Network


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

Hotspot


Dive into the research topics where Jaume Pons is active.

Publication


Featured researches published by Jaume Pons.


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.


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.


FEBS Letters | 1995

Crystal structure of Bacillus licheniformis 1,3-1,4-β-d-glucan 4-glucanohydrolase at 1.8 Å resolution

Michael Hahn; Jaume Pons; Antoni Planas; Enrique Querol; Udo Heinemann

The crystal structure of the 1,3‐1,4‐β‐d‐glucan 4‐glucanohydrolase from Bacillus licheniformis is solved at a resolution of 1.8 Å and refined to R = 16.5%. The protein has a similar β‐sandwich structure as the homologous enzyme from Bacillus macerans and the hybrid H(A16‐M). This demonstrates that the jellyroll fold of these proteins is remarkably rigid and only weakly influenced by crystal contacts. The crystal structure permits to extend mechanistic considerations derived for the B. licheniformis enzyme to the entire class of bacterial 1,3‐1,4‐β‐d‐glucan 4‐glucanohydrolases.


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.


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.


Nature Biotechnology | 2015

Site-specific conjugation improves therapeutic index of antibody drug conjugates with high drug loading

Pavel Strop; Kathy Delaria; Davide Foletti; Jody Melton Witt; Adela Hasa-Moreno; Kris Poulsen; Meritxell Galindo Casas; Magdalena Grazyna Dorywalska; Santiago E. Farias; Ariel Pios; Victor Lui; Russell Dushin; Dahui Zhou; Thayalan Navaratnam; Thomas-Toan Tran; Janette Sutton; Kevin Lindquist; Bora Han; Shu-Hui Liu; David L. Shelton; Jaume Pons; Arvind Rajpal

Site-specific conjugation improves therapeutic index of antibody drug conjugates with high drug loading


Molecular Cancer Therapeutics | 2016

Molecular basis of valine-citrulline-PABC linker instability in site-specific ADCs and its mitigation by linker design

Magdalena Grazyna Dorywalska; Russell Dushin; Ludivine Moine; Santiago E. Farias; Dahui Zhou; Thayalan Navaratnam; Victor Lui; Adela Hasa-Moreno; Meritxell Galindo Casas; Thomas-Toan Tran; Kathy Delaria; Shu-Hui Liu; Davide Foletti; Christopher J. O'Donnell; Jaume Pons; David L. Shelton; Arvind Rajpal; Pavel Strop

The degree of stability of antibody–drug linkers in systemic circulation, and the rate of their intracellular processing within target cancer cells are among the key factors determining the efficacy of antibody–drug conjugates (ADC) in vivo. Previous studies demonstrated the susceptibility of cleavable linkers, as well as auristatin-based payloads, to enzymatic cleavage in rodent plasma. Here, we identify Carboxylesterase 1C as the enzyme responsible for the extracellular hydrolysis of valine-citrulline-p-aminocarbamate (VC-PABC)-based linkers in mouse plasma. We further show that the activity of Carboxylesterase 1C towards VC-PABC–based linkers, and consequently the stability of ADCs in mouse plasma, can be effectively modulated by small chemical modifications to the linker. While the introduced modifications can protect the VC-PABC–based linkers from extracellular cleavage, they do not significantly alter the intracellular linker processing by the lysosomal protease Cathepsin B. The distinct substrate preference of the serum Carboxylesterase 1C offers the opportunity to modulate the extracellular stability of cleavable ADCs without diminishing the intracellular payload release required for ADC efficacy. Mol Cancer Ther; 15(5); 958–70. ©2016 AACR.


Journal of Biological Chemistry | 1997

MUTATIONAL ANALYSIS OF THE MAJOR LOOP OF BACILLUS 1,3-1,4-BETA -D-GLUCAN 4-GLUCANOHYDROLASES : EFFECTS ON PROTEIN STABILITY AND SUBSTRATE BINDING

Jaume Pons; Enrique Querol; Antoni Planas

The carbohydrate-binding cleft of Bacillus licheniformis 1,3-1,4-β-d-glucan 4-glucanohydrolase is partially covered by the surface loop between residues 51 and 67, which is linked to β-strand-(87–95) of the minor β-sheet III of the protein core by a single disulfide bond at Cys61–Cys90. An alanine scanning mutagenesis approach has been applied to analyze the role of loop residues from Asp51 to Arg64 in substrate binding and stability by means of equilibrium urea denaturation, enzyme thermotolerance, and kinetics. The ΔΔG Ubetween oxidized and reduced forms is approximately constant for all mutants, with a contribution of 5.3 ± 0.2 kcal·mol−1 for the disulfide bridge to protein stability. A good correlation is observed between ΔG U values by reversible unfolding and enzyme thermotolerance. The N57A mutant, however, is more thermotolerant than the wild-type enzyme, whereas it is slightly less stable to reversible urea denaturation. Mutants with a <2-fold increase inK m correspond to mutations at residues not involved in substrate binding, for which the reduction in catalytic efficiency (k cat/K m ) is proportional to the loss of stability relative to the wild-type enzyme. Y53A, N55A, F59A, and W63A, on the other hand, show a pronounced effect on catalytic efficiency, with K m > 2-fold andk cat < 5% of the wild-type values. These mutated residues are directly involved in substrate binding or in hydrophobic packing of the loop. Interestingly, the mutation M58A yields an enzyme that is more active than the wild-type enzyme (7-fold increase in k cat), but it is slightly less stable.


Bioconjugate Chemistry | 2017

Improved Lysosomal Trafficking Can Modulate the Potency of Antibody Drug Conjugates

Rachel M. DeVay; Kathy Delaria; Guoyun Zhu; Charles Holz; Davide Foletti; Janette Sutton; Gary Louis Bolton; Russell Dushin; Christine Bee; Jaume Pons; Arvind Rajpal; Hong Liang; David L. Shelton; Shu-Hui Liu; Pavel Strop

Antibody drug conjugates (ADCs) provide an efficacious and relatively safe means by which chemotherapeutic agents can be specifically targeted to cancer cells. In addition to the selection of antibody targets, ADCs offer a modular design that allows selection of ADC characteristics through the choice of linker chemistries, toxins, and conjugation sites. Many studies have indicated that release of toxins bound to antibodies via noncleavable linker chemistries relies on the internalization and intracellular trafficking of the ADC. While this can make noncleavable ADCs more stable in the serum, it can also result in lower efficacy when their respective targets are not internalized efficiently or are recycled back to the cell surface following internalization. Here, we show that a lysosomally targeted ADC against the protein APLP2 mediates cell killing, both in vitro and in vivo, more effectively than an ADC against Trop2, a protein with less efficient lysosomal targeting. We also engineered a bispecific ADC with one arm targeting HER2 for the purpose of directing the ADC to tumors, and the other arm targeting APLP2, whose purpose is to direct the ADC to lysosomes for toxin release. This proof-of-concept bispecific ADC demonstrates that this technology can be used to shift the intracellular trafficking of a constitutively recycled target by directing one arm of the antibody against a lysosomally delivered protein. Our data also show limitations of this approach and potential future directions for development.

Collaboration


Dive into the Jaume Pons's collaboration.

Top Co-Authors

Avatar

Arvind Rajpal

University of California

View shared research outputs
Top Co-Authors

Avatar

Pavel Strop

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antoni Planas

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Hong Liang

California Institute for Quantitative Biosciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge