Network


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

Hotspot


Dive into the research topics where Laura L. Stafman is active.

Publication


Featured researches published by Laura L. Stafman.


Translational Oncology | 2016

Effect of Repeat Dosing of Engineered Oncolytic Herpes Simplex Virus on Preclinical Models of Rhabdomyosarcoma

Alicia M. Waters; Laura L. Stafman; Evan F. Garner; Smitha Mruthyunjayappa; Jerry E. Stewart; Gregory K. Friedman; Jennifer M. Coleman; James M. Markert; G. Yancey Gillespie; Elizabeth A. Beierle

Rhabdomyosarcoma (RMS), a tumor of skeletal muscle origin, is the most common sarcoma of childhood. Despite multidrug chemotherapy regimens, surgical intervention, and radiation treatment, outcomes remain poor, especially in advanced disease, and novel therapies are needed for the treatment of these aggressive malignancies. Genetically engineered oncolytic viruses, such as herpes simplex virus-1 (HSV), are currently being explored as treatments for pediatric tumors. M002, an oncolytic HSV, has both copies of the γ134.5 gene deleted, enabling replication in tumor cells but thwarting infection of normal, postmitotic cells. We hypothesized that M002 would infect human RMS tumor cells and lead to decreased tumor cell survival in vitro and impede tumor growth in vivo. In the current study, we demonstrated that M002 could infect, replicate in, and decrease cell survival in both embryonal (ERMS) and alveolar rhabdomyosarcoma (ARMS) cells. Additionally, M002 reduced xenograft tumor growth and increased animal survival in both ARMS and ERMS. Most importantly, we showed for the first time that repeated dosing of oncolytic virus coupled with low-dose radiation provided improved tumor response in RMS. These findings provide support for the clinical investigation of oncolytic HSV in pediatric RMS.


Cancers | 2016

Cell Proliferation in Neuroblastoma

Laura L. Stafman; Elizabeth A. Beierle

Neuroblastoma, the most common extracranial solid tumor of childhood, continues to carry a dismal prognosis for children diagnosed with advanced stage or relapsed disease. This review focuses upon factors responsible for cell proliferation in neuroblastoma including transcription factors, kinases, and regulators of the cell cycle. Novel therapeutic strategies directed toward these targets in neuroblastoma are discussed.


Translational Oncology | 2016

Targeting Focal Adhesion Kinase Suppresses the Malignant Phenotype in Rhabdomyosarcoma Cells

Alicia M. Waters; Laura L. Stafman; Evan F. Garner; Smitha Mruthyunjayappa; Jerry E. Stewart; Elizabeth Mroczek-Musulman; Elizabeth A. Beierle

Despite the tremendous advances in the treatment of childhood solid tumors, rhabdomyosarcoma (RMS) continues to provide a therapeutic challenge. Children with metastatic or relapsed disease have a disease-free survival rate under 30%. Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase that is important in many facets of tumorigenesis. Signaling pathways both upstream and downstream to FAK have been found to be important in sarcoma tumorigenesis, leading us to hypothesize that FAK would be present in RMS and would impact cellular survival. In the current study, we showed that FAK was present and phosphorylated in pediatric alveolar and embryonal RMS tumor specimens and cell lines. We also examined the effects of FAK inhibition upon two RMS cell lines utilizing parallel approaches including RNAi and small molecule inhibitors. FAK inhibition resulted in decreased cellular survival, invasion, and migration and increased apoptosis. Furthermore, small molecule inhibition of FAK led to decreased tumor growth in a nude mouse RMS xenograft model. The findings from this study will help to further our understanding of the regulation of tumorigenesis in RMS and may provide desperately needed novel therapeutic strategies for these difficult-to-treat tumors.


Translational Oncology | 2019

Investigation of PP2A and Its Endogenous Inhibitors in Neuroblastoma Cell Survival and Tumor Growth

Adele P. Williams; Evan F. Garner; Alicia M. Waters; Laura L. Stafman; Jamie Aye; Hooper Markert; Jerry E. Stewart; Elizabeth A. Beierle

High-risk neuroblastoma continues to carry a poor prognosis. Nearly 50% of these tumors relapse following extensive treatment regimens. Protein phosphatase 2A (PP2A), a tumor suppressor, has been shown to be downregulated in many human cancers via multiple mechanisms including upregulation of its endogenous inhibitors, I2PP2A or CIP2A. We hypothesized that inhibition of the endogenous PP2A inhibitors or activation of PP2A would decrease tumorigenicity in human neuroblastoma cells. Four human neuroblastoma cell lines were utilized. Expression of PP2A and its endogenous inhibitors I2PP2A and CIP2A was confirmed by immunoblotting. PP2A activation was measured via phosphatase activation assay. Multiple parallel methods including siRNA inhibition of the endogenous PP2A inhibitors and pharmacologic activation of PP2A were utilized. Cell viability, proliferation, migration, and invasion assays were performed. In vivo studies were utilized to determine the effects of PP2A activation on neuroblastoma tumor growth. Inhibition of the endogenous inhibitors of PP2A or pharmacologic activation of PP2A with the PP2A activator FTY720 led to decreased neuroblastoma cell viability, proliferation, migration, and invasion. Treatment of mice bearing SK-N-AS or SK-N-BE(2) neuroblastoma tumors with FTY720 resulted in a significant decrease in tumor growth compared to vehicle-treated animals. In conclusion, activation of PP2A may provide a novel therapeutic target for neuroblastoma.


Scientific Reports | 2018

FTY720 Decreases Tumorigenesis in Group 3 Medulloblastoma Patient-Derived Xenografts

Evan F. Garner; Adele P. Williams; Laura L. Stafman; Jamie Aye; Elizabeth Mroczek-Musulman; Blake P. Moore; Jerry E. Stewart; Gregory K. Friedman; Elizabeth A. Beierle

Group 3 tumors account for 28% of medulloblastomas and have the worst prognosis. FTY720, an immunosuppressant currently approved for treatment of multiple sclerosis, has shown antitumor effects in several human cancer cell lines. We hypothesized that treatment with FTY720 (fingolimod) would decrease tumorigenicity in medulloblastoma patient-derived xenografts (PDXs). Three Group 3 medulloblastoma PDXs (D341, D384 and D425) were utilized. Expression of PP2A and its endogenous inhibitors I2PP2A and CIP2A was detected by immunohistochemistry and immunoblotting. PP2A activation was measured via phosphatase activation kit. Cell viability, proliferation, migration and invasion assays were performed after treatment with FTY720. Cell cycle analysis was completed using flow cytometry. A flank model using D425 human medulloblastoma PDX cells was used to assess the in vivo effects of FTY720. FTY720 activated PP2A and led to decreased medulloblastoma PDX cell viability, proliferation, migration and invasion and G1 cell cycle arrest in all three PDXs. FTY720 treatment of mice bearing D425 medulloblastoma PDX tumors resulted in a significant decrease in tumor growth compared to vehicle treated animals. FTY720 decreased viability, proliferation, and motility in Group 3 medulloblastoma PDX cells and significantly decreased tumor growth in vivo. These results suggest that FTY720 should be investigated further as a potential therapeutic agent for medulloblastoma.


Oncotarget | 2018

Targeting PIM kinase as a therapeutic strategy in human hepatoblastoma

Laura L. Stafman; Smitha Mruthyunjayappa; Alicia M. Waters; Evan F. Garner; Jamie Aye; Jerry E. Stewart; Karina J. Yoon; Kimberly Whelan; Elizabeth Mroczek-Musulman; Elizabeth A. Beierle

Increasing incidence coupled with poor prognosis and treatments that are virtually unchanged over the past 20 years have made the need for the development of novel therapeutics for hepatoblastoma imperative. PIM kinases have been implicated as drivers of tumorigenesis in multiple cancers, including hepatocellular carcinoma. We hypothesized that PIM kinases, specifically PIM3, would play a role in hepatoblastoma tumorigenesis and that PIM kinase inhibition would affect hepatoblastoma in vitro and in vivo. Parameters including cell survival, proliferation, motility, and apoptosis were assessed in human hepatoblastoma cells following PIM3 knockdown with siRNA or treatment with the PIM inhibitor AZD1208. An in vivo model of human hepatoblastoma was utilized to study the effects of PIM inhibition alone and in combination with cisplatin. PIM kinases were found to be present in the human hepatoblastoma cell line, HuH6, and in a human hepatoblastoma patient-derived xenograft, COA67. PIM3 knockdown or inhibition with AZD1208 decreased cell survival, attachment independent growth, and motility. Additionally, inhibition of tumor growth was observed in a hepatoblastoma xenograft model in mice treated with AZD1208. Combination therapy with AZD1208 and cisplatin resulted in a significant increase in animal survival when compared to either treatment alone. The current studies showed that PIM kinase inhibition decreased human hepatoblastoma tumorigenicity both in vitro and in vivo, implying that PIM inhibitors may be useful as a novel therapeutic for children with hepatoblastoma.


Journal of Pediatric Surgery | 2018

Corruption of Neuroblastoma Patient Derived Xenografts with Human T Cell Lymphoma

Adele P. Williams; Jerry E. Stewart; Laura L. Stafman; Jamie Aye; Elizabeth Mroczek-Musulman; Changchun Ren; Karina Yoon; Kimberly Whelan; Elizabeth A. Beierle

BACKGROUND Patient derived xenografts (PDXs) provide a unique opportunity for investigators to study tumor cell activity, response to therapeutics, and resistance patterns without exposing the human patient to experimental compounds, and thereby play a crucial role in pre-clinical evaluation of new therapies. It has been reported that PDXs may undergo a transformation to lymphoma, most commonly associated with Epstein Barr virus (EBV). If the character of a xenograft becomes compromised and remains undetected, it could have a detrimental impact on the research community as a whole. Our lab has established a number of pediatric solid tumor PDXs which accurately recapitulate the human tumors following several passages. One particular neuroblastoma PDX was noted to grow quickly and with an unusual phenotype, leading us to hypothesize that this PDX had undergone a transformation. METHODS The PDX in question was investigated with histology, immunohistochemistry (IHC), EBER in situ hybridization, and PCR to determine its identity. RESULTS Histology on the tumor revealed a small, round blue cell tumor similar to the original neuroblastoma from which it was derived. IHC staining showed that the tumor was composed of lymphocytes that were CD3 positive, <5% CD4 positive, and CD20 negative. The cells were Epstein Barr virus negative. PCR demonstrated that the tumor was human and not murine in origin. CONCLUSION These findings indicate that a human T Cell lymphoma developed in place of this neuroblastoma PDX. Changes in PDX identity such as this one will significantly impact studies utilizing pediatric PDXs and the mechanism by which this occurred warrants further investigation.


Journal of Neuro-oncology | 2018

UAB30, a novel RXR agonist, decreases tumorigenesis and leptomeningeal disease in group 3 medulloblastoma patient-derived xenografts

Evan F. Garner; Laura L. Stafman; Adele P. Williams; Jamie Aye; Caroline Goolsby; Venkatram R. Atigadda; Blake P. Moore; Li Nan; Jerry E. Stewart; Anita B. Hjelmeland; Gregory K. Friedman; Elizabeth A. Beierle

BackgroundGroup 3 tumors account for approximately 25–30% of medulloblastomas and have the worst prognosis. UAB30 is a novel synthetic rexinoid shown to have limited toxicities in humans and significant efficacy in the pediatric neuroectodermal tumor, neuroblastoma. We hypothesized that treatment with UAB30 would decrease tumorigenicity in medulloblastoma patient-derived xenografts (PDXs).MethodsThree group 3 medulloblastoma PDXs (D341, D384 and D425) were utilized. Cell viability, proliferation, migration and invasion assays were performed after treatment with UAB30 or 13-cis-retinoic acid (RA). Cell cycle analysis was completed using flow cytometry. A flank model, a cerebellar model, and a model of leptomeningeal metastasis using human medulloblastoma PDX cells was used to assess the in vivo effects of UAB30 and RA.ResultsUAB30 treatment led to cell differentiation and decreased medulloblastoma PDX cell viability, proliferation, migration and invasion and G1 cell cycle arrest in all three PDXs similar to RA. UAB30 and RA treatment of mice bearing medulloblastoma PDX tumors resulted in a significant decrease in tumor growth and metastasis compared to vehicle treated animals.ConclusionsUAB30 decreased viability, proliferation, and motility in group 3 medulloblastoma PDX cells and significantly decreased tumor growth in vivo in a fashion similar to RA, suggesting that further investigations into the potential therapeutic application of UAB30 for medulloblastoma are warranted.


Journal of Surgical Research | 2018

Socioeconomic disparities affect survival in malignant ovarian germ cell tumors in AYA population

Laura V. Bownes; Laura L. Stafman; Ilan I. Maizlin; Matthew Dellinger; Kenneth W. Gow; Adam B. Goldin; Melanie Goldfarb; Monica Langer; Mehul V. Raval; John J. Doski; Jed G. Nuchtern; Sanjeev A. Vasudevan; Elizabeth A. Beierle


Journal of The American College of Surgeons | 2018

FTY720 Enhances the Anti-Tumor Effects of Doxorubicin on Neuroblastoma Cells in Vitro

Adele P. Williams; Laura L. Stafman; Jamie Aye; Jerry E. Stewart; Elizabeth A. Beierle

Collaboration


Dive into the Laura L. Stafman's collaboration.

Top Co-Authors

Avatar

Elizabeth A. Beierle

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Jerry E. Stewart

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Evan F. Garner

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Jamie Aye

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Adele P. Williams

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Smitha Mruthyunjayappa

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Alicia M. Waters

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Blake P. Moore

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Gregory K. Friedman

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Karina J. Yoon

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge