Network


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

Hotspot


Dive into the research topics where Anish Suri is active.

Publication


Featured researches published by Anish Suri.


Bioanalysis | 2012

Pellet digestion: a simple and efficient sample preparation technique for LC–MS/MS quantification of large therapeutic proteins in plasma

Zheng Ouyang; Michael T. Furlong; Steven Wu; Bogdan Sleczka; James Tamura; Haiqing Wang; Suzanne J. Suchard; Anish Suri; Timothy Olah; Adrienne A. Tymiak; Mohammed Jemal

BACKGROUND There is a need for a simple and efficient sample preparation technique for LC-MS/MS quantification of large therapeutic proteins in plasma. RESULTS The sample preparation technique presented here is based upon trypsin digestion of the pellet obtained following precipitation of the protein analyte from plasma. The pellet digestion technique was shown to facilitate efficient digestion of large therapeutic proteins, with concomitant removal of a substantial amount of potentially problematic plasma phospholipids. The technique was successfully applied to a pharmacokinetic study of a large therapeutic protein. CONCLUSION This simple sample preparation approach will be beneficial to bioanalytical laboratories engaged in the LC-MS/MS quantification of large therapeutic proteins in biological matrices.


Journal of Immunology | 2014

Engineering of a Novel Anti-CD40L Domain Antibody for Treatment of Autoimmune Diseases

Jenny Xie; Aaron P. Yamniuk; Virna Borowski; Robert Kuhn; Vojkan Susulic; Sandra Rex-Rabe; Xiaoxia Yang; Xiadi Zhou; Yifan Zhang; Kathleen M. Gillooly; Ruth Brosius; Rathna Ravishankar; Kimberly Waggie; Kathy Mink; Laura A. Price; Robert Rehfuss; James Tamura; Yongmi An; Lin Cheng; Bozena Abramczyk; Olga Ignatovich; Philip Drew; Steven Grant; James W. Bryson; Suzanne J. Suchard; Luisa Salter-Cid; Steven G. Nadler; Anish Suri

CD40–CD40L interactions play a critical role in regulating immune responses. Blockade of CD40L by Abs, such as the anti-CD40L Ab 5c8, demonstrated positive clinical effects in patients with autoimmune diseases; however, incidents of thromboembolism (TE) precluded further development of these molecules. In this study, we examined the role of the Fc domain interaction with FcγRs in modulating platelet activation and potential for TE. Our results show that the interaction of the 5c8 wild-type IgG1 Fc domain with FcγRs is responsible for platelet activation, as measured by induction of PAC-1 and CD62P. A version of 5c8 with a mutated IgG1 tail was identified that showed minimal FcγR binding and platelet activation while maintaining full binding to CD40L. To address whether Fc effector function is required for immunosuppression, a potent Ab fragment, termed a “domain Ab” (dAb), against murine CD40L was identified and fused to a murine IgG1 Fc domain containing a D265A mutation that lacks Fc effector function. In vitro, this dAb–Fc demonstrated comparable potency to the benchmark mAb MR-1 in inhibiting B cell and dendritic cell activation. Furthermore, the anti-CD40L dAb–Fc exhibited a notable efficacy comparable to MR-1 in various preclinical models, such as keyhole limpet hemocyanin–induced Ab responses, alloantigen-induced T cell proliferation, “heart-to-ear” transplantation, and NZB × NZW F1 spontaneous lupus. Thus, our data show that immunosuppression and TE can be uncoupled and that a CD40L dAb with an inert Fc tail is expected to be efficacious for treating autoimmune diseases, with reduced risk for TE.


American Journal of Transplantation | 2013

An anti-CD154 domain antibody prolongs graft survival and induces Foxp3(+) iTreg in the absence and presence of CTLA-4 Ig.

D. F. Pinelli; Maylene E. Wagener; Danya Liu; Aaron P. Yamniuk; J. Tamura; S. Grant; Christian P. Larsen; Anish Suri; Steven G. Nadler; Mandy L. Ford

The use of monoclonal antibodies targeting the CD154 molecule remains one of the most effective means of promoting graft tolerance in animal models, but thromboembolic complications during early clinical trials have precluded their use in humans. Furthermore, the role of Fc‐mediated deletion of CD154‐expressing cells in the observed efficacy of these reagents remains controversial. Therefore, determining the requirements for anti‐CD154‐induced tolerance will instruct the development of safer but equally efficacious treatments. To investigate the mechanisms of action of anti‐CD154 therapy, two alternative means of targeting the CD40–CD154 pathway were used: a nonagonistic anti‐CD40 antibody and an Fc‐silent anti‐CD154 domain antibody. We compared these therapies to an Fc‐intact anti‐CD154 antibody in both a fully allogeneic model and a surrogate minor antigen model in which the fate of alloreactive cells could be tracked. Results indicated that anti‐CD40 mAbs as well as Fc‐silent anti‐CD154 domain antibodies were equivalent to Fc‐intact anti‐CD154 mAbs in their ability to inhibit alloreactive T cell expansion, attenuate cytokine production of antigen‐specific T cells and promote the conversion of Foxp3+ iTreg. Importantly, iTreg conversion observed with Fc‐silent anti‐CD154 domain antibodies was preserved in the presence of CTLA4‐Ig, suggesting that this therapy is a promising candidate for translation to clinical use.


Autoimmunity | 2010

Targeting lymphocyte co-stimulation: From bench to bedside

Nathan J. Felix; Anish Suri; Luisa Salter-Cid; Steven G. Nadler; Sheila Gujrathi; Michael Corbo; Richard Aranda

T and B lymphocytes are central regulators and effectors of immune responses and are believed to have a key role in many autoimmune diseases. Targeting the activation or effector function of lymphocytes is a potentially effective approach to treat autoimmunity. Typically, T-cell activation occurs after engagement of the T-cell receptor with its cognate peptide-major histocompatibility complex (signal 1) and subsequent engagement of co-stimulatory molecules (signal 2). This “second signal” contributes to T-cell activation by promoting proliferation, survival, and effector function. In general, activation in the absence of co-stimulation leads to a reduced immune response, anergy, or even tolerance. B-cell activation similarly requires co-stimulation for the development of complete effector function. The most potent co-stimulatory molecules identified to date are CD28 for T-cells and CD40 for B-cells. Both molecules are recognized for their potential as immune modulators; however, thus far neither molecule has been successfully targeted directly for the treatment of autoimmune disease. The only current therapy to target either of these pathways is cytotoxic T-lymphocyte antigen-4 (CTLA-4-Ig), which indirectly blocks CD28 signaling and has proven efficacy in rheumatoid arthritis and juvenile idiopathic arthritis patients. In addition to CD28 and CD40, an array of other co-stimulatory as well as inhibitory pathways has recently been identified and scientists are just beginning to understand how these different signaling pathways interact to regulate lymphocyte activation. In the more than two decades since the discovery of the first co-stimulatory molecule, the full clinical potential of these pathways is yet to be realized. In this review, we will primarily focus on CD28 and CD40 which are the most clinically validated co-stimulatory pathways, and briefly summarize and discuss some of the other T-cell co-stimulatory molecules.


American Journal of Transplantation | 2017

Fc-Silent Anti-CD154 Domain Antibody Effectively Prevents Nonhuman Primate Renal Allograft Rejection

Steven C. Kim; Walter Wakwe; Laura Higginbotham; David V. Mathews; Cynthia P. Breeden; Allison Stephenson; Joe Jenkins; Elizabeth Strobert; Karen D. Price; Laura A. Price; Robert Kuhn; Haiqing Wang; Aaron P. Yamniuk; Suzanne J. Suchard; Alton B. Farris; Thomas C. Pearson; Christian P. Larsen; Mandy L. Ford; Anish Suri; Steven G. Nadler; Andrew B. Adams

The advent of costimulation blockade provides the prospect for targeted therapy with improved graft survival in transplant patients. Perhaps the most effective costimulation blockade in experimental models is the use of reagents to block the CD40/CD154 pathway. Unfortunately, successful clinical translation of anti‐CD154 therapy has not been achieved. In an attempt to develop an agent that is as effective as previous CD154 blocking antibodies but lacks the risk of thromboembolism, we evaluated the efficacy and safety of a novel anti‐human CD154 domain antibody (dAb, BMS‐986004). The anti‐CD154 dAb effectively blocked CD40‐CD154 interactions but lacked crystallizable fragment (Fc) binding activity and resultant platelet activation. In a nonhuman primate kidney transplant model, anti‐CD154 dAb was safe and efficacious, significantly prolonging allograft survival without evidence of thromboembolism (Median survival time 103 days). The combination of anti‐CD154 dAb and conventional immunosuppression synergized to effectively control allograft rejection (Median survival time 397 days). Furthermore, anti‐CD154 dAb treatment increased the frequency of CD4+CD25+Foxp3+ regulatory T cells. This study demonstrates that the use of a novel anti‐CD154 dAb that lacks Fc binding activity is safe without evidence of thromboembolism and is equally as potent as previous anti‐CD154 agents at prolonging renal allograft survival in a nonhuman primate preclinical model.


Journal of Molecular Biology | 2016

Functional Antagonism of Human CD40 Achieved by Targeting a Unique Species-Specific Epitope.

Aaron P. Yamniuk; Anish Suri; Stanley R. Krystek; James Tamura; Vidhyashankar Ramamurthy; Robert Kuhn; Karen Carroll; Catherine A. Fleener; Rolf Ryseck; Lin Cheng; Yongmi An; Philip Drew; Steven Grant; Suzanne J. Suchard; Steven G. Nadler; James W. Bryson; Steven Sheriff

Current clinical anti-CD40 biologic agents include both antagonist molecules for the treatment of autoimmune diseases and agonist molecules for immuno-oncology, yet the relationship between CD40 epitope and these opposing biological outcomes is not well defined. This report describes the identification of potent antagonist domain antibodies (dAbs) that bind to a novel human CD40-specific epitope that is divergent in the CD40 of nonhuman primates. A similarly selected anti-cynomolgus CD40 dAb recognizing the homologous epitope is also a potent antagonist. Mutagenesis, biochemical, and X-ray crystallography studies demonstrate that the epitope is distinct from that of CD40 agonists. Both the human-specific and cynomolgus-specific molecules remain pure antagonists even when formatted as bivalent Fc-fusion proteins, making this an attractive therapeutic format for targeting hCD40 in autoimmune indications.


Archive | 2012

Antibody polypeptides that antagonize cd40

Anish Suri; Steven Sheriff; Suzanne J. Suchard; Aaron P. Yamniuk; Stanley R. Krystek; James Tamura; James W. Bryson; Steven Grant; Philip Drew


Archive | 2017

Methods of treating transplant rejection using a domain antibody directed against CD40L

Anish Suri; Steven G. Nadler; Christian P. Larsen; Andrew Briane Adams


Archive | 2012

Animal Models for Type 1 Diabetes

Anish Suri; Matteo Levisetti


Archive | 2016

polipeptídeos de anticorpo que antagonizam cd40l

Aaron P. Yamniuk; Anish Suri; James Tamura; James W. Bryson; Laura A. Price; Olga Ignatovich; Philip Drew; Robert Rehfuss; Steven G. Nadler; Steven Grant; Suzanne J. Suchard

Collaboration


Dive into the Anish Suri's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Olga Ignatovich

Laboratory of Molecular Biology

View shared research outputs
Researchain Logo
Decentralizing Knowledge