Network


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

Hotspot


Dive into the research topics where Brad St. Croix is active.

Publication


Featured researches published by Brad St. Croix.


Current Opinion in Oncology | 2004

Tumor endothelial markers: new targets for cancer therapy.

Akash Nanda; Brad St. Croix

Purpose of review Targeting the endothelial cells that line tumor infiltrating blood vessels is a new anticancer strategy that has gained widespread support from biologists and clinicians. Here we highlight different approaches currently being used to target tumor endothelium and discuss new avenues for intervention that have been opened through the recent identification of tumor endothelial markers (TEMs). Recent findings The ability of Avastin to prolong survival in a Phase III clinical trial of human colorectal cancer has established the validity of the anti-angiogenic approach. However, realization of the full potential of a vascular targeting strategy may require the exploitation of molecules which are highly restricted in expression to tumor endothelium. Here we explore the potential of TEMs as new targets for cancer therapy. Current knowledge of these markers and their relation to other family members in the context of tumor angiogenesis is discussed. In particular, we highlight those molecules which, by virtue of their structure, cell-surface location and expression pattern, appear to hold promise as targets for future drug development. The identification of TEM8 as the anthrax toxin receptor and the successful targeting of this receptor in preclinical tumor models make this molecule a particularly attractive candidate for future vascular targeting studies. Summary Technological advances in cellular fractionation and genomics enabled the identification of several markers preferentially expressed on human tumor endothelium. Studies of these TEMs are expected to aid in our understanding of angiogenesis and could lead to the development of new imaging and diagnostic agents for cancer.


Current Opinion in Oncology | 1997

Cell adhesion and drug resistance in cancer.

Brad St. Croix; Robert S. Kerbel

The impact of cell adhesion in the pathobiology of tumors has been studied almost exclusively in the context of invasion, angiogenesis, and metastasis. Here we review data supporting a major role for cell-cell adhesion in the regulation of intrinsic or acquired resistance of solid tumors to various anticancer therapeutics. Cell-cell interactions are known to protect cells from apoptosis, and may help to explain chemoresistance of solid tumors. Recent data implicates p27KIP1, a cyclin-depen-dent kinase inhibitor, as a possible mediator of adhesion-dependent drug resistance. A model is described whereby cell-cell interactions signal the upregulation of p27, which in turn causes growth arrest in the G1 phase of the cell cycle and resistance to apoptosis induced by anticancer agents that target rapidly dividing cells. We focus on E-cadherin, a homophilic cell-cell adhesion molecule capable of upregulating p27, as one potential mediator of intrinsic resistance of carcinomas. A clearer understanding of how cell-cell adhesion suppresses cell growth and apoptosis should aid in the development of novel, more effective anticancer strategies.


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.


Molecular Cancer Therapeutics | 2013

A human single-domain antibody elicits potent antitumor activity by targeting an epitope in mesothelin close to the cancer cell surface.

Zhewei Tang; Mingqian Feng; Wei Gao; Yen Phung; Weizao Chen; Amit Chaudhary; Brad St. Croix; Min Qian; Dimiter S. Dimitrov; Mitchell Ho

Monoclonal antibodies against mesothelin are being evaluated for the treatment of mesothelioma and multiple forms of cancers, and show great promise for clinical development for solid cancers. Antibodies against mesothelin have been shown to act via immunotoxin-based inhibition of tumor growth and induction of antibody-dependent cell-mediated cytotoxicity (ADCC). However, complement-dependent cytotoxicity (CDC), considered an important additional mechanism of therapeutic antibodies against tumors, is inactive for such antibodies. Here, we used phage display antibody engineering technology and synthetic peptide screening to identify SD1, a human single-domain antibody to mesothelin. SD1 recognizes a conformational epitope at the C-terminal end (residues 539–588) of mesothelin close to the cell surface. To investigate SD1 as a potential therapeutic agent, we generated a recombinant human Fc (SD1-hFc) fusion protein. Interestingly, the SD1-hFc protein exhibits strong CDC activity, in addition to ADCC, against mesothelin-expressing tumor cells. Furthermore, it causes growth inhibition of human tumor xenografts in nude mice as a single agent. SD1 is the first human single-domain antibody targeting mesothelin-expressing tumors, shows potential as a cancer therapeutic candidate, and may improve current antibody therapy targeting mesothelin-expressing tumors. Mol Cancer Ther; 12(4); 416–26. ©2013 AACR.


Cell Cycle | 2012

Selective blockade of tumor angiogenesis.

Amit Chaudhary; Brad St. Croix

Blocking tumor angiogenesis is an important goal of cancer therapy, but clinically approved anti-angiogenic agents suffer from limited efficacy and adverse side effects, fueling the need to identify alternative angiogenesis regulators. Tumor endothelial marker 8 (TEM8) is a highly conserved cell surface receptor overexpressed on human tumor vasculature. Genetic disruption of Tem8 in mice revealed that TEM8 is important for promoting tumor angiogenesis and tumor growth but dispensable for normal development and wound healing. The induction of TEM8 in cultured endothelial cells by nutrient or growth factor deprivation suggests that TEM8 may be part of a survival response pathway that is activated by tumor microenvironmental stress. In preclinical studies, antibodies targeted against the extracellular domain of TEM8 inhibited tumor angiogenesis and blocked the growth of multiple human tumor xenografts. Anti-TEM8 antibodies augmented the activity of other anti-angiogenic agents, vascular targeting agents and conventional chemotherapeutic agents and displayed no detectable toxicity. Thus, anti-TEM8 antibodies provide a promising new tool for selective blockade of neovascularization associated with cancer and possibly other angiogenesis-dependent diseases.


Molecular Pharmaceutics | 2014

Immuno-PET Imaging of Tumor Endothelial Marker 8 (TEM8)

Frank Kuo; Stephanie Histed; Biying Xu; Veerendra Bhadrasetty; Lawrence P. Szajek; Mark R. Williams; Karen Wong; Haitao Wu; Kelly Lane; Vincent Coble; Olga Vasalatiy; Gary Griffiths; Chang H. Paik; Osama Elbuluk; Christopher S. Szot; Amit Kumar Chaudhary; Brad St. Croix; Peter L. Choyke; Elaine M. Jagoda

Tumor endothelial marker 8 (TEM8) is a cell surface receptor that is highly expressed in a variety of human tumors and promotes tumor angiogenesis and cell growth. Antibodies targeting TEM8 block tumor angiogenesis in a manner distinct from the VEGF receptor pathway. Development of a TEM8 imaging agent could aid in patient selection for specific antiangiogenic therapies and for response monitoring. In these studies, L2, a therapeutic anti-TEM8 monoclonal IgG antibody (L2mAb), was labeled with 89Zr and evaluated in vitro and in vivo in TEM8 expressing cells and mouse xenografts (NCI-H460, DLD-1) as a potential TEM8 immuno-PET imaging agent. 89Zr-df–L2mAb was synthesized using a desferioxamine–L2mAb conjugate (df–L2mAb); 125I-L2mAb was labeled directly. In vitro binding studies were performed using human derived cell lines with high, moderate, and low/undetectable TEM8 expression. 89Zr-df–L2mAb in vitro autoradiography studies and CD31 IHC staining were performed with cryosections from human tumor xenografts (NCI-H460, DLD-1, MKN-45, U87-MG, T-47D, and A-431). Confirmatory TEM8 Western blots were performed with the same tumor types and cells. 89Zr-df–L2mAb biodistribution and PET imaging studies were performed in NCI-H460 and DLD-1 xenografts in nude mice. 125I-L2mAb and 89Zr-df–L2mAb exhibited specific and high affinity binding to TEM8 that was consistent with TEM8 expression levels. In NCI-H460 and DLD-1 mouse xenografts nontarget tissue uptake of 89Zr-df–L2mAb was similar; the liver and spleen exhibited the highest uptake at all time points. 89Zr-L2mAb was highly retained in NCI-H460 tumors with <10% losses from day 1 to day 3 with the highest tumor to muscle ratios (T:M) occurring at day 3. DLD-1 tumors exhibited similar pharmacokinetics, but tumor uptake and T:M ratios were reduced ∼2-fold in comparison to NCI-H460 at all time points. NCI-H460 and DLD-1 tumors were easily visualized in PET imaging studies despite low in vitro TEM8 expression in DLD-1 cells indicating that in vivo expression might be higher in DLD-1 tumors. From in vitro autoradiography studies 89Zr-df–L2mAb specific binding was found in 6 tumor types (U87-MG, NCI-H460, T-47D MKN-45, A-431, and DLD-1) which highly correlated to vessel density (CD31 IHC). Westerns blots confirmed the presence of TEM8 in the 6 tumor types but found undetectable TEM8 levels in DLD-1 and MKN-45 cells. This data would indicate that TEM8 is associated with the tumor vasculature rather than the tumor tissue, thus explaining the increased TEM8 expression in DLD-1 tumors compared to DLD-1 cell cultures. 89Zr-df–L2mAb specifically targeted TEM8 in vitro and in vivo although the in vitro expression was not necessarily predictive of in vivo expression which seemed to be associated with the tumor vasculature. In mouse models, 89Zr-df–L2mAb tumor uptakes and T:M ratios were sufficient for visualization during PET imaging. These results would suggest that a TEM8 targeted PET imaging agent, such as 89Zr-df–L2mAb, may have potential clinical, diagnostic, and prognostic applications by providing a quantitative measure of tumor angiogenesis and patient selection for future TEM8 directed 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.


Cancer Research | 2016

Abstract 2312: TEM8/ANTXR1 specific T cells co-target tumor stem cells and tumor vasculature in triple-negative breast cancer

Tiara Byrd; Kristen Fousek; Antonella Pignata; Christopher S. Szot; Kevin Bielamowicz; Steven Seaman; Daniel Landi; Nino Rainusso; Poul H. Sorensen; Joachim Koch; Winfried S. Wels; Bradley Fletcher; Meenakshi Hegde; Brad St. Croix; Nabil Ahmed

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with no approved targeted therapies. Tumor endothelial marker 8 (TEM8), initially identified as a marker of tumor endothelial cells in colorectal cancer and other solid tumors has recently been shown to be upregulated in TNBC and breast cancer stem cells (BCSCs). We investigated whether TEM8 specific chimeric antigen receptor (CAR) T cells recognize and kill both tumor endothelial cells as well as TNBC tumor cells. TEM8 specific CAR molecules were generated using single chain variable fragment derived from the monoclonal antibody, L2. L2 CAR T cells selectively recognized TEM8, secreted immunostimulatory cytokines and effectively killed both TEM8 positive TNBC and tumor endothelial cell lines. Moreover, L2 CAR T cells targeted breast cancer stem cells significantly reducing the number of mammospheres relative to non-transduced T cells. In vivo, adoptive transfer of L2 CAR T cells induced regression of established vascularized TNBC xenografts. Hence, TEM8 may serve as an attractive target for immunotherapy of TNBC. Citation Format: Tiara Byrd, Kristen Fousek, Antonella Pignata, Christopher Szot, Kevin Bielamowicz, Steven Seaman, Daniel Landi, Nino Rainusso, Poul Sorensen, Joachim Koch, Winfried Wels, Bradley Fletcher, Meenakshi Hegde, Brad St Croix, Nabil Ahmed. TEM8/ANTXR1 specific T cells co-target tumor stem cells and tumor vasculature in triple-negative breast cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2312.


Journal for ImmunoTherapy of Cancer | 2014

Dual targeting of the tumor and its associated vasculature using a single bispecific chimeric antigen receptor molecule

Tiara Byrd; Kristen Fousek; Antonella Pignata; Amanda Wakefield; Zakaria Grada; Kevin Aviles-Padilla; Bradley S. Fletcher; Meenakshi Hegde; Brad St. Croix; Nabil Ahmed

Meeting abstracts We have previously demonstrated the efficacy of HER2 specific chimeric antigen receptor (CAR) T cells in animal models of human cancer. While HER2 CAR T cells induced tumor regression, tumors recurred in a subset of animals. Lytic co-targeting of the tumor endothelium could


Journal for ImmunoTherapy of Cancer | 2014

TEM8 specific T cells target the tumor cells and tumor-associated vasculature in triple negative breast cancer

Tiara Byrd; Kristen Fousek; Antonella Pignata; Amanda Wakefield; Brad St. Croix; Bradley S. Fletcher; Meenakshi Hegde; Nabil Ahmed

Meeting abstracts Tumor endothelium marker 8 (TEM8) was discovered by St Croix, et. al. as one of nine gene products preferentially upregulated in the tumor-associated vs. normal endothelium [[1][1]]. Interestingly, TEM8 has also been identified as a tumor restricted antigen in triple negative

Collaboration


Dive into the Brad St. Croix's collaboration.

Top Co-Authors

Avatar

Enrique Zudaire

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dimiter S. Dimitrov

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Steven Seaman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Antonella Pignata

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kristen Fousek

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Meenakshi Hegde

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Nabil Ahmed

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Tiara Byrd

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Zhongyu Zhu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Saurabh Saha

Johns Hopkins University

View shared research outputs
Researchain Logo
Decentralizing Knowledge