishian; Randie H Kim; Eva Hernando; David Polsky; Doug Hanniford; Richard L. Shapiro; Russell S. Berman; Anna C. Pavlick; Melissa Wilson; Tomas Kirchhoff; Jeffrey S. Weber; Judy Zhong; Iman Osman

BACKGROUND Two primary histologic subtypes, superficial spreading melanoma (SSM) and nodular melanoma (NM), comprise the majority of all cutaneous melanomas. NM is associated with worse outcomes, which have been attributed to increased thickness at presentation, and it is widely expected that NM and SSM would exhibit similar behavior once metastasized. Herein, we tested the hypothesis that primary histologic subtype is an independent predictor of survival and may impact response to treatment in the metastatic setting. METHODS We examined the most recent Surveillance, Epidemiology, and End Results (SEER) cohort (n = 118 508) and the New York University (NYU) cohort (n = 1621) with available protocol-driven follow-up. Outcomes specified by primary histology were studied in both the primary and metastatic settings with respect to BRAF-targeted therapy and immunotherapy. We characterized known driver mutations and examined a 140-gene panel in a subset of NM and SSM cases using next-generation sequencing. All statistical tests were two-sided. RESULTS NM was an independent risk factor for death in both the SEER (hazard ratio [HR] = 1.55, 95% confidence interval [CI] = 1.41 to 1.70, P < .001) and NYU (HR = 1.47, 95% CI = 1.05, 2.07, P = .03) cohorts, controlling for thickness, ulceration, stage, and other variables. In the metastatic setting, NM remained an independent risk factor for death upon treatment with BRAF-targeted therapy (HR = 3.33, 95% CI = 1.06 to 10.47, P = .04) but showed no statistically significant difference with immune checkpoint inhibition. NM was associated with a higher rate of NRAS mutation (P < .001), and high-throughput sequencing revealed NM-specific genomic alterations in NOTCH4, ANK3, and ZNF560, which were independently validated. CONCLUSIONS Our data reveal distinct clinical and biological differences between NM and SSM that support revisiting the prognostic and predictive impact of primary histology subtype in the management of cutaneous melanoma.


Journal of Clinical Oncology | 2017

Mutation burden as a potential prognostic marker of melanoma progression and survival.

Danny Simpson; Robert Ferguson; Carlos N Martinez; Esther Kazlow; Una Moran; Adriana Heguy; Douglas Hanniford; Eva Hernando; Iman Osman; Tomas Kirchhoff


Journal of Clinical Oncology | 2018

Autoimmune genetic variants as germline biomarkers of response in melanoma immunotherapy treatment.

Vylyny Chat; Robert Ferguson; Danny Simpson; Esther Kazlow; Rebecca Lax; Una Moran; Melissa Wilson; Anna C. Pavlick; Ryan J. Sullivan; Keith T. Flaherty; Iman Osman; Jeffrey S. Weber; Tomas Kirchhoff


Journal of Clinical Oncology | 2018

Mutation burden in conjunction with MAPK-pathway mutation status as a prognostic biomarker of overall melanoma survival.

John Cadley; Danny Simpson; Robert Ferguson; Adur Pandya; Taymor Hekal; Taj Richards; Kareem Ibrahim; Jeffrey S. Weber; Iman Osman; Tomas Kirchhoff


Annals of Oncology | 2018

71PImmunomodulatory germline variation impacts the development of multiple primary melanoma (MPM)

Robert Ferguson; A Archambault; Danny Simpson; Esther Kazlow; R Lax; Una Moran; Melissa Wilson; Richard L. Shapiro; Anna C. Pavlick; Iman Osman; David Polsky; Tomas Kirchhoff


Annals of Oncology | 2018

1182PAnti-CTLA4 toxicity associates with genetic variation correlating with serum antibody diversity

Danny Simpson; Robert Ferguson; Michael Gowen; Keith M. Giles; Jeremy Tchack; Hua Zhou; Una Moran; Zarmeena Dawood; Anna C. Pavlick; S Hu; Melissa Wilson; Hua Zhong; Michelle Krogsgaard; Jeffrey S. Weber; Iman Osman; Tomas Kirchhoff

Collaboration


Dive into the Danny Simpson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge