Network


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

Hotspot


Dive into the research topics where Steven Seaman is active.

Publication


Featured researches published by Steven Seaman.


Cancer Research | 2004

TEM8 Interacts with the Cleaved C5 Domain of Collagen α3(VI)

Akash Nanda; Eleanor B. Carson-Walter; Steven Seaman; Thomas D. Barber; Jason Stampfl; Bert Vogelstein; Kenneth W. Kinzler; Brad St. Croix

Tumor endothelial marker (TEM)8 was uncovered as a gene expressed predominantly in tumor endothelium, and its protein product was recently identified as the receptor for anthrax toxin. Here, we demonstrate that TEM8 protein is preferentially expressed in endothelial cells of neoplastic tissue. We used the extracellular domain of TEM8 to search for ligands and identified the α3 subunit of collagen VI as an interacting partner. The TEM8-interacting region on collagen α3(VI) was mapped to its COOH-terminal C5 domain. Remarkably, collagen α3(VI) is also preferentially expressed in tumor endothelium in a pattern concordant with that of TEM8. These results suggest that the TEM8/C5 interaction may play an important biological role in tumor angiogenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2011

GPR124, an orphan G protein-coupled receptor, is required for CNS-specific vascularization and establishment of the blood–brain barrier

Mike Cullen; Mohammed K. Elzarrad; Steven Seaman; Enrique Zudaire; Janine Stevens; Mi Young Yang; Xiujie Li; Amit Chaudhary; Lihong Xu; Mary Beth Hilton; Daniel Logsdon; Emily Hsiao; Erica V. Stein; Frank Cuttitta; Diana C. Haines; Kunio Nagashima; Lino Tessarollo; Brad St. Croix

Every organ in the body requires blood vessels for efficient delivery of oxygen and nutrients, but independent vascular beds are highly specialized to meet the individual needs of specific organs. The vasculature of the brain is tightly sealed, with blood–brain barrier (BBB) properties developing coincident with neural vascularization. G protein-coupled receptor 124 (GPR124) (tumor endothelial marker 5, TEM5), an orphan member of the adhesion family of G protein-coupled receptors, was previously identified on the basis of its overexpression in tumor vasculature. Here, we show that global deletion or endothelial-specific deletion of GPR124 in mice results in embryonic lethality associated with abnormal angiogenesis of the forebrain and spinal cord. Expression of GPR124 was found to be required for invasion and migration of blood vessels into neuroepithelium, establishment of BBB properties, and expansion of the cerebral cortex. Thus, GPR124 is an important regulator of neurovasculature development and a potential drug target for cerebrovascular diseases.


Cancer Cell | 2012

TEM8/ANTXR1 Blockade Inhibits Pathological Angiogenesis and Potentiates Tumoricidal Responses against Multiple Cancer Types

Amit Chaudhary; Mary Beth Hilton; Steven Seaman; Diana C. Haines; Susan C. Stevenson; Peter LeMotte; William R. Tschantz; Xiaoyan M. Zhang; Saurabh Saha; Tony Fleming; Brad St. Croix

Current antiangiogenic agents used to treat cancer only partially inhibit neovascularization and cause normal tissue toxicities, fueling the need to identify therapeutic agents that are more selective for pathological angiogenesis. Tumor endothelial marker 8 (TEM8), also known as anthrax toxin receptor 1 (ANTXR1), is a highly conserved cell-surface protein overexpressed on tumor-infiltrating vasculature. Here we show that genetic disruption of Tem8 results in impaired growth of human tumor xenografts of diverse origin including melanoma, breast, colon, and lung cancer. Furthermore, antibodies developed against the TEM8 extracellular domain blocked anthrax intoxication, inhibited tumor-induced angiogenesis, displayed broad antitumor activity, and augmented the activity of clinically approved anticancer agents without added toxicity. Thus, TEM8 targeting may allow selective inhibition of pathological angiogenesis.


Cell Reports | 2015

GPR124 Functions as a WNT7-Specific Coactivator of Canonical β-Catenin Signaling

Ekaterina Posokhova; Animesh Shukla; Steven Seaman; Suresh Volate; Mary Beth Hilton; Bofan Wu; Holly Morris; Deborah A. Swing; Ming Zhou; Enrique Zudaire; Jeffrey S. Rubin; Brad St. Croix

G protein-coupled receptor 124 (GPR124) is an orphan receptor in the adhesion family of GPCRs, and previous global or endothelial-specific disruption of Gpr124 in mice led to defective CNS angiogenesis and blood-brain barriergenesis. Similar developmental defects were observed following dual deletion of Wnt7a/Wnt7b or deletion of β-catenin in endothelial cells, suggesting a possible relationship between GPR124 and canonical WNT signaling. Here, we show using in vitro reporter assays, mutation analysis, and genetic interaction studies in vivo that GPR124 functions as a WNT7A/WNT7B-specific costimulator of β-catenin signaling in brain endothelium. WNT7-stimulated β-catenin signaling was dependent upon GPR124s intracellular PDZ binding motif and a set of leucine-rich repeats in its extracellular domain. This study reveals a vital role for GPR124 in potentiation of WNT7-induced canonical β-catenin signaling with important implications for understanding and manipulating CNS-specific angiogenesis and blood-brain barrier-genesis.


Cancer Research | 2004

Identification of a Binding Partner for the Endothelial Cell Surface Proteins TEM7 and TEM7R

Akash Nanda; Phillip Buckhaults; Steven Seaman; Nishant Agrawal; Paula Boutin; Srinivas Shankara; Mariana Nacht; Beverly A. Teicher; Jason Stampfl; Bert Vogelstein; Kenneth W. Kinzler; Brad St. Croix

Tumor endothelial marker 7 (TEM7) was recently identified as an mRNA transcript overexpressed in the blood vessels of human solid tumors. Here, we identify several new variants of TEM7, derived by alternative splicing, that are predicted to be intracellular (TEM7-I), secreted (TEM7-S), or on the cell surface membrane (TEM7-M) of tumor endothelium. Using new antibodies against the TEM7 protein, we confirmed the predicted expression of TEM7 on the cell surface and demonstrated that TEM7-M protein, like its mRNA, is overexpressed on the endothelium of various tumor types. We then used an affinity purification strategy to search for TEM7-binding proteins and identified cortactin as a protein capable of binding to the extracellular region of both TEM7 and its closest homologue, TEM7-related (TEM7R), which is also expressed in tumor endothelium. The binding domain of cortactin was mapped to a unique nine-amino acid region in its plexin-like domain. These studies establish the overexpression of TEM7 protein in tumor endothelium and provide new opportunities for the delivery of therapeutic and imaging agents to the vessels of solid tumors.


Biochimica et Biophysica Acta | 2011

The Cell Surface Structure of Tumor Endothelial Marker 8 (TEM8) is Regulated by the Actin Cytoskeleton

Mi Young Yang; Amit Chaudhary; Steven Seaman; Jill Dunty; Janine Stevens; Mohammed K. Elzarrad; Arthur E. Frankel; Brad St. Croix

Tumor endothelial marker 8 (TEM8) is an integrin-like cell surface protein upregulated on tumor blood vessels and a potential vascular target for cancer therapy. Here, we found that the ability of an anti-TEM8 antibody, clone SB5, to recognize the extracellular domain of TEM8 on the cell surface depends on other host-cell factors. By taking advantage of SB5s ability to distinguish different forms of cell surface TEM8, we identified alpha-smooth muscle actin and transgelin, an actin binding protein, as intracellular factors able to alter TEM8 cell surface structure. Overexpression of either of these proteins in cells converted TEM8 from an SB5-exposed to an SB5-masked form and protected cells from SB5-saporin immunotoxins. Because the predominant form of TEM8 on the cell surface is not recognized by SB5, we also developed a new monoclonal antibody, called AF334, which is able to recognize both the SB5-exposed and the SB5-masked forms of TEM8. AF334-saporin selectively killed TEM8-positive cells independent of TEM8 cell surface structure. These studies reveal that TEM8 exists in different forms at the cell surface, a structure dependent on interactions with components of the actin cytoskeleton, and should aid in the rational design of the most effective diagnostic and therapeutic anti-TEM8 monoclonal antibodies.


Cancer Cell | 2017

Eradication of Tumors through Simultaneous Ablation of CD276/B7-H3-Positive Tumor Cells and Tumor Vasculature

Steven Seaman; Zhongyu Zhu; Saurabh Saha; Xiaoyan M. Zhang; Mi Young Yang; Mary Beth Hilton; Karen Morris; Christopher S. Szot; Holly Morris; Deborah A. Swing; Lino Tessarollo; Sean Smith; Sylvia Degrado; Dmitry Borkin; Nareshkumar Jain; Julia Scheiermann; Yang Feng; Yanping Wang; Jinyu Li; Dean Welsch; Gary A Decrescenzo; Amit Kumar Chaudhary; Enrique Zudaire; Kimberly D. Klarmann; Jonathan R. Keller; Dimiter S. Dimitrov; Brad St. Croix

Targeting the tumor vasculature with antibody-drug conjugates (ADCs) is a promising anti-cancer strategy that in order to be realized must overcome several obstacles, including identification of suitable targets and optimal warheads. Here, we demonstrate that the cell-surface protein CD276/B7-H3 is broadly overexpressed by multiple tumor types on both cancer cells and tumor-infiltrating blood vessels, making it a potentially ideal dual-compartment therapeutic target. In preclinical studies CD276 ADCs armed with a conventional MMAE warhead destroyed CD276-positive cancer cells, but were ineffective against tumor vasculature. In contrast, pyrrolobenzodiazepine-conjugated CD276 ADCs killed both cancer cells and tumor vasculature, eradicating large established tumors and metastases, and improving long-term overall survival. CD276-targeted dual-compartment ablation could aid in the development of highly selective broad-acting anti-cancer therapies.


Cancer Research | 2018

TEM8/ANTXR1-specific CAR T cells as a targeted therapy for triple-negative breast cancer

Tiara Byrd; Kristen Fousek; Antonella Pignata; Christopher S. Szot; Heba Samaha; Steven Seaman; Lacey E. Dobrolecki; Vita Salsman; Htoo Zarni Oo; Kevin Bielamowicz; Daniel Landi; Nino Rainusso; John Hicks; Suzanne Z. Powell; Matthew L. Baker; Winfried S. Wels; Joachim Koch; Poul H. Sorensen; Benjamin Deneen; Matthew J. Ellis; Michael T. Lewis; Meenakshi Hegde; Bradley S. Fletcher; Brad St. Croix; Nabil Ahmed

Triple-negative breast cancer (TNBC) is an aggressive disease lacking targeted therapy. In this study, we developed a CAR T cell-based immunotherapeutic strategy to target TEM8, a marker initially defined on endothelial cells in colon tumors that was discovered recently to be upregulated in TNBC. CAR T cells were developed that upon specific recognition of TEM8 secreted immunostimulatory cytokines and killed tumor endothelial cells as well as TEM8-positive TNBC cells. Notably, the TEM8 CAR T cells targeted breast cancer stem-like cells, offsetting the formation of mammospheres relative to nontransduced T cells. Adoptive transfer of TEM8 CAR T cells induced regression of established, localized patient-derived xenograft tumors, as well as lung metastatic TNBC cell line-derived xenograft tumors, by both killing TEM8+ TNBC tumor cells and targeting the tumor endothelium to block tumor neovascularization. Our findings offer a preclinical proof of concept for immunotherapeutic targeting of TEM8 as a strategy to treat TNBC.Significance: These findings offer a preclinical proof of concept for immunotherapeutic targeting of an endothelial antigen that is overexpressed in triple-negative breast cancer and the associated tumor vasculature. Cancer Res; 78(2); 489-500. ©2017 AACR.


Journal of Clinical Investigation | 2018

Tumor stroma–targeted antibody-drug conjugate triggers localized anticancer drug release

Christopher S. Szot; Saurabh Saha; Xiaoyan M. Zhang; Zhongyu Zhu; Mary Beth Hilton; Karen Morris; Steven Seaman; James M. Dunleavey; Kuo-Sheng Hsu; Guo-Jun Yu; Holly Morris; Deborah A. Swing; Diana C. Haines; Yanping Wang; Jennifer Hwang; Yang Feng; Dean Welsch; Gary A Decrescenzo; Amit Kumar Chaudhary; Enrique Zudaire; Dimiter S. Dimitrov; Brad St. Croix

&NA; Although nonmalignant stromal cells facilitate tumor growth and can occupy up to 90% of a solid tumor mass, better strategies to exploit these cells for improved cancer therapy are needed. Here, we describe a potent MMAE‐linked antibody‐drug conjugate (ADC) targeting tumor endothelial marker 8 (TEM8, also known as ANTXR1), a highly conserved transmembrane receptor broadly overexpressed on cancer‐associated fibroblasts, endothelium, and pericytes. Anti‐TEM8 ADC elicited potent anticancer activity through an unexpected killing mechanism we term DAaRTS (drug activation and release through stroma), whereby the tumor microenvironment localizes active drug at the tumor site. Following capture of ADC prodrug from the circulation, tumor‐associated stromal cells release active MMAE free drug, killing nearby proliferating tumor cells in a target‐independent manner. In preclinical studies, ADC treatment was well tolerated and induced regression and often eradication of multiple solid tumor types, blocked metastatic growth, and prolonged overall survival. By exploiting TEM8+ tumor stroma for targeted drug activation, these studies reveal a drug delivery strategy with potential to augment therapies against multiple cancer types.


bioRxiv | 2018

High-resolution structural genomics reveals new therapeutic vulnerabilities in glioblastoma

Michael J Johnston; Ana Nikolic; Nicoletta Ninkovic; Paul Guilhamon; Florence Mg Cavalli; Steven Seaman; Franz J. Zemp; John Lee; Aly Abdelkareem; Katrina Ellestad; Alex Murison; Michelle M Kushida; Fiona J Coutinho; Yussanne Ma; Andrew J. Mungall; Richard A. Moore; Marco A. Marra; Michael D. Taylor; Peter Dirks; Trevor J. Pugh; Sorana Morrissy; Bradley St Croix; Douglas J. Mahoney; Mathieu Lupien; Marco Gallo

We investigated the role of 3D genome architecture in instructing functional properties of glioblastoma stem cells (GSCs) by generating the highest-resolution 3D genome maps to-date for this cancer. Integration of DNA contact maps with chromatin and transcriptional profiles identified specific mechanisms of gene regulation, including individual physical interactions between regulatory regions and their target genes. Residing in structurally conserved regions in GSCs was CD276, a gene known to play a role in immuno-modulation. We show that, unexpectedly, CD276 is part of a stemness network in GSCs and can be targeted with an antibody-drug conjugate to curb self-renewal, a key stemness property. Our results demonstrate that integrated structural genomics datasets can be employed to rationally identify therapeutic vulnerabilities in self-renewing cells. SIGNIFICANCE In adult GBM, GSCs act as therapy-resistant reservoirs to nucleate tumor recurrence. New therapeutic approaches that target these cell populations hold the potential of significantly improving patient care and overall prognosis for this always-lethal cancer. Our work describes new links between 3D genome architecture and stemness properties in GSCs. In particular, through integration of multiple genomics and structural genomics datasets, we found an unexpected connection between immune-related genes and self-renewal programs in GBM. Among these, we show that targeting CD276 with knockdown strategies or specific antibody-drug conjugates achieve suppression of self-renewal. Strategies to target CD276+ cells are currently in clinical trials for solid tumors. Our results indicate that CD276-targeting agents could be deployed in GBM to specifically target GSC populations. HIGHLIGHTS We generated high (sub-5 kb) resolution Hi-C maps for stem-like cells from GBM patients. Integration of Hi-C and genomics datasets dissects mechanisms of gene regulation. 3D genomes poise immune-related genes, including CD276, for expression. Targeting CD276 curbs self-renewal properties of GBM cells.

Collaboration


Dive into the Steven Seaman's collaboration.

Top Co-Authors

Avatar

Brad St. Croix

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mary Beth Hilton

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Brad St. Croix

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Saurabh Saha

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Antonella Pignata

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar

Dimiter S. Dimitrov

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Enrique Zudaire

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kevin Bielamowicz

Center for Cell and Gene Therapy

View shared research outputs
Researchain Logo
Decentralizing Knowledge