Network


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

Hotspot


Dive into the research topics where Crystal McKnight is active.

Publication


Featured researches published by Crystal McKnight.


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

High-throughput combinatorial screening identifies drugs that cooperate with ibrutinib to kill activated B-cell-like diffuse large B-cell lymphoma cells.

Lesley A. Mathews Griner; Rajarshi Guha; Paul Shinn; Ryan M. Young; Jonathan M. Keller; Dongbo Liu; Ian S. Goldlust; Adam Yasgar; Crystal McKnight; Matthew B. Boxer; Damien Y. Duveau; Jian-kang Jiang; Sam Michael; Tim Mierzwa; Wenwei Huang; Martin J. Walsh; Bryan T. Mott; Paresma R. Patel; William Leister; David J. Maloney; Christopher A. LeClair; Ganesha Rai; Ajit Jadhav; Brian D. Peyser; Christopher P. Austin; Scott E. Martin; Anton Simeonov; Marc Ferrer; Louis M. Staudt; Craig J. Thomas

Significance The treatment of cancer is highly reliant on drug combinations. Next-generation, targeted therapeutics are demonstrating interesting single-agent activities in clinical trials; however, the discovery of companion drugs through iterative clinical trial-and-error is not a tenable mechanism to prioritize clinically important combinations for these agents. Herein we describe the results of a large, high-throughput combination screen of the Bruton’s tyrosine kinase inhibitor ibrutinib versus a library of nearly 500 approved and investigational drugs. Multiple ibrutinib combinations were discovered through this study that can be prioritized for clinical examination. The clinical development of drug combinations is typically achieved through trial-and-error or via insight gained through a detailed molecular understanding of dysregulated signaling pathways in a specific cancer type. Unbiased small-molecule combination (matrix) screening represents a high-throughput means to explore hundreds and even thousands of drug–drug pairs for potential investigation and translation. Here, we describe a high-throughput screening platform capable of testing compounds in pairwise matrix blocks for the rapid and systematic identification of synergistic, additive, and antagonistic drug combinations. We use this platform to define potential therapeutic combinations for the activated B-cell–like subtype (ABC) of diffuse large B-cell lymphoma (DLBCL). We identify drugs with synergy, additivity, and antagonism with the Bruton’s tyrosine kinase inhibitor ibrutinib, which targets the chronic active B-cell receptor signaling that characterizes ABC DLBCL. Ibrutinib interacted favorably with a wide range of compounds, including inhibitors of the PI3K-AKT-mammalian target of rapamycin signaling cascade, other B-cell receptor pathway inhibitors, Bcl-2 family inhibitors, and several components of chemotherapy that is the standard of care for DLBCL.


Scientific Reports | 2015

High-throughput matrix screening identifies synergistic and antagonistic antimalarial drug combinations

Bryan T. Mott; Richard T. Eastman; Rajarshi Guha; Katy S. Sherlach; Amila Siriwardana; Paul Shinn; Crystal McKnight; Sam Michael; Norinne Lacerda-Queiroz; Paresma Patel; Pwint Khine; Hongmao Sun; Monica Kasbekar; Nima Aghdam; Shaun D. Fontaine; Dongbo Liu; Tim Mierzwa; Lesley Mathews-Griner; Marc Ferrer; Adam R. Renslo; James Inglese; Jing Yuan; Paul D. Roepe; Xin-Zhuan Su; Craig J. Thomas

Drug resistance in Plasmodium parasites is a constant threat. Novel therapeutics, especially new drug combinations, must be identified at a faster rate. In response to the urgent need for new antimalarial drug combinations we screened a large collection of approved and investigational drugs, tested 13,910 drug pairs, and identified many promising antimalarial drug combinations. The activity of known antimalarial drug regimens was confirmed and a myriad of new classes of positively interacting drug pairings were discovered. Network and clustering analyses reinforced established mechanistic relationships for known drug combinations and identified several novel mechanistic hypotheses. From eleven screens comprising >4,600 combinations per parasite strain (including duplicates) we further investigated interactions between approved antimalarials, calcium homeostasis modulators, and inhibitors of phosphatidylinositide 3-kinases (PI3K) and the mammalian target of rapamycin (mTOR). These studies highlight important targets and pathways and provide promising leads for clinically actionable antimalarial therapy.


Journal of Biological Chemistry | 2016

Small Molecule Inhibition of the Ubiquitin-specific Protease USP2 Accelerates cyclin D1 Degradation and Leads to Cell Cycle Arrest in Colorectal Cancer and Mantle Cell Lymphoma Models

Mindy I. Davis; Rajan Pragani; Jennifer T. Fox; Min Shen; Kalindi Parmar; Emily Gaudiano; Li Liu; Cordelle Tanega; Lauren McGee; Matthew D. Hall; Crystal McKnight; Paul Shinn; Henrike Nelson; Debasish Chattopadhyay; Alan D. D'Andrea; Douglas S. Auld; Larry DeLucas; Zhuyin Li; Matthew B. Boxer; Anton Simeonov

Deubiquitinases are important components of the protein degradation regulatory network. We report the discovery of ML364, a small molecule inhibitor of the deubiquitinase USP2 and its use to interrogate the biology of USP2 and its putative substrate cyclin D1. ML364 has an IC50 of 1.1 μm in a biochemical assay using an internally quenched fluorescent di-ubiquitin substrate. Direct binding of ML364 to USP2 was demonstrated using microscale thermophoresis. ML364 induced an increase in cellular cyclin D1 degradation and caused cell cycle arrest as shown in Western blottings and flow cytometry assays utilizing both Mino and HCT116 cancer cell lines. ML364, and not the inactive analog 2, was antiproliferative in cancer cell lines. Consistent with the role of cyclin D1 in DNA damage response, ML364 also caused a decrease in homologous recombination-mediated DNA repair. These effects by a small molecule inhibitor support a key role for USP2 as a regulator of cell cycle, DNA repair, and tumor cell growth.


PLOS Neglected Tropical Diseases | 2017

The phosphatidylinositol-3-phosphate 5-kinase inhibitor apilimod blocks filoviral entry and infection

Elizabeth A. Nelson; Julie Dyall; Thomas Hoenen; Alyson B. Barnes; Huanying Zhou; Janie Y. Liang; Julia Michelotti; William H. Dewey; Lisa Evans DeWald; Richard S. Bennett; Patrick J. Morris; Rajarshi Guha; Carleen Klumpp-Thomas; Crystal McKnight; Yu-Chi Chen; Xin Xu; Amy Wang; Emma Hughes; Scott E. Martin; Craig J. Thomas; Peter B. Jahrling; Lisa E. Hensley; Gene G. Olinger; Judith M. White

Phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) is a lipid kinase involved in endosome maturation that emerged from a haploid genetic screen as being required for Ebola virus (EBOV) infection. Here we analyzed the effects of apilimod, a PIKfyve inhibitor that was reported to be well tolerated in humans in phase 2 clinical trials, for its effects on entry and infection of EBOV and Marburg virus (MARV). We first found that apilimod blocks infections by EBOV and MARV in Huh 7, Vero E6 and primary human macrophage cells, with notable potency in the macrophages (IC50, 10 nM). We next observed that similar doses of apilimod block EBOV-glycoprotein-virus like particle (VLP) entry and transcription-replication competent VLP infection, suggesting that the primary mode of action of apilimod is as an entry inhibitor, preventing release of the viral genome into the cytoplasm to initiate replication. After providing evidence that the anti-EBOV action of apilimod is via PIKfyve, we showed that it blocks trafficking of EBOV VLPs to endolysosomes containing Niemann-Pick C1 (NPC1), the intracellular receptor for EBOV. Concurrently apilimod caused VLPs to accumulate in early endosome antigen 1-positive endosomes. We did not detect any effects of apilimod on bulk endosome acidification, on the activity of cathepsins B and L, or on cholesterol export from endolysosomes. Hence by antagonizing PIKfyve, apilimod appears to block EBOV trafficking to its site of fusion and entry into the cytoplasm. Given the drug’s observed anti-filoviral activity, relatively unexplored mechanism of entry inhibition, and reported tolerability in humans, we propose that apilimod be further explored as part of a therapeutic regimen to treat filoviral infections.


PLOS ONE | 2016

Evaluation of the Activity of Lamivudine and Zidovudine against Ebola Virus

Yu Cong; Julie Dyall; Brit J. Hart; Lisa Evans DeWald; Joshua C. Johnson; Elena Postnikova; Huanying Zhou; Robin Gross; Oscar Rojas; Isis Alexander; Nicole Josleyn; Tengfei Zhang; Julia Michelotti; Krisztina Janosko; Pamela J. Glass; Mike Flint; Laura K. McMullan; Christina F. Spiropoulou; Tim Mierzwa; Rajarshi Guha; Paul Shinn; Sam Michael; Carleen Klumpp-Thomas; Crystal McKnight; Craig J. Thomas; Ann E. Eakin; Kathleen O’Loughlin; Carol E. Green; Paul Catz; Jon C. Mirsalis

In the fall of 2014, an international news agency reported that patients suffering from Ebola virus disease (EVD) in Liberia were treated successfully with lamivudine, an antiviral drug used to treat human immunodeficiency virus-1 and hepatitis B virus infections. According to the report, 13 out of 15 patients treated with lamivudine survived and were declared free from Ebola virus disease. In this study, the anti-Ebola virus (EBOV) activity of lamivudine and another antiretroviral, zidovudine, were evaluated in a diverse set of cell lines against two variants of wild-type EBOV. Variable assay parameters were assessed to include different multiplicities of infection, lengths of inoculation times, and durations of dosing. At a multiplicity of infection of 1, lamivudine and zidovudine had no effect on EBOV propagation in Vero E6, Hep G2, or HeLa cells, or in primary human monocyte-derived macrophages. At a multiplicity of infection of 0.1, zidovudine demonstrated limited anti-EBOV activity in Huh 7 cells. Under certain conditions, lamivudine had low anti-EBOV activity at the maximum concentration tested (320 μM). However, lamivudine never achieved greater than 30% viral inhibition, and the activity was not consistently reproducible. Combination of lamivudine and zidovudine showed no synergistic antiviral activity. Independently, a set of in vitro experiments testing lamivudine and zidovudine for antiviral activity against an Ebola-enhanced green fluorescent protein reporter virus was performed at the Centers for Disease Control and Prevention. No antiviral activity was observed for either compound. A study evaluating the efficacy of lamivudine in a guinea pig model of EVD found no survival benefit. This lack of benefit was observed despite plasma lamivudine concentrations in guinea pig of about 4 μg/ml obtained in a separately conducted pharmacokinetics study. These studies found no evidence to support the therapeutic use of lamivudine for the treatment of EVD.


Cell Death and Disease | 2016

Large-scale pharmacological profiling of 3D tumor models of cancer cells

Lesley A. Mathews Griner; Xiaohu Zhang; Rajarshi Guha; Crystal McKnight; Ian S. Goldlust; Madhu Lal-Nag; Kelli Wilson; Sam Michael; Steve Titus; Paul Shinn; Craig J. Thomas; Marc Ferrer

The discovery of chemotherapeutic agents for the treatment of cancer commonly uses cell proliferation assays in which cells grow as two-dimensional (2D) monolayers. Compounds identified using 2D monolayer assays often fail to advance during clinical development, most likely because these assays do not reproduce the cellular complexity of tumors and their microenvironment in vivo. The use of three-dimensional (3D) cellular systems have been explored as enabling more predictive in vitro tumor models for drug discovery. To date, small-scale screens have demonstrated that pharmacological responses tend to differ between 2D and 3D cancer cell growth models. However, the limited scope of screens using 3D models has not provided a clear delineation of the cellular pathways and processes that differentially regulate cell survival and death in the different in vitro tumor models. Here we sought to further understand the differences in pharmacological responses between cancer tumor cells grown in different conditions by profiling a large collection of 1912 chemotherapeutic agents. We compared pharmacological responses obtained from cells cultured in traditional 2D monolayer conditions with those responses obtained from cells forming spheres versus cells already in 3D spheres. The target annotation of the compound library screened enabled the identification of those key cellular pathways and processes that when modulated by drugs induced cell death in all growth conditions or selectively in the different cell growth models. In addition, we also show that many of the compounds targeting these key cellular functions can be combined to produce synergistic cytotoxic effects, which in many cases differ in the magnitude of their synergism depending on the cellular model and cell type. The results from this work provide a high-throughput screening framework to profile the responses of drugs both as single agents and in pairwise combinations in 3D sphere models of cancer cells.


Current protocols in chemical biology | 2013

Multiplexing High‐Content Flow (HCF) and Quantitative High‐Throughput Screening (qHTS) to Identify Compounds Capable of Decreasing Cell Viability, Activating Caspase 3/7, Expressing Annexin V, and Changing Mitochondrial Membrane Integrity

Lesley A. Mathews; Jonathan M. Keller; Crystal McKnight; Sam Michael; Paul Shinn; Dongbo Liu; Louis M. Staudt; Craig J. Thomas; Marc Ferrer

High‐content flow (HCF) screening systems, such as the iQue Screener and HTFC Screening System from IntelliCyt, have facilitated the implementation of flow cytometry assays for high‐throughput screening. HCF screening systems enable the use of smaller sample volumes and multiplexed assays to simultaneously assess different cellular parameters from a single well. This becomes invaluable when working with cells or compounds that are available in limited quantities or when conducting large‐scale screens. When assays can be miniaturized to a 384‐ or 1536‐well microplate format, it is possible to implement dose‐response‐based high‐throughput screens, also known as quantitative HTS or qHTS. This article describes how qHTS at the new National Center for Advancing Translational Science (NCATS) has been systematically coupled with the HTFC Screening System and Multimetric Apoptosis Screening Kit from IntelliCyt to biologically validate active compounds from primary cell proliferation screens using a model of diffuse large B cell lymphoma (DLBCL). Curr. Protoc. Chem. Biol. 5:195‐212


The Journal of Infectious Diseases | 2018

Identification of Combinations of Approved Drugs With Synergistic Activity Against Ebola Virus in Cell Cultures

Julie Dyall; Elizabeth A. Nelson; Lisa Evans DeWald; Rajarshi Guha; Brit J. Hart; Huanying Zhou; Elena Postnikova; James Logue; Walter M Vargas; Robin Gross; Julia Michelotti; Nicole Deiuliis; Richard S. Bennett; Ian Crozier; Patrick J. Morris; Carleen Klumpp-Thomas; Crystal McKnight; Tim Mierzwa; Paul Shinn; Pamela J. Glass; Lisa M. Johansen; Peter B. Jahrling; Lisa E. Hensley; Gene G. Olinger; Craig J. Thomas; Judith M. White

Background A need to develop therapeutics to treat Ebola virus disease patients in remote and resource-challenged settings remains in the wake of the 2013-2016 epidemic in West Africa. Toward this goal, we screened drugs under consideration as treatment options and other drugs of interest, most being small molecules approved by the Food and Drug Administration. Drugs demonstrating in vitro antiviral activity were advanced for evaluation in combinations because of advantages often provided by drug cocktails. Methods Drugs were screened for blockade of Ebola virus infection in cultured cells. Twelve drugs were tested in all (78 pair-wise) combinations, and 3 were tested in a subset of combinations. Results Multiple synergistic drug pairs emerged, with the majority comprising 2 entry inhibitors. For the pairs of entry inhibitors studied, synergy was demonstrated at the level of virus entry into host cells. Highly synergistic pairs included aripiprazole/piperacetazine, sertraline/toremifene, sertraline/bepridil, and amodiaquine/clomiphene. Conclusions Our study shows the feasibility of identifying pairs of approved drugs that synergistically block Ebola virus infection in cell cultures. We discuss our findings in terms of the theoretic ability of these or alternate combinations to reach therapeutic levels. Future research will assess selected combinations in small-animal models of Ebola virus disease.


SLAS TECHNOLOGY: Translating Life Sciences Innovation | 2018

Mutation Profiles in Glioblastoma 3D Oncospheres Modulate Drug Efficacy

Kelli Wilson; Lesley Mathews-Griner; Tara Williamson; Rajarshi Guha; Lu Chen; Paul Shinn; Crystal McKnight; Sam Michael; Carleen Klumpp-Thomas; Zev A. Binder; Marc Ferrer; Gary L. Gallia; Craig J. Thomas; Gregory J. Riggins

Glioblastoma (GBM) is a lethal brain cancer with a median survival time of approximately 15 months following treatment. Common in vitro GBM models for drug screening are adherent and do not recapitulate the features of human GBM in vivo. Here we report the genomic characterization of nine patient-derived, spheroid GBM cell lines that recapitulate human GBM characteristics in orthotopic xenograft models. Genomic sequencing revealed that the spheroid lines contain alterations in GBM driver genes such as PTEN, CDKN2A, and NF1. Two spheroid cell lines, JHH-136 and JHH-520, were utilized in a high-throughput drug screen for cell viability using a 1912-member compound library. Drug mechanisms that were cytotoxic in both cell lines were Hsp90 and proteasome inhibitors. JHH-136 was uniquely sensitive to topoisomerase 1 inhibitors, while JHH-520 was uniquely sensitive to Mek inhibitors. Drug combination screening revealed that PI3 kinase inhibitors combined with Mek or proteasome inhibitors were synergistic. However, animal studies to test these drug combinations in vivo revealed that Mek inhibition alone was superior to the combination treatments. These data show that these GBM spheroid lines are amenable to high-throughput drug screening and that this dataset may deliver promising therapeutic leads for future GBM preclinical studies.


Malaria Journal | 2018

A single nucleotide polymorphism in the Plasmodium falciparum atg18 gene associates with artemisinin resistance and confers enhanced parasite survival under nutrient deprivation

Kimberly F. Breglio; Roberto Amato; Richard C. Eastman; Pharath Lim; Juliana M. Sá; Rajarshi Guha; Sundar Ganesan; David W. Dorward; Carleen Klumpp-Thomas; Crystal McKnight; Rick M. Fairhurst; David J. Roberts; Craig J. Thomas; Anna Katharina Simon

BackgroundArtemisinin-resistant Plasmodium falciparum has been reported throughout the Greater Mekong subregion and threatens to disrupt current malaria control efforts worldwide. Polymorphisms in kelch13 have been associated with clinical and in vitro resistance phenotypes; however, several studies suggest that the genetic determinants of resistance may involve multiple genes. Current proposed mechanisms of resistance conferred by polymorphisms in kelch13 hint at a connection to an autophagy-like pathway in P. falciparum.ResultsA SNP in autophagy-related gene 18 (atg18) was associated with long parasite clearance half-life in patients following artemisinin-based combination therapy. This gene encodes PfAtg18, which is shown to be similar to the mammalian/yeast homologue WIPI/Atg18 in terms of structure, binding abilities, and ability to form puncta in response to stress. To investigate the contribution of this polymorphism, the atg18 gene was edited using CRISPR/Cas9 to introduce a T38I mutation into a k13-edited Dd2 parasite. The presence of this SNP confers a fitness advantage by enabling parasites to grow faster in nutrient-limited settings. The mutant and parent parasites were screened against drug libraries of 6349 unique compounds. While the SNP did not modulate the parasite’s susceptibility to any of the anti-malarial compounds using a 72-h drug pulse, it did alter the parasite’s susceptibility to 227 other compounds.ConclusionsThese results suggest that the atg18 T38I polymorphism may provide additional resistance against artemisinin derivatives, but not partner drugs, even in the absence of kelch13 mutations, and may also be important in parasite survival during nutrient deprivation.

Collaboration


Dive into the Crystal McKnight's collaboration.

Top Co-Authors

Avatar

Craig J. Thomas

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Rajarshi Guha

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Paul Shinn

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Sam Michael

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Marc Ferrer

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Carleen Klumpp-Thomas

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Bryan T. Mott

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dongbo Liu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ian S. Goldlust

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kelli Wilson

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge