Network


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

Hotspot


Dive into the research topics where Kelly S. Clark is active.

Publication


Featured researches published by Kelly S. Clark.


Cell | 2013

Monitoring tumorigenesis and senescence in vivo with a p16 INK4a-luciferase model

Christin E. Burd; Jessica A. Sorrentino; Kelly S. Clark; David B. Darr; Janakiraman Krishnamurthy; Allison M. Deal; Nabeel Bardeesy; Diego H. Castrillon; David Beach; Norman E. Sharpless

Monitoring cancer and aging in vivo remains experimentally challenging. Here, we describe a luciferase knockin mouse (p16(LUC)), which faithfully reports expression of p16(INK4a), a tumor suppressor and aging biomarker. Lifelong assessment of luminescence in p16(+/LUC) mice revealed an exponential increase with aging, which was highly variable in a cohort of contemporaneously housed, syngeneic mice. Expression of p16(INK4a) with aging did not predict cancer development, suggesting that the accumulation of senescent cells is not a principal determinant of cancer-related death. In 14 of 14 tested tumor models, expression of p16(LUC) was focally activated by early neoplastic events, enabling visualization of tumors with sensitivity exceeding other imaging modalities. Activation of p16(INK4a) was noted in the emerging neoplasm and surrounding stromal cells. This work suggests that p16(INK4a) activation is a characteristic of all emerging cancers, making the p16(LUC) allele a sensitive, unbiased reporter of neoplastic transformation.


Nature Medicine | 2016

An oncogenic Ezh2 mutation induces tumors through global redistribution of histone 3 lysine 27 trimethylation.

George P. Souroullas; William R. Jeck; Joel S. Parker; Jeremy M. Simon; Jie Yu Liu; Joshiawa Paulk; Jessie Xiong; Kelly S. Clark; Yuri Fedoriw; Jun Qi; Christin E. Burd; James E. Bradner; Norman E. Sharpless

B cell lymphoma and melanoma harbor recurrent mutations in the gene encoding the EZH2 histone methyltransferase (EZH2), but the carcinogenic role of these mutations is unclear. Here we describe a mouse model in which the most common somatic Ezh2 gain-of-function mutation (EZH2Y646F in human; Ezh2Y641F in mouse) is conditionally expressed. Expression of Ezh2Y641F in mouse B cells or melanocytes caused high-penetrance lymphoma or melanoma, respectively. Overexpression of the anti-apoptotic protein Bcl2, but not the oncoprotein Myc, or loss of the tumor suppressor protein p53 (encoded by Trp53 in mice) further accelerated lymphoma progression. Expression of the mutant Braf but not the mutant Nras oncoprotein further accelerated melanoma progression. Although expression of Ezh2Y641F globally increased the abundance of trimethylated Lys27 of histone H3 (H3K27me3), it also caused a widespread redistribution of this repressive mark, including a loss of H3K27me3 that was associated with increased transcription at many loci. These results suggest that Ezh2Y641F induces lymphoma and melanoma through a vast reorganization of chromatin structure, inducing both repression and activation of polycomb-regulated loci.B-cell lymphoma and melanoma harbor recurrent mutations in the gene encoding the EZH2 histone methyltransferase, but the carcinogenic role of these mutations is unclear. Here we describe a mouse model in which the most common somatic EZH2 gain-of-function mutation (Y646F in human, Y641F in the mouse) can be conditionally expressed. Expression of Ezh2Y641F in mouse B-cells or melanocytes caused high-penetrance lymphoma or melanoma, respectively. Bcl2 overexpression or p53 loss, but not c-Myc overexpression, further accelerated lymphoma progression, and expression of mutant B-Raf but not mutant N-Ras further accelerated melanoma progression. Although expression of Ezh2Y641F increased abundance of global H3K27 trimethylation (H3K27me3), it also caused a widespread redistribution of this repressive mark, including a loss of H3K27me3 associated with increased transcription at many loci. These results suggest that Ezh2Y641F induces lymphoma and melanoma through a vast reorganization of chromatin structure inducing both repression and activation of polycomb-regulated loci.


Oncogene | 2014

Canonical and non-canonical NF-κB signaling promotes breast cancer tumor-initiating cells.

Megan F. Kendellen; Jennifer W. Bradford; Cortney L. Lawrence; Kelly S. Clark; Albert S. Baldwin

Tumor-initiating cells (TICs) are a sub-population of cells that exhibit a robust ability to self-renew and contribute to the formation of primary tumors, the relapse of previously treated tumors and the development of metastases. TICs have been identified in various tumors including those of the breast, and are particularly enriched in the basal-like and claudin-low subtypes of breast cancer. The signaling pathways that contribute to the function and maintenance of TICs are under intense study. We explored the potential involvement of the nuclear factor-κB (NF-κB) family of transcription factors in TICs in cell lines that are representative of basal-like and claudin-low breast cancer. NF-κB was found to be activated in breast cancer cells that form tumorspheres efficiently. Moreover, both canonical and non-canonical NF-κB signaling is required for these cells to self-renew in vitro and to form xenograft tumors efficiently in vivo using limiting dilutions of cells. Consistent with this fact, canonical and non-canonical NF-κB signaling is activated in TICs isolated from breast cancer cell lines. Experimental results indicate that NF-κB promotes the function of TICs by stimulating epithelial-to-mesenchymal transition and by upregulating the expression of the inflammatory cytokines interleukin-1β and interleukin-6. The results suggest the use of NF-κB inhibitors for clinical therapy of certain breast cancers.


Journal of Clinical Investigation | 2014

Erythropoietin promotes breast tumorigenesis through tumor-initiating cell self-renewal.

Bing Zhou; Jeffrey S. Damrauer; Sean T. Bailey; Tanja Hadzic; Youngtae Jeong; Kelly S. Clark; Cheng Fan; Laura Murphy; Cleo Lee; Melissa A. Troester; C. Ryan Miller; Jian Jin; David B. Darr; Charles M. Perou; Ross L. Levine; Maximilian Diehn; William Y. Kim

Erythropoietin (EPO) is a hormone that induces red blood cell production. In its recombinant form, EPO is the one of most prescribed drugs to treat anemia, including that arising in cancer patients. In randomized trials, EPO administration to cancer patients has been associated with decreased survival. Here, we investigated the impact of EPO modulation on tumorigenesis. Using genetically engineered mouse models of breast cancer, we found that EPO promoted tumorigenesis by activating JAK/STAT signaling in breast tumor-initiating cells (TICs) and promoted TIC self renewal. We determined that EPO was induced by hypoxia in breast cancer cell lines, but not in human mammary epithelial cells. Additionally, we demonstrated that high levels of endogenous EPO gene expression correlated with shortened relapse-free survival and that pharmacologic JAK2 inhibition was synergistic with chemotherapy for tumor growth inhibition in vivo. These data define an active role for endogenous EPO in breast cancer progression and breast TIC self-renewal and reveal a potential application of EPO pathway inhibition in breast cancer therapy.


Cancer Research | 2015

Abstract 2649: Inhibition of mTOR, but not PI3K, is required for the anti-tumor efficacy in breast cancer of dual PI3K/mTOR inhibitors given in combination with MEK inhibitors

Aleisha M. Smith; Jessie Xiong; Lucas Hunter; Jamie L. Jordan; Kelly S. Clark; David B. Darr; Sharpless Norman; Charles M. Perou; William Y. Kim

Background: The mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) pathways are deregulated in many cancers including breast cancer. Additionally, studies have shown that significant crosstalk exists between these pathways; often making single pathway inhibition ineffective. While, combined PI3K/mTOR and MEK inhibition appears highly active in pre-clinical models, the combination has been reported to be toxic in human use. In an effort to produce a more targeted regimen, we examined whether both PI3K and mTOR inhibition are required for pre-clinical anti-tumor efficacy in combination with a MEK inhibitor. Methods: In vivo studies assessed overall survival and best tumor response upon administration of dual PI3K/mTOR (BEZ235 and GSK806) versus allosteric mTOR inhibition (Everolimus) in combination with MEK inhibition (AZD244 and GSK212) in genetically engineered murine models (GEMM) of breast cancer [T11 and C(3)Tag]. Activation and response of the PI3K/mTOR and MEK/ERK pathways to dual PI3K/mTOR, allosteric mTOR inhibition, and MEK inhibition were assessed in vitro in murine cell lines derived from the above GEM models. Results: Single agent regimens of MEK, allosteric mTOR, or dual PI3K/mTOR inhibition were minimally effective in C(3)Tag or T11 tumors in vivo. As previously reported, combined PI3K/mTOR and MEK inhibition showed potent pre-clinical activity in both the T11 and C3Tag models, prolonging overall survival relative to both vehicle or single-agent treated mice. Surprisingly, combinations of MEK inhibitors with allosteric mTOR inhibitors were as active as the MEK/PI3K/mTOR combination. In vitro studies in cell lines derived from these tumor models showed that mTORC1 inhibition, but not PI3K inhibition, resulted in compensatory activation of MEK. Conclusions: Combined targeting of the mTOR and MEK pathways results in improved response and survival in faithful GEM models of breast cancer. The combination of dual PI3K/mTOR catalytic inhibitors with MEK inhibition does not afford benefit over combined allosteric mTOR and MEK inhibition Citation Format: Aleisha M. Smith, Jessie Xiong, Lucas Hunter, Jamie Jordan, Kelly Clark, David B. Darr, Sharpless Norman, Charles M. Perou, William Y. Kim. Inhibition of mTOR, but not PI3K, is required for the anti-tumor efficacy in breast cancer of dual PI3K/mTOR inhibitors given in combination with MEK inhibitors. [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 2649. doi:10.1158/1538-7445.AM2015-2649


Cancer Research | 2014

Abstract 947: A small molecule Mer tyrosine kinase inhibitor (UNC MerTKi) effectively inhibits growth of murine melanoma

H. Shelton Earp; David B. Darr; Albert G. Zimmermann; Kelly S. Clark; Norman E. Sharpless; Wolfgang Bergmeier; Weihe Zhang; Xiaodong Wang; Deborah DeRyckere; Stephen V. Frye; Douglas K. Graham

The Mer Receptor Tyrosine Kinase (RTK) is overexpressed in hematologic and epithelial malignancies. Mer is not a proliferative driver but rather produces a cancer cell-intrinsic survival signal. In addition, tumor-associated macrophages (TAMs) express Mer, which upon binding ligand-associated with apoptotic material, triggers engulfment (effercytosis). The Mer signal also polarizes macrophages towards an M2-like, wound healing, anti-inflammatory state, calming the innate immune response when ingesting “self”. We have previously shown that syngeneic polyoma middle T (PyVmT) breast cancer cells demonstrate markedly attenuated growth when orthotopically implanted into Mer knock-out mice, with Mer -/- TAMs of PyVmT tumors producing M1-like inflammatory cytokines (JCI 123:3231, 2013). This result suggests that Mer inhibition could enhance innate anti-tumor responses, and toward that end, we examine the activity of first-in-class, orally bioavailable MerTK inhibitor on tumor growth in autochthonous murine tumor models. UNC MerTKi is 5-fold selective for Mer vs. Axl/Tyro3 and has favorable pharmacokinetics. Once daily, oral dosing inhibits the growth of Mer-expressing leukemia and NSCLC xenografts. UNC MerTKi was assessed in immune-competent, genetically engineered murine models (GEMMs) in the UNC Lineberger Mouse Phase 1 Unit (MP1U). After dose-finding studies in wild-type mice established an MTD, the inhibitor was given at 120 mpk/d in mouse chow, a dose which did not cause weight loss and produced a measurable effect (i.e. inhibition of second phase platelet aggregation, a known Mer pharmacodynamics marker). This dose did not exhibit single agent activity in a murine model of breast cancer (C3TAg), but exhibited pronounced single agent activity in RAS-driven, INK4a/Arf null melanoma GEMM (TRIA). The MP1U has previously reported the efficacy of 15 chemotherapeutic and/or targeted regimens in a large (>220) cohort of TRIA mice (CCR 18:5290, 2012). The overall response was 10% (partial responses and stable disease). There were no complete responses. A combination of MEK (AZD 6244) and PI3K/mTOR (BEZ235) inhibitors were the most active previous regimen (responses seen in 9/18 mice = 50%, with 0 CRs) with moderate toxicity. UNC MerTKi exhibited greater activity (6/8 mice = 75%, with 3 CRs) with mild, well-tolerated toxicity in the TRIA model. TRIA cell lines do not express Mer, suggesting that UNC MerTKi as a monotherapy may induce responses via Mer inhibition in TAMs and the tumor microenvironment, or via inhibition of Axl, Tyro or an unknown target. In summary, a potent and selective Mer inhibitor exhibited greater pre-clinical efficacy in a highly faithful model of RAS-mutant melanoma than any other drug tested to date, including several compounds that are FDA approved for use in metastatic melanoma. Citation Format: H Shelton Earp, David Darr, Albert Zimmermann, Kelly Clark, Norman E. Sharpless, Wolfgang Bergmeier, Weihe Zhang, Xiaodong Wang, Deborah DeRyckere, Stephen Frye, Douglas Graham. A small molecule Mer tyrosine kinase inhibitor (UNC MerTKi) effectively inhibits growth of murine melanoma. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 947. doi:10.1158/1538-7445.AM2014-947


Seminars in Perinatology | 2016

What we have learned about best practices for recruitment and retention in multicenter pregnancy studies

Ashley Salazar; Susan Tolivaisa; D. Allard; Tammy Sinclair Bishop; Sabine Bousleiman; Kelly S. Clark; Wendy Dalton; Stacy L. Harris; Kathy Hale; Kim Hill; Francee Johnson; Gail Mallett; Lisa Moseley; F. Ortiz; Cynthia Willson; Elizabeth Thom

For 30 years, the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Maternal-Fetal Medicine Units (MFMU) Network has had significant impact on clinical practice in obstetrics. The MFMU Network has conducted 50 randomized clinical trials and observational studies designed to improve pregnancy outcomes for mothers and children. Each center has a designated clinical research nurse coordinator who coordinates the day-to-day operations of each trial and leads a research team that is responsible for recruitment and retention of participants. Some of the lessons learned by the nurse coordinators over the past 30 years are described with examples from recent studies. Best practices that we have amassed from our experience are also described.


Cancer Research | 2014

Abstract 4943: Visualization of endogenous melanoma initiation and progression using intravital microscopy

James E. Bear; Hailey E. Brighton; Norman E. Sharpless; David B. Darr; Kelly S. Clark

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA The death rates for malignant metastatic melanoma continue to rise because melanoma is largely refractory to existing therapies. Loss or inactivation of the tumor suppressor PTEN (Phosphatase and Tensin Homolog on Chromosome Ten) is observed in 40-50% of melanoma, and BRAF and PTEN mutations exist coincidentally in approximately 20% of melanoma cases. In mouse GEMM models, these mutations cooperate to promote metastatic melanomagenesis. However, existing methods to activate these lesions with Tyr-CreERT2 and topical tamoxifen application yields multifocal primary tumors whose origins cannot be accurately determined. To improve this model, we combined a tdTomatoLSL allele that serves as a visual marker of Cre recombination with Tyr-CreERT2;BrafCA;Ptenlox/lox to produce an animal where we can reproducibly initiate tumorigenesis in a spatiotemporally controllable manner by transiently applying extremely low doses (<20 nM) of tamoxifen to the dermis of the ear. Due to melanocyte-specific expression of the fluorescent protein tdTomato, we can track tumor progression from endogenous cells using two-photon intravital imaging. Starting from a single primary tumor on the ear, we can detect distant metastases in cervical lymph nodes and lungs via macroscopic fluorescent imaging, qRT-PCR and histology. We are using this model in combination with targeted therapy to investigate the effects of selective molecular inhibitors on the growth and metastasis of endogenous BRAFV600E/PTEN-/- metastatic melanoma at the cellular level. Preliminary studies using intravital imaging and a selective MEK1/2 inhibitor, GSK212 (Trametinib, FDA approved treatment for metastatic melanoma) show that targeted therapy causes heterogeneous reorganization of the tumor cells as early as 3 days post-treatment. After several weeks of continuous treatment, the remaining, resistant tumor cells are highly correlated spatially with bundled collagen structures detected by second harmonic generation signal, suggesting that cellular milieu strongly influences drug response. In addition, GSK212 treated animals still had lung macro-metastatic tumors, indicating that despite continuous MEK1/2 inhibition, metastatic tumor growth in the lung was still possible. In summary, we have developed an inducible endogenous model of melanoma metastasis that can be used to directly investigate both heterogeneous effects of targeted therapy at the cellular level and the efficacy of targeted therapy against metastasis. Citation Format: James E. Bear, Hailey E. Brighton, Norman E. Sharpless, David B. Darr, Kelly S. Clark. Visualization of endogenous melanoma initiation and progression using intravital microscopy. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4943. doi:10.1158/1538-7445.AM2014-4943


Cancer Research | 2014

Abstract 5024: mDX400, the murine analog against the anti-PD1 antibody MK-347 is active in immunocompetent, autochthonous murine models of melanoma and breast cancer

David B. Darr; Kelly S. Clark; Joseph H. Phillips; Elaine M. Pinheiro; Venkataraman Sriram; Jessie Xiong; Jamie L. Jordan; Norman E. Sharpless; Charles M. Perou; Stergios J. Moschos

Although effective therapies exist for BRAF-mutant metastatic melanomas (MM) and ER+/PR+/HER2+ breast cancers, fewer options are available for the more aggressive triple negative breast cancers (TNBC) and Ras-mutant MM. Immune infiltration is frequently observed in patient subsets with MM or TNBC. An anti-tumor host immune response may be restrained by the expression of immune checkpoint proteins, such as the programmed death 1 (PD1) protein. We evaluated mDX400, a murine version of the anti-PD-1 antibody MK-3475 that is currently in human clinical trials, in genetically engineered murine models (GEMMs) of melanoma and breast cancer (BC). PD1 antibody was used alone and in combination. Models studied were for basal-like, TNBC (C3TAg), Claudin-Low breast cancer p53null, luminal BC (MMTV-Her2/Neu), RAS-driven melanoma (Tyr-H-Ras and null for Ink4a/Arf (TRIA)) and BRaf-mutant, Pten-deficient melanoma (BRaf/Pten). Mice were housed, treated, and evaluated in the UNC Lineberger Comprehensive Cancer Center Mouse Phase I Unit (MP1U). mDX400 and isotype antibody (10mg/kg IP qw) were supplied by Merck & Co., Inc. Murine cohorts were assessed weekly and therapeutic intervention began once tumors reached 40-64mm3. TNBC models were treated with Carboplatin (C, 50mg/kg IP qwk) in combination with mDX400. mDX400 was administered as a single agent in Melanoma GEMMs. Endpoints were maximal response at ≥21 days and median overall survival (OS). In the C3TAg BC model, mDX400 did not exhibit single-agent activity, but substantial synergy was observed in combination with C [partial response (PR) or CR in 12 of 15 vs. 0 of 19 in non-treated animals, Fisher9s exact p=0.006 or vs. C alone 5 of 13 animals, p=0.05] and prolonged OS (41 vs. 28 days in NT, log-rank p=0.006 or C only 28 days, p=0.006) in the C3TAg model. Treatment with single-agent mDX400 induced a marked response in the RAS-driven TRIA melanoma model that is resistant to multiple systemic treatments. mDX400-treated TRIA mice enjoyed an almost tripling of OS [median 56 vs. 21 days, p=0.006]. BRaf/Pten melanomas did not respond to mDX400. Likewise mDX400 plus C provided no benefit to the p53null or MMTV-Her2/Neu BC GEMMs vs. C alone. Our data show differential response of PD-1 antibody therapies to various melanoma and BC models, the latter in combination with C. The responsiveness of a RAS-driven melanoma model to mDX400 is consistent with earlier reports about the clinical benefit of patients with NRAS-mutant melanoma to Ipilimumab, and the lack of PD-L1 expression and immune infiltrates of PTEN-deficient melanomas. At the meeting we will report expression of PD-L1 and other immune checkpoint proteins by cancer cells. The basis whereby carboplatin potentiates the activity of anti-PD1 in TNBC is an area of ongoing study. Citation Format: David Darr, Kelly S. Clark, Joseph H. Phillips, Elaine Pinheiro, Venkataraman Sriram, Jessie Xiong, Jamie Jordan, Norman E. Sharpless, Charles Perou, Stergios Moschos. mDX400, the murine analog against the anti-PD1 antibody MK-347 is active in immunocompetent, autochthonous murine models of melanoma and breast cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 5024. doi:10.1158/1538-7445.AM2014-5024


Cancer Research | 2013

Abstract 1087: Differential oncogenicity of N-RAS mutations in melanoma.

Christin E. Burd; William R. Jeck; Kailing Fu; Kelly S. Clark; Jessie Xiong; George P. Souroullas; Norman E. Sharpless

In human cancer, N-RAS mutations resulting in constitutive, oncogenic signaling are predominately localized to codons 12, 13 or 61. Traditionally, activating RAS mutations have been considered oncogenically equivalent, yet recent studies suggest important clinical distinctions between colon cancers containing K-RAS codon 12 vs. codon 13 mutations. More than 20% of human melanomas harbor N-RAS mutations, the vast majority of which target codon 61, as opposed to N-RAS mutations of codon 12 or 13, which are common in other tumor types. To address this issue, we characterized syngeneic knock-in mouse models conditionally expressing either N-Ras G12D or N-Ras Q61R under the control of the endogenous locus. In primary melanocyte cultures, activation of either N-Ras allele reduced cellular proliferation even in the presence of concomitant p16 INK4a deletion. Correspondingly, melanocyte-specific N-Ras G12D expression combined with p16 INK4a loss failed to efficiently promote melanoma formation in mice. In contrast, melanocyte-specific N-Ras Q61R induction readily combined with p16 INK4a loss to promote melanoma formation in vivo with short latency and high penetrance. These results provide a first murine model of melanoma featuring knock-in N-Ras mutation Q61R , and suggest that the preference for codon 61 mutations in human tumors is conserved in mice. Citation Format: Christin Burd, William R. Jeck, Kailing Fu, Kelly S. Clark, Jessie C. Xiong, George P. Souroullas, Norman E. Sharpless. Differential oncogenicity of N-RAS mutations in melanoma. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 1087. doi:10.1158/1538-7445.AM2013-1087

Collaboration


Dive into the Kelly S. Clark's collaboration.

Top Co-Authors

Avatar

Norman E. Sharpless

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

David B. Darr

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jessie Xiong

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Charles M. Perou

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

William R. Jeck

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jamie L. Jordan

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Kailing Fu

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

William Y. Kim

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge