Network


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

Hotspot


Dive into the research topics where Montserrat Carrasco-Triguero is active.

Publication


Featured researches published by Montserrat Carrasco-Triguero.


Bioanalysis | 2013

Bioanalytical assay strategies for the development of antibody–drug conjugate biotherapeutics

Surinder Kaur; Keyang Xu; Ola Saad; Randall Dere; Montserrat Carrasco-Triguero

Antibody-drug conjugates (ADCs) are monoclonal antibodies with covalently bound cytotoxic drugs. They are designed to target tumor antigens selectively and offer the hope of cancer treatment without the debilitating side-effects of conventional therapies. The concept of ADCs is not new; however, development of these therapeutics is challenging and only recently are promising clinical data emerging. These challenges include ADC bioanalysis, such as quantifying in serum/plasma for PK studies and strategies for assessing immunogenicity. ADCs have complex molecular structures incorporating large- and small-molecule characteristics and require diverse analytical methods, including ligand-binding assays and MS-based methods. ADCs are typically mixtures with a range of drug-to-antibody ratios. Biotransformations in vivo can lead to additional changes in drug-to-antibody ratios resulting in dynamically changing mixtures. Thus, a standard calibration curve consisting of the reference standard may not be appropriate for quantification of analytes in vivo and represents a unique challenge. This paper will share our perspective on why ADC bioanalysis is so complex and describe the strategies and rationale that we have used for ADCs, with highlights of original data from a variety of nonclinical and clinical case studies. Our strategy has involved novel protein structural characterization tools to help understand ADC biotransformations in vivo and use of the analyte knowledge gained to guide the development of quantitative bioanalytical assays.


Bioanalysis | 2013

Immunogenicity assays for antibody–drug conjugates: case study with ado-trastuzumab emtansine

Montserrat Carrasco-Triguero; Joo-Hee Yi; Randall Dere; Zhihua Julia Qiu; Corinna Lei; Yanhong Li; Connie Mahood; Bei Wang; Douglas D. Leipold; Kirsten Achilles Poon; Surinder Kaur

BACKGROUND Antibody-drug conjugates (ADCs) such as Kadcyla™ (ado-trastuzumab emtansine [T-DM1]) present covalently bound cytotoxic drugs, which may influence their immunogenicity potential compared with antibody therapies. Therefore, ADCs require assay strategies that allow measurement of responses to all the molecular components. RESULTS The immunogenicity strategy for T-DM1 used a risk-based, tiered approach that included screening and titration to detect antitherapeutic antibodies; confirmation of positive responses; and characterization to assess whether the immune response is primarily to the antibody or to the linker-drug and/or new epitopes in trastuzumab resulting from conjugation. CONCLUSION The tiered immunogenicity assay strategy for T-DM1 allowed detection of antitherapeutic antibodies to all components of the ADC in multiple nonclinical and clinical studies. Characterization strategies implemented in clinical studies provided additional insights into the specificity of the immune response.


Aaps Journal | 2015

Immunogenicity of Antibody Drug Conjugates: Bioanalytical Methods and Monitoring Strategy for a Novel Therapeutic Modality

M. Benjamin Hock; Karen Thudium; Montserrat Carrasco-Triguero; Nikolai F. Schwabe

Immunogenicity (the development of an adaptive immune response reactive with a therapeutic) is a well-described but unwanted facet of biotherapeutic development. There are commonly applied procedures for immunogenicity risk assessment, testing strategies, and bioanalysis. With some modifications, these can be applied to new biotherapeutic modalities. For novel therapies such as antibody-drug conjugates (ADCs), the unique structural components may contribute additional complexities to both immunologic responses and bioanalytical methods. US product inserts (USPIs) for two commercially available ADCs detail the incidence of immunogenicity; however, the body of literature on immunogenicity of ADCs is limited. We recently participated in a conference session on this topic (Annual meeting of the American Association of Pharmaceutical Scientists, held November 2013 in San Antonio, TX, USA. The meeting featured the Symposium: Immunogenicity Assessment for Novel Antibody Drug Conjugates, Nonclinical to Clinical) which prompted an effort to share our perspectives on how immunogenicity risk assessment, testing strategies, and bioanalytical methods can be adapted to reflect the complexity of ADC therapeutics.


Bioanalysis | 2012

Overcoming soluble target interference in an anti-therapeutic antibody screening assay for an antibody-drug conjugate therapeutic

Montserrat Carrasco-Triguero; Connie Mahood; Marija Milojic-Blair; Caroline Amaya; Jane Ruppel; Kyu Hong; Joo-Hee Yi; Surinder Kaur

BACKGROUND The standard safety evaluation of biotherapeutics includes assessment of immunogenicity. Anti-therapeutic antibodies (ATA) can be detected in serum using immunoassays with a bridging format. However, these assays can be subject to interference. RESULTS In the bridging ATA assay for 3A5 TDC, an antibody-drug conjugate that binds to the multimeric extracellular domain of MUC16 (CA125), soluble CA125 in the serum caused false-positive results by binding to the ATA assay reagents. This interaction was blocked by wheat germ agglutinin lectin as it binds to the glycans in CA125; thus, the specificity of the assay improved. CONCLUSION The assay development and validation results showed that the addition of wheat germ agglutinin eliminates the interference from circulating CA125 without impacting the ability to detect ATA.


mAbs | 2016

Pharmacokinetics and pharmacodynamics of DSTA4637A: A novel THIOMAB™ antibody antibiotic conjugate against Staphylococcus aureus in mice

Chenguang Zhou; Sophie M. Lehar; Johnny Gutierrez; Carrie M. Rosenberger; Nina Ljumanovic; Jb Dinoso; Neelima Koppada; Kyu Hong; Amos Baruch; Montserrat Carrasco-Triguero; Ola Saad; Sanjeev Mariathasan; Amrita V. Kamath

ABSTRACT DSTA4637A, a novel THIOMAB™ antibody antibiotic conjugate (TAC) against Staphylococcus aureus (S. aureus), is currently being investigated as a potential therapy against S. aureus infections. Structurally, TAC is composed of an anti-S. aureus antibody linked to a potent antibiotic, dmDNA31. The goal of the current study was to characterize the pharmacokinetics (PK) of TAC in mice, assess the effect of S. aureus infection on its PK, and evaluate its pharmacodynamics (PD) by measuring the bacterial load in various organs at different timepoints following TAC treatment. Plasma concentrations of 3 analytes, total antibody (TAb), antibody-conjugated dmDNA31 (ac-dmDNA31), and unconjugated dmDNA31, were measured in these studies. In non-infected mice (target antigen absent), following intravenous (IV) administration of a single dose of TAC, systemic concentration-time profiles of both TAb and ac-dmDNA31 were bi-exponential and characterized by a short distribution phase and a long elimination phase as expected for a monoclonal antibody-based therapeutic. Systemic exposures of both TAb and ac-dmDNA31 were dose proportional over the dose range tested (5 to 50 mg/kg). In a mouse model of systemic S. aureus infection (target antigen present), a single IV dose of TAC demonstrated PK behavior similar to that in the non-infected mice, and substantially reduced bacterial load in the heart, kidney, and bones on 7 and 14 d post dosing. These findings have increased our understanding of the PK and PK/PD of this novel molecule, and have shown that at efficacious dose levels the presence of S. aureus infection had minimal effect on TAC PK.


Clinical & Developmental Immunology | 2016

Application of a Plug-and-Play Immunogenicity Assay in Cynomolgus Monkey Serum for ADCs at Early Stages of Drug Development

Montserrat Carrasco-Triguero; Helen Davis; Yuda Zhu; Daniel Coleman; Denise Nazzal; Paul Vu; Surinder Kaur

Immunogenicity assessment during early stages of nonclinical biotherapeutic development is not always warranted. It is rarely predictive for clinical studies and evidence for the presence of anti-drug antibodies (ADAs) may be inferred from the pharmacokinetic (PK) profile. However, collecting and banking samples during the course of the study are prudent for confirmation and a deeper understanding of the impact on PK and safety. Biotherapeutic-specific ADA assays commonly developed can require considerable time and resources. In addition, the ADA assay may not be ready when needed if the study of PK and safety data triggers assay development. During early stages of drug development for antibody-drug conjugates (ADCs), there is the added complication of the potential inclusion of several molecular variants in a study, differing in the linker and/or drug components. To simplify analysis of ADAs at this stage, we developed plug-and-play generic approaches for both the assay format and the data analysis steps. Firstly, the assay format uses generic reagents to detect ADAs. Secondly, we propose a cut point methodology based on animal specific baseline variability instead of a population data approach. This assay showed good sensitivity, drug tolerance, and reproducibility across a variety of antibody-derived biotherapeutics without the need for optimization across molecules.


Bioanalysis | 2015

Insights on the immunogenicity of antibody–drug conjugates

Montserrat Carrasco-Triguero

Biotherapeutics can elicit an immune response that may alter their PK, efficacy and safety. Novel biotherapeutics such as antibody– drug conjugates (ADCs) contain chemical domains and have hapten-like structures that may increase their immunogenicity risk. ADCs are composed of a cytotoxic drug linked to a monoclonal antibody (mAb) which recognizes a tumor-associated antigen. The mechanism of action involves targeted delivery of the cytotoxic drug into cancer cells. Compared with traditional chemotherapeutics, ADCs offer the promise of reduced systemic exposure, lower off-target side effects and a higher therapeutic index. Although the concept of ADCs is not new, only two examples are approved for the US market: brentuximab vedotin (Adcetris, Seattle Genetics) and ado-trastuzumab emtansine (Kadcyla, Genentech-Roche). Another ADC that gained approval but was later removed from the US market is gentuzumab ozogamicin (Mylotarg, Pfizer). These ADCs and most of those in clinical development [1] use conventional lysine or cysteine conjugation chemistries that result in a product with heterogeneous drug–antibody ratios, the complexity of which further increases in vivo [2]. Companies are working to make ADCs with reduced drug–antibody ratio heterogeneity, improved conjugate stability and the possibility of an increased therapeutic window [1]. From an immunogenicity perspective, conventional and next-generation ADCs have a hapten-like structure with potentially increased immunogenicity compared with the stand-alone corresponding mAb. The cytotoxic drug component in ADCs is the hapten while the mAb is the carrier. When administered by itself, a hapten is unlikely to induce immunogenicity, but may do so when coupled to a carrier [3]. Humoral immune responses to ADCs can include anti-therapeutic antibodies (ATAs) against all components of the ADC, including the cytotoxic drug, the mAb and neoepitopes formed in the mAb as a result of the conjugation [3]. ATAs against different domains of an ADC could have different effects on PK, efficacy and safety; this is an important point to consider when developing immunogenicity strategies. Immunogenicity strategies followed for other biotherapeutics are appropriate for ADCs, but with additional characterization to better understand the ATA specificity [4–6]. These strategies should include a risk-based assessment and assays that are consistent with industry best practices [7].


Bioanalysis | 2016

Commercial biomarker assays: friend and foe

Saloumeh Kadkhodayan Fischer; Montserrat Carrasco-Triguero; Kyu Hong; Jeremy Good; Teresa Davancaze; Trung Nguy; Jessica Li; Walter C. Darbonne; Alyssa Morimoto; Yuda Zhu

AIM Commercial kits can provide a convenient solution for measuring circulating biomarkers to support drug development. However, their suitability should be assessed in the disease matrix of interest. METHODOLOGY Twelve biomarkers were evaluated in samples from patients with rheumatoid arthritis. We used immunoassay kits from five vendors on three multiplexed platforms. Kit suitability was evaluated on the basis of detectability and prespecified performance acceptance criteria. RESULTS Assays had varying levels of sensitivity and susceptibility to interference by matrix components. Only a few assays in the multiplexed kits were found to be suitable. In general, kits for analytes that passed our assay criteria showed good correlation between vendors. CONCLUSION The data from this study demonstrate that the majority of assays on multiplexed kits evaluated either lacked sensitivity and/or had poor performance, which diminishes the utility of the multiplexing approach.


Aaps Journal | 2017

Development and Translational Application of an Integrated, Mechanistic Model of Antibody-Drug Conjugate Pharmacokinetics.

Siddharth Sukumaran; Crystal Zhang; Douglas D. Leipold; Ola Saad; Keyang Xu; Kapil Gadkar; Divya Samineni; Bei Wang; Marija Milojic-Blair; Montserrat Carrasco-Triguero; Bonnee Rubinfeld; Paul J. Fielder; Kedan Lin; Saroja Ramanujan

Antibody drug conjugates (ADC), in which small molecule cytotoxic agents are non-specifically linked to antibodies, can enable targeted delivery of chemotherapeutics to tumor cells. ADCs are often produced and administered as a mixture of conjugated antibodies with different drug to antibody ratios (DAR) resulting in complex and heterogeneous disposition kinetics. We developed a mechanism-based platform model that can describe and predict the complex pharmacokinetic (PK) behavior of ADCs with protease-cleavable valine-citrulline (VC) linker linked to Monomethylmonomethyl auristatin F/E by incorporating known mechanisms of ADC disposition. The model includes explicit representation of all DAR species; DAR-dependent sequential deconjugation of the drug, resulting in the conversion of higher DAR to lower DAR species; and DAR-dependent antibody/ADC clearance. PK profiles of multiple analytes (total antibody, drug-conjugated antibody, and/or antibody-conjugated drug) for different ADC molecules and targets in rodents and cynomolgus monkeys were used for model development. The integrated cross-species model was successful in capturing the multi-analyte PK profiles after administration of purified ADCs with defined DAR species and ADCs with mixtures of DAR. Human PK predictions for DSTP3086S (anti-STEAP1-vc-MMAE) with the platform model agreed well with PK (total antibody and antibody-conjugated drug concentrations) measurements in the dose-ranging phase I clinical study. The integrated model is applicable to various other ADCs with different formats, conjugated drugs, and linkers, and provides a valuable tool for the exploration of mechanisms governing disposition of ADCs and enables translational predictions.


mAbs | 2018

Preclinical pharmacokinetics and pharmacodynamics of DCLL9718A: An antibody-drug conjugate for the treatment of acute myeloid leukemia

Douglas D. Leipold; Isabel Figueroa; Shabkhaiz Masih; Brandon Latifi; Victor Yip; Ben-Quan Shen; Randall Dere; Montserrat Carrasco-Triguero; M. Violet Lee; Ola Saad; Luna Liu; Jintang He; Dian Su; Keyang Xu; Brian R. Vuillemenot; Steven T. Laing; Melissa Schutten; Katherine R. Kozak; Bing Zheng; Andrew G. Polson; Amrita V. Kamath

ABSTRACT Few treatment options are available for acute myeloid leukemia (AML) patients. DCLL9718A is an antibody-drug conjugate that targets C-type lectin-like molecule-1 (CLL-1). This receptor is prevalent on monocytes, neutrophils, and AML blast cells, and unlike CD33, is not expressed on hematopoietic stem cells, thus providing possible hematopoietic recovery. DCLL9718A comprises an anti-CLL-1 IgG1 antibody (MCLL0517A) linked to a pyrrolobenzodiazepine (PBD) dimer payload, via a cleavable disulfide-labile linker. Here, we characterize the in vitro and in vivo stability, the pharmacokinetics (PK) and pharmacodynamics (PD) of DCLL9718A and MCLL0517A in rodents and cynomolgus monkeys. Three key PK analytes were measured in these studies: total antibody, antibody-conjugated PBD dimer and unconjugated PBD dimer. In vitro, DCLL9718A, was stable with most (> 80%) of the PBD dimer payload remaining conjugated to the antibody over 96 hours. This was recapitulated in vivo with antibody-conjugated PBD dimer clearance estimates similar to DCLL9718A total antibody clearance. Both DCLL9718A and MCLL0517A showed linear PK in the non-binding rodent species, and non-linear PK in cynomolgus monkeys, a binding species. The PK data indicated minimal impact of conjugation on the disposition of DCLL9718A total antibody. Finally, in cynomolgus monkey, MCLL0517A showed target engagement at all doses tested (0.5 and 20 mg/kg) as measured by receptor occupancy, and DCLL9718A (at doses of 0.05, 0.1 and 0.2 mg/kg) showed strong PD activity as evidenced by notable reduction in monocytes and neutrophils.

Collaboration


Dive into the Montserrat Carrasco-Triguero'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
Researchain Logo
Decentralizing Knowledge