Network


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

Hotspot


Dive into the research topics where Arvind Rajpal is active.

Publication


Featured researches published by Arvind Rajpal.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Precise determination of the diversity of a combinatorial antibody library gives insight into the human immunoglobulin repertoire

Jacob Glanville; Wenwu Zhai; Jan Berka; Dilduz Telman; Gabriella Huerta; Gautam R. Mehta; Irene Ni; Li Mei; Purnima Sundar; Giles M. R. Day; David Cox; Arvind Rajpal; Jaume Pons

Antibody repertoire diversity, potentially as high as 1011 unique molecules in a single individual, confounds characterization by conventional sequence analyses. In this study, we present a general method for assessing human antibody sequence diversity displayed on phage using massively parallel pyrosequencing, a novel application of Kabat column-labeled profile Hidden Markov Models, and translated complementarity determining region (CDR) capture-recapture analysis. Pyrosequencing of domain amplicon and RCA PCR products generated 1.5 × 106 reads, including more than 1.9 × 105 high quality, full-length sequences of antibody variable fragment (Fv) variable domains. Novel methods for germline and CDR classification and fine characterization of sequence diversity in the 6 CDRs are presented. Diverse germline contributions to the repertoire with random heavy and light chain pairing are observed. All germline families were found to be represented in 1.7 × 104 sequences obtained from repeated panning of the library. While the most variable CDR (CDR-H3) presents significant length and sequence variability, we find a substantial contribution to total diversity from somatically mutated germline encoded CDRs 1 and 2. Using a capture-recapture method, the total diversity of the antibody library obtained from a human donor Immunoglobulin M (IgM) pool was determined to be at least 3.5 × 1010. The results provide insights into the role of IgM diversification, display library construction, and productive germline usages in antibody libraries and the humoral repertoire.


Journal of Biological Chemistry | 2012

Increasing Serum Half-life and Extending Cholesterol Lowering in Vivo by Engineering Antibody with pH-sensitive Binding to PCSK9

Javier Chaparro-Riggers; Hong Liang; Rachel M. DeVay; Lanfang Bai; Janette Sutton; Wei Chen; Tao Geng; Kevin Lindquist; Meritxell Galindo Casas; Leila Marie Boustany; Colleen Brown; Jeffrey R. Chabot; Bruce Charles Gomes; Pamela D. Garzone; Andrea Rossi; Pavel Strop; Dave Shelton; Jaume Pons; Arvind Rajpal

Background: An antagonistic anti-PCSK9 antibody exhibits target-mediated clearance, resulting in a dose-dependent PK. Results: Engineering of an antibody with pH-sensitive binding to PCSK9 decreases target-mediated clearance, resulting in increased PK and efficacy in vivo. Conclusion: pH-sensitive anti-PCSK9 antibodies are excellent candidates for therapeutic development. Significance: pH-sensitive antibodies may enable less frequent or lower dosing of antibodies hampered by target-mediated clearance and high antigen load. Target-mediated clearance and high antigen load can hamper the efficacy and dosage of many antibodies. We show for the first time that the mouse, cynomolgus, and human cross-reactive, antagonistic anti-proprotein convertase substilisin kexin type 9 (PCSK9) antibodies J10 and the affinity-matured and humanized J16 exhibit target-mediated clearance, resulting in dose-dependent pharmacokinetic profiles. These antibodies prevent the degradation of low density lipoprotein receptor, thus lowering serum levels of LDL-cholesterol and potently reducing serum cholesterol in mice, and selectively reduce LDL-cholesterol in cynomolgus monkeys. In order to increase the pharmacokinetic and efficacy of this promising therapeutic for hypercholesterolemia, we engineered pH-sensitive binding to mouse, cynomolgus, and human PCSK9 into J16, resulting in J17. This antibody shows prolonged half-life and increased duration of cholesterol lowering in two species in vivo by binding to endogenous PCSK9 in mice and cynomolgus monkeys, respectively. The proposed mechanism of this pH-sensitive antibody is that it binds with high affinity to PCSK9 in the plasma at pH 7.4, whereas the antibody-antigen complex dissociates at the endosomal pH of 5.5–6.0 in order to escape from target-mediated degradation. Additionally, this enables the antibody to bind to another PCSK9 and therefore increase the antigen-binding cycles. Furthermore, we show that this effect is dependent on the neonatal Fc receptor, which rescues the dissociated antibody in the endosome from degradation. Engineered pH-sensitive antibodies may enable less frequent or lower dosing of antibodies hampered by target-mediated clearance and high antigen load.


The EMBO Journal | 2003

Transcriptional activation of known and novel apoptotic pathways by Nur77 orphan steroid receptor.

Arvind Rajpal; Yuri A. Cho; Biana Yelent; Petra Koza-Taylor; Dongling Li; Elaine Chen; Michael Whang; Chulho Kang; Thomas G. Turi; Astar Winoto

Nur77 is a nuclear orphan steroid receptor that has been implicated in negative selection. Expression of Nur77 in thymocytes and cell lines leads to apoptosis through a mechanism that remains unclear. In some cell lines, Nur77 was reported to act through a transcription‐independent mechanism involving translocation to mitochondria, leading to cytochrome c release. However, we show here that Nur77‐mediated apoptosis in thymocytes does not involve cytoplasmic cytochrome c release and cannot be rescued by Bcl‐2. Microarray analysis shows that Nur77 induces many genes, including two novel genes (NDG1, NDG2) and known apoptotic genes FasL and TRAIL. Characterization of NDG1 and NDG2 indicates that NDG1 initiates a novel apoptotic pathway in a Bcl‐2‐independent manner. Thus Nur77‐mediated apoptosis in T cells involves Bcl‐2 independent transcriptional activation of several known and novel apoptotic pathways.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Naive antibody gene-segment frequencies are heritable and unaltered by chronic lymphocyte ablation.

Jacob Glanville; Tracy Chia-Chien Kuo; H.-Christian von Büdingen; Lin Guey; Jan Berka; Purnima Sundar; Gabriella Huerta; Gautam R. Mehta; Jorge R. Oksenberg; Stephen L. Hauser; D. R. Cox; Arvind Rajpal; Jaume Pons

A diverse antibody repertoire is essential for an effective adaptive immune response to novel molecular surfaces. Although past studies have observed common patterns of V-segment use, as well as variation in V-segment use between individuals, the relative contributions to variance from genetics, disease, age, and environment have remained unclear. Using high-throughput sequence analysis of monozygotic twins, we show that variation in naive VH and DH segment use is strongly determined by an individuals germ-line genetic background. The inherited segment-use profiles are resilient to differential environmental exposure, disease processes, and chronic lymphocyte depletion therapy. Signatures of the inherited profiles were observed in class switched germ-line use of each individual. However, despite heritable segment use, the rearranged complementarity-determining region-H3 repertoires remained highly specific to the individual. As it has been previously demonstrated that certain V-segments exhibit biased representation in autoimmunity, lymphoma, and viral infection, we anticipate our findings may provide a unique mechanism for stratifying individual risk profiles in specific diseases.


Journal of Pharmacology and Experimental Therapeutics | 2012

Proprotein Convertase Substilisin/Kexin Type 9 Antagonism Reduces Low-Density Lipoprotein Cholesterol in Statin-Treated Hypercholesterolemic Nonhuman Primates

Hong Liang; Javier Chaparro-Riggers; Pavel Strop; Tao Geng; Janette Sutton; Daniel Tsai; Lanfang Bai; Yasmina Noubia Abdiche; Jeanette Dilley; Jessica Yu; Si Wu; Sherman M Chin; Nicole A Lee; Andrea Rossi; John C. Lin; Arvind Rajpal; Jaume Pons; David L. Shelton

Proprotein convertase substilisin/kexin type 9 (PCSK9) promotes the degradation of low-density lipoprotein (LDL) receptor (LDLR) and thereby increases serum LDL-cholesterol (LDL-C). We have developed a humanized monoclonal antibody that recognizes the LDLR binding domain of PCSK9. This antibody, J16, and its precursor mouse antibody, J10, potently inhibit PCSK9 binding to the LDLR extracellular domain and PCSK9-mediated down-regulation of LDLR in vitro. In vivo, J10 effectively reduces serum cholesterol in C57BL/6 mice fed normal chow. J16 reduces LDL-C in healthy and diet-induced hypercholesterolemic cynomologous monkeys, but does not significantly affect high-density lipoprotein-cholesterol. Furthermore, J16 greatly lowered LDL-C in hypercholesterolemic monkeys treated with the HMG-CoA reductase inhibitor simvastatin. Our data demonstrate that anti-PCSK9 antibody is a promising LDL-C-lowering agent that is both efficacious and potentially additive to current therapies.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Anti–IL-7 receptor-α reverses established type 1 diabetes in nonobese diabetic mice by modulating effector T-cell function

Li-Fen Lee; Kathryn Logronio; Guang Huan Tu; Wenwu Zhai; Irene Ni; Li Mei; Jeanette Dilley; Jessica Yu; Arvind Rajpal; Colleen Brown; Charles Takeshi Appah; Sherman Michael Chin; Bora Han; Timothy Affolter; John C. Lin

Genetic variation in the IL-7 receptor-α (IL-7R) gene is associated with susceptibility to human type 1 diabetes (T1D). Here we investigate the therapeutic efficacy and mechanism of IL-7Rα antibody in a mouse model of T1D. IL-7Rα antibody induces durable, complete remission in newly onset diabetic mice after only two to three injections. IL-7 increases, whereas IL-7Rα antibody therapy reduces, the IFN-γ–producing CD4+ (TH1) and IFN-γ–producing CD8+ T cells. Conversely, IL-7 decreases and IL-7Rα antibody enhances the inhibitory receptor Programmed Death 1 (PD-1) expression in the effector T cells. Programmed Death 1 blockade reversed the immune tolerance mediated by the IL-7Rα antibody therapy. Furthermore, IL-7Rα antibody therapy increases the frequency of regulatory T cells without affecting their suppressor activity. The durable efficacy and the multipronged tolerogenic mechanisms of IL-7Rα antibody therapy suggest a unique disease-modifying approach to T1D.


Bioconjugate Chemistry | 2015

Effect of attachment site on stability of cleavable antibody drug conjugates.

Magdalena Grazyna Dorywalska; Pavel Strop; Jody A. Melton-Witt; Adela Hasa-Moreno; Santiago E. Farias; Meritxell Galindo Casas; Kathy Delaria; Victor Lui; Kris Poulsen; Carole M. Loo; Stellanie Krimm; Gary Louis Bolton; Ludivine Moine; Russell Dushin; Thomas-Toan Tran; Shu-Hui Liu; Mathias Rickert; Davide Foletti; David L. Shelton; Jaume Pons; Arvind Rajpal

The systemic stability of the antibody-drug linker is crucial for delivery of an intact antibody-drug conjugate (ADC) to target-expressing tumors. Linkers stable in circulation but readily processed in the target cell are necessary for both safety and potency of the delivered conjugate. Here, we report a range of stabilities for an auristatin-based payload site-specifically attached through a cleavable valine-citrulline-p-aminobenzylcarbamate (VC-PABC) linker across various sites on an antibody. We demonstrate that the conjugation site plays an important role in determining VC-PABC linker stability in mouse plasma, and that the stability of the linker positively correlates with ADC cytotoxic potency both in vitro and in vivo. Furthermore, we show that the VC-PABC cleavage in mouse plasma is not mediated by Cathepsin B, the protease thought to be primarily responsible for linker processing in the lysosomal degradation pathway. Although the VC-PABC cleavage is not detected in primate plasma in vitro, linker stabilization in the mouse is an essential prerequisite for designing successful efficacy and safety studies in rodents during preclinical stages of ADC programs. The divergence of linker metabolism in mouse plasma and its intracellular cleavage offers an opportunity for linker optimization in the circulation without compromising its efficient payload release in the target cell.


Current Opinion in Immunology | 2003

Apoptosis during lymphoid development.

Sue J. Sohn; Arvind Rajpal; Astar Winoto

Recent investigations have provided important insights into how signaling through the antigen receptors determines whether a cell survives or dies. In T cells, Grb2 and MAP kinases play essential roles in differentiating between apoptotic and survival signals. The PTEN phosphatase and Bim, a pro-apoptotic Bcl-2 family member, regulate apoptosis in both T and B cells. In B cells, antigen receptor-mediated death can be rescued by co-stimulation, in which the roles of protein kinase C and BAFF, a TNF family member, have been recently elucidated. In a recently identified mechanism of regulating inflammation, receptors such as c-mer and glycoproteins such as MFG-E8 were found to participate in the clearance of apoptotic cells.


mAbs | 2015

The neonatal Fc receptor (FcRn) binds independently to both sites of the IgG homodimer with identical affinity.

Yasmina Noubia Abdiche; Yik Andy Yeung; Javier Chaparro-Riggers; Ishita Barman; Pavel Strop; Sherman Michael Chin; Amber Pham; Gary Louis Bolton; Dan McDonough; Kevin Lindquist; Jaume Pons; Arvind Rajpal

The neonatal Fc receptor (FcRn) is expressed by cells of epithelial, endothelial and myeloid lineages and performs multiple roles in adaptive immunity. Characterizing the FcRn/IgG interaction is fundamental to designing therapeutic antibodies because IgGs with moderately increased binding affinities for FcRn exhibit superior serum half-lives and efficacy. It has been hypothesized that 2 FcRn molecules bind an IgG homodimer with disparate affinities, yet their affinity constants are inconsistent across the literature. Using surface plasmon resonance biosensor assays that eliminated confounding experimental artifacts, we present data supporting an alternate hypothesis: 2 FcRn molecules saturate an IgG homodimer with identical affinities at independent sites, consistent with the symmetrical arrangement of the FcRn/Fc complex observed in the crystal structure published by Burmeister et al. in 1994. We find that human FcRn binds human IgG1 with an equilibrium dissociation constant (KD) of 760 ± 60 nM (N = 14) at 25°C and pH 5.8, and shows less than 25% variation across the other human subtypes. Human IgG1 binds cynomolgus monkey FcRn with a 2-fold higher affinity than human FcRn, and binds both mouse and rat FcRn with a 10-fold higher affinity than human FcRn. FcRn/IgG interactions from multiple species show less than a 2-fold weaker affinity at 37°C than at 25°C and appear independent of an IgGs variable region. Our in vivo data in mouse and rat models demonstrate that both affinity and avidity influence an IgGs serum half-life, which should be considered when choosing animals, especially transgenic systems, as surrogates.


Bioconjugate Chemistry | 2014

Mass Spectrometric Characterization of Transglutaminase Based Site-Specific Antibody–Drug Conjugates

Santiago E. Farias; Pavel Strop; Kathy Delaria; Meritxell Galindo Casas; Magdalena Grazyna Dorywalska; David L. Shelton; Jaume Pons; Arvind Rajpal

Antibody drug conjugates (ADCs) are becoming an important new class of therapeutic agents for the treatment of cancer. ADCs are produced through the linkage of a cytotoxic small molecule (drug) to monoclonal antibodies that target tumor cells. Traditionally, most ADCs rely on chemical conjugation methods that yield heterogeneous mixtures of varying number of drugs attached at different positions. The potential benefits of site-specific drug conjugation in terms of stability, manufacturing, and improved therapeutic index has recently led to the development of several new site-specific conjugation technologies. However, detailed characterization of the degree of site specificity is currently lacking. In this study we utilize mass spectrometry to characterize the extent of site-specificity of an enzyme-based site-specific antibody-drug conjugation technology that we recently developed. We found that, in addition to conjugation of the engineered site, a small amount of aglycosylated antibody present in starting material led to conjugation at position Q295, resulting in approximately 1.3% of off-target conjugation. Based on our detection limits, we show that Q295N mutant eliminates the off-target conjugation yielding highly homogeneous conjugates that are better than 99.8% site-specific. Our study demonstrates the importance of detailed characterization of ADCs and describes methods that can be utilized to characterize not only our enzyme based conjugates, but also ADCs generated by other conjugation technologies.

Collaboration


Dive into the Arvind Rajpal's collaboration.

Top Co-Authors

Avatar

Pavel Strop

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jaume Pons

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julien Valton

University of Copenhagen

View shared research outputs
Researchain Logo
Decentralizing Knowledge