discover-glo-2025

May 27, 2025 in Barcelona
9:00 am - 6:00 pm CEST in Barcelona

Advancing Science and Translational Research
with Bioluminescent Technologies

Free registration
Parc Científic de Barcelona
Sala Dolores Aleu
c/Baldiri Reixac 15
08028 Barcelona

Barcelona, a vibrant hub of innovation, is easily accessible, seamlessly combining modern convenience with its rich cultural heritage. Located within the renowned Parc Científic de Barcelona, Sala Dolores Aleu sits at the heart of the city’s scientific community. Just a short distance from key research institutions and dynamic city attractions, this venue offers the perfect setting for collaboration and discovery.

Whether you're arriving by train, plane, or public transport, reaching c/Baldiri Reixac 15, 08028 Barcelona is straightforward. And if you prefer to drive, there are several local parking options available nearby.

9:00 AM – Registration and Welcome Breakfast
Guests will be greeted with coffee and a welcome pack


9:30 AM – Event Opening by Gijs J. Jochems, Ph.D. (General Manager, Promega Biotech Ibérica, S.L.)
Promega team will deliver a welcome address introducing luminescence applications and their relevance in modern research contextualizing the agenda. 


10:00 AM – 11:30 AM  Session I: Cutting-edge Biologics
Integrative technologies for biologics encompass advanced platforms and reagents that streamline the development of therapeutic proteins and antibodies. These solutions support a seamless workflow, from precise molecular characterization to functional assays that evaluate potency, stability and safety. 

  • "Application of Lumit® Technology for Rapid Potency Testing of Tumor Infiltrating Lymphocytes (TILs)"
    Gemma Arán, Ph.D., Cell Therapy Quality Control Specialist, Banc de Sang i Teixits
  • "Analytical Characterization of Monoclonal Antibodies Using Luminescence-Based and Cell-Based Bioassays."
    Anna Fernández, Ph.D., Analytical Development Scientist, mAbxience
  • "Advancing CAR-T cell manufacturing through tight control of cell activation in stirred-tank bioreactors"
    Margarida Costa, Ph.D. Student, Bioengineering for Cell-based Therapies, iBET
  • ROUND TABLE: Speakers + Moderator: Jokin Carrillo, Ph.D. (Director of Drug Discovery, CIMA-UNAV)

11:30 AM – 12:00 PM – Coffee Break


12:00 PM – 1:30 PM – Session II: How bright can GloMax® Galaxy shine the cellular universe in Real Time assays?
Introducing the GloMax® Galaxy Bioluminescence Imager, a compact, high-sensitivity platform that couples luminescence, fluorescence, BRET and brightfield imaging to reveal rare proteins, spatial biology and kinetic cellular events in real time through intuitive and multimodal workflows.

  • "Dual-Color Bioluminescence Analysis Using a High-Efficiency Resonance Energy Transfer Fusion of NanoLuc and HaloTag"
    Thomas Machleidt, Ph.D., Director of Advanced Technologies, R&D, Promega Corp. 
  • "SPAchip® technology for sensing and filming live-cell physiology"
    Julio Martín, Ph.D., Senior Strategic Liaison, Arrays for Cell Nanodevices, S.L.

1:30 PM – 3:00 PM – Lunch / Networking / Hands-on GloMax® Galaxy


3:00 PM – 4:30 PM – Session III:  Glow-driven pathways to light up innovative approaches

How latest advancements in bioluminescent technologies are being harnessed to interrogate and modulate complex disease-relevant signaling pathways will be explored, highlighting luminescence as a powerful tool accelerating biomolecular discovery and therapeutic innovation across biomedical research.

  • "Luminescence-based approaches to target RAF1 degradation via disruption of KRAS and CDC37 interactions in lung adenocarcinoma."
    Lucía Lomba and Gonzalo Aizpurua de Arteche, Predoctoral Researchers, Spanish National Cancer Research Centre (CNIO)
  • "Non-coding RNAs as Central Regulators of Gene Expression: Insights from Bioluminescence-Based Assays"
    Eric Rovira, Ph.D., Postdoctoral Researcher, Centro de Investigación Médica Aplicada (CIMA)
  • "Understanding GPCR5B function and dysfunction in leukodystrophy and multiple sclerosis using NanoLuc complementation methods"
    Raúl Estévez, Ph.D., Full Professor of Physiology. Faculty of Medicine. Principal Researcher at University of Barcelona/IDIBELL/CIBERER
  • ROUND TABLE. Speakers + Moderator: Israel Ramos, Ph.D. (Drug Screening Coordinator, IRB Barcelona)

4:30 PM – Event Closure
Thank-you note by Promega team.


4:45 PM – 6:00 PM – Farewell Cocktail and Hands-on GloMax® Galaxy

Informal space for attendees and the Promega team to connect and touch the instrument.

Secure your seat and register today (free of charge)
mcosta-ibet

Margarida Costa

Ph.D. Student
iBET - Instituto de Biologia Experimental e Tecnológica
Portugal
Bio

Margarida completed the Integrated MSc in Biomedical Engineering in 2021 by Instituto Superior Técnico, University of Lisbon, Portugal. She developed her master thesis at Instituto de Biologia Experimental e Tecnológica (iBET) under the scope of the international project CARDIOPATCH, which focused on developing scalable and integrated bioprocesses for manufacturing of mesenchymal stromal cells (MSC) and functional MSC-derived extracellular vesicles with augmented cardiac properties. In 2023, she started her PhD at iBET, aligned with the CARTool project funded by the National Portuguese Foundation for Science and Technology. Her research is centered on implementing a tightly controlled process to manufacture specific subsets of more persistent CAR T cells in a scalable and reproducible manner.

Abstract
Advancing CAR T cell manufacturing through tight control of cell activation in stirred-tank bioreactors

Chimeric Antigen Receptor (CAR) T-cell therapies have shown significant promise in treating hematological malignancies. Still, current production methods lack scalability and a clear understanding of how manufacturing parameters impact CAR T-cell phenotype. To address these limitations, we propose an integrated manufacturing process using stirred-tank bioreactors (STB) to enhance control over T-cell activation and expansion, aiming to increase process yields and produce specific subsets of more persistent CAR T cells. T cells expanded in STB demonstrated significantly greater expansion, higher viability, and reduced exhaustion compared to static cultures. Notably, higher proportion of CD8+ T cells, important for directly killing tumour cells, were generated in STB. As a proof-of-concept of a scalable process to manufacture CAR T cells targeting solid tumours, we have generated CAR T cells against HER2+ breast cancer cells. These results highlight the potential of scalable and tightly controlled manufacturing processes for generating CAR T cells with improved potency.
llomba

Lucía Lomba Riego

Predoctoral Researcher
Spanish National Cancer Research Centre (CNIO)
Spain
Bio
Lucía Lomba obtained her bachelor’s degree in Chemistry and Biology from the University of Coruña. She then completed her MSc in Biomedicine and Biochemistry at the Complutense University of Madrid. For the past two years, she has been working as a predoctoral researcher thanks to “La Caixa” Foundation fellowship at the Spanish National Cancer Research Centre. Her research focuses on developing nanobodies as a novel therapeutic approach for the treatment of KRAS-driven tumors. As part of this project, she employs luminescence-based techniques to interrogate protein-protein interactions (PPIs).
Abstract

Luminescence-based approaches to target RAF1 degradation via disruption of KRAS and CDC37 interactions in lung adenocarcinoma.

RAF1 is a critical effector of KRAS and plays an essential role in tumor progression. Our laboratory has demonstrated that genetic ablation of RAF1 induces lung tumor regression in murine models without causing toxicities, highlighting its therapeutic potential.
In this study, we explore two innovative strategies to promote RAF1 degradation. First, we target the KRAS-RAF1 interaction using a nanobody-based approach. Second, we aim to disrupt the RAF1-CDC37 interaction, which is critical for RAF1 stability, through the use of small molecules.
To identify compounds capable of targeting RAF1, we have developed medium-throughput luminiscence-based screening assays to monitor the disruption of these protein-protein interactions using NanoBRET® technology. Additionally, we used a cell line expressing a RAF1-HiBiT fusion protein to assess RAF1 degradation in a cellular context.
Our findings provide new insights into the regulation of RAF1 stability and its oncogenic functions, potentially paving the way for novel therapeutic strategies against KRAS-driven tumors.

gaizpurua

Gonzalo Aizpurua de Arteche

Predoctoral Researcher
Spanish National Cancer Research Centre (CNIO)
Spain
Bio
Gonzalo has a degree in Biomedicine from Universidad Francisco de Vitoria, where he completed his final project at the Rothlin-Ghosh Lab at Yale University (USA). During his studies, he undertook research internships at the Centro Nacional de Investigaciones Cardiovasculares (CNIC) in Dr. Ibáñez’s lab through the CICERONE program, and at the Centro Nacional de Biotecnología (CNB-CSIC) in Dr. Barber´s lab with a JAE-INTRO grant. He earned his Master’s degree in Molecular Biomedicine from Universidad Autónoma de Madrid, where he joined the Experimental Oncology group at the CNIO, under the supervision of Dr. Mariano Barbacid and Dr. Sara García to perform his Master´s thesis. Currently, he is conducting his PhD in the same laboratory thanks to a La Caixa Foundation Fellowship, focusing on the development of RAF1 degraders as a novel therapeutic strategy against KRAS-driven tumors.
Abstract

Luminescence-based approaches to target RAF1 degradation via disruption of KRAS and CDC37 interactions in lung adenocarcinoma.

RAF1 is a critical effector of KRAS and plays an essential role in tumor progression. Our laboratory has demonstrated that genetic ablation of RAF1 induces lung tumor regression in murine models without causing toxicities, highlighting its therapeutic potential.
In this study, we explore two innovative strategies to promote RAF1 degradation. First, we target the KRAS-RAF1 interaction using a nanobody-based approach. Second, we aim to disrupt the RAF1-CDC37 interaction, which is critical for RAF1 stability, through the use of small molecules.
To identify compounds capable of targeting RAF1, we have developed medium-throughput luminiscence-based screening assays to monitor the disruption of these protein-protein interactions using NanoBRET® technology. Additionally, we used a cell line expressing a RAF1-HiBiT fusion protein to assess RAF1 degradation in a cellular context.
Our findings provide new insights into the regulation of RAF1 stability and its oncogenic functions, potentially paving the way for novel therapeutic strategies against KRAS-driven tumors.

tm

Thomas Machleidt, Ph.D.

Director of Advanced Technologies, R&D
Promega Corp.
USA
Bio

Dr. Machleidt currently leads the Advanced Technology Group with in Promega R&D. His main interest is the development and application of novel technology platforms for the analysis of protein dynamics in cells. Dr. Machleidt earned his M.S. in Biology from the Eberhardt Karls University in Tuebingen. He received his Ph.D. in Immunology for his work on the signaling mechanisms of the pro-inflammatory cytokine TNF in Prof. Kronke’s lab at the Technical University of Munich. For his postdoc he joined the lab of Prof. Richard Anderson at UT Southwestern Medical Center where he investigated the structure and function of the caveolin protein family. He subsequently held R&D positions at Johnson & Johnson and Ansata Therapeutics. Prior to joining Promega in 2011 he worked for as a group leader for 7 years at Life Technologies. Dr. Machleidt has authored and co-authored over 60 peer reviewed publications and book chapters.

Abstract
Dual-Color Bioluminescence Analysis Using a High-Efficiency Resonance Energy Transfer Fusion of NanoLuc® and HaloTag®

NanoLuc® and its complementation technologies (e.g., NanoBiT®, LgBiT®/HiBiT®) are widely used in functional biology for their high sensitivity, broad dynamic range, and suitability for monitoring kinetics of biological processes. These systems have proven valuable in applications such as target engagement, protein degradation, and protein–protein interaction studies. However, a major limitation of NanoLuc® is its restriction to a single output channel, with peak emission at 460 nm, hindering its use in multiplexed assays.
To address this, previous strategies have fused NanoLuc® to fluorescent proteins to shift emission spectra via resonance energy transfer. Building on this concept, we developed a high-efficiency bioluminescence resonance energy transfer (BRET) system by inserting circularly permuted NanoLuc® (or LgBiT) into a surface loop of the self-labeling protein HaloTag®. This engineered cpNLuc-HaloTag® and cpLgBiT-HaloTag® (cpNLuc-HT and cpLgBiT-HT/HiBiT®) chimeras achieve exceptionally high BRET efficiency (>90%) by optimally positioning NanoLuc®/NanoBIT® near a fluorophore covalently bound to HaloTag®, such as JF549.
By pairing cpNLuc-HT /JF549 with unmodified NLuc, we created a dual-channel bioluminescent reporter platform with strong signal intensities and excellent spectral separation (>100 nm), enabling simultaneous real-time analyses. Here, we demonstrate this system in the context of PROTAC-induced degradation, allowing parallel analysis of both target and a control protein within the same well, using either plate-based assays or bioluminescence imaging. This dual-color technology offers a robust and convenient solution for internal normalization, assessment of off-target effects as well as opportunity to monitor two cellular events at the same time.
jmartin

Julio Martín, Ph.D.

Senior Strategic Liaison
Arrays for Cell Nanodevices, S.L.
Spain
Bio

Throughout 29 years of experience in the R&D of a big pharma (GSK), I developed skills to combine Science and Business. PhD in Biochemistry, Enzymologist by training and discoverer of innovative medicines by professional career and vocation. My motivation: explaining biological complexity and making innovative discoveries that transform society's quality of life.
My areas of expertise are preclinical drug discovery and management of innovation. Experience in highly collaborative “open innovation” environments within Global Health. Once I left GSK, I have worked as freelance, consultant and advisor, and CSO at a biotech start-up. Likewise, I have collaborated with different societies and foundations, such as European Ambassador of SLAS. Currently, Senior Strategic Liaison at A4cell.

The Sapiens secret to success is large-scale flexible cooperation” (Yuval Noah Harari)

Abstract
SPAchip® technology for sensing and filming live-cell physiology

Imagine monitoring live cells in real time, non-invasively and over long periods of time. Just like having an eye inside the cell. Traditional fluorescent probes often suffer from cytotoxicity and instability, limiting their use in live-cell imaging. To overcome this, SPAchip® offers a breakthrough solution, advancing cell imaging from simple morphological snapshots to dynamic physiological insights. SPAchip® enables fluorescence-based intracellular analysis of pH, Ca²⁺, ROS, and O₂ in single living cells. Built on functionalized silicon chips, it allows covalent attachment of chemical or biological probes. After non-invasive uptake, the chips remain stable and non-toxic within the cytosol. Their signal can be quantified via fluorescence microscopy or flow cytometry. Compatible with both 2D and 3D cell models, SPAchip® opens new possibilities for high-content, real-time cellular analytics. It is also fully compatible with the GloMax® Galaxy Bioluminescence Imager from Promega, enhancing its integration into existing imaging workflows.
restevez

Raúl Estévez, Ph.D.

Full Professor of Physiology. Faculty of Medicine. Principal Researcher
University of Barcelona/IDIBELL/CIBERER
Spain
Bio

I obtained my degree in Biochemistry at the UB in 1994 and a PhD in Biochemistry in 2000, both with highest honors. I also completed an MBA in 1999. Then, I did a postdoctoral stay at the ZMNH in Germany. In 2003, I returned to the UB with a Ramón y Cajal contract. In 2007 I gained a position as Professor of Physiology in the Faculty of Medicine (Bellvitge Campus, UB), working mainly on the leukodystrophy MLC, a rare disease affecting the myelin. I have been awarded two consecutive times with the ICREA Academia Prize. I belong to CIBERER since 2007, to the Institute of Neurosciences of UB and to IDIBELL. I am a vocal member of the SEBBM.

Abstract
Understanding GPCR5B function and dysfunction in leukodystrophy and multiple sclerosis using NanoLuc complementation methods

Our group is working on the molecular basis of a rare type of leukodystrophy named MLC, caused by mutations in MLC1, GLIALCAM, GPRC5B or AQP4, characterized by the presence of myelin vacuoles. Importantly, autoantibodies against GlialCAM and MLC1 are found in patients with multiple sclerosis, an autoimmune demyelinating disorder of the central nervous system. We have been using NanoLuc complementation methods to unravel the relationships between these proteins and to quantify the surface expression of pathogenic mutations. In addition, the simplicity of these assays was used to find compounds modulating the activity of these proteins. Our results have implications to understand the pathophysiology of these disorders and to provide new therapeutical solutions for these disorders affecting the myelin.
garan

Gemma Arán, Ph.D.

Cell Therapy Quality Control Specialist
Banc de Sang i Teixits
Spain
Bio

I have a background in Biomedical Science, with a Master’s in Advanced Immunology and a PhD focused on Innate Immunity. After completing my doctoral studies, I worked for two years as a postdoctoral researcher, finishing ongoing projects. I’m currently a Quality Control Specialist at the Blood and Tissue Bank in Barcelona, where I’m responsible for the quality control of various cell therapy products, including blood components, stem cell apheresis, cord blood, and ATMPs under GMP complience. I also support the development of new cell-based products for both conventional and advanced therapies.

Abstract
Application of Lumit® Technology for Rapid Potency Testing of Tumor Infiltrating Lymphocytes (TILs)

The release of advanced therapy medicinal products (ATMPs) requires the performance of potency assays to demonstrate the biological activity of the product. These assays are often time-consuming and involve complex experimental procedures. In the case of Tumor Infiltrating Lymphocytes (TIL) products, that are products freshly infused, there is a need for rapid potency testing due to its direct impact on timely clinical decision-making. To address this need, we have validated a rapid assay based on TNFα and IFNγ detection in the cell supernatant after stimulation, using Lumit® technology, which offers a significantly shorter turnaround time compared to conventional ELISA methods, and allows a faster release of the potency of TIL products.
afdez

Anna Fernández, Ph.D.

Analytical Development Scientist
mAbxience
Spain
Bio

I obtained a Biology degree from the University of Girona (2002) and completed my PhD at IIBB-CSIC under Prof. José Carlos Fernández-Checa, focusing on the mitochondrial regulation of cell death. In 2013, I joined the Department of Biomedical Sciences at the University of León under Prof. Javier González-Gallego, conducting research on hepatocellular carcinoma. Since 2015, I have been part of the R&D Analytical Development department at mAbxience, where we develop and qualify analytical methods for the biological characterization of monoclonal antibodies. Our work supports R&D activities and includes method transfer to Quality Control for GMP-compliant batch release. We specialize in cell-based assays, binding assays, and luminescence-based techniques to evaluate potency, mechanism of action, and binding affinity.

Abstract
Analytical Characterization of Monoclonal Antibodies Using Luminescence-Based and Cell-Based Bioassays.

The Analytical Development department at mAbxience R&D specializes in the development, optimization, and qualification of bioassays for the detailed biological characterization of monoclonal antibodies. These assays evaluate critical quality attributes such as potency, mechanism of action—including Fab- and Fc-mediated functions—and binding affinity. The department also manages technology transfer to Quality Control laboratories for GMP-compliant batch release. A range of luminescence- and cell-based assays from Promega is employed to assess specific antibody functions. The PD-1/PD-L1 Blockade Bioassay quantifies immune checkpoint inhibition, while the RANKL Bioassay measures RANK/RANKL pathway modulation. Fc-mediated effector functions, including antibody-dependent cellular cytotoxicity (ADCC), are evaluated using the Fc Effector Reporter Bioassay. Additionally, the Lumit® FcRn Binding Immunoassay characterizes FcRn interactions crucial for pharmacokinetics and half-life. These assays are robust, precise, and sensitive, supporting biosimilar development and their potential use as release assays to ensure consistent product quality and efficacy.
erovira

Eric Rovira, Ph.D.

Postdoctoral Researcher
Centro de Investigación Médica Aplicada (CIMA)
Spain
Bio

Dr. Eric Rovira Barreira is a geneticist who graduated from the Universitat Autònoma de Barcelona (UAB). He earned his MSc in Cell and Gene Therapy from University College London (UCL) in 2017 and completed his PhD in Biomedical Research at Universidad de Navarra (UNAV) in 2022. Dr. Rovira currently focuses on RNA structure and RNA engineering, with a particular interest in the regulation of gene expression in human cells. His multidisciplinary background spans genetics, immunology, cell and gene therapy, and biomedical research, equipping him with expertise in both fundamental and applied aspects of gene regulation.

Abstract
Non-coding RNAs as Central Regulators of Gene Expression: Insights from Bioluminescence-Based Assays

At our lab, we aim to harness the therapeutic potential of non-coding RNA (ncRNA). To address this objective, we pursue two complementary strategies. On the one hand, we engineer synthetic ncRNAs, known as riboswitches, which are designed to sense exogenous small molecules and modulate gene expression in a dose-dependent manner. On the other hand, we investigate ncRNAs that are deregulated in tumors to elucidate their previously uncharacterized roles in tumorigenesis and to identify novel druggable targets. Within this diverse landscape of tumor-associated ncRNAs, those capable of expressing microproteins are of particular interest, as these microproteins may serve as novel targets and/or tumor-specific neoantigens for vaccine development. Across all projects, bioluminescence assays - especially employing a split version of NanoLuc - has proven invaluable as quantitative readouts for gene expression regulation and as sensitive surrogates for detecting endogenous microproteins, which are typically expressed at low levels.