Network


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

Hotspot


Dive into the research topics where Meritxell Galindo Casas is active.

Publication


Featured researches published by Meritxell Galindo Casas.


Chemistry & Biology | 2013

Location Matters: Site of Conjugation Modulates Stability and Pharmacokinetics of Antibody Drug Conjugates

Pavel Strop; Shu-Hui Liu; Magdalena Grazyna Dorywalska; Kathy Delaria; Russell Dushin; Thomas-Toan Tran; Wei-Hsien Ho; Santiago E. Farias; Meritxell Galindo Casas; Yasmina Noubia Abdiche; Dahui Zhou; Ramalakshmi Y. Chandrasekaran; Caroline Samain; Carole M. Loo; Andrea Rossi; Mathias Rickert; Stellanie Krimm; Teresa Wong; Sherman Michael Chin; Jessica Yu; Jeanette Dilley; Javier Chaparro-Riggers; Gary Frederick Filzen; Christopher J. O’Donnell; Fang Wang; Jeremy Myers; Jaume Pons; David L. Shelton; Arvind Rajpal

Antibody drug conjugates (ADCs) are a therapeutic class offering promise for cancer therapy. The attachment of cytotoxic drugs to antibodies can result in an effective therapy with better safety potential than nontargeted cytotoxics. To understand the role of conjugation site, we developed an enzymatic method for site-specific antibody drug conjugation using microbial transglutaminase. This allowed us to attach diverse compounds at multiple positions and investigate how the site influences stability, toxicity, and efficacy. We show that the conjugation site has significant impact on ADC stability and pharmacokinetics in a species-dependent manner. These differences can be directly attributed to the position of the linkage rather than the chemical instability, as was observed with a maleimide linkage. With this method, it is possible to produce homogeneous ADCs and tune their properties to maximize the therapeutic window.


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 Molecular Biology | 2012

Generating Bispecific Human IgG1 and IgG2 Antibodies from Any Antibody Pair

Pavel Strop; Wei-Hsien Ho; Leila Marie Boustany; Yasmina Noubia Abdiche; Kevin Lindquist; Santiago E. Farias; Mathias Rickert; Charles Takeshi Appah; Edward Derrick Pascua; Teresa Radcliffe; Janette Sutton; Javier Chaparro-Riggers; Wei Chen; Meritxell Galindo Casas; Sherman Michael Chin; Oi Kwan Wong; Shu-Hui Liu; German J. Vergara; Dave Shelton; Arvind Rajpal; Jaume Pons

Bispecific antibodies and antibody fragments are a new class of therapeutics increasingly utilized in the clinic for T cell recruitment (catumaxomab anti-EpCAM/CD3 and blinatumomab anti-CD19/CD3), increase in the selectivity of targeting, or simultaneous modulation of multiple cellular pathways. While the clinical potential for certain bispecific antibody formats is clear, progress has been hindered because they are often difficult to manufacture, may suffer from suboptimal pharmacokinetic properties, and may be limited due to potential immunogenicity issues. Current state-of-the-art human IgG-like bispecific technologies require co-expression of two heavy chains with a single light chain, use crossover domains to segregate light chains, or utilize scFv (single-chain fragment variable)-Fc fusion. We have engineered both human IgG1 and IgG2 subtypes, with minimal point mutations, to form full-length bispecific human antibodies with high efficiency and in high purity. In our system, the two antibodies of interest can be expressed and purified separately, mixed together under appropriate redox conditions, resulting in a formation of a stable bispecific antibody with high yields. With this approach, it is not necessary to generate new antibodies that share a common light chain, therefore allowing the immediate use of an existing antibody regardless of whether it has been generated via standard hybridoma or display methods. We demonstrate the generality of the approach and show that these bispecific antibodies have properties similar to those of wild-type IgGs, and we further demonstrate the utility of the technology with an example of a CD3/CD20 bispecific antibody that effectively depletes B cells in vitro and in vivo.


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.


Journal of Molecular Biology | 2013

Mechanism of action and in vivo efficacy of a human-derived antibody against Staphylococcus aureus α-hemolysin.

Davide Foletti; Pavel Strop; Lee Shaughnessy; Adela Hasa-Moreno; Meritxell Galindo Casas; Marcella Russell; Christine Bee; Si Wu; Amber Pham; Zhilan Zeng; Jaume Pons; Arvind Rajpal; Dave Shelton

The emergence and spread of multi-drug-resistant strains of Staphylococcus aureus in hospitals and in the community emphasize the urgency for the development of novel therapeutic interventions. Our approach was to evaluate the potential of harnessing the human immune system to guide the development of novel therapeutics. We explored the role of preexisting antibodies against S. aureus α-hemolysin in the serum of human individuals by isolating and characterizing one antibody with a remarkably high affinity to α-hemolysin. The antibody provided protection in S. aureus pneumonia, skin, and bacteremia mouse models of infection and also showed therapeutic efficacy when dosed up to 18 h post-infection in the pneumonia model. Additionally, in pneumonia and bacteremia animal models, the therapeutic efficacy of the α-hemolysin antibody appeared additive to the antibiotic linezolid. To better understand the mechanism of action of this isolated antibody, we solved the crystal structure of the α-hemolysin:antibody complex. To our knowledge, this is the first report of the crystal structure of the α-hemolysin monomer. The structure of the complex shows that the antibody binds α-hemolysin between the cap and the rim domains. In combination with biochemical data, the structure suggests that the antibody neutralizes the activity of the toxin by preventing binding to the plasma membrane of susceptible host cells. The data presented here suggest that protective antibodies directed against S. aureus molecules exist in some individuals and that such antibodies have a therapeutic potential either alone or in combination with antibiotics.


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 | 2016

Engineering Highly Potent and Selective Microproteins against Nav1.7 Sodium Channel for Treatment of Pain.

Anatoly Shcherbatko; Andrea Rossi; Davide Foletti; Guoyun Zhu; Oren Bogin; Meritxell Galindo Casas; Mathias Rickert; Adela Hasa-Moreno; Victor V. Bartsevich; Andreas Crameri; Alexander Steiner; Robert Henningsen; Avinash Gill; Jaume Pons; David L. Shelton; Arvind Rajpal; Pavel Strop

The prominent role of voltage-gated sodium channel 1.7 (Nav1.7) in nociception was revealed by remarkable human clinical and genetic evidence. Development of potent and subtype-selective inhibitors of this ion channel is crucial for obtaining therapeutically useful analgesic compounds. Microproteins isolated from animal venoms have been identified as promising therapeutic leads for ion channels, because they naturally evolved to be potent ion channel blockers. Here, we report the engineering of highly potent and selective inhibitors of the Nav1.7 channel based on tarantula ceratotoxin-1 (CcoTx1). We utilized a combination of directed evolution, saturation mutagenesis, chemical modification, and rational drug design to obtain higher potency and selectivity to the Nav1.7 channel. The resulting microproteins are highly potent (IC50 to Nav1.7 of 2.5 nm) and selective. We achieved 80- and 20-fold selectivity over the closely related Nav1.2 and Nav1.6 channels, respectively, and the IC50 on skeletal (Nav1.4) and cardiac (Nav1.5) sodium channels is above 3000 nm. The lead molecules have the potential for future clinical development as novel therapeutics in the treatment of pain.


PLOS ONE | 2015

Site-Dependent Degradation of a Non-Cleavable Auristatin-Based Linker-Payload in Rodent Plasma and Its Effect on ADC Efficacy

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

The efficacy of an antibody-drug conjugate (ADC) is dependent on the properties of its linker-payload which must remain stable while in systemic circulation but undergo efficient processing upon internalization into target cells. Here, we examine the stability of a non-cleavable Amino-PEG6-based linker bearing the monomethyl auristatin D (MMAD) payload site-specifically conjugated at multiple positions on an antibody. Enzymatic conjugation with transglutaminase allows us to create a stable amide linkage that remains intact across all tested conjugation sites on the antibody, and provides us with an opportunity to examine the stability of the auristatin payload itself. We report a position-dependent degradation of the C terminus of MMAD in rodent plasma that has a detrimental effect on its potency. The MMAD cleavage can be eliminated by either modifying the C terminus of the toxin, or by selection of conjugation site. Both approaches result in improved stability and potency in vitro and in vivo. Furthermore, we show that the MMAD metabolism in mouse plasma is likely mediated by a serine-based hydrolase, appears much less pronounced in rat, and was not detected in cynomolgus monkey or human plasma. Clarifying these species differences and controlling toxin degradation to optimize ADC stability in rodents is essential to make the best ADC selection from preclinical models. The data presented here demonstrate that site selection and toxin susceptibility to mouse plasma degradation are important considerations in the design of non-cleavable ADCs, and further highlight the benefits of site-specific conjugation methods.

Collaboration


Dive into the Meritxell Galindo Casas's collaboration.

Researchain Logo
Decentralizing Knowledge