Network


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

Hotspot


Dive into the research topics where Xiaoyan M. Zhang is active.

Publication


Featured researches published by Xiaoyan M. Zhang.


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.


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.


Nuclear Medicine and Biology | 2016

[124I]FIAU: Human dosimetry and infection imaging in patients with suspected prosthetic joint infection

Xiaoyan M. Zhang; Halle H. Zhang; Patrick McLeroth; Richard D. Berkowitz; Michael A. Mont; Michael G. Stabin; Barry A. Siegel; Abass Alavi; T. Marc Barnett; Jeffrey Gelb; Chantal Petit; John Spaltro; Steve Y. Cho; Martin G. Pomper; James J. Conklin; Chetan Bettegowda; Saurabh Saha

INTRODUCTION Fialuridine (FIAU) is a nucleoside analog that is a substrate for bacterial thymidine kinase (TK). Once phosphorylated by TK, [(124)I]FIAU becomes trapped within bacteria and can be detected with positron emission tomography/computed tomography (PET/CT). [(124)I]FIAU PET/CT has been shown to detect bacteria in patients with musculoskeletal bacterial infections. Accurate diagnosis of prosthetic joint infections (PJIs) has proven challenging because of the lack of a well-validated reference. In the current study, we assessed biodistribution and dosimetry of [(124)I]FIAU, and investigated whether [(124)I]FIAU PET/CT can diagnose PJIs with acceptable accuracy. METHODS To assess biodistribution and dosimetry, six subjects with suspected hip or knee PJI and six healthy subjects underwent serial PET/CT after being dosed with 74MBq (2mCi) [(124)I]FIAU intravenously (IV). Estimated radiation doses were calculated with the OLINDA/EXM software. To determine accuracy of [(124)I]FIAU, 22 subjects with suspected hip or knee PJI were scanned at 2-6 and 24-30h post IV injection of 185MBq (5mCi) [(124)I]FIAU. Images were interpreted by a single reader blinded to clinical information. Representative cases were reviewed by 3 additional readers. The utility of [(124)I]FIAU to detect PJIs was assessed based on the correlation of the patients infection status with imaging results as determined by an independent adjudication board (IAB). RESULTS The kidney, liver, spleen, and urinary bladder received the highest radiation doses of [(124)I]FIAU. The effective dose was 0.16 to 0.20mSv/MBq and doses to most organs ranged from 0.11 to 0.76mGy/MBq. PET image quality obtained from PJI patients was confounded by metal artifacts from the prostheses and pronounced FIAU uptake in muscle. Consequently, a correlation with infection status and imaging results could not be established. CONCLUSIONS [(124)I]FIAU was well-tolerated in healthy volunteers and subjects with suspected PJI, and had acceptable dosimetry. However, the utility of [(124)I]FIAU for the clinical detection of PJIs is limited by poor image quality and low specificity.


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.


Molecular Cancer Therapeutics | 2015

Abstract LB-B15: A novel PI3K gamma isoform selective small molecule kinase inhibitor demonstrates single agent anti-tumor activity and enhanced combination activity with checkpoint blockade in syngeneic mouse models of cancer

Saurabh Saha; Linping Zhang; Thomas Hoock; Alex Aronov; Sudipta Mahajan; Michael J. Boyd; Jon H. Come; Veronique Damagnez; Wojciech Dworakowski; Suvarna Khare-Pandit; Arnaud LeTiran; Cameron Stuver Moody; Harwin O'Dowd; Joseph Prezioso; Setu Roday; Xiaoyan M. Zhang

Immune checkpoint inhibitors have generated impressive clinical responses, however, a number of patients and cancer types remain resistant to checkpoint blockade. Immunosuppressive cells such as Tregs and MDSCs in the tumor microenvironment are hypothesized to mediate this resistance. The gamma (γ) isoform of PI3K has long been known to modulate myeloid and lymphocyte cell migration and function, including trafficking of monocytic and granulocytic cells into inflamed tissues such as tumors. Here we report the identification and characterization of BVD-723, a PI3Kγ selective small molecule kinase inhibitor which demonstrated single agent and enhanced combination anti-tumor activity[xx]with either an anti-PD-1 or anti-CTLA-4 antibody. In a mouse syngeneic colon cancer model,[xx]BVD-723 in combination with anti-PD-1 or anti-CTLA-4 resulted in complete regression[xx]of tumors in 35% (7/20) and 60% (12/20) of animals treated with each antibody respectively [versus 0% (0/20) and 5% (1/20) with either antibody alone]. Interestingly, the anti-tumor activity was coupled with a decrease in tumor infiltrating Tregs, granulocytic MDSCs, CD4+ T cells and an increase in the CD8+/Treg ratio, suggesting that inhibition of PI3Kγ by BVD-723 promotes immune-reactivation in the tumor microenvironment. Additionally, BVD-723 demonstrated potent synergy with an anti-PD-1 antibody in a syngeneic mouse model of lymphoma. Single agent BVD-723 activity was also observed in syngeneic mouse models of pancreas, colon, lymphoma and bladder cancers. Results from completed safety pharmacology, toxicology and in vivo efficacy studies support clinical evaluation of BVD-723[xx]as a monotherapy or in combination with the checkpoint inhibitors in cancer.[xx] Citation Format: Saurabh Saha, Linping Zhang, Thomas Hoock, Alex Aronov, Sudipta Mahajan, Michael Boyd, Jon Come, Veronique Damagnez, Wojciech Dworakowski, Suvarna Khare-Pandit, Arnaud LeTiran, Cameron Moody, Harwin O9Dowd, Joseph Prezioso, Setu Roday, Xiaoyan M. Zhang. A novel PI3K gamma isoform selective small molecule kinase inhibitor demonstrates single agent anti-tumor activity and enhanced combination activity with checkpoint blockade in syngeneic mouse models of cancer. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr LB-B15.


Archive | 2015

COMBINATION THERAPIES USING AGENTS THAT TARGET TUMOR-ASSOCIATED STROMA OR TUMOR CELLS AND TUMOR VASCULATURE

Saurabh Saha; Xiaoyan M. Zhang; Dimiter S. Dimitrov; Zhongyu Zhu; Croix Brad; Enrique Zudaire


Archive | 2015

COMBINATION THERAPIES TARGETING TUMOR-ASSOCIATED STROMA OR TUMOR CELLS

Saurabh Saha; Xiaoyan M. Zhang; Dimiter S. Dimitrov; Zhongyu Zhu; Croix Brad; Enrique Zudaire


Archive | 2015

COMBINATION THERAPIES USING AGENTS THAT TARGET TUMOR-ASSOCIATED STROMA OR TUMOR CELLS AND ALKYLATING AGENTS

Saurabh Saha; Xiaoyan M. Zhang; Dimiter S. Dimitrov; Zhongyu Zhu; Brad St. Croix; Enrique Zudaire


Archive | 2015

Polythérapies utilisant des agents de platine et des agent ciblant le stroma associé à une tumeur ou des cellules tumorales

Saurabh Saha; Xiaoyan M. Zhang; Dimiter S. Dimitrov; Zhongyu Zhu; Croix Brad; Enrique Zudaire


Archive | 2015

COMBINATION THERAPIES USING AGENTS THAT TARGET TUMOR-ASSOCIATED STROMA OR TUMOR CELLS AND OTHER PATHWAYS

Saurabh Saha; Xiaoyan M. Zhang; Dimiter S. Dimitrov; Zhongyu Zhu; Croix Brad; Enrique Zudaire

Collaboration


Dive into the Xiaoyan M. Zhang's collaboration.

Top Co-Authors

Avatar

Saurabh Saha

Johns Hopkins University

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

Zhongyu Zhu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Croix Brad

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Brad St. Croix

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Mary Beth Hilton

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

Brad St. Croix

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge