Network


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

Hotspot


Dive into the research topics where Catherine Burkhart is active.

Publication


Featured researches published by Catherine Burkhart.


Cancer Research | 2009

Small-Molecule Multidrug Resistance–Associated Protein 1 Inhibitor Reversan Increases the Therapeutic Index of Chemotherapy in Mouse Models of Neuroblastoma

Catherine Burkhart; Fujiko Watt; Jayne Murray; Marina Pajic; Anatoly Prokvolit; Chengyuan Xue; Claudia Flemming; Janice Smith; Andrei Purmal; Nadezhda Isachenko; Pavel G. Komarov; Katerina V. Gurova; Alan C. Sartorelli; Glenn M. Marshall; Murray D. Norris; Andrei V. Gudkov; Michelle Haber

The multidrug resistance-associated protein 1 (MRP1) has been closely linked to poor treatment response in several cancers, most notably neuroblastoma. Homozygous deletion of the MRP1 gene in primary murine neuroblastoma tumors resulted in increased sensitivity to MRP1 substrate drugs (vincristine, etoposide, and doxorubicin) compared with tumors containing both copies of wild-type MRP1, indicating that MRP1 plays a significant role in the drug resistance in this tumor type and defining this multidrug transporter as a target for pharmacologic suppression. A cell-based readout system was created to functionally determine intracellular accumulation of MRP1 substrates using a p53-responsive reporter as an indicator of drug-induced DNA damage. Screening of small-molecule libraries in this readout system revealed pyrazolopyrimidines as a prominent structural class of potent MRP1 inhibitors. Reversan, the lead compound of this class, increased the efficacy of both vincristine and etoposide in murine models of neuroblastoma (syngeneic and human xenografts). As opposed to the majority of inhibitors of multidrug transporters, Reversan was not toxic by itself nor did it increase the toxicity of chemotherapeutic drug exposure in mice. Therefore, Reversan represents a new class of nontoxic MRP1 inhibitor, which may be clinically useful for the treatment of neuroblastoma and other MRP1-overexpressing drug-refractory tumors by increasing their sensitivity to conventional chemotherapy.


Medical and Pediatric Oncology | 2000

Expression of N-myc and MRP genes and their relationship to N-myc gene dosage and tumor formation in a murine neuroblastoma model.

Murray D. Norris; Catherine Burkhart; Glenn M. Marshall; William A. Weiss; Michelle Haber

BACKGROUND Although the association between N-myc gene amplification and poor clinical outcome in neuroblastoma is well established, the mechanism by which amplification influences prognosis is not well defined. PROCEDURE We used a human N-myc transgenic mouse model to investigate the role of N-myc in neuroblastoma, including its relationship to the multidrug-resistance-associated protein (MRP) gene. We developed a rapid real-time PCR method to distinguish homozygous and hemizygous N-myc mice that is comparable to Southern analysis. RESULTS A highly significant correlation (P < 0.0001) between N-myc and MRP expression was demonstrated in murine tumors. Amplification of the transgene was observed in the majority of tumors, highlighting the clinical relevance of this model. However, no correlation between N-myc expression and transgene dosage or tumor latency was observed. CONCLUSIONS The data suggest that increased N-myc dosage contributes to increased tumor incidence and decreased latency by mechanisms independent of N-myc expression.


Journal of Biochemical and Biophysical Methods | 2002

A simple method for the isolation of genomic DNA from mouse tail free of real-time PCR inhibitors.

Catherine Burkhart; Murray D. Norris; Michelle Haber

Although real-time PCR is a rapid, quantitative method for the analysis of gene and RNA levels, the presence of inhibitors in samples is an obstacle to its successful use. We have found that genomic DNA isolated from mouse tail tips using a standard proteinase K digestion method caused marked inhibition of real-time PCR. Inhibition was specific for mouse tail DNA since genomic DNA isolated from other tissue sources using the same methodology was readily amplified. We have therefore developed a nonproteinase K DNA isolation method involving the use of Chelex 100 resin. This method produces mouse tail DNA that is free of real-time PCR inhibitors.


Science Translational Medicine | 2015

Therapeutic targeting of the MYC signal by inhibition of histone chaperone FACT in neuroblastoma

Daniel Carter; Jayne Murray; Belamy B. Cheung; Laura Gamble; Jessica Koach; Joanna Tsang; Selina Sutton; Heyam Kalla; Sarah Syed; Andrew J. Gifford; Natalia Issaeva; Asel Biktasova; Bernard Atmadibrata; Yuting Sun; Nicolas Sokolowski; Dora Ling; Patrick Y. Kim; Hannah Webber; Ashleigh Clark; Michelle Ruhle; Bing Liu; André Oberthuer; Matthias Fischer; Jennifer A. Byrne; Federica Saletta; Le M. Thwe; Andrei Purmal; Gary Haderski; Catherine Burkhart; Frank Speleman

Histone chaperone FACT acts in a positive feedback loop with MYCN and is a therapeutic target in neuroblastoma. Uncovering the FACTs in neuroblastoma Neuroblastoma is a common pediatric cancer of the nervous system. It is often difficult to treat, and tumors with amplifications of the MYC oncogene are particularly aggressive. Carter et al. have identified a histone chaperone called FACT as a mediator of MYC signaling in neuroblastoma and demonstrated its role in a feedback loop that allows tumor cells to maintain a high expression of both MYC and FACT. The authors then used curaxins, which are drugs that inhibit FACT, to break the vicious cycle. They demonstrated that curaxins work in synergy with standard genotoxic chemotherapy to kill cancer cells and treat neuroblastoma in mouse models. Amplification of the MYCN oncogene predicts treatment resistance in childhood neuroblastoma. We used a MYC target gene signature that predicts poor neuroblastoma prognosis to identify the histone chaperone FACT (facilitates chromatin transcription) as a crucial mediator of the MYC signal and a therapeutic target in the disease. FACT and MYCN expression created a forward feedback loop in neuroblastoma cells that was essential for maintaining mutual high expression. FACT inhibition by the small-molecule curaxin compound CBL0137 markedly reduced tumor initiation and progression in vivo. CBL0137 exhibited strong synergy with standard chemotherapy by blocking repair of DNA damage caused by genotoxic drugs, thus creating a synthetic lethal environment in MYCN-amplified neuroblastoma cells and suggesting a treatment strategy for MYCN-driven neuroblastoma.


Cancer Research | 2012

N-Myc Regulates Expression of the Detoxifying Enzyme Glutathione Transferase GSTP1, a Marker of Poor Outcome in Neuroblastoma

Jamie I. Fletcher; Samuele Gherardi; Jayne Murray; Catherine Burkhart; Amanda J. Russell; Emanuele Valli; Janice Smith; Andr e Oberthuer; Lesley J. Ashton; Wendy B. London; Glenn M. Marshall; Murray D. Norris; Giovanni Perini; Michelle Haber

Amplification of the transcription factor MYCN is associated with poor outcome and a multidrug-resistant phenotype in neuroblastoma. N-Myc regulates the expression of several ATP-binding cassette (ABC) transporter genes, thus affecting global drug efflux. Because these transporters do not confer resistance to several important cytotoxic agents used to treat neuroblastoma, we explored the prognostic significance and transcriptional regulation of the phase II detoxifying enzyme, glutathione S-transferase P1 (GSTP1). Using quantitative real-time PCR, GSTP1 gene expression was assessed in a retrospective cohort of 51 patients and subsequently in a cohort of 207 prospectively accrued primary neuroblastomas. These data along with GSTP1 expression data from an independent microarray study of 251 neuroblastoma samples were correlated with established prognostic indicators and disease outcome. High levels of GSTP1 were associated with decreased event-free and overall survival in all three cohorts. Multivariable analyses, including age at diagnosis, tumor stage, and MYCN amplification status, were conducted on the two larger cohorts, independently showing the prognostic significance of GSTP1 expression levels in this setting. Mechanistic investigations revealed that GSTP1 is a direct transcriptional target of N-Myc in neuroblastoma cells. Together, our findings reveal that N-Myc regulates GSTP1 along with ABC transporters that act to control drug metabolism and efflux. Furthermore, they imply that strategies to jointly alter these key multidrug resistance mechanisms may have therapeutic implications to manage neuroblastomas and other malignancies driven by amplified Myc family genes.


Neuro-oncology | 2016

Anticancer drug candidate CBL0137, which inhibits histone chaperone FACT, is efficacious in preclinical orthotopic models of temozolomide-responsive and -resistant glioblastoma

Tara A. Barone; Catherine Burkhart; Gary Haderski; Katerina V. Gurova; Andrei A. Purmal; Andrei V. Gudkov; Robert J. Plunkett

Background The survival rate for patients with glioblastoma (GBM) remains dismal. New therapies targeting molecular pathways dysregulated in GBM are needed. One such clinical-stage drug candidate, CBL0137, is a curaxin, small molecules which simultaneously downregulate nuclear factor-kappaB (NF-ĸB) and activate p53 by inactivating the chromatin remodeling complex, Facilitates Chromatin Transcription (FACT). Methods We used publicly available databases to establish levels of FACT subunit expression in GBM. In vitro, we evaluated the toxicity and effect of CBL0137 on FACT, p53, and NF-ĸB on U87MG and A1207 human GBM cells. In vivo, we implanted the cells orthotopically in nude mice and administered CBL0137 in various dosing regimens to assess brain and tumor accumulation of CBL0137, its effect on tumor cell proliferation and apoptosis, and on survival of mice with and without temozolomide (TMZ). Results FACT subunit expression was elevated in GBM compared with normal brain. CBL0137 induced loss of chromatin-unbound FACT, activated p53, inhibited NF-ĸB-dependent transcription, and was toxic to GBM cells. The drug penetrated the blood-brain barrier and accumulated in orthotopic tumors significantly more than normal brain tissue. It increased apoptosis and suppressed proliferation in both U87MG and A1207 tumors. Intravenous administration of CBL0137 significantly increased survival in models of early- through late-stage TMZ-responsive and -resistant GBM, with a trend toward significantly increasing the effect of TMZ in TMZ-responsive U87MG tumors. Conclusion CBL0137 targets GBM according to its proposed mechanism of action, crosses the blood-brain barrier, and is efficacious in both TMZ-responsive and -resistant orthotopic models, making it an attractive new therapy for GBM.


eLife | 2018

TRAIN (Transcription of Repeats Activates INterferon) in response to chromatin destabilization induced by small molecules in mammalian cells

Katerina I. Leonova; Elimelech Nesher; Poorva Sandlesh; Rachel Pratt; Catherine Burkhart; Brittany C. Lipchick; Ilya Gitlin; Costakis Frangou; Igor Koman; Jianmin Wang; Kirill I. Kirsanov; Marianna G. Yakubovskaya; Andrei V. Gudkov; Katerina V. Gurova

Cellular responses to the loss of genomic stability are well-established, while how mammalian cells respond to chromatin destabilization is largely unknown. We previously found that DNA demethylation on p53-deficient background leads to transcription of repetitive heterochromatin elements, followed by an interferon response, a phenomenon we named TRAIN (Transcription of Repeats Activates INterferon). Here, we report that curaxin, an anticancer small molecule, destabilizing nucleosomes via disruption of histone/DNA interactions, also induces TRAIN. Furthermore, curaxin inhibits oncogene-induced transformation and tumor growth in mice in an interferon-dependent manner, suggesting that anticancer activity of curaxin, previously attributed to p53-activation and NF-kappaB-inhibition, may also involve induction of interferon response to epigenetic derepression of the cellular ‘repeatome’. Moreover, we observed that another type of drugs decondensing chromatin, HDAC inhibitor, also induces TRAIN. Thus, we proposed that TRAIN may be one of the mechanisms ensuring epigenetic integrity of mammalian cells via elimination of cells with desilenced chromatin.


bioRxiv | 2017

TRAIN in response to treatment with anti-cancer small molecule destabilizing chromatin

Yekaterina Leonova; Elimelech Nesher; Poorva Sandlesh; Rachel Pratt; Catherine Burkhart; Brittany C. Lipchick; Costakis Frangou; Igor Koman; Jianmin Wang; Kirill I. Kirsanov; Marianna G. Yakubovskaya; Andrei V. Gudkov; Katerina V. Gurova

Genome stability is in the focus of research for many decades, while stability and integrity of chromatin is far less studied. Cell identity in multicellular organism is completely dependent on chromatin stability, therefore there should be mechanisms ensuring maintenance of epigenetic integrity. Previously, we have found that loss of DNA methylation in the absence of p53 leads to the transcription of silenced repetitive elements, such as pericentromeric repeats and endogenous viruses, what causes activation of IFN response, similarly to viral invasion, and IFN-dependent cell death. We named this phenomenon TRAIN (Transcription of Repeats Activates INterferon). Now we found that small molecule, curaxin, which destabilizes nucleosome via binding to DNA and deforming helix shape, causes TRAIN independently on p53 status. Curaxin demonstrated activity as cancer treatment and preventive agent via established previously p53 activating and NF-kappaB inhibiting activities. Here we showed that activation of IFN response is an additional mechanism of inhibition of oncogene-induced transformation by curaxin. Our data suggest that TRAIN is a response to the loss of chromatin stability and one of the mechanisms which prevents oncogene-induced transformation.


Cancer Research | 2016

Abstract 2070: Gemcitabine-loaded microparticles promote cancer cell death in subcutaneous pancreatic cancer xenografts

Maria Munoz-Sagastibelza; Vadim Kurbatov; Sophia Dynes; Jennifer Caceres; Michael Chen; Raavi Gupta; Catherine Burkhart; Laura Martello-Rooney

Pancreatic cancer is the fourth leading cause of cancer death in the United States with only 7% of diagnosed patients surviving 5 years. Most pancreatic cancer patients are not surgical candidates due to advanced stage at diagnosis. Current systemic chemotherapies, while exposing patients to the adverse side effects of treatment, have not been very effective at decreasing tumor burden primarily due to poor systemic drug uptake resulting from the dense stromal nature of pancreatic tumors. Poly(lactic-co-glycolic acid)-based (PLGA) microparticles (MPs) are a promising tool for localized drug delivery within the tumor due to their high biocompatibility, flexibility in the encapsulation of different drugs and extended drug release inside the tumor. The present study investigated whether gemcitabine-loaded microparticles (GMPs) in the range of 10-50 microns, in comparison with blank (no drug) MPs (BMPs), saline intraperitoneal injection (SIP) and gemcitabine intraperitoneal injection (GIP) as controls, are able to promote cancer cell killing effects in vivo. In vitro studies with PANC-1 and MIAPaCa-2 human pancreatic adenocarcinoma cell lines treated with different PLGA co-polymer ratios used to encapsulate gemcitabine showed enhanced cell killing and decreased colony formation in the longer release co-polymer ratio after 2 weeks of treatment. Subsequently, the in vivo efficacy of GMPs was tested by direct injection of GMPs into established subcutaneous MIAPaCa-2 tumors in nude mice. Treatment commenced when tumor volume was approximately 250 mm3. Following two weeks of treatment, there was a trending decrease in tumor volume in the GMPs-injected MIAPaCa-2 tumors compared to the BMPs-injected tumors. When comparing the SIP to GIP groups, there was no difference in final tumor volume emphasizing the lack of effective penetration of systemic gemcitabine into the tumor. In addition, we observed less tumor progression in the GMPs group compared to the others. At the endpoint, the tumors were excised, frozen in OCT compound and sectioned to visualize fluorescent MPs and to detect apoptosis by immunofluorescence. Interestingly, we observed a significant increase in apoptosis in the tumors treated with GMPs compared to the BMP tumors (p In conclusion, our data suggest that gemcitabine-loaded MPs could decrease tumor volume and increase local pancreatic tumor cell death. Further studies are needed to optimize the MPs loading and injection to confirm its efficacy. The described drug delivery method has the potential to be a more efficient local treatment modality than systemic gemcitabine against pancreatic cancer. Citation Format: Maria Munoz-Sagastibelza, Vadim Kurbatov, Sophia Dynes, Jennifer Caceres, Michael Chen, Raavi Gupta, Catherine Burkhart, Laura Martello-Rooney. Gemcitabine-loaded microparticles promote cancer cell death in subcutaneous pancreatic cancer xenografts. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2070.


Cancer Research | 2016

Preclinical Validation of a Single-Treatment Infusion Modality That Can Eradicate Extremity Melanomas

Minhyung Kim; Nickolay Neznanov; Chandler Wilfong; Daria Fleyshman; Andrei A. Purmal; Gary Haderski; Patricia Stanhope-Baker; Catherine Burkhart; Katerina V. Gurova; Andrei V. Gudkov; Joseph J. Skitzki

Isolated limb perfusion (ILP) with the chemotherapeutic agent melphalan is an effective treatment option for extremity in-transit melanoma but is toxic and technically challenging to deliver locoregionally. CBL0137 is an experimental clinical drug with broad anticancer activity in animal models, owing to its ability to bind DNA in a nongenotoxic manner and inactivate the FACT chromatin modulator essential for tumor cell viability. Here, we report that CBL0137 delivered by ILP in a murine melanoma model is as efficacious as melphalan, displaying antitumor activity at doses corresponding to only a fraction of the systemic MTD of CBL0137. The ability to bind DNA quickly combined with a favorable safety profile made it possible to substitute CBL0137 in the ILP protocol, using an intra-arterial infusion method, to safely achieve effective tumor suppression. Our findings of a preclinical proof of concept for CBL0137 and its administration via intra-arterial infusion as a superior treatment compared with melphalan ILP allows for locoregional treatment anywhere a catheter can be placed. Cancer Res; 76(22); 6620-30. ©2016 AACR.

Collaboration


Dive into the Catherine Burkhart's collaboration.

Top Co-Authors

Avatar

Andrei V. Gudkov

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Michelle Haber

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Murray D. Norris

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Katerina V. Gurova

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Glenn M. Marshall

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jayne Murray

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Andrei Purmal

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge