75 results on '"Compte, Marta"'
Search Results
52. Efficient production of single-chain fragment variable-based N-terminal trimerbodies in Pichia pastoris
- Author
-
Blanco-Toribio, Ana, primary, Lacadena, Javier, additional, Nuñez-Prado, Natalia, additional, Álvarez-Cienfuegos, Ana, additional, Villate, Maider, additional, Compte, Marta, additional, Sanz, Laura, additional, Blanco, Francisco J, additional, and Álvarez-Vallina, Luis, additional
- Published
- 2014
- Full Text
- View/download PDF
53. Functional comparison of single-chain and two-chain anti-CD3-based bispecific antibodies in gene immunotherapy applications
- Author
-
Compte, Marta, primary, Álvarez-Cienfuegos, Ana, additional, Nuñez-Prado, Natalia, additional, Sainz-Pastor, Noelia, additional, Blanco-Toribio, Ana, additional, Pescador, Nuria, additional, Sanz, Laura, additional, and Álvarez-Vallina, Luis, additional
- Published
- 2014
- Full Text
- View/download PDF
54. Lipopolysaccharide Activates Toll-like Receptor 4 (TLR4)-mediated NF-κB Signaling Pathway and Proinflammatory Response in Human Pericytes
- Author
-
Guijarro-Muñoz, Irene, primary, Compte, Marta, additional, Álvarez-Cienfuegos, Ana, additional, Álvarez-Vallina, Luis, additional, and Sanz, Laura, additional
- Published
- 2014
- Full Text
- View/download PDF
55. Basement membrane-rich Organoids with functional human blood vessels are permissive niches for human breast cancer metastasis
- Author
-
Fernández-Periáñez, Rodrigo, Molina-Privado, Irene, Rojo, Federico, Guijarro-Muñoz, Irene, Alonso-Camino, Vanesa, Zazo, Sandra, Compte, Marta, Álvarez-Cienfuegos, Ana, Cuesta, Ángel M., Sánchez-Martín, David, Álvarez-Méndez, Ana M., Sanz, Laura, Álvarez-Vallina, Luis, Fernández-Periáñez, Rodrigo, Molina-Privado, Irene, Rojo, Federico, Guijarro-Muñoz, Irene, Alonso-Camino, Vanesa, Zazo, Sandra, Compte, Marta, Álvarez-Cienfuegos, Ana, Cuesta, Ángel M., Sánchez-Martín, David, Álvarez-Méndez, Ana M., Sanz, Laura, and Álvarez-Vallina, Luis
- Abstract
Metastasic breast cancer is the leading cause of death by malignancy in women worldwide. Tumor metastasis is a multistep process encompassing local invasion of cancer cells at primary tumor site, intravasation into the blood vessel, survival in systemic circulation, and extravasation across the endothelium to metastasize at a secondary site. However, only a small percentage of circulating cancer cells initiate metastatic colonies. This fact, together with the inaccessibility and structural complexity of target tissues has hampered the study of the later steps in cancer metastasis. In addition, most data are derived from in vivo models where critical steps such as intravasation/extravasation of human cancer cells are mediated by murine endothelial cells. Here, we developed a new mouse model to study the molecular and cellular mechanisms underlying late steps of the metastatic cascade. We have shown that a network of functional human blood vessels can be formed by co-implantation of human endothelial cells and mesenchymal cells, embedded within a reconstituted basement membrane-like matrix and inoculated subcutaneously into immunodeficient mice. The ability of circulating cancer cells to colonize these human vascularized organoids was next assessed in an orthotopic model of human breast cancer by bioluminescent imaging, molecular techniques and immunohistological analysis. We demonstrate that disseminated human breast cancer cells efficiently colonize organoids containing a functional microvessel network composed of human endothelial cells, connected to the mouse circulatory system. Human breast cancer cells could be clearly detected at different stages of the metastatic process: initial arrest in the human microvasculature, extravasation, and growth into avascular micrometastases. This new mouse model may help us to map the extravasation process with unprecedented detail, opening the way for the identification of relevant targets for therapeutic intervention.
- Published
- 2013
56. Antibody Gene Therapy: Getting Closer to Clinical Application?
- Author
-
Guijarro-Munoz, Irene, primary, Compte, Marta, additional, Alvarez-Vallina, Luis, additional, and Sanz, Laura, additional
- Published
- 2013
- Full Text
- View/download PDF
57. Basement Membrane-Rich Organoids with Functional Human Blood Vessels Are Permissive Niches for Human Breast Cancer Metastasis
- Author
-
Fernández-Periáñez, Rodrigo, primary, Molina-Privado, Irene, additional, Rojo, Federico, additional, Guijarro-Muñoz, Irene, additional, Alonso-Camino, Vanesa, additional, Zazo, Sandra, additional, Compte, Marta, additional, Álvarez-Cienfuegos, Ana, additional, Cuesta, Ángel M., additional, Sánchez-Martín, David, additional, Álvarez-Méndez, Ana M., additional, Sanz, Laura, additional, and Álvarez-Vallina, Luis, additional
- Published
- 2013
- Full Text
- View/download PDF
58. In Vivo Secretion of Bispecific Antibodies Recruiting Lymphocytic Effector Cells
- Author
-
Compte, Marta, primary, Nuñez-Prado, Natalia, additional, Sanz, Laura, additional, and Alvarez-Vallina, Luis, additional
- Published
- 2013
- Full Text
- View/download PDF
59. CARbodies: Human Antibodies Against Cell Surface Tumor Antigens Selected From Repertoires Displayed on T Cell Chimeric Antigen Receptors
- Author
-
Alonso-Camino, Vanesa, primary, Sánchez-Martín, David, additional, Compte, Marta, additional, Nuñez-Prado, Natalia, additional, Diaz, Rosa M, additional, Vile, Richard, additional, and Alvarez-Vallina, Luis, additional
- Published
- 2013
- Full Text
- View/download PDF
60. Lymphocyte Display: A Novel Antibody Selection Platform Based on T Cell Activation
- Author
-
Alonso-Camino, Vanesa, primary, Sánchez-Martín, David, additional, Compte, Marta, additional, and Álvarez-Vallina, Laura Sanz, Luis, additional
- Published
- 2009
- Full Text
- View/download PDF
61. In Vivo Tumor Targeting and Imaging with Engineered Trivalent Antibody Fragments Containing Collagen-Derived Sequences
- Author
-
Cuesta, Ángel M., primary, Sánchez-Martín, David, additional, Sanz, Laura, additional, Bonet, Jaume, additional, Compte, Marta, additional, Kremer, Leonor, additional, Blanco, Francisco J., additional, Oliva, Baldomero, additional, and Álvarez-Vallina, Luis, additional
- Published
- 2009
- Full Text
- View/download PDF
62. Tumor Immunotherapy Using Gene-Modified Human Mesenchymal Stem Cells Loaded into Synthetic Extracellular Matrix Scaffolds
- Author
-
Compte, Marta, primary, Cuesta, Ángel M., additional, Sánchez-Martín, David, additional, Alonso-Camino, Vanesa, additional, Vicario, José Luís, additional, Sanz, Laura, additional, and Álvarez-Vallina, Luís, additional
- Published
- 2009
- Full Text
- View/download PDF
63. Long-term in vivo imaging of human angiogenesis: Critical role of bone marrow-derived mesenchymal stem cells for the generation of durable blood vessels
- Author
-
Sanz, Laura, primary, Santos-Valle, Patricia, additional, Alonso-Camino, Vanesa, additional, Salas, Clara, additional, Serrano, Antonio, additional, Vicario, José Luís, additional, Cuesta, Ángel M., additional, Compte, Marta, additional, Sánchez-Martín, David, additional, and Álvarez-Vallina, Luís, additional
- Published
- 2008
- Full Text
- View/download PDF
64. Microencapsulation of therapeutic bispecific antibodies producing cells: immunotherapeutic organoids for cancer management.
- Author
-
Saenz del Burgo, Laura, Compte, Marta, Aceves, Mónica, Hernández, Rosa María, Sanz, Laura, Álvarez-Vallina, Luis, and Pedraz, Jose Luis
- Subjects
- *
CANCER treatment , *MICROENCAPSULATION , *ALGINATES , *IMMUNOTHERAPY , *NANOMEDICINE - Abstract
Regardless of the important therapeutic advances developed over the last years for the management of cancer, the fact is that many patients still suffer from a tremendous reduction on their quality of life due to lack of complete selectivity of conventionally administered chemotherapeutic drugs. In the search of more efficacious tumor-targeted therapies, the use of bispecific antibodies (bsAbs) capable of simultaneous binding to tumor-associated antigens and to an activating receptor, such as CD3, has emerged as a promising approach. With the intention to complementing and improving this cancer immunotherapy, human HEK-293 cells have been genetically modified ex vivo to secrete a recombinant anti-CEA (carcinoembryonic antigen) × anti-CD3 bsAb. After encapsulation in alginate-poly- l-lysine microcapsules, bsAb-secreting HEK-293 cells were monitorized for several weeks. This system has proved to be feasible for the maintenance of cell growth and recombinant antibody production giving proof-of-concept of its use as immunotherapeutic organoids in cancer treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
65. Enhanced antiangiogenic therapy with antibody-collagen XVIII NC1 domain fusion proteins engineered to exploit matrix remodeling events
- Author
-
Sánchez-Arévalo Lobo, Víctor J., primary, Cuesta, Ángel M., additional, Sanz, Laura, additional, Compte, Marta, additional, García, Pascal, additional, Prieto, Jesús, additional, Blanco, Francisco J., additional, and Álvarez-Vallina, Luis, additional
- Published
- 2006
- Full Text
- View/download PDF
66. ATTACK, a novel bispecific T cell-recruiting antibody with trivalent EGFR binding and monovalent CD3 binding for cancer immunotherapy.
- Author
-
Harwood, Seandean Lykke, Alvarez-Cienfuegos, Ana, Nuñez-Prado, Natalia, Compte, Marta, Hernández-Pérez, Sara, Merino, Nekane, Bonet, Jaume, Navarro, Rocio, Van Bergen en Henegouwen, Paul M. P., Lykkemark, Simon, Mikkelsen, Kasper, Mølgaard, Kasper, Jabs, Frederic, Sanz, Laura, Blanco, Francisco J., Roda-Navarro, Pedro, and Alvarez-Vallina, Luis
- Subjects
CANCER immunotherapy ,T cells ,BISPECIFIC antibodies ,THERAPEUTICS - Abstract
The redirection of T cell activity using bispecific antibodies is one of the most promising cancer immunotherapy approaches currently in development, but it is limited by cytokine storm-related toxicities, as well as the pharmacokinetics and tumor-penetrating capabilities of current bispecific antibody formats. Here, we have engineered the ATTACK (Asymmetric Tandem Trimerbody for T cell Activation and Cancer Killing), a novel T cell-recruiting bispecific antibody which combines three EGFR-binding single-domain antibodies (VHH; clone EgA1) with a single CD3-binding single-chain variable fragment (scFv; clone OKT3) in an intermediate molecular weight package. The two specificities are oriented in opposite directions in order to simultaneously engage cancer cells and T cell effectors, and thereby promote immunological synapse formation. EgA1 ATTACK was expressed as a homogenous, non-aggregating, soluble protein by mammalian cells and demonstrated an enhanced binding to EGFR, but not CD3, when compared to the previously characterized tandem bispecific antibody which has one EgA1 VHHand one OKT3 scFv per molecule. EgA1 ATTACK induced synapse formation and early signaling pathways downstream of TCR engagement at lower concentrations than the tandem VHH-scFv bispecific antibody. Furthermore, it demonstrated extremely potent, dose-dependent cytotoxicity when retargeting human T cells towards EGFR-expressing cells, with an efficacy over 15-fold higher than that of the tandem VHH-scFv bispecific antibody. These results suggest that the ATTACK is an ideal format for the development of the next-generation of T cell-redirecting bispecific antibodies. [ABSTRACT FROM PUBLISHER]
- Published
- 2018
- Full Text
- View/download PDF
67. Tumor targeted 4-1BB agonist antibody-albumin fusions with high affinity to FcRn induce anti-tumor immunity without toxicity
- Author
-
Oana Hangiu, Marta Compte, Anders Dinesen, Rocio Navarro, Antonio Tapia-Galisteo, Ole A. Mandrup, Ainhoa Erce-Llamazares, Rodrigo Lázaro-Gorines, Daniel Nehme-Álvarez, Carmen Domínguez-Alonso, Seandean L. Harwood, Carlos Alfonso, Belen Blanco, Laura Rubio-Pérez, Anaïs Jiménez-Reinoso, Laura Díez-Alonso, Francisco J. Blanco, Laura Sanz, Kenneth A. Howard, Luis Álvarez-Vallina, Ministerio de Ciencia e Innovación (España), European Commission, Instituto de Salud Carlos III, Asociación Española Contra el Cáncer, Fundación CRIS contra el Cáncer, Fundación BBVA, La Caixa, Novo Nordisk Foundation, Center for Multifunctional Biomolecular Drug Design (Denmark), Comunidad de Madrid, Ministerio de Ciencia, Innovación y Universidades (España), Hangiu, Oana, Compte, Marta, Navarro, Rocío, Tapia-Galisteo, Antonio, Mandrup, Ole A., Lázaro-Gorines, Rodrigo, Nehme-Álvarez, Daniel, Domínguez-Alonso, Carmen, Harwood, Seandean Lykke, Alfonso, Carlos, Jiménez-Reinoso, Anaïs, Díez-Alonso, Laura, Blanco, Francisco J., Sanz, Laura, Alvarez-Vallina, Luis, Hangiu, Oana [0000-0002-2641-8531], Compte, Marta [0000-0002-7138-9266], Navarro, Rocío [0000-0002-0083-7711], Tapia-Galisteo, Antonio [0000-0002-0507-8435], Mandrup, Ole A. [0000-0002-9700-4328], Lázaro-Gorines, Rodrigo [0000-0002-6885-3486], Nehme-Álvarez, Daniel [0000-0001-5054-5373], Domínguez-Alonso, Carmen [0000-0002-0446-9629], Harwood, Seandean Lykke [0000-0003-4654-8832], Alfonso, Carlos [0000-0001-7165-4800], Jiménez-Reinoso, Anaïs [0000-0001-8229-1881], Díez-Alonso, Laura [0000-0002-9545-6910], Blanco, Francisco J. [0000-0003-2545-4319], Sanz, Laura [0000-0002-3119-3218], and Alvarez-Vallina, Luis [0000-0003-3053-6757]
- Subjects
Multidisciplinary ,Immunology ,Immunological methods ,Immune response ,Cancer - Abstract
17 p.-4 fig.-1 tab.-1 grph. abst., Costimulation of tumor-infiltrating T lymphocytes by anti-4-1BB monoclonal antibodies (mAbs) has shown anti-tumor activity in human trials, but can be associated with significant off-tumor toxicities involving FcγR interactions. Here, we introduce albumin-fused mouse and human bispecific antibodies with clinically favorable pharmacokinetics designed to confine 4-1BB costimulation to the tumor microenvironment. These Fc-free 4-1BB agonists consist of an EGFR-specific VHH antibody, a 4-1BB-specific scFv, and a human albumin sequence engineered for high FcRn binding connected in tandem (LiTCo-Albu). We demonstrate in vitro cognate target engagement, EGFR-specific costimulatory activity, and FcRn-driven cellular recycling similar to non-fused FcRn high-binding albumin. The mouse LiTCo-Albu exhibited a prolonged circulatory half-life and in vivo tumor inhibition, with no indication of 4-1BB mAb-associated toxicity. Furthermore, we show a greater therapeutic effect when used in combination with PD-1-blocking mAbs. These findings demonstrate the feasibility of tumor-specific LiTCo-Albu antibodies for safe and effective costimulatory strategies in cancer immunotherapy., Financial support for this work was obtained from the MCIN/AEI/10.13039/501100011033 (SAF2017-89437-P and PDC2021-121711-100 to LA-V, PID2019-104544GB-I00 to CA, and PID2020-113225GB-I00 to FJB), partially supported by the European Regional Development Fund (ERDF); the Carlos III Health Institute (ISCIII) (PI19/00132 to LS; PI20/01030 to BB), partially supported by the ERDF; the ISCIII-RICORS within the Next Generation EU program (plan de Recuperación, Transformación y Resilencia); the Spanish Association Against Cancer (AECC 19084 to LA-V); the CRIS Cancer Foundation (FCRIS-2018-0042 and FCRIS-2021-0090 to LA-V), the BBVA Foundation (Ayudas Fundación BBVA a Equipos de Investigación Científica SARS-CoV-2 years COVID-19 to LA-V); and the Fundació “La Caixa” (HR21-00761 project IL7R_LungCan to LA-V). AD, OAM, and KAH were funded by the Novo Nordisk Foundation, Grant; CEMBID (Center for Multifunctional Biomolecular Drug Design, Grant Number: NNF17OC0028070). OH was supported by an industrial PhD fellowship from the Comunidad de Madrid (IND2020/BMD-17668). AE-L was supported industrial PhD fellowship from the Carlos III Health Institute (IFI18/00045). CD-A was supported by a predoctoral fellowship from the Spanish Ministry of Science Innovation and Universities (PRE2018-083445). LR-P was supported by a predoctoral fellowship from the Immunology Chair, Universidad Francisco de Vitoria/Merck. LD-A was supported by a Rio Hortega fellowship from the Carlos III Health Institute (CM20/00004).
- Published
- 2022
68. Trispecific T-cell engagers for dual tumor-targeting of colorectal cancer
- Author
-
Antonio Tapia-Galisteo, Íñigo Sánchez Rodríguez, Oscar Aguilar-Sopeña, Seandean Lykke Harwood, Javier Narbona, Mariola Ferreras Gutierrez, Rocío Navarro, Laura Martín-García, Cesáreo Corbacho, Marta Compte, Javier Lacadena, Francisco J. Blanco, Patrick Chames, Pedro Roda-Navarro, Luis Álvarez-Vallina, Laura Sanz, Hospital Universitario Puerta de Hierro-Majadahonda [Madrid, Spain], Universidad Complutense de Madrid = Complutense University of Madrid [Madrid] (UCM), Aarhus University [Aarhus], Centro de Investigaciones Biológicas Margarita Salas, Leadartis Sl, Centre de Recherche en Cancérologie de Marseille (CRCM), Aix Marseille Université (AMU)-Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Aix Marseille Université (AMU), Hospital Universitario 12 de Octubre [Madrid], Chames, Patrick, Instituto de Salud Carlos III, Comunidad de Madrid, Ministerio de Economía y Competitividad (España), Ministerio de Ciencia e Innovación (España), Asociación Española Contra el Cáncer, Fundación CRIS contra el Cáncer, Tapia-Galisteo, Antonio, Sánchez Rodríguez, Íñigo, Aguilar-Sopeña, Óscar, Harwood, Seandean Lykke, Ferreras-Gutiérrez, Mariola, Navarro, Rocío, Corbacho, Cesáreo, Compte, Marta, Lacadena, Javier, Blanco, Francisco J., Roda-Navarro, Pedro, Álvarez-Vallina, Luis, Sanz, Laura, Tapia-Galisteo, Antonio [0000-0002-0507-8435], Sánchez Rodríguez, Íñigo [0000-0002-6440-0922], Aguilar-Sopeña, Óscar [0000-0002-2435-8598], Harwood, Seandean Lykke [0000-0003-4654-8832], Ferreras-Gutiérrez, Mariola [0000-0003-4421-3158], Navarro, Rocío [0000-0002-0083-7711], Corbacho, Cesáreo [0000-0002-6644-3475], Compte, Marta [0000-0002-7138-9266], Lacadena, Javier [0000-0002-7314-0333], Blanco, Francisco J. [0000-0003-2545-4319], Chames, Patrick [0000-0002-6104-6286], Roda-Navarro, Pedro [0000-0003-3799-8823], Álvarez-Vallina, Luis [0000-0003-3053-6757], and Sanz, Laura [0000-0002-3119-3218]
- Subjects
Mammals ,cancer immunotherapy ,T-Lymphocytes ,[SDV]Life Sciences [q-bio] ,single-domain antibodies ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,colorectal cancer ,RC581-607 ,Epithelial Cell Adhesion Molecule ,Lymphocyte Activation ,scFv ,scfv ,ErbB Receptors ,singledomain antibodies ,[SDV] Life Sciences [q-bio] ,Oncology ,Trispecific antibodies ,tandem antibodies ,Animals ,Immunology and Allergy ,Immunologic diseases. Allergy ,Colorectal Neoplasms ,trispecific antibodies ,RC254-282 - Abstract
14 p.-6 fig., Retargeting of T lymphocytes toward cancer cells by bispecific antibodies has demonstrated its therapeutic potential, with one such antibody approved for the treatment of acute lymphoblastic leukemia (blinatumomab) and several other in clinical trials. However, improvement of their efficacy and selectivity for solid tumors is still required. Here, we describe a novel tandem T-cell recruiting trispecific antibody for the treatment of colorectal cancer (CRC). This construct, termed trispecific T-cell engager (TriTE), consists of a CD3-specific single-chain Fv (scFv) flanked by anti-epidermal growth factor receptor (EGFR) and anti-epithelial cell adhesion molecule (EpCAM) single-domain VHH antibodies. The TriTE was well expressed in mammalian and yeast cells, bound the cognate antigens of the three parental antibodies, and enabled the specific cytolysis of EGFR- and/or EpCAM-expressing cancer cells, without inducing T cell activation and cytoxicity against double-negative (EGFR−EpCAM−) cancer cells. Bivalent bispecific targeting of double-positive HCT116 cells by TriTE improved in vitro potency up to 100-fold compared to single-positive cells and significantly prolonged survival in vivo. In addition, it was less efficient at killing single-positive target cells than the corresponding bispecific controls, leading to potentially enhanced tumor specificity. Moreover, dual targeting of two tumor-associated antigens may contribute toward preventing the tumor escape by antigen loss caused by selective pressures from conventional single-targeting T-cell engagers, and may help to overcome antigenic heterogeneity., This study was funded by grants from Instituto de Salud Carlos III PI16/00357, PI19/00132), partially supported by the European Regional Development Fund (ERDF), Comunidad Autónoma de Madrid (S2010-BMD-2312), and Ministerio de Economía y Competitividad (RTC-2016-5118-1) to L.S.; and from Ministerio de Ciencia e Innovación (SAF2017-89437-P and PID2020-117323RB-I00), partially supported by ERDF, the Spanish Association Against Cancer (AECC 19084) and the CRIS Cancer Foundation FCRIS-2018-0042, FCRIS-2021-0090 (FCRIS-2018-0042 and FCRIS-2021-0090) to L.A-V. A.T-G. was supported by a predoctoral fellowship from Comunidad Autónoma de Madrid (PEJD-2018-PRE/BMD-8314);Spanish Ministry of Science and Innovation [SAF2017-89437-P, PID2020-117323RB-I00].
- Published
- 2022
69. An Fc-free EGFR-specific 4-1BB-agonistic Trimerbody Displays Broad Antitumor Activity in Humanized Murine Cancer Models without Toxicity
- Author
-
Francisco J. Blanco, Miguel Ángel Morcillo, Ainhoa Erce-Llamazares, Nekane Merino, Eva M. Garrido-Martin, Luis Álvarez-Vallina, Ignacio Melero, Laura Sanz, Antonio Tapia-Galisteo, Marta Oteo, Ángel Ramírez-Fernández, Irene Ferrer, Seandean Lykke Harwood, Eduardo Romero, Belén Blanco, Manuela Zonca, Carmen Domínguez-Alonso, Oana Hangiu, Inés G. Muñoz, Maria C. Ochoa, Marta Compte, Luis Paz-Ares, Ana Belén Enguita, Daniel Nehme-Álvarez, José Luis Rodríguez-Peralto, European Commission, Ministerio de Ciencia, Innovación y Universidades (España), Ministerio de Economía y Competitividad (España), Instituto de Salud Carlos III, Fundación CRIS contra el Cáncer, Fundación Científica Asociación Española Contra el Cáncer, Compte, Marta [0000-0002-7138-9266], Harwood, Seandean Lykke [0000-0003-4654-8832], Tapia-Galisteo, Antonio [0000-0002-0507-8435], Garrido-Martin, Eva M. [0000-0002-8094-2668], Ochoa, Maria Carmen [0000-0001-9920-2144], Oteo, Marta [0000-0002-2855-3403], Merino, Nekane [0000-0003-0721-4813], Nehme-Álvarez, Daniel [0000-0001-5054-5373], Hangiu, Oana [0000-0002-2641-8531], Domínguez-Alonso, Carmen [0000-0002-0446-9629], Zonca, Manuela [0000-0003-3868-1975], Blanco, Francisco J. [0000-0003-2545-4319], Muñoz, Inés G. [0000-0001-6732-4059], Rodríguez-Peralto, J. L. [0000-0002-6578-7153], Paz-Ares, Luis [0000-0002-1947-3364], Sanz, Laura [0000-0002-3119-3218], Alvarez-Vallina, Luis [0000-0003-3053-6757], Compte, Marta, Harwood, Seandean Lykke, Tapia-Galisteo, Antonio, Garrido-Martin, Eva M., Ochoa, Maria Carmen, Oteo, Marta, Merino, Nekane, Nehme-Álvarez, Daniel, Hangiu, Oana, Domínguez-Alonso, Carmen, Zonca, Manuela, Blanco, Francisco J., Muñoz, Inés G., Rodríguez-Peralto, J. L., Paz-Ares, Luis, Sanz, Laura, and Alvarez-Vallina, Luis
- Subjects
0301 basic medicine ,PD-L1 ,Cancer Research ,Lung Neoplasms ,Bispecific antibody ,T-Lymphocytes ,Cell ,T cells ,Breast Neoplasms ,Mice, Transgenic ,Lymphocyte Activation ,4-1BB ,Cell Line ,03 medical and health sciences ,Tumor Necrosis Factor Receptor Superfamily, Member 9 ,0302 clinical medicine ,In vivo ,Checkpoint blockade ,Carcinoma, Non-Small-Cell Lung ,medicine ,Animals ,biology ,business.industry ,Trimerbody ,Cancer ,medicine.disease ,Fragment crystallizable region ,ErbB Receptors ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Systemic administration ,Female ,Immunotherapy ,Antibody ,business ,CD8 - Abstract
32 p.-4 fig., Purpose: The induction of 4-1BB signaling by agonistic antibodies can drive the activation and proliferation of effector T cells and thereby enhance a T-cell–mediated antitumor response. Systemic administration of anti-4-1BB–agonistic IgGs, although effective preclinically, has not advanced in clinical development due to their severe hepatotoxicity., Experimental Design: Here, we generated a humanized EGFR-specific 4-1BB-agonistic trimerbody, which replaces the IgG Fc region with a human collagen homotrimerization domain. It was characterized by structural analysis and in vitro functional studies. We also assessed pharmacokinetics, antitumor efficacy, and toxicity in vivo., Results: In the presence of a T-cell receptor signal, the trimerbody provided potent T-cell costimulation that was strictly dependent on 4-1BB hyperclustering at the point of contact with a tumor antigen-displaying cell surface. It exhibits significant antitumor activity in vivo, without hepatotoxicity, in a wide range of human tumors including colorectal and breast cancer cell-derived xenografts, and non–small cell lung cancer patient-derived xenografts associated with increased tumor-infiltrating CD8+ T cells. The combination of the trimerbody with a PD-L1 blocker led to increased IFNγ secretion in vitro and resulted in tumor regression in humanized mice bearing aggressive triple-negative breast cancer., Conclusions: These results demonstrate the nontoxic broad antitumor activity of humanized Fc-free tumor-specific 4-1BB-agonistic trimerbodies and their synergy with checkpoint blockers, which may provide a way to elicit responses in most patients with cancer while avoiding Fc-mediated adverse reactions., This work was supported by grants from the European Union [IACT Project (602262), H2020-iNEXT (1676)]; the Spanish Ministry of Science, Innovation and Universities and the Spanish Ministry of Economy and Competitiveness (SAF2017-89437-P, CTQ2017-83810-R, RTC-2016-5118-1, RTC-2017-5944-1), partially supported by the European Regional Development Fund; the Carlos III Health Institute (PI16/00357), co-founded by the Plan Nacional de Investigación and the European Union; the CRIS Cancer Foundation (FCRIS-IFI-2018); and the Spanish Association Against Cancer (AECC, 19084). C. Domínguez-Alonso was supported by a predoctoral fellowship from the Spanish Ministry of Science, Innovation and Universities (PRE2018-083445). M. Zonca was supported by the Torres Quevedo Program from the Spanish Ministry of Economy and Competitiveness, co-founded by the European Social Fund (PTQ-16-08340).
- Published
- 2020
70. A PD-L1/EGFR bispecific antibody combines immune checkpoint blockade and direct anti-cancer action for an enhanced anti-tumor response
- Author
-
Laura Rubio-Pérez, Rodrigo Lázaro-Gorines, Seandean L. Harwood, Marta Compte, Rocío Navarro, Antonio Tapia-Galisteo, Jaume Bonet, Belén Blanco, Simon Lykkemark, Ángel Ramírez-Fernández, Mariola Ferreras-Gutiérrez, Carmen Domínguez-Alonso, Laura Díez-Alonso, Alejandro Segura-Tudela, Oana Hangiu, Ainhoa Erce-Llamazares, Francisco J. Blanco, Cruz Santos, José L. Rodríguez-Peralto, Laura Sanz, Luis Álvarez-Vallina, Ministerio de Ciencia e Innovación (España), Instituto de Salud Carlos III, Fundación CRIS contra el Cáncer, Asociación Española Contra el Cáncer, Fundación 'La Caixa', Fundación de Investigación Biomédica Hospital 12 de Octubre, Universidad Francisco de Vitoria, Comunidad de Madrid, Rubio-Pérez, Laura, Lázaro-Gorines, Rodrigo, Harwood, Seandean Lykke, Compte, Marta, Navarro, Rocío, Tapia-Galisteo, Antonio, Bonet, Jaume, Blanco, Belén, Lykkemark, Simon, Ramírez-Fernández, Ángel, Ferreras-Gutiérrez, Mariola, Domínguez-Alonso, Carmen, Díez-Alonso, Laura, Segura-Tudela, Alejandro, Hangiu, Oana, Erce-Llamazares, Ainhoa, Blanco, Francisco J., Santos, Cruz, Rodríguez-Peralto, J. L., Sanz, Laura, and Alvarez-Vallina, Luis
- Subjects
combination ,cancer immunotherapy ,family ,receptor ,Immunology ,persistent activity ,bispecific antibody ,igg1 ,Oncology ,cetuximab ,cells ,Immunology and Allergy ,activation ,escape ,Cancer immunotherapy, bispecific antibody ,Dual action ,Immune checkpoint blockade ,Epithelial growth factor receptor - Abstract
Immune checkpoint blockade (ICB) with antibodies has shown durable clinical responses in a wide range of cancer types, but the overall response rate is still limited. Other effective therapeutic modalities to increase the ICB response rates are urgently needed. New bispecific antibody (bsAb) formats combining the ICB effect and a direct action on cancer cells could improve the efficacy of current immunotherapies. Here, we report the development of a PD-L1/EGFR symmetric bsAb by fusing a dual-targeting tandem trimmer body with the human IgG1 hinge and Fc regions. The bsAb was characterized in vitro and the antitumor efficacy was evaluated in humanized mice bearing xenografts of aggressive triple-negative breast cancer and lung cancer. The IgG-like hexavalent bsAb, designated IgTT-1E, was able to simultaneously bind both EGFR and PD-L1 antigens, inhibit EGF-mediated proliferation, effectively block PD-1/PD-L1 interaction, and induce strong antigen-specific antibody-dependent cellular cytotoxicity activity in vitro. Potent therapeutic efficacies of IgTT-1E in two different humanized mouse models were observed, where tumor growth control was associated with a significantly increased proportion of CD8+ T cells. These results support the development of IgTT-1E for the treatment of EGFR+ cancers., L.A-V. was supported by grants from the MCIN/AEI/10.13039/ 501100011033 (PID2020-117323RB-100 and PDC2021-121711-100), the Instituto de Salud Carlos III (DTS20/00089), the CRIS Cancer Foundation (FCRIS-2021-0090), the Spanish Association Against Cancer (PROYE19084ALVA), the Fundación ‘‘La Caixa’’ (HR21-00761 project IL7R_LungCan) and the Fundación de Investigación Biomédica 12 de Octubre Programa Investiga (2022-0082). B.B and L.S. were supported by grants PI20/01030 and PI19/00132 from the Instituto de Salud Carlos III (PI20/01030). FJB and MF-G were supported by grants PID2020- 113225GB-I00 and PRE2018-085788 funded by MCIN/AEI/10.13039/ 501100011033. L.R-P. was supported by a predoctoral fellowship from the Immunology Chair, Universidad Francisco de Vitoria/Merck. C. D-A. was supported by a predoctoral fellowship from the MCIN/AEI/ 10.13039/501100011033 (PRE2018-083445). L.D-A. was supported by a Rio Hortega fellowship from the Instituto de Salud Carlos III (CM20/ 00004). O.H. was supported by an industrial PhD fellowship from the Comunidad de Madrid (IND2020/BMD-17668). AE-L was supported industrial PhD fellowship from the Instituto de Salud Carlos III (IFI18/ 00045)
- Published
- 2023
71. Applications of trimerbodies in cancer immunotherapy.
- Author
-
Compte M, Sanz L, and Álvarez-Vallina L
- Subjects
- Humans, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents, Immunological therapeutic use, Immunotherapy, Neoplasms
- Abstract
Trimerbodies, with their unique structural and functional properties, are the basis of a new generation of therapeutic antibodies, which due to their small size and plasticity are ideal for the generation of novel biological protein drugs with multiple competitive advantages over conventional full-length monoclonal antibodies. Since their emergence, trimerbodies have been used in preclinical cancer diagnosis and therapy. Trimerbodies are highly adaptable molecules, as they allow target-specific modulation of T cell-mediated anti-tumor immunity to enhance preexisting responses or to generate de novo immune responses. In fact, a tumor-specific humanized 4-1BB-agonistic trimerbody has shown a rather impressive safety and efficacy profile in preclinical studies making it a realistic option for clinical development. Moreover, thanks to the avidity effect they are endowed with considerable therapeutic potential as carriers to deliver cytotoxic payloads to tumors. In addition, molecular imaging studies could benefit from some intermediate-sized trivalent trimerbodies as promising candidates for targeted therapy and tumor imaging., Competing Interests: Competing interests MC is current employee of Leadartis S.L. LS and LA-V are co-founders of Leadartis S.L., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
72. ATTACK, a novel bispecific T cell-recruiting antibody with trivalent EGFR binding and monovalent CD3 binding for cancer immunotherapy.
- Author
-
Harwood SL, Alvarez-Cienfuegos A, Nuñez-Prado N, Compte M, Hernández-Pérez S, Merino N, Bonet J, Navarro R, Van Bergen En Henegouwen PMP, Lykkemark S, Mikkelsen K, Mølgaard K, Jabs F, Sanz L, Blanco FJ, Roda-Navarro P, and Alvarez-Vallina L
- Abstract
The redirection of T cell activity using bispecific antibodies is one of the most promising cancer immunotherapy approaches currently in development, but it is limited by cytokine storm-related toxicities, as well as the pharmacokinetics and tumor-penetrating capabilities of current bispecific antibody formats. Here, we have engineered the ATTACK ( Asymmetric Tandem Trimerbody for T cell Activation and Cancer Killing ), a novel T cell-recruiting bispecific antibody which combines three EGFR-binding single-domain antibodies (V
HH ; clone EgA1) with a single CD3-binding single-chain variable fragment (scFv; clone OKT3) in an intermediate molecular weight package. The two specificities are oriented in opposite directions in order to simultaneously engage cancer cells and T cell effectors, and thereby promote immunological synapse formation. EgA1 ATTACK was expressed as a homogenous, non-aggregating, soluble protein by mammalian cells and demonstrated an enhanced binding to EGFR, but not CD3, when compared to the previously characterized tandem bispecific antibody which has one EgA1 VHH and one OKT3 scFv per molecule. EgA1 ATTACK induced synapse formation and early signaling pathways downstream of TCR engagement at lower concentrations than the tandem VHH -scFv bispecific antibody. Furthermore, it demonstrated extremely potent, dose-dependent cytotoxicity when retargeting human T cells towards EGFR-expressing cells, with an efficacy over 15-fold higher than that of the tandem VHH -scFv bispecific antibody. These results suggest that the ATTACK is an ideal format for the development of the next-generation of T cell-redirecting bispecific antibodies.- Published
- 2017
- Full Text
- View/download PDF
73. CARbodies: Human Antibodies Against Cell Surface Tumor Antigens Selected From Repertoires Displayed on T Cell Chimeric Antigen Receptors.
- Author
-
Alonso-Camino V, Sánchez-Martín D, Compte M, Nuñez-Prado N, Diaz RM, Vile R, and Alvarez-Vallina L
- Abstract
A human single-chain variable fragment (scFv) antibody library was expressed on the surface of human T cells after transduction with lentiviral vectors (LVs). The repertoire was fused to a first-generation T cell receptor ζ (TCRζ)-based chimeric antigen receptor (CAR). We used this library to isolate antibodies termed CARbodies that recognize antigens expressed on the tumor cell surface in a proof-of-principle system. After three rounds of activation-selection there was a clear repertoire restriction, with the emergence dominant clones. The CARbodies were purified from bacterial cultures as soluble and active proteins. Furthermore, to validate its potential application for adoptive cell therapy, human T cells were transduced with a LV encoding a second-generation costimulatory CAR (CAR(v2)) bearing the selected CARbodies. Transduced human primary T cells expressed significant levels of the CARbodies-based CAR(v2) fusion protein on the cell surface, and importantly could be specifically activated, after stimulation with tumor cells. This approach is a promising tool for the generation of antibodies fully adapted to the display format (CAR) and the selection context (cell synapse), which could extend the scope of current adoptive cell therapy strategies with CAR-redirected T cells.Molecular Therapy-Nucleic Acids (2013) 2, e93; doi:10.1038/mtna.2013.19; published online 21 May 2013.
- Published
- 2013
- Full Text
- View/download PDF
74. The efficacy versus toxicity profile of combination virotherapy and TLR immunotherapy highlights the danger of administering TLR agonists to oncolytic virus-treated mice.
- Author
-
Rommelfanger DM, Compte M, Grau MC, Diaz RM, Ilett E, Alvarez-Vallina L, Thompson JM, Kottke TJ, Melcher A, and Vile RG
- Subjects
- Animals, Antigens, Neoplasm immunology, Cell Line, Tumor, Combined Modality Therapy adverse effects, Combined Modality Therapy methods, Genetic Therapy, Genetic Vectors, Immunity, Innate, Immunotherapy, Interferon-alpha metabolism, Interleukin-6 blood, Interleukin-6 immunology, Lipopolysaccharides metabolism, Mice, Mice, Inbred C57BL, Oncolytic Viruses genetics, Signal Transduction, T-Lymphocytes immunology, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 metabolism, Tumor Necrosis Factor-alpha blood, Tumor Necrosis Factor-alpha immunology, Vesiculovirus immunology, Oncolytic Virotherapy adverse effects, Oncolytic Virotherapy methods, Toll-Like Receptor 4 agonists, Vesiculovirus genetics
- Abstract
Injection of oncolytic vesicular stomatitis virus (VSV) into established B16ova melanomas results in tumor regression, in large part by inducing innate immune reactivity against the viral infection, mediated by MyD88- and type III interferon (IFN)-, but not TLR-4-, signaling. We show here that intratumoral (IT) treatment with lipopolysaccharide (LPS), a TLR-4 agonist, significantly enhanced the local therapy induced by VSV by combining activation of different innate immune pathways. Therapy was further enhanced by co-recruiting a potent antitumor, adaptive T-cell response by using a VSV engineered to express the ovalbumin tumor-associated antigen ova, in combination with LPS. However, the combination of IT LPS with systemically delivered VSV resulted in rapid morbidity and mortality in the majority of mice. Decreasing the intravenous (IV) dose of VSV to levels at which toxicity was ameliorated did not enhance therapy compared with IT LPS alone. Toxicity of the systemic VSV + IT LPS regimen was associated with rapidly elevated levels of serum tumor necrosis factor-α (TNF-α) and interleukin (IL)-6, which neither systemic VSV, nor IT LPS, alone induced. These data show that therapy associated with direct IT injections of oncolytic viruses can be significantly enhanced by combination with agonists of innate immune activation pathways, which are not themselves activated by the virus alone. Importantly, they also highlight possible, unforeseen dangers of combination therapies in which an immunotherapy, even delivered locally at the tumor site, may systemically sensitize the patient to a cytokine shock-like response triggered by IV delivery of oncolytic virus.
- Published
- 2013
- Full Text
- View/download PDF
75. Immunotherapeutic organoids: a new approach to cancer treatment.
- Author
-
Compte M, Nuñez-Prado N, Sanz L, and Alvarez-Vallina L
- Subjects
- Antibodies, Monoclonal genetics, Genetic Therapy, Humans, Immunotherapy, Injections, Subcutaneous, Mesenchymal Stem Cell Transplantation methods, Neoplasms pathology, Tissue Scaffolds, Antibodies, Monoclonal therapeutic use, Mesenchymal Stem Cells metabolism, Neoplasms therapy, Organoids injuries
- Abstract
Therapeutic monoclonal antibodies have revolutionized the treatment of cancer and other diseases. However, several limitations of antibody-based treatments, such as the cost of therapy and the achievement of sustained plasma levels, should be still addressed for their widespread use as therapeutics. The use of cell and gene transfer methods offers additional benefits by producing a continuous release of the antibody with syngenic glycosylation patterns, which makes the antibody potentially less immunogenic. In vivo secretion of therapeutic antibodies by viral vector delivery or ex vivo gene modified long-lived autologous or allogeneic human mesenchymal stem cells may advantageously replace repeated injection of clinical-grade antibodies. Gene-modified autologous mesenchymal stem cells can be delivered subcutaneously embedded in a non-immunogenic synthetic extracellular matrix-based scaffold that guarantees the survival of the cell inoculum. The scaffold would keep cells at the implantation site, with the therapeutic protein acting at distance (immunotherapeutic organoid), and could be retrieved once the therapeutic effect is fulfilled. In the present review we highlight the practical importance of living cell factories for in vivo secretion of recombinant antibodies.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.