Network


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

Hotspot


Dive into the research topics where David B. Powers is active.

Publication


Featured researches published by David B. Powers.


Clinical Cancer Research | 2008

CS1, a Potential New Therapeutic Antibody Target for the Treatment of Multiple Myeloma

Eric D. Hsi; Roxanne Steinle; Balaji Balasa; Susann Szmania; Aparna Draksharapu; Benny Shum; Mahrukh Huseni; David B. Powers; Amulya Nanisetti; Yin Zhang; Audie Rice; Anne van Abbema; Melanie Wong; Gao Liu; Fenghuang Zhan; Myles Dillon; Shihao Chen; Susan Rhodes; Franklin Fuh; Naoya Tsurushita; Shankar Kumar; Vladimir Vexler; John D. Shaughnessy; Bart Barlogie; Frits van Rhee; Mohamad A. Hussein; Daniel E. H. Afar; Marna Williams

Purpose: We generated a humanized antibody, HuLuc63, which specifically targets CS1 (CCND3 subset 1, CRACC, and SLAMF7), a cell surface glycoprotein not previously associated with multiple myeloma. To explore the therapeutic potential of HuLuc63 in multiple myeloma, we examined in detail the expression profile of CS1, the binding properties of HuLuc63 to normal and malignant cells, and the antimyeloma activity of HuLuc63 in preclinical models. Experimental Design: CS1 was analyzed by gene expression profiling and immunohistochemistry of multiple myeloma samples and numerous normal tissues. HuLuc63-mediated antimyeloma activity was tested in vitro in antibody-dependent cellular cytotoxicity (ADCC) assays and in vivo using the human OPM2 xenograft model in mice. Results: CS1 mRNA was expressed in >90% of 532 multiple myeloma cases, regardless of cytogenetic abnormalities. Anti-CS1 antibody staining of tissues showed strong staining of myeloma cells in all plasmacytomas and bone marrow biopsies. Flow cytometric analysis of patient samples using HuLuc63 showed specific staining of CD138+ myeloma cells, natural killer (NK), NK-like T cells, and CD8+ T cells, with no binding detected on hematopoietic CD34+ stem cells. HuLuc63 exhibited significant in vitro ADCC using primary myeloma cells as targets and both allogeneic and autologous NK cells as effectors. HuLuc63 exerted significant in vivo antitumor activity, which depended on efficient Fc-CD16 interaction as well as the presence of NK cells in the mice. Conclusions: These results suggest that HuLuc63 eliminates myeloma cells, at least in part, via NK-mediated ADCC and shows the therapeutic potential of targeting CS1 with HuLuc63 for the treatment of multiple myeloma.


Journal of Translational Medicine | 2007

A function blocking anti-mouse integrin α5β1 antibody inhibits angiogenesis and impedes tumor growth in vivo

Vinay Bhaskar; Dong-Ping Zhang; Melvin Fox; Pui Seto; Melanie Hl Wong; Pauline Wales; David B. Powers; Debra T. Chao; Robert B. Dubridge; Vanitha Ramakrishnan

BackgroundIntegrins are important adhesion molecules that regulate tumor and endothelial cell survival, proliferation and migration. The integrin α5β1 has been shown to play a critical role during angiogenesis. An inhibitor of this integrin, volociximab (M200), inhibits endothelial cell growth and movement in vitro, independent of the growth factor milieu, and inhibits tumor growth in vivo in the rabbit VX2 carcinoma model. Although volociximab has already been tested in open label, pilot phase II clinical trials in melanoma, pancreatic and renal cell cancer, evaluation of the mechanism of action of volociximab has been limited because this antibody does not cross-react with murine α5β1, precluding its use in standard mouse xenograft models.MethodsWe generated a panel of rat-anti-mouse α5β1 antibodies, with the intent of identifying an antibody that recapitulated the properties of volociximab. Hybridoma clones were screened for analogous function to volociximab, including specificity for α5β1 heterodimer and blocking of integrin binding to fibronectin. A subset of antibodies that met these criteria were further characterized for their capacities to bind to mouse endothelial cells, inhibit cell migration and block angiogenesis in vitro. One antibody that encompassed all of these attributes, 339.1, was selected from this panel and tested in xenograft models.ResultsA panel of antibodies was characterized for specificity and potency. The affinity of antibody 339.1 for mouse integrin α5β1 was determined to be 0.59 nM, as measured by BIAcore. This antibody does not significantly cross-react with human integrin, however 339.1 inhibits murine endothelial cell migration and tube formation and elicits cell death in these cells (EC50 = 5.3 nM). In multiple xenograft models, 339.1 inhibited the growth of established tumors by 40–60% (p < 0.05) and this inhibition correlates with a concomitant decrease in vessel density.ConclusionThe results herein demonstrate that 339.1, like volociximab, exhibits potent anti-α5β1 activity and confirms that inhibition of integrin α5β1 impedes angiogenesis and slows tumor growth in vivo.


Clinical Cancer Research | 2010

Antibodies to TWEAK Receptor Inhibit Human Tumor Growth through Dual Mechanisms

Patricia A. Culp; Donghee Choi; Yongke Zhang; Johnny Yin; Pui Seto; Suzanne Ybarra; Mian Su; Mien Sho; Roxanne Steinle; Melanie H.L. Wong; Ferdinand Evangelista; Jennifer Grove; Marie Cardenas; Marjorie James; Eric D. Hsi; Debra T. Chao; David B. Powers; Vanitha Ramakrishnan; Robert B. Dubridge

Purpose: Targeted therapeutics have significantly changed the outcome for patients diagnosed with cancer. Still, effective therapeutic intervention does not exist for many cancers and much remains to be done. The objective of this study was to identify novel genes that potentially regulate tumor growth, to target these gene products with monoclonal antibodies, and to examine the therapeutic potential of these antibodies. Experimental Design: Using cDNA microarray analysis, we identified genes overexpressed in several solid malignancies. We generated a mouse monoclonal antibody, 19.2.1, and its humanized counterpart, PDL192, to one such target, TweakR (TWEAK receptor, Fn14, TNFRSF12A, CD266), and characterized the antitumor activities in vitro and in mouse xenograft models. Results: Both 19.2.1 (mouse IgG2a) and PDL192 (human IgG1), like TWEAK, the natural ligand of TweakR, inhibited the growth of several TweakR-expressing cancer cell lines in anchorage-dependent and anchorage-independent assays in vitro. Both antibodies showed significant antitumor activity in multiple mouse xenograft models. PDL192 and 19.2.1 also induced antibody-dependent cellular cytotoxicity (ADCC) of cancer cell lines in vitro. A chimeric version of 19.2.1 containing the mouse IgG1 Fc region (19.2.1×G1) exhibited significantly less ADCC than 19.2.1. However, 19.2.1×G1 showed differential activity in vivo, with activity equivalent to 19.2.1 in one model, but significantly less efficacy than 19.2.1 in a second model. These results indicate that PDL192 and 19.2.1 mediate their antitumor effects by signaling through TweakR, resulting in reduced tumor cell proliferation, and by ADCC. Clin Cancer Res; 16(2); 497–508


mAbs | 2013

Deep mutational scanning of an antibody against epidermal growth factor receptor using mammalian cell display and massively parallel pyrosequencing

Charles M. Forsyth; Veronica Juan; Yoshiko Akamatsu; Robert B. Dubridge; Minhtam Doan; Alexander V. Ivanov; Zhiyuan Ma; Dixie Polakoff; Jennifer Razo; Keith S. Wilson; David B. Powers

We developed a method for deep mutational scanning of antibody complementarity-determining regions (CDRs) that can determine in parallel the effect of every possible single amino acid CDR substitution on antigen binding. The method uses libraries of full length IgGs containing more than 1000 CDR point mutations displayed on mammalian cells, sorted by flow cytometry into subpopulations based on antigen affinity and analyzed by massively parallel pyrosequencing. Higher, lower and neutral affinity mutations are identified by their enrichment or depletion in the FACS subpopulations. We applied this method to a humanized version of the anti-epidermal growth factor receptor antibody cetuximab, generated a near comprehensive data set for 1060 point mutations that recapitulates previously determined structural and mutational data for these CDRs and identified 67 point mutations that increase affinity. The large-scale, comprehensive sequence-function data sets generated by this method should have broad utility for engineering properties such as antibody affinity and specificity and may advance theoretical understanding of antibody-antigen recognition.


Journal of Immunology | 2012

Affinity and Cross-Reactivity Engineering of CTLA4-Ig To Modulate T Cell Costimulation

Zhenghai Xu; Veronica Juan; Alexander V. Ivanov; Zhiyuan Ma; Dixie Polakoff; David B. Powers; Robert B. Dubridge; Keith T. Wilson; Yoshiko Akamatsu

CTLA4-Ig is an Fc fusion protein containing the extracellular domain of CTLA-4, a receptor known to deliver a negative signal to T cells. CTLA4-Ig modulates T cell costimulatory signals by blocking the CD80 and CD86 ligands from binding to CD28, which delivers a positive T cell costimulatory signal. To engineer CTLA4-Ig variants with altered binding affinity to CD80 and CD86, we employed a high-throughput protein engineering method to map the ligand binding surface of CTLA-4. The resulting mutagenesis map identified positions critical for the recognition of each ligand on the three CDR-like loops of CTLA-4, consistent with the published site-directed mutagenesis and x-ray crystal structures of the CTLA-4/CD80 and CTLA-4/CD86 complexes. A number of single amino acid substitutions were identified that equally affected the binding affinity of CTLA4-Ig for both ligands as well as those that differentially affected binding. All of the high-affinity variants showed improved off-rates, with the best one being a 17.5-fold improved off-rate over parental CTLA4-Ig binding to CD86. Allostimulation of human CD4+ T cells showed that improvement of CD80 and CD86 binding activity augmented inhibition of naive and primed T cell activation. In general, increased affinity for CD86 resulted in more potent inhibition of T cell response than did increased affinity for CD80. Optimization of the affinity balance to CD80 and CD86 to particular disease settings may lead to development of a CTLA4-Ig molecule with improved efficacy and safety profiles.


Molecular Cancer Therapeutics | 2004

Preclinical validation of anti-TMEFF2-auristatin E–conjugated antibodies in the treatment of prostate cancer

Daniel E. H. Afar; Vinay Bhaskar; Eric Ibsen; Danna Breinberg; Susan M. Henshall; James G. Kench; Marija Drobnjak; Rick Powers; Melanie Wong; Ferdinand Evangelista; Chris O'Hara; David B. Powers; Robert B. DuBridge; Ingrid Caras; Ruth Winter; Terri Anderson; Nanette Solvason; Carlos Cordon-Cardo; Howard I. Scher; John J. Grygiel; Robert L. Sutherland; Richard Murray; Vanitha Ramakrishnan; Debbie A. Law


Archive | 2007

Chimeric and humanized antibodies to α5β1 integrin that modulate angiogenesis

Vanitha Ramakrishnan; David B. Powers; Dale E. Johnson; Ursula Jeffry; Vinay Bhaskar


Archive | 2003

Antibodies Against Cancer Antigen TMEFF2 and Uses Thereof

Vinay Bhaskar; Agustin de la Calle; Debbie Law; Ingrid Caras; Vanitha Ramakrishnan; Richard Murray; Daniel E. H. Afar; David B. Powers


Archive | 2010

Anti-vegf antibodies and their uses

Fiona A. Harding; Yoshiko Akamatsu; Robert B. Dubridge; David B. Powers


Archive | 2010

Anti-her2 antibodies and their uses

Fiona A. Harding; Yoshiko Akamatsu; Robert B. Dubridge; David B. Powers

Collaboration


Dive into the David B. Powers's collaboration.

Top Co-Authors

Avatar

Robert B. Dubridge

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Gao Liu

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Debbie Law

University of California

View shared research outputs
Top Co-Authors

Avatar

Ingrid Caras

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge