Network


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

Hotspot


Dive into the research topics where Russell A. Hammond is active.

Publication


Featured researches published by Russell A. Hammond.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2000

Vaccine-Induced Antibodies Inhibit CETP Activity In Vivo and Reduce Aortic Lesions in a Rabbit Model of Atherosclerosis

Charles W. Rittershaus; David P. Miller; Lawrence J. Thomas; Michele Picard; Christopher M. Honan; Constance D. Emmett; Carolyn L. Pettey; Hedy Adari; Russell A. Hammond; David T. Beattie; Allan D. Callow; Henry C. Marsh; Una S. Ryan

Using a vaccine approach, we immunized New Zealand White rabbits with a peptide containing a region of cholesteryl ester transfer protein (CETP) known to be required for neutral lipid transfer function. These rabbits had significantly reduced plasma CETP activity and an altered lipoprotein profile. In a cholesterol-fed rabbit model of atherosclerosis, the fraction of plasma cholesterol in HDL was 42% higher and the fraction of plasma cholesterol in LDL was 24% lower in the CETP-vaccinated group than in the control-vaccinated group. Moreover, the percentage of the aorta surface exhibiting atherosclerotic lesion was 39.6% smaller in the CETP-vaccinated rabbits than in controls. The data reported here demonstrate that CETP activity can be reduced in vivo by vaccination with a peptide derived from CETP and support the concept that inhibition of CETP activity in vivo can be antiatherogenic. In addition, these studies suggest that vaccination against a self-antigen is a viable therapeutic strategy for disease management.


Journal of The American Society of Nephrology | 2013

Soluble CR1 Therapy Improves Complement Regulation in C3 Glomerulopathy

Yuzhou Zhang; Carla M. Nester; Danniele G. Holanda; Henry C. Marsh; Russell A. Hammond; Lawrence J. Thomas; Nicole C. Meyer; Lawrence G. Hunsicker; Sanjeev Sethi; Richard J.H. Smith

Dense deposit disease (DDD) and C3 glomerulonephritis (C3GN) are widely recognized subtypes of C3 glomerulopathy. These ultra-rare renal diseases are characterized by fluid-phase dysregulation of the alternative complement pathway that leads to deposition of complement proteins in the renal glomerulus. Disease triggers are unknown and because targeted treatments are lacking, progress to end stage renal failure is a common final outcome. We studied soluble CR1, a potent regulator of complement activity, to test whether it restores complement regulation in C3 glomerulopathy. In vitro studies using sera from patients with DDD showed that soluble CR1 prevents dysregulation of the alternative pathway C3 convertase, even in the presence of C3 nephritic factors. In mice deficient in complement factor H and transgenic for human CR1, soluble CR1 therapy stopped alternative pathway activation, resulting in normalization of serum C3 levels and clearance of iC3b from glomerular basement membranes. Short-term use of soluble CR1 in a pediatric patient with end stage renal failure demonstrated its safety and ability to normalize activity of the terminal complement pathway. Overall, these data indicate that soluble CR1 re-establishes regulation of the alternative complement pathway and provide support for a limited trial to evaluate soluble CR1 as a treatment for DDD and C3GN.


Human Vaccines | 2009

Co-administration of a CpG adjuvant (VaxImmuneTM, CPG 7909) with CETP vaccines increased immunogenicity in rabbits and mice

Lawrence J. Thomas; Russell A. Hammond; Eric M. Forsberg; Kathleen M. Geoghegan-Barek; Brad H. Karalius; Henry C. Marsh; Charles W. Rittershaus

Cholesteryl ester transfer protein (CETP) is a plasma glycoprotein that facilitates the transfer of neutral lipids and phospholipids between lipoproteins and contributes to the regulation of the plasma concentration of high density lipoprotein cholesterol (HDL-C). Vaccines have been developed that elicit antibodies that bind to and reduce the lipid transfer function of CETP as a way to increase the plasma concentration of HDL-C and prevent or treat atherosclerosis. This study assessed the immunogenicity of two vaccine peptides. The first, CETi-1, is a dimerized synthetic peptide, including residues 461-476 of human CETP and residues 830-843 of tetanus toxoid, TT(830-843). The second, PADRE-CETP, is a monomeric peptide, in which a PADRE T cell epitope (aK-Cha-VAAWTLKAa) replaces the TT(830-843) T cell epitope of CETi-1. Both peptides were formulated with aluminum-containing adjuvants (Alhydrogel®), and tested in mice and rabbits with or without the co-administration of the investigational TLR9 agonist VaxImmuneTM (CPG 7909). In both mice and rabbits, the vaccine peptide utilizing the PADRE T cell epitope elicited stronger anti-CETP antibody responses than the CETi-1 vaccine. Also, co-administration of VaxImmune enhanced the anti-CETP antibody responses to both vaccines. Isotype analysis of the murine anti-CETP antibody response to both vaccines demonstrated a switch from IgG1 to IgG2a upon co-administration of VaxImmune. We conclude that 1) the PADRE T cell epitope is more potent than the TT(830-843) epitope in providing help for the anti-CETP antibody response; and 2) co-administration of VaxImmune with either vaccine increased immunogenicity as measured by antibody response.


Molecular Cancer Therapeutics | 2016

Development of a Novel Antibody-drug Conjugate for the Potential Treatment of Ovarian, Lung and Renal Cell Carcinoma Expressing TIM-1

Lawrence J. Thomas; Laura Vitale; Thomas O'Neill; Ree Y. Dolnick; Paul K. Wallace; Hans Minderman; Gergel Le; Forsberg Em; Boyer Jm; Storey; Pilsmaker Cd; Russell A. Hammond; Jenifer Widger; Karuna Sundarapandiyan; Andrea Crocker; Henry C. Marsh; Tibor Keler

T-cell immunoglobulin and mucin domain 1 (TIM-1) is a type I transmembrane protein that was originally described as kidney injury molecule 1 (KIM-1) due to its elevated expression in kidney and urine after renal injury. TIM-1 expression is also upregulated in several human cancers, most notably in renal and ovarian carcinomas, but has very restricted expression in healthy tissues, thus representing a promising target for antibody-mediated therapy. To this end, we have developed a fully human monoclonal IgG1 antibody specific for the extracellular domain of TIM-1. This antibody was shown to bind purified recombinant chimeric TIM-1-Fc protein and TIM-1 expressed on a variety of transformed cell lines, including Caki-1 (human renal clear cell carcinoma), IGROV-1 (human ovarian adenocarcinoma), and A549 (human lung carcinoma). Internalization studies using confocal microscopy revealed the antibody was rapidly internalized by cells in vitro, and internalization was confirmed by quantitative imaging flow cytometry. An antibody–drug conjugate (ADC) was produced with the anti-TIM-1 antibody covalently linked to the potent cytotoxin, monomethyl auristatin E (MMAE), and designated CDX-014. The ADC was shown to exhibit in vitro cytostatic or cytotoxic activity against a variety of TIM-1–expressing cell lines, but not on TIM-1–negative cell lines. Using the Caki-1, IGROV-1, and A549 xenograft mouse models, CDX-014 showed significant antitumor activity in a clinically relevant dose range. Safety evaluation in nonhuman primates has demonstrated a good profile and led to the initiation of clinical studies of CDX-014 in renal cell carcinoma and potentially other TIM-1–expressing tumors. Mol Cancer Ther; 15(12); 2946–54. ©2016 AACR.


Journal for ImmunoTherapy of Cancer | 2013

Combination therapies augment the anti-tumor activity of agonist CD27 mAb in human CD27 transgenic mouse models

Li-Zhen He; Lawrence J. Thomas; James Testa; Jeffrey Weidlick; Crystal Sisson; Russell A. Hammond; Laura Vitale; Henry C. Marsh; Tibor Keler

CDX-1127 is a fully human antibody to CD27, a TNF receptor superfamily member expressed on the majority of T cells and subsets of NK cells and B cells. We have previously characterized the co-stimulatory activities of CDX-1127 with human T cell cultures and a human CD27 transgenic mouse model (hCD27-Tg). Similar to the findings originally shown by M.J. Glennie and colleagues using an agonist anti-mouse CD27 mAb, CDX-1127 has potent antitumor activity as monotherapy in several syngeneic tumor models in hCD27-Tg mice. In the current studies, we sought to enhance the anti-tumor efficacy of CDX-1127 in challenging tumor settings by combination with clinically relevant therapies. Specifically, we focused on therapies that could decrease or control tumor growth while providing a source of antigen to drive anti-tumor immunity (e.g. chemotherapy or targeted therapy) and immune modifiers that may allow the CD27 driven T cell response to overcome self-regulation (e.g. checkpoint inhibitors or immune activators). In the EG7 delayed treatment model (average tumor size of ~ 50 mm3 when treatment initiated), the combination of CDX-1127 with cyclophosphamide significantly improved survival (>70% survival) compared to either agent alone (<30 % survival). Notably, we found that the combination therapy was associated with increases in the ratio of effector to regulatory T cells in the tumors compared to either single agent group. Additional combination studies with various agents are on-going, and initial studies with CDX-1127 combined with anti-CTLA-4 mAb has shown superior anti-tumor activity (median survival 36.5 days in combination versus 20 days with either single agent). These studies, along with the good safety profile of CDX-1127 reported in a Phase 1 clinical trial, supports the design of future combination studies in patients with cancer.


Journal of Molecular Biology | 2008

The Partly Folded Back Solution Structure Arrangement of the 30 SCR Domains in Human Complement Receptor Type 1 (CR1) Permits Access to its C3b and C4b Ligands

Patricia B. Furtado; Chen Y. Huang; Demvihin Ihyembe; Russell A. Hammond; Henry C. Marsh; Stephen J. Perkins


Glycobiology | 2004

Production of a complement inhibitor possessing sialyl Lewis X moieties by in vitro glycosylation technology

Lawrence J. Thomas; Krishnasamy Panneerselvam; David T. Beattie; Michele Picard; Bi Xu; Charles W. Rittershaus; Henry C. Marsh; Russell A. Hammond; Jun Qian; Tom Stevenson; David Zopf; Robert Bayer


Molecular Immunology | 2007

The partly-folded back solution structure arrangement of the 30 SCR domains in human complement receptor type 1 (CR1) indicates inter-SCR flexibility

Patricia B. Furtado; Chen Y. Huang; Demvihin Ihyembe; Russell A. Hammond; Henry C. Marsh; Stephen J. Perkins


Molecular Cancer Therapeutics | 2018

Abstract B194: Identification and characterization of monoclonal antibodies targeting the Tyro3, Axl and MerTK (TAM) family of receptor tyrosine kinases

Diego Alvarado; Laura Vitale; Michael Murphy; Thomas O'Neill; Andrew Proffitt; Jay S. Lillquist; Gwenda F. Ligon; Craig Polson; James Storey; Jenifer Widger; Laura Mills-Chen; Karuna Sundarapandiyan; Andrea Crocker; Colleen Patterson; Biwei Zhao; Russell A. Hammond; Li-Zhen He; Venky Ramakrishna; Joel Goldstein; Lawrence J. Thomas; Henry C. Marsh; Tibor Keler; Richard Gedrich


Cancer Research | 2014

Abstract 2649: Development of an antibody-drug conjugate targeting TIM-1 for the treatment of ovarian and renal cell carcinoma

Lawrence J. Thomas; Laura Vitale; Thomas O'Neill; Ree Y. Dolnick; Paul K. Wallace; Hans Minderman; Lauren E. Gergel; Eric Forsberg; James M. Boyer; James Storey; Russell A. Hammond; Jennifer Widger; Karuna Sundarapandiyan; Andrea Crocker; Henry C. Marsh; Tibor Keler

Collaboration


Dive into the Russell A. Hammond's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Laura Vitale

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Danniele G. Holanda

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge