Network


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

Hotspot


Dive into the research topics where Gary L. Clayman is active.

Publication


Featured researches published by Gary L. Clayman.


Journal of Clinical Oncology | 1998

Adenovirus-mediated p53 gene transfer in patients with advanced recurrent head and neck squamous cell carcinoma.

Gary L. Clayman; Adel K. El-Naggar; Scott M. Lippman; Ying C. Henderson; Mitchell J. Frederick; James Merritt; Louis A. Zumstein; Therese M. Timmons; Ta Jen Liu; Lawrence E. Ginsberg; Jack A. Roth; Waun Ki Hong; Patricia Bruso; Helmuth Goepfert

PURPOSE Standard therapies of head and neck squamous cell carcinoma (HNSCC) often cause profound morbidity and have not significantly improved survival over the last 30 years. Preclinical studies showed that adenoviral vector delivery of the wild-type p53 gene reduced tumor growth in mouse xenograft models. Our purpose was to ascertain the safety and therapeutic potential of adenoviral (Ad)-p53 in advanced HNSCC. PATIENTS AND METHODS Patients with incurable recurrent local or regionally metastatic HNSCC received multiple intratumoral injections of Ad-p53, either with or without tumor resection. Patients were monitored for adverse events and antiadenoviral antibodies, tumors were monitored for response and p53 expression, and body fluids were analyzed for Ad-p53. RESULTS Tumors of 33 patients were injected with doses of up to 1 x 10(11) plaque-forming units (pfu). No dose-limiting toxicity or serious adverse events were noted. p53 expression was detected in tumor biopsies despite antibody responses after Ad-p53 injections. Clinical efficacy could be evaluated in 17 patients with nonresectable tumors: two patients showed objective tumor regressions of greater than 50%, six patients showed stable disease for up to 3.5 months, and nine patients showed progressive disease. One resectable patient was considered a complete pathologic response. Ad-p53 was detected in blood and urine in a dose-dependent fashion, and in sputum. CONCLUSION Patients were safely injected intratumorally with Ad-p53. Objective antitumor activity was detected in several patients. The infectious Ad-p53 in body fluids was asymptomatic, and suggests that systemic or regional treatment may be tolerable. These results suggest the further investigation of Ad-p53 as a therapeutic agent for patients with HNSCC.


Journal of Biological Chemistry | 2006

A POTENTIAL ROLE FOR MULTIPLE TISSUE KALLIKREIN SERINE PROTEASES IN EPIDERMAL DESQUAMATION

Carla A. Borgoño; Iacovos P. Michael; Nahoko Komatsu; Arumugam Jayakumar; Ravi M. Kapadia; Gary L. Clayman; Georgia Sotiropoulou; Eleftherios P. Diamandis

Desquamation of the stratum corneum is a serine protease-dependent process. Two members of the human tissue kallikrein (KLK) family of (chymo)tryptic-like serine proteases, KLK5 and KLK7, are implicated in desquamation by digestion of (corneo)desmosomes and inhibition by desquamation-related serine protease inhibitors (SPIs). However, the epidermal localization and specificity of additional KLKs also supports a role for these enzymes in desquamation. This study aims to delineate the probable contribution of KLK1, KLK5, KLK6, KLK13, and KLK14 to desquamation by examining their interactions, in vitro, with: 1) colocalized SPI, lympho-epithelial Kazal-type-related inhibitor (LEKTI, four recombinant fragments containing inhibitory domains 1–6 (rLEKTI(1–6)), domains 6–8 and partial domain 9 (rLEKTI(6–9′)), domains 9–12 (rLEKTI(9–12)), and domains 12–15 (rLEKTI(12–15)), secretory leukocyte protease inhibitor, and elafin and 2) their ability to digest the (corneo)desmosomal cadherin, desmoglein 1. KLK1 was not inhibited by any SPI tested. KLK5, KLK6, KLK13, and KLK14 were potently inhibited by rLEKTI(1–6), rLEKTI(6–9′), and rLEKTI(9–12) with Ki values in the range of 2.3–28.4 nm, 6.1–221 nm, and 2.7–416 nm for each respective fragment. Only KLK5 was inhibited by rLEKTI(12–15) (Ki = 21.8 nm). No KLK was inhibited by secretory leukocyte protease inhibitor or elafin. Apart from KLK13, all KLKs digested the ectodomain of desmoglein 1 within cadherin repeats, Ca2+ binding sites, or in the juxtamembrane region. Our study indicates that multiple KLKs may participate in desquamation through cleavage of desmoglein 1 and regulation by LEKTI. These findings may have clinical implications for the treatment of skin disorders in which KLK activity is elevated.


Laryngoscope | 2005

Lymph Node Metastases from Cutaneous Squamous Cell Carcinoma of the Head and Neck

Brian A. Moore; Randal S. Weber; Victor G. Prieto; Adel K. El-Naggar; F. Christopher Holsinger; Xian Zhou; J. Jack Lee; Scott M. Lippman; Gary L. Clayman

Objective/Hypothesis: Cutaneous squamous cell carcinoma (CSCC) has been reported to metastasize to parotid and cervical lymph nodes. Few prospective investigations of associated clinical and histopathologic findings and their effect on patient outcomes exist. We seek to identify risk factors for nodal metastases in CSCC and determine the impact of lymphatic spread on survival and recurrence.


Cancer Research | 2004

BRAK/CXCL14 Is a Potent Inhibitor of Angiogenesis and a Chemotactic Factor for Immature Dendritic Cells

Thomas D. Shellenberger; Mary Wang; Manu Gujrati; Arumugam Jayakumar; Robert M. Strieter; Marie D. Burdick; Constantin G. Ioannides; Clayton L. Efferson; Adel K. El-Naggar; Dianna B. Roberts; Gary L. Clayman; Mitchell J. Frederick

BRAK/CXCL14 is a CXC chemokine constitutively expressed at the mRNA level in certain normal tissues but absent from many established tumor cell lines and human cancers. Although multiple investigators cloned BRAK, little is known regarding the physiologic function of BRAK or the reason for decreased expression in cancer. To understand the possible significance associated with loss of BRAK mRNA in tumors, we examined the pattern of BRAK protein expression in normal and tumor specimens from patients with squamous cell carcinoma (SCC) of the tongue and used recombinant BRAK (rBRAK) to investigate potential biological functions. Using a peptide-specific antiserum, abundant expression of BRAK protein was found in suprabasal layers of normal tongue mucosa but consistently was absent in tongue SCC. Consistent with previous in situ mRNA studies, BRAK protein also was expressed strongly by stromal cells adjacent to tumors. In the rat corneal micropocket assay, BRAK was a potent inhibitor of in vivo angiogenesis stimulated by multiple angiogenic factors, including interleukin 8, basic fibroblast growth factor, and vascular endothelial growth factor. In vitro, rBRAK blocked endothelial cell chemotaxis at concentrations as low as 1 nmol/L, suggesting this was a major mechanism for angiogenesis inhibition. Although only low affinity receptors for BRAK could be found on endothelial cells, human immature monocyte-derived dendritic cells (iDCs) bound rBRAK with high affinity (i.e., Kd, ∼2 nmol/L). Furthermore, rBRAK was chemotactic for iDCs at concentrations ranging from 1 to 10 nmol/L. Our findings support a hypothesis that loss of BRAK expression from tumors may facilitate neovascularization and possibly contributes to immunologic escape.


Head and Neck-journal for The Sciences and Specialties of The Head and Neck | 1999

Selective neck dissections for squamous carcinoma of the upper aerodigestive tract: patterns of regional failure.

Robert M. Byers; Gary L. Clayman; Diane McGill; Todd Andrews; Ramon Paul Kare; Dianna B. Roberts; Helmuth Goepfert

Surgeons have been using selective neck dissections in the treatment of squamous carcinoma of the upper aerodigestive tract for over 20 years. To date, no data is available that can answer the question “What are the patterns of failure in the neck following a selective neck dissection and is a selective neck dissection a reliable procedure for metastatic disease?”


Oncogene | 1997

Frequent inactivation of p16(INK4α) in oral premalignant lesions

Vali Papadimitrakopoulou; Julie Izzo; Scott M. Lippman; Jin Soo Lee; You Hong Fan; Gary L. Clayman; Jay Y. Ro; Walter N. Hittelman; Reuben Lotan; Waun Ki Hong; Li Mao

Head and neck carcinogenesis is believed to be a multistep process, whereby genetic events accumulate in the carcinogen-exposed field at risk, resulting in distinct phenotypic premalignant changes that eventually evolve into invasive cancer. Frequent loss of heterozygosity (LOH) at the chromosome 9p21 region and inactivation of p16INK4a by different mechanisms have been described in head and neck squamous cell carcinoma (HNSCC). Recently, we reported that loss of 9p21 is also frequent in oral premalignant lesions. To investigate potential inactivation of p16INK4a in these premalignant lesions, we analysed 74 biopsies from 36 patients by immunohistochemistry (IHC) for expression of the p16 protein. Loss of p16 expression was found in 28 (38%) of the lesion biopsies from 17 patients (47%). LOH at the D9s171, a marker in the 9p21 region, was observed in 19 lesion biopsies from 12 cases and correlated with absence of p16 by IHC in 11 (92%) of the 12 comparable cases and 15 (79%) of 19 lesion biopsies. By direct sequencing of ten lesion biopsies from ten individuals with LOH at D9s171 for p16INK4a exon 2, one non-sense mutation at codon 88 (GGA→TGA) was identified. Our data suggest that inactivation of p16INK4a may play an important role in early head and neck cancer development.


American Journal of Pathology | 2000

In vivo expression of the novel CXC chemokine BRAK in normal and cancerous human tissue.

Mitchell J. Frederick; Ying C. Henderson; Xiao Chun Xu; Michael T. Deavers; Aysegul A. Sahin; Hong Wu; Dorothy E. Lewis; Adel K. El-Naggar; Gary L. Clayman

Using differential display, we cloned a gene with reduced expression in short-term explants of head and neck squamous cell carcinoma (HNSCC) tumors compared to cultured normal oral epithelial cells. The differentially expressed gene was identical to the recently cloned CXC chemokine BRAK, which is ubiquitously expressed in normal tissue extracts but is absent from many tumor cell lines in vitro. To define the cell populations expressing BRAK in vivo, in situ mRNA hybridization was performed on normal and cancerous tissues from six different histological sites. The predominant normal cell type constitutively expressing BRAK in vivo was squamous epithelium. Expression in tumors was heterogeneous, with the majority of HNSCCs and some cervical squamous cell carcinomas (SCCs) showing loss of BRAK mRNA. Although absent in unstimulated peripheral blood mononuclear cells, high levels of BRAK were consistently found in infiltrating inflammatory cells (with lymphocyte morphology) in nearly all cancers examined. Furthermore, BRAK expression was demonstrated in B cells and monocytes, after stimulation of peripheral blood mononuclear cells with lipopolysaccharide. This study demonstrates for the first time up-regulation of BRAK mRNA by inflammatory cells in the tumor microenvironment and lost expression from certain cancers in vivo. The data suggest that BRAK may have a role in host-tumor interactions.


Head and Neck-journal for The Sciences and Specialties of The Head and Neck | 2004

Parathyroid carcinoma: A 22-year experience

Naifa L. Busaidy; Camilo Jimenez; Mouhammed Amir Habra; Pamela N. Schultz; Adel K. El-Naggar; Gary L. Clayman; Joshua A. Asper; Eduardo M. Diaz; Douglas B. Evans; Robert F. Gagel; Adam S. Garden; Ana O. Hoff; Jeffrey E. Lee; William H. Morrison; David I. Rosenthal; Steven I. Sherman; Erich M. Sturgis; Steven G. Waguespack; Randal S. Weber; Kelly L. Wirfel; Rena Vassilopoulou-Sellin

Because parathyroid carcinoma is rare, clear consensus is not available regarding the optimal management of patients with this condition. Treatment strategies generally derive from clinical and anecdotal experiences. We report our experience with this entity.


Clinical Cancer Research | 2011

Assembly and Initial Characterization of a Panel of 85 Genomically Validated Cell Lines from Diverse Head and Neck Tumor Sites

Mei Zhao; Daisuke Sano; Curtis R. Pickering; Samar A. Jasser; Ying C. Henderson; Gary L. Clayman; Erich M. Sturgis; Thomas J. Ow; Reuben Lotan; Thomas E. Carey; Peter G. Sacks; Jennifer R. Grandis; David Sidransky; Nils Erik Heldin; Jeffrey N. Myers

Purpose: Human cell lines are useful for studying cancer biology and preclinically modeling cancer therapy, but can be misidentified and cross-contamination is unfortunately common. The purpose of this study was to develop a panel of validated head and neck cell lines representing the spectrum of tissue sites and histologies that could be used for studying the molecular, genetic, and phenotypic diversity of head and neck cancer. Methods: A panel of 122 clinically and phenotypically diverse head and neck cell lines from head and neck squamous cell carcinoma, thyroid cancer, cutaneous squamous cell carcinoma, adenoid cystic carcinoma, oral leukoplakia, immortalized primary keratinocytes, and normal epithelium was assembled from the collections of several individuals and institutions. Authenticity was verified by carrying out short tandem repeat analysis. Human papillomavirus (HPV) status and cell morphology were also determined. Results: Eighty-five of the 122 cell lines had unique genetic profiles. HPV-16 DNA was detected in 2 cell lines. These 85 cell lines included cell lines from the major head and neck primary tumor sites, and close examination shows a wide range of in vitro phenotypes. Conclusions: This panel of 85 genomically validated head and neck cell lines represents a valuable resource for the head and neck cancer research community that can help advance understanding of the disease by providing a standard reference for cell lines that can be used for biological as well as preclinical studies. Clin Cancer Res; 17(23); 7248–64. ©2011 AACR.


Clinical Cancer Research | 2004

Gene Expression Profiling of Differentiated Thyroid Neoplasms Diagnostic and Clinical Implications

Sylvie Chevillard; Nicolas Ugolin; Philippe Vielh; Katherine Ory; Céline Levalois; Danielle D. Elliott; Gary L. Clayman; Adel K. El-Naggar

Purpose: The purpose of this research was to identify novel genes that can be targeted as diagnostic and clinical markers of differentiated thyroid tumors. Experimental Design: Gene expression analysis using microarray platform was performed on 6 pathologically normal thyroid samples and 12 primary follicular and papillary thyroid neoplasms. Microarrays containing probes for 5,760 human full-length cDNAs were used for hybridization with total RNA from normal and tumor thyroid samples labeled with Cy3-dUTP and Cy5-dUTP, respectively. Scanned array images were recorded, and data analysis was performed. Selected sets of differentially expressed genes were analyzed using quantitative real-time reverse transcription-PCR for verification. Results: We identified 155 genes that differentiate histologically normal thyroid tissues from benign and malignant thyroid neoplasms. Of these 75 genes were differentiated between follicular neoplasms (adenoma and carcinoma) and the follicular variant of papillary carcinoma. Purely follicular neoplasms (adenomas and carcinomas) shared many genetic profiles, and only 43 genes were distinctly different between these tumors. Hierarchical cluster analysis also differentiated conventional papillary carcinoma from its follicular variant and follicular tumors. The differentially expressed genes were composed of members of cell differentiation, adhesion, immune response, and proliferation associated pathways. Quantitative real-time reverse transcription-PCR analysis of selected genes corroborated the microarray expression results. Conclusions: Our study show the following: (1) differences in gene expression between tumor and nontumor bearing normal thyroid tissue can be identified, (2) a set of genes differentiate follicular neoplasm from follicular variant of papillary carcinoma, (3) follicular adenoma and carcinoma share many of the differentiated genes, and (4) gene expression differences identify conventional papillary carcinoma from the follicular variant.

Collaboration


Dive into the Gary L. Clayman's collaboration.

Top Co-Authors

Avatar

Adel K. El-Naggar

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ying C. Henderson

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Scott M. Lippman

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Mitchell J. Frederick

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Helmuth Goepfert

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Randal S. Weber

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Waun Ki Hong

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Arumugam Jayakumar

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Steven I. Sherman

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Adam S. Garden

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge