Network


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

Hotspot


Dive into the research topics where Gregory K. Friedman is active.

Publication


Featured researches published by Gregory K. Friedman.


Journal of Neuro-oncology | 2014

Expression of PRMT5 correlates with malignant grade in gliomas and plays a pivotal role in tumor growth in vitro

Xiaosi Han; Rong Li; Wenbin Zhang; Xiuhua Yang; Crystal G. Wheeler; Gregory K. Friedman; Paula Province; Qiang Ding; Zhiying You; Hassan M. Fathallah-Shaykh; G. Yancey Gillespie; Xinyang Zhao; Peter H. King; L. Burt Nabors

Abstract Protein arginine methyltransferase 5 (PRMT5) catalyzes the formation of ω-NG,N′G-symmetric dimethylarginine residues on histones as well as other proteins. These modifications play an important role in cell differentiation and tumor cell growth. However, the role of PRMT5 in human glioma cells has not been characterized. In this study, we assessed protein expression profiles of PRMT5 in control brain, WHO grade II astrocytomas, anaplastic astrocytomas, and glioblastoma multiforme (GBM) by immunohistochemistry. PRMT5 was low in glial cells in control brain tissues and low grade astrocytomas. Its expression increased in parallel with malignant progression, and was highly expressed in GBM. Knockdown of PRMT5 by small hairpin RNA caused alterations of p-ERK1/2 and significantly repressed the clonogenic potential and viability of glioma cells. These findings indicate that PRMT5 is a marker of malignant progression in glioma tumors and plays a pivotal role in tumor growth.


Cancers | 2011

Cancer Stem Cells and Pediatric Solid Tumors

Gregory K. Friedman; G. Yancey Gillespie

Recently, a subpopulation of cells, termed tumor-initiating cells or tumor stem cells (TSC), has been identified in many different types of solid tumors. These TSC, which are typically more resistant to chemotherapy and radiation compared to other tumor cells, have properties similar to normal stem cells including multipotency and the ability to self-renew, proliferate, and maintain the neoplastic clone. Much of the research on TSC has focused on adult cancers. With considerable differences in tumor biology between adult and pediatric cancers, there may be significant differences in the presence, function and behavior of TSC in pediatric malignancies. We discuss what is currently known about pediatric solid TSC with specific focus on TSC markers, tumor microenvironment, signaling pathways, therapeutic resistance and potential future therapies to target pediatric TSC.


Molecular Therapy | 2009

Herpes Simplex Virus Oncolytic Therapy for Pediatric Malignancies

Gregory K. Friedman; Joseph G. Pressey; Alyssa T. Reddy; James M. Markert; G. Yancey Gillespie

Despite improving survival rates for children with cancer, a subset of patients exist with disease resistant to traditional therapies such as surgery, chemotherapy, and radiation. These patients require newer, targeted treatments used alone or in combination with more traditional approaches. Oncolytic herpes simplex virus (HSV) is one of these newer therapies that offer promise for several difficult to treat pediatric malignancies. The potential benefit of HSV therapy in pediatric solid tumors including brain tumors, neuroblastomas, and sarcomas is reviewed along with the many challenges that need to be addressed prior to moving oncolytic HSV therapy from the laboratory to the beside in the pediatric population.


Pediatric Blood & Cancer | 2010

Complete response to carboplatin, gemcitabine, and paclitaxel in a patient with advanced metastatic renal medullary carcinoma.

Alexandra Walsh; David R. Kelly; Yoginder N. Vaid; Lee Hilliard; Gregory K. Friedman

Renal medullary carcinoma (RMC) is a rare and aggressive malignancy seen primarily in patients with sickle‐cell trait. We report a complete response to carboplatin, paclitaxel, and gemcitabine in a patient with advanced metastatic RMC. Pediatr Blood Cancer. 2010;55:1217–1220.


Pediatric Research | 2012

Targeting Pediatric Cancer Stem Cells with Oncolytic Virotherapy

Gregory K. Friedman; Kevin A. Cassady; Elizabeth A. Beierle; James M. Markert; G. Yancey Gillespie

Cancer stem cells (CSCs), also termed “cancer-initiating cells” or “cancer progenitor cells,” which have the ability to self-renew, proliferate, and maintain the neoplastic clone, have recently been discovered in a wide variety of pediatric tumors. These CSCs are thought to be responsible for tumorigenesis and tumor maintenance, aggressiveness, and recurrence due to inherent resistance to current treatment modalities such as chemotherapy and radiation. Oncolytic virotherapy offers a novel, targeted approach for eradicating pediatric CSCs using mechanisms of cell killing that differ from conventional therapies. Moreover, oncolytic viruses have the ability to target specific features of CSCs such as cell-surface proteins, transcription factors, and the CSC microenvironment. Through genetic engineering, a wide variety of foreign genes may be expressed by oncolytic viruses to augment the oncolytic effect. We review the current data regarding the ability of several types of oncolytic viruses (herpes simplex virus-1, adenovirus, reovirus, Seneca Valley virus, vaccinia virus, Newcastle disease virus, myxoma virus, vesicular stomatitis virus) to target and kill both CSCs and tumor cells in pediatric tumors. We highlight advantages and limitations of each virus and potential ways in which next-generation engineered viruses may target resilient CSCs.


Pediatric Blood & Cancer | 2007

Changing trends of research and treatment in infant neuroblastoma.

Gregory K. Friedman; Robert P. Castleberry

Neuroblastoma is the most common malignancy in infants and 40% of neuroblastomas are diagnosed in the first year of life. While generally neuroblastoma behaves less aggressively in this age group, tumors that have adverse biologic characteristics do not differ in their behavior from counterparts in older children. Clinical and biologic behavior of neuroblastoma in children up to 460 days of age is similar to that in children less than 1 year of age. Thus the categorization of children up to 18 months of age into risk category is critically dependent on biologic characterization and assignment to appropriate treatment intensity categories. Pediatr Blood Cancer 2007;49:1060–1065.


Pediatric Blood & Cancer | 2013

CD133 Marks a Myogenically Primitive Subpopulation in Rhabdomyosarcoma Cell Lines that are Relatively Chemoresistant but Sensitive to Mutant HSV

Joseph G. Pressey; Marilyn C. Haas; Christine S. Pressey; Virginia M. Kelly; Jacqueline N. Parker; G. Yancey Gillespie; Gregory K. Friedman

Rhabdomyosarcoma (RMS) is characterized by features of skeletal muscle and is comprised of two major histological subtypes, embryonal (E‐RMS), and alveolar (A‐RMS). Subsets of each RMS subtype demonstrate resistance to multimodal therapy leading to treatment failure. Cancer stem cells or cancer‐initiating cells (CIC) represent a theorized population of cells that give rise to tumors and are responsible for treatment resistance.


Neurotherapeutics | 2017

Oncolytic Virotherapy for the Treatment of Malignant Glioma

Paul M. Foreman; Gregory K. Friedman; Kevin A. Cassady; James M. Markert

Malignant glioma is the most common primary brain tumor and carries a grim prognosis, with a median survival of just over 14 months. Given the poor outcomes with standard-of-care treatments, novel treatment strategies are needed. The concept of virotherapy for the treatment of malignant tumors dates back more than a century and can be divided into replication-competent oncolytic viruses and replication-deficient viral vectors. Oncolytic viruses are designed to selectively target, infect, and replicate in tumor cells, while sparing surrounding normal brain. A host of oncolytic viruses has been evaluated in early phase human trials with promising safety results, but none has progressed to phase III trials. Despite the 25 years that has passed since the initial publication of genetically engineered oncolytic viruses for the treatment of glioma, much remains to be learned about the use of this therapy, including its mechanism of action, optimal treatment paradigm, appropriate targets, and integration with adjuvant agents. Oncolytic viral therapy for glioma remains promising and will undoubtedly impact the future of patient care.


Neuro-oncology | 2016

Pediatric medulloblastoma xenografts including molecular subgroup 3 and CD133+ and CD15+ cells are sensitive to killing by oncolytic herpes simplex viruses.

Gregory K. Friedman; Blake P. Moore; Li Nan; Virginia M. Kelly; Tina Etminan; Catherine P. Langford; Hui Xu; Xiaosi Han; James M. Markert; Elizabeth A. Beierle; G. Yancey Gillespie

BACKGROUND Childhood medulloblastoma is associated with significant morbidity and mortality that is compounded by neurotoxicity for the developing brain caused by current therapies, including surgery, craniospinal radiation, and chemotherapy. Innate therapeutic resistance of some aggressive pediatric medulloblastoma has been attributed to a subpopulation of cells, termed cancer-initiating cells or cancer stemlike cells (CSCs), marked by the surface protein CD133 or CD15. Brain tumors characteristically contain areas of pathophysiologic hypoxia, which has been shown to drive the CSC phenotype leading to heightened invasiveness, angiogenesis, and metastasis. Novel therapies that target medulloblastoma CSCs are needed to improve outcomes and decrease toxicity. We hypothesized that oncolytic engineered herpes simplex virus (oHSV) therapy could effectively infect and kill pediatric medulloblastoma cells, including CSCs marked by CD133 or CD15. METHODS Using 4 human pediatric medulloblastoma xenografts, including 3 molecular subgroup 3 tumors, which portend worse patient outcomes, we determined the expression of CD133, CD15, and the primary HSV-1 entry molecule nectin-1 (CD111) by fluorescence activated cell sorting (FACS) analysis. Infectability and cytotoxicity of clinically relevant oHSVs (G207 and M002) were determined in vitro and in vivo by FACS, immunofluorescent staining, cytotoxicity assays, and murine survival studies. RESULTS We demonstrate that hypoxia increased the CD133+ cell fraction, while having the opposite effect on CD15 expression. We established that all 4 xenografts, including the CSCs, expressed CD111 and were highly sensitive to killing by G207 or M002. CONCLUSIONS Pediatric medulloblastoma, including Group 3 tumors, may be an excellent target for oHSV virotherapy, and a clinical trial in medulloblastoma is warranted.


Oncolytic Virotherapy | 2014

Oncolytic viral therapy: targeting cancer stem cells

Tyrel T Smith; Justin C. Roth; Gregory K. Friedman; G. Yancey Gillespie

Cancer stem cells (CSCs) are defined as rare populations of tumor-initiating cancer cells that are capable of both self-renewal and differentiation. Extensive research is currently underway to develop therapeutics that target CSCs for cancer therapy, due to their critical role in tumorigenesis, as well as their resistance to chemotherapy and radiotherapy. To this end, oncolytic viruses targeting unique CSC markers, signaling pathways, or the pro-tumor CSC niche offer promising potential as CSCs-destroying agents/therapeutics. We provide a summary of existing knowledge on the biology of CSCs, including their markers and their niche thought to comprise the tumor microenvironment, and then we provide a critical analysis of the potential for targeting CSCs with oncolytic viruses, including herpes simplex virus-1, adenovirus, measles virus, reovirus, and vaccinia virus. Specifically, we review current literature regarding first-generation oncolytic viruses with their innate ability to replicate in CSCs, as well as second-generation viruses engineered to enhance the oncolytic effect and CSC-targeting through transgene expression.

Collaboration


Dive into the Gregory K. Friedman's collaboration.

Top Co-Authors

Avatar

James M. Markert

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

G. Yancey Gillespie

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Elizabeth A. Beierle

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

Jerry E. Stewart

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Li Nan

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Virginia M. Kelly

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

George Yancey Gillespie

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

Alyssa T. Reddy

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge