Network


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

Hotspot


Dive into the research topics where Dingguo Liu is active.

Publication


Featured researches published by Dingguo Liu.


Clinical Cancer Research | 2011

Specifically Targeting Angiopoietin-2 Inhibits Angiogenesis, Tie2-Expressing Monocyte Infiltration, and Tumor Growth

Hanhua Huang; Jing-Yu Lai; Janet Do; Dingguo Liu; Lingna Li; Joselyn Del Rosario; Venkata Ramana Doppalapudi; Steven Pirie-Shepherd; Nancy Levin; Curt W. Bradshaw; Gary Woodnutt; Rodney W. Lappe; Abhijit Bhat

Purpose: Angiopoietin-1 (Ang1) plays a key role in maintaining stable vasculature, whereas in a tumor Ang2 antagonizes Ang1s function and promotes the initiation of the angiogenic switch. Specifically targeting Ang2 is a promising anticancer strategy. Here we describe the development and characterization of a new class of biotherapeutics referred to as CovX-Bodies, which are created by chemical fusion of a peptide and a carrier antibody scaffold. Experimental Design: Various linker tethering sites on peptides were examined for their effect on CovX-Body in vitro potency and pharmacokinetics. Ang2 CovX-Bodies with low nmol/L IC50s and significantly improved pharmacokinetics were tested in tumor xenograft studies alone or in combination with standard of care agents. Tumor samples were analyzed for target engagement, via Ang2 protein level, CD31-positive tumor vasculature, and Tie2 expressing monocyte penetration. Results: Bivalent Ang2 CovX-Bodies selectively block the Ang2–Tie2 interaction (IC50 < 1 nmol/L) with dramatically improved pharmacokinetics (T½ > 100 hours). Using a staged Colo-205 xenograft model, significant tumor growth inhibition (TGI) was observed (40%–63%, P < 0.01). Ang2 protein levels were reduced by approximately 50% inside tumors (P < 0.01), whereas tumor microvessel density (P < 0.01) and intratumor proangiogenic Tie2+CD11b+ cells (P < 0.05) were significantly reduced. When combined with sunitinib, sorafenib, bevacizumab, irinotecan, or docetaxel, Ang2 CovX-Bodies produced even greater efficacy (∼80% TGI, P < 0.01). Conclusion: CovX-Bodies provide an elegant solution to overcome the pharmacokinetic–pharmacodynamic problems of peptides. Long-acting Ang2 specific CovX-Bodies will be useful as single agents and in combination with standard-of-care agents. Clin Cancer Res; 17(5); 1001–11. ©2011 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Chemical generation of bispecific antibodies

Venkata Ramana Doppalapudi; Jie Huang; Dingguo Liu; Ping Jin; Bin Liu; Lingna Li; Joel Desharnais; Crystal Hagen; Nancy Levin; Michael J. Shields; Michelle Parish; Robert E. Murphy; Joselyn Del Rosario; Bryan Oates; Jing-Yu Lai; Marla J. Matin; Zemeda W. Ainekulu; Abhijit Bhat; Curt W. Bradshaw; Gary Woodnutt; Richard A. Lerner; Rodney W. Lappe

Bispecific antibodies (BsAbs) are regarded as promising therapeutic agents due to their ability to simultaneously bind two different antigens. Several bispecific modalities have been developed, but their utility is limited due to problems with stability and manufacturing complexity. Here we report a versatile technology, based on a scaffold antibody and pharmacophore peptide heterodimers, that enables rapid generation and chemical optimization of bispecific antibodies, which are termed bispecific CovX-Bodies. Two different peptides are joined together using a branched azetidinone linker and fused to the scaffold antibody under mild conditions in a site-specific manner. Whereas the pharmacophores are responsible for functional activities, the antibody scaffold imparts long half-life and Ig-like distribution. The pharmacophores can be chemically optimized or replaced with other pharmacophores to generate optimized or unique bispecific antibodies. As a prototype, we developed a bispecific antibody that binds both vascular endothelial growth factor (VEGF) and angiopoietin-2 (Ang2) simultaneously, inhibits their function, shows efficacy in tumor xenograft studies, and greatly augments the antitumor effects of standard chemotherapy. This unique antiangiogenic bispecific antibody is in phase-1 clinical trials.


Bioorganic & Medicinal Chemistry Letters | 2012

Kappa agonist CovX-Bodies.

Lee R. Roberts; Kevin Brady; Amy Brown; Doreen Davey; Lijin Feng; Hanhua Huang; Dingguo Liu; Laia Malet; Gordon McMurray; Anne Phelan; Ken Saunders; Abhijit Bhat

Small peptidic kappa agonists were covalently linked to the reactive lysine of the CovX antibody to create compounds having potent activity at the kappa receptor with greatly extended half-life when compared to the parent peptide as exemplified by compound 20.


Cancer Research | 2011

Abstract LB-396: FGFR4-antagonistic CovX-Body™ inhibits tumor growth

Allan Kaspar; Dingguo Liu; Janet Do; Oscar Betancourt; Bin Liu; Joel Desharnais; Joselyn Del Rosario; Lingna Li; Steven Pirie-Shepherd; Nancy Levin; Abhijit Bhat; Gary Woodnutt; Rodney W. Lappe

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL FGFR4 is the receptor for FGF19. Published data has shown FGF19 transgenic animals develop hepatocellular carcinoma (HCC), suggesting that the FGFR4:FGF19 axis may play a role in HCC. FGFR4 upregulation has been shown to be a negative prognostic indicator in multiple tumor types. CVX-63, an FGFR4-specific CovX-Body , was produced by fusing a chemically modified peptide to the specific Fab binding site of a specially designed antibody. CVX-63 was shown to inhibit FGF19 binding to FGFR4 in a competition ELISA with an IC50 of 10 nM, and had no cross reactivity with FGFR1–3. CVX-63 has a beta half life of 55 hrs in the mouse. The anti-tumor activity of CVX-63 was demonstrated in a staged Colo-205 xenograft model, with CVX-63 administered i.v. weekly. Maximal activity of 30% tumor growth inhibition was seen at dosing 10 mg/kg. Immunohistochemistry analysis of treated tumors shows a >90% reduction in phospho-ERK and 50% downregulation of FGFR4 on tumor cells. Ki67, a marker of proliferation was reduced, but active caspase 3 was not found in treated tumors. A patient-derived HCC xenograft was identified which expresses both FGFR4 and FGF19. When tested in this HCC model, CVX-63 did not show any effect inhibiting tumor growth. These data demonstrate that CVX-63 effectively inhibits FGFR4 activity in xenografts. FGFR4 inhibition has a modest effect on human tumor progression and FGFR4 does not seem to be a critical driver of HCC. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr LB-396. doi:10.1158/1538-7445.AM2011-LB-396


Archive | 2009

Anti-Angiogenic Compounds

Venkata Ramana Doppalapudi; Jing-Yu Lai; Bin Liu; Dingguo Liu; Joel Desharnais; Abhijit S. Bhat; Yanwen Fu; Bryan Oates; Gang Chen; Curt W. Bradshaw


ACS Medicinal Chemistry Letters | 2016

Design, Synthesis, and Cytotoxic Evaluation of Novel Tubulysin Analogues as ADC Payloads.

Carolyn A. Leverett; Sai Chetan K. Sukuru; Beth Cooper Vetelino; Sylvia Musto; Kevin Parris; Jayvardhan Pandit; Frank Loganzo; Alison H. Varghese; Guoyun Bai; Bin Liu; Dingguo Liu; Sarah Hudson; Venkata Ramana Doppalapudi; Joseph Stock; Christopher J. O’Donnell; Chakrapani Subramanyam


Archive | 2014

POLYNUCLEOTIDE CONSTRUCTS HAVING DISULFIDE GROUPS

Curt W. Bradshaw; Laxman Eltepu; Ayman Kabakibi; Son Lam; Bin Liu; Dingguo Liu; Bryan R. Meade; Sukumar Sakamuri


Archive | 2015

POLYNUCLEOTIDE CONSTRUCTS HAVING BIOREVERSIBLE AND NON-BIOREVERSIBLE GROUPS

Curt W. Bradshaw; Sukumar Sakamuri; Laxman Eltepu; Son Lam; Dingguo Liu; Bryan R. Meade; Giuseppe Dello Iacono; Joseph Stock; Bin Liu


Cancer Research | 2008

Angiopoietin-2 specific CVX-060 inhibits tumor growth cooperatively with chemotherapy

Hanhua Huang; Janet Do; Lingna Li; Jing-Yu Lai; Dingguo Liu; Steven Pirie-Shepherd; Nancy Levin; Curt W. Bradshaw; Gary Woodnutt; Rodney W. Lappe; Abhijit Bhat


Archive | 2017

POLYNUCLEOTIDE CONSTRUCTS HAVING AN AUXILIARY MOIETY NON-BIOREVERSIBLY LINKED TO AN INTERNUCLEOSIDE PHOSPHATE OR PHOSPHOROTHIOATE

Bradshaw, Curt, W.; Sukumar Sakamuri; Meade, Bryan, R.; Laxman Eltepu; Iacono, Giuseppe, Dello; Son Lam; Joseph Stock; Dingguo Liu; Bin Liu

Collaboration


Dive into the Dingguo Liu's collaboration.

Top Co-Authors

Avatar

Curt W. Bradshaw

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nancy Levin

Wayne State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge