30 results on '"Harwood, Seandean Lykke"'
Search Results
2. Recent cryogenic electron microscopy structures of human A2M may not be representative of the native protein
- Author
-
Harwood, Seandean Lykke, Andersen, Gregers Rom, and Enghild, Jan J.
- Published
- 2022
3. The conformational change of the protease inhibitor α2-macroglobulin is triggered by the retraction of the cleaved bait region from a central channel
- Author
-
Harwood, Seandean Lykke, Diep, Khang, Nielsen, Nadia Sukusu, Jensen, Kathrine Tejlgård, and Enghild, Jan J.
- Published
- 2022
- Full Text
- View/download PDF
4. Cryo-EM structures of human A2ML1 elucidate the protease-inhibitory mechanism of the A2M family
- Author
-
Nielsen, Nadia Sukusu, Zarantonello, Alessandra, Harwood, Seandean Lykke, Jensen, Kathrine Tejlgård, Kjøge, Katarzyna, Thøgersen, Ida B., Schauser, Leif, Karlsen, Jesper Lykkegaard, Andersen, Gregers R., and Enghild, Jan J.
- Published
- 2022
- Full Text
- View/download PDF
5. Development of selective protease inhibitors via engineering of the bait region of human α2-macroglobulin
- Author
-
Harwood, Seandean Lykke, Nielsen, Nadia Sukusu, Diep, Khang, Jensen, Kathrine Tejlgård, Nielsen, Peter Kresten, Yamamoto, Kazuhiro, and Enghild, Jan J.
- Published
- 2021
- Full Text
- View/download PDF
6. Mutation-induced dimerization of transforming growth factor-β–induced protein may drive protein aggregation in granular corneal dystrophy
- Author
-
Nielsen, Nadia Sukusu, Gadeberg, Trine A.F., Poulsen, Ebbe Toftgaard, Harwood, Seandean Lykke, Weberskov, Christian E., Pedersen, Jan Skov, Andersen, Gregers R., and Enghild, Jan J.
- Published
- 2021
- Full Text
- View/download PDF
7. Structural Investigations of Human A2M Identify a Hollow Native Conformation That Underlies Its Distinctive Protease-Trapping Mechanism
- Author
-
Harwood, Seandean Lykke, Lyngsø, Jeppe, Zarantonello, Alessandra, Kjøge, Katarzyna, Nielsen, Peter Kresten, Andersen, Gregers Rom, Pedersen, Jan Skov, and Enghild, Jan J.
- Published
- 2021
- Full Text
- View/download PDF
8. α2-Macroglobulin-like protein 1 can conjugate and inhibit proteases through their hydroxyl groups, because of an enhanced reactivity of its thiol ester
- Author
-
Harwood, Seandean Lykke, Nielsen, Nadia Sukusu, Jensen, Kathrine Tejlgård, Nielsen, Peter Kresten, Thøgersen, Ida B., and Enghild, Jan J.
- Published
- 2020
- Full Text
- View/download PDF
9. Dendritic Cell-Mediated Cross-Priming by a Bispecific Neutralizing Antibody Boosts Cytotoxic T Cell Responses and Protects Mice against SARS-CoV-2
- Author
-
European Commission, Fundación BBVA, Ministerio de Ciencia e Innovación (España), Instituto de Salud Carlos III, Asociación Española Contra el Cáncer, Fundación CRIS contra el Cáncer, La Caixa, Comunidad de Madrid, Atresmedia, Banco Santander, Consejo Superior de Investigaciones Científicas (España), Universidad Francisco de Vitoria, Fundación Merck Salud, Lázaro-Gorines, Rodrigo [0000-0002-6885-3486], Pérez, Patricia [0000-0001-8983-6784], Heras-Murillo, Ignacio [0000-0002-8797-8786], Adán-Barrientos, Irene [0000-0003-4442-9645], Albericio, Guillermo [0000-0003-0190-4848], Astorgano, David [0000-0002-2969-1840], Luczkowiak, Joanna [0000-0001-6950-9372], Labiod, Nuria [0000-0001-7297-1162], Harwood, Seandean Lykke [0000-0003-4654-8832], Segura-Tudela, Alejandro [0000-0002-5506-0153], Rubio-Pérez, Laura [0000-0002-2877-6092], Nugraha, Yudhi [0000-0003-1186-4093], Shang, Xiaoran [0000-0002-3986-4557], Li, Yuxing [0000-0001-9785-2960], Alfonso, Carlos [0000-0001-7165-4800], Abeyawardhane, Dinendra L. [0000-0003-3002-7792], Navarro, Rocío [0000-0002-0083-7711], Compte, Marta [0000-0002-7138-9266], Sanz, Laura [0000-0002-3119-3218], Weber, David J. [0000-0002-8824-1110], Blanco, Francisco J. [0000-0003-2545-4319], Esteban, Mariano [0000-0003-0846-2827], Pozharski, Edwin [0000-0001-7012-5376], Godoy-Ruiz, Raquel [0000-0003-4569-0781], Muñoz, Inés G. [0000-0001-6732-4059], Delgado, Rafael [0000-0002-6912-4736], Sancho, David [0000-0003-2890-3984], García-Arriaza, Juan [0000-0002-5167-5724], Álvarez-Vallina, Luis [0000-0003-3053-6757], Lázaro-Gorines, Rodrigo, Pérez Ramírez, Patricia, Heras-Murillo, Ignacio, Adán-Barrientos, Irene, Albericio, Guillermo, Astorgano, David, Flores, Sara, Luczkowiak, Joanna, Labiod, Nuria, Harwood, Seandean Lykke, Segura-Tudela, Alejandro, Rubio-Pérez, Laura, Nugraha, Yudhi, Shang, Xiaoran, Li, Yuxing, Alfonso, Carlos, Adipietro, Kaylin A., Abeyawardhane, Dinendra L., Navarro, Rocío, Compte, Marta, Yu, Wenbo, MacKerell Jr., Alexander D., Sanz, Laura, Weber, David J., Blanco, Francisco J., Esteban, Mariano, Pozharski, Edwin, Godoy-Ruiz, Raquel, Muñoz, Inés G., Delgado, Rafael, Sancho, David, García-Arriaza, Juan, Álvarez-Vallina, Luis, European Commission, Fundación BBVA, Ministerio de Ciencia e Innovación (España), Instituto de Salud Carlos III, Asociación Española Contra el Cáncer, Fundación CRIS contra el Cáncer, La Caixa, Comunidad de Madrid, Atresmedia, Banco Santander, Consejo Superior de Investigaciones Científicas (España), Universidad Francisco de Vitoria, Fundación Merck Salud, Lázaro-Gorines, Rodrigo [0000-0002-6885-3486], Pérez, Patricia [0000-0001-8983-6784], Heras-Murillo, Ignacio [0000-0002-8797-8786], Adán-Barrientos, Irene [0000-0003-4442-9645], Albericio, Guillermo [0000-0003-0190-4848], Astorgano, David [0000-0002-2969-1840], Luczkowiak, Joanna [0000-0001-6950-9372], Labiod, Nuria [0000-0001-7297-1162], Harwood, Seandean Lykke [0000-0003-4654-8832], Segura-Tudela, Alejandro [0000-0002-5506-0153], Rubio-Pérez, Laura [0000-0002-2877-6092], Nugraha, Yudhi [0000-0003-1186-4093], Shang, Xiaoran [0000-0002-3986-4557], Li, Yuxing [0000-0001-9785-2960], Alfonso, Carlos [0000-0001-7165-4800], Abeyawardhane, Dinendra L. [0000-0003-3002-7792], Navarro, Rocío [0000-0002-0083-7711], Compte, Marta [0000-0002-7138-9266], Sanz, Laura [0000-0002-3119-3218], Weber, David J. [0000-0002-8824-1110], Blanco, Francisco J. [0000-0003-2545-4319], Esteban, Mariano [0000-0003-0846-2827], Pozharski, Edwin [0000-0001-7012-5376], Godoy-Ruiz, Raquel [0000-0003-4569-0781], Muñoz, Inés G. [0000-0001-6732-4059], Delgado, Rafael [0000-0002-6912-4736], Sancho, David [0000-0003-2890-3984], García-Arriaza, Juan [0000-0002-5167-5724], Álvarez-Vallina, Luis [0000-0003-3053-6757], Lázaro-Gorines, Rodrigo, Pérez Ramírez, Patricia, Heras-Murillo, Ignacio, Adán-Barrientos, Irene, Albericio, Guillermo, Astorgano, David, Flores, Sara, Luczkowiak, Joanna, Labiod, Nuria, Harwood, Seandean Lykke, Segura-Tudela, Alejandro, Rubio-Pérez, Laura, Nugraha, Yudhi, Shang, Xiaoran, Li, Yuxing, Alfonso, Carlos, Adipietro, Kaylin A., Abeyawardhane, Dinendra L., Navarro, Rocío, Compte, Marta, Yu, Wenbo, MacKerell Jr., Alexander D., Sanz, Laura, Weber, David J., Blanco, Francisco J., Esteban, Mariano, Pozharski, Edwin, Godoy-Ruiz, Raquel, Muñoz, Inés G., Delgado, Rafael, Sancho, David, García-Arriaza, Juan, and Álvarez-Vallina, Luis
- Abstract
Administration of neutralizing antibodies (nAbs) has proved to be effective by providing immediate protection against SARS-CoV-2. However, dual strategies combining virus neutralization and immune response stimulation to enhance specific cytotoxic T cell responses, such as dendritic cell (DC) cross-priming, represent a promising field but have not yet been explored. Here, a broadly nAb, TNT, are first generated by grafting an anti-RBD biparatopic tandem nanobody onto a trimerbody scaffold. Cryo-EM data show that the TNT structure allows simultaneous binding to all six RBD epitopes, demonstrating a high-avidity neutralizing interaction. Then, by C-terminal fusion of an anti-DNGR-1 scFv to TNT, the bispecific trimerbody TNTDNGR-1 is generated to target neutralized virions to type 1 conventional DCs (cDC1s) and promote T cell cross-priming. Therapeutic administration of TNTDNGR-1, but not TNT, protects K18-hACE2 mice from a lethal SARS-CoV-2 infection, boosting virus-specific humoral responses and CD8+ T cell responses. These results further strengthen the central role of interactions with immune cells in the virus-neutralizing antibody activity and demonstrate the therapeutic potential of the Fc-free strategy that can be used advantageously to provide both immediate and long-term protection against SARS-CoV-2 and other viral infections.
- Published
- 2023
10. A PD-L1/EGFR bispecific antibody combines immune checkpoint blockade and direct anti-cancer action for an enhanced anti-tumor response
- Author
-
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 para la Investigación Biomédica del Hospital Universitario 12 de Octubre, Universidad Francisco de Vitoria, Comunidad de Madrid, Rubio-Pérez, Laura [0000-0002-2877-6092], Lázaro-Gorines, Rodrigo [0000-0002-6885-3486], Harwood, Seandean Lykke [0000-0003-4654-8832], Compte, Marta [0000-0002-7138-9266], Navarro, Rocío [0000-0002-0083-7711], Tapia-Galisteo, Antonio [0000-0002-0507-8435], Bonet, Jaume [0000-0001-5210-4387], Blanco, Belén [0000-0001-5085-7756], Lykkemark, Simon [0000-0001-8920-6926], Ramírez-Fernández, Ángel [0000-0002-3265-6878], Ferreras-Gutiérrez, Mariola [0000-0003-4421-3158], Domínguez-Alonso, Carmen [0000-0002-0446-9629], Díez-Alonso, Laura [0000-0002-9545-6910], Segura-Tudela, Alejandro [0000-0002-5506-0153], Hangiu, Oana [0000-0002-2641-8531], Erce-Llamazares, Ainhoa [0000-0002-3656-1913], Blanco, Francisco J. [0000-0003-2545-4319], Santos, Cruz [0000-0001-5164-5050], Rodríguez-Peralto, J. L. [0000-0002-6578-7153], Sanz, Laura [0000-0002-3119-3218], Alvarez-Vallina, Luis [0000-0003-3053-6757], 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, Álvarez-Vallina, Luis, 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 para la Investigación Biomédica del Hospital Universitario 12 de Octubre, Universidad Francisco de Vitoria, Comunidad de Madrid, Rubio-Pérez, Laura [0000-0002-2877-6092], Lázaro-Gorines, Rodrigo [0000-0002-6885-3486], Harwood, Seandean Lykke [0000-0003-4654-8832], Compte, Marta [0000-0002-7138-9266], Navarro, Rocío [0000-0002-0083-7711], Tapia-Galisteo, Antonio [0000-0002-0507-8435], Bonet, Jaume [0000-0001-5210-4387], Blanco, Belén [0000-0001-5085-7756], Lykkemark, Simon [0000-0001-8920-6926], Ramírez-Fernández, Ángel [0000-0002-3265-6878], Ferreras-Gutiérrez, Mariola [0000-0003-4421-3158], Domínguez-Alonso, Carmen [0000-0002-0446-9629], Díez-Alonso, Laura [0000-0002-9545-6910], Segura-Tudela, Alejandro [0000-0002-5506-0153], Hangiu, Oana [0000-0002-2641-8531], Erce-Llamazares, Ainhoa [0000-0002-3656-1913], Blanco, Francisco J. [0000-0003-2545-4319], Santos, Cruz [0000-0001-5164-5050], Rodríguez-Peralto, J. L. [0000-0002-6578-7153], Sanz, Laura [0000-0002-3119-3218], Alvarez-Vallina, Luis [0000-0003-3053-6757], 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 Álvarez-Vallina, Luis
- 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.
- Published
- 2023
11. Proteolytic cleavage of the TGFβ co‐receptor CD109 changes its conformation, resulting in protease inhibition via activation of its thiol ester, and dissociation from the cell membrane.
- Author
-
Jensen, Kathrine Tejlgård, Nielsen, Nadia Sukusu, Viana Almeida, Ana, Thøgersen, Ida B., Enghild, Jan J., and Harwood, Seandean Lykke
- Subjects
MEMBRANE glycoproteins ,TRANSFORMING growth factors ,ESTERS ,PEPTIDE bonds ,PROTEOLYTIC enzymes - Abstract
The glycosylphosphatidylinositol (GPI)‐anchored protein cluster of differentiation 109 (CD109) is expressed on many human cell types and modulates the transforming growth factor β (TGF‐β) signaling network. CD109 belongs to the alpha‐macroglobulin family of proteins, known for their protease‐triggered conformational changes. However, the effect of proteolysis on CD109 and its conformation are unknown. Here, we investigated the interactions of CD109 with proteases. We found that a diverse selection of proteases cleaved peptide bonds within the predicted bait region of CD109, inducing a conformational change that activated the thiol ester of CD109. We show CD109 was able to conjugate proteases with this thiol ester and decrease their activity toward protein substrates, demonstrating that CD109 is a protease inhibitor. We additionally found that CD109 has a unique mechanism whereby its GPI‐anchored macroglobulin 8 (MG8) domain dissociates during its conformational change, allowing proteases to release CD109 from the cell surface by a precise mechanism and not unspecific shedding. We conclude that proteolysis of the CD109 bait region affects both its structure and location, and that interactions between CD109 and proteases may be important to understanding its functions, for example, as a TGF‐β co‐receptor. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Trispecific T-cell engagers for dual tumor-targeting of colorectal cancer
- Author
-
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 [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], Sanz, Laura [0000-0002-3119-3218], Tapia-Galisteo, Antonio, Sánchez Rodríguez, Íñigo, Aguilar-Sopeña, Óscar, Harwood, Seandean Lykke, Narbona, Javier, Ferreras-Gutiérrez, Mariola, Navarro, Rocío, Martín-García, Laura, Corbacho, Cesáreo, Compte, Marta, Lacadena, Javier, Blanco, Francisco J., Chames, Patrick, Roda-Navarro, Pedro, Álvarez-Vallina, Luis, Sanz, Laura, 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 [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], Sanz, Laura [0000-0002-3119-3218], Tapia-Galisteo, Antonio, Sánchez Rodríguez, Íñigo, Aguilar-Sopeña, Óscar, Harwood, Seandean Lykke, Narbona, Javier, Ferreras-Gutiérrez, Mariola, Navarro, Rocío, Martín-García, Laura, Corbacho, Cesáreo, Compte, Marta, Lacadena, Javier, Blanco, Francisco J., Chames, Patrick, Roda-Navarro, Pedro, Álvarez-Vallina, Luis, and Sanz, Laura
- Abstract
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.
- Published
- 2022
13. Characterization of a novel cold-adapted intracellular serine protease from the extremophile Planococcus halocryophilus Or1
- Author
-
Rasmussen, Casper Bøjer, primary, Scavenius, Carsten, additional, Thøgersen, Ida B., additional, Harwood, Seandean Lykke, additional, Larsen, Øivind, additional, Bjerga, Gro Elin Kjaereng, additional, Stougaard, Peter, additional, Enghild, Jan J., additional, and Thøgersen, Mariane Schmidt, additional
- Published
- 2023
- Full Text
- View/download PDF
14. Characterization of a novel cold-adapted intracellular serine protease from the extremophile Planococcus halocryophilus Or1
- Author
-
Rasmussen, Casper Bøjer, Scavenius, Carsten, Thøgersen, Ida B, Harwood, Seandean Lykke, Larsen, Øivind, Bjerga, Gro Elin Kjaereng, Stougaard, Peter, Enghild, Jan J, and Thøgersen, Mariane Schmidt
- Subjects
Planococcus ,Microbiology (medical) ,calcium ,protein chemistry ,maturation ,cold adaptation ,characterization ,Microbiology ,intracellular subtilisin protease - Abstract
The enzymes of microorganisms that live in cold environments must be able to function at ambient temperatures. Cold-adapted enzymes generally have less ordered structures that convey a higher catalytic rate, but at the cost of lower thermodynamic stability. In this study, we characterized P355, a novel intracellular subtilisin protease (ISP) derived from the genome of Planococcus halocryophilus Or1, which is a bacterium metabolically active down to −25°C. P355′s stability and activity at varying pH values, temperatures, and salt concentrations, as well as its temperature-dependent kinetics, were determined and compared to an uncharacterized thermophilic ISP (T0099) from Parageobacillus thermoglucosidasius, a previously characterized ISP (T0034) from Planococcus sp. AW02J18, and Subtilisin Carlsberg (SC). The results showed that P355 was the most heat-labile of these enzymes, closely followed by T0034. P355 and T0034 exhibited catalytic constants (kcat) that were much higher than those of T0099 and SC. Thus, both P355 and T0034 demonstrate the characteristics of the stability-activity trade-off that has been widely observed in cold-adapted proteases.
- Published
- 2023
- Full Text
- View/download PDF
15. A tumor-targeted trimeric 4-1BB-agonistic antibody induces potent anti-tumor immunity without systemic toxicity
- Author
-
Compte, Marta, Harwood, Seandean Lykke, Muñoz, Ines G., Navarro, Rocio, Zonca, Manuela, Perez-Chacon, Gema, Erce-Llamazares, Ainhoa, Merino, Nekane, Tapia-Galisteo, Antonio, Cuesta, Angel M., Mikkelsen, Kasper, Caleiras, Eduardo, Nuñez-Prado, Natalia, Aznar, M. Angela, Lykkemark, Simon, Martínez-Torrecuadrada, Jorge, Melero, Ignacio, Blanco, Francisco J., Bernardino de la Serna, Jorge, Zapata, Juan M., Sanz, Laura, and Alvarez-Vallina, Luis
- Published
- 2018
- Full Text
- View/download PDF
16. Trispecific T-cell engagers for dual tumor-targeting of colorectal cancer
- Author
-
Tapia-Galisteo, Antonio, primary, Sánchez Rodríguez, Íñigo, additional, Aguilar-Sopeña, Oscar, additional, Harwood, Seandean Lykke, additional, Narbona, Javier, additional, Ferreras Gutierrez, Mariola, additional, Navarro, Rocío, additional, Martín-García, Laura, additional, Corbacho, Cesáreo, additional, Compte, Marta, additional, Lacadena, Javier, additional, Blanco, Francisco J., additional, Chames, Patrick, additional, Roda-Navarro, Pedro, additional, Álvarez-Vallina, Luis, additional, and Sanz, Laura, additional
- Published
- 2022
- Full Text
- View/download PDF
17. Running title: Non-toxic broad anti-tumor activity of an EGFR×4-1BB bispecific trimerbod
- Author
-
Compte, Marta, Harwood, Seandean Lykke, Erce-Llamazares, Ainhoa, Tapia-Galisteo, Antonio, Romero, Eduardo, Ferrer, Irene, Garrido-Martin, Eva M., Enguita, Ana Belén, Ochoa, Maria Carmen, Blanco, Belén, Oteo, Marta, Merino, Nekane, Nehme-Álvarez, Daniel, Hangiu, Oana, Domínguez-Alonso, Carmen, Zonca, Manuela, Ramírez-Fernández, Ángel, Blanco, Francisco J., Morcillo, Miguel Ángel, Muñoz, Inés G., Melero, Ignacio, Rodríguez-Peralto, J. L., Paz-Ares, Luis, Sanz, Laura, Álvarez-Vallina, Luis, 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], and Alvarez-Vallina, Luis [0000-0003-3053-6757]
- Subjects
PD-L1 ,Trimerbody ,Checkpoint blockade ,Bispecific antibody ,T cells ,4-1BB - 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
- 2021
- Full Text
- View/download PDF
18. Correction: α2-Macroglobulin-like protein 1 can conjugate and inhibit proteases through their hydroxyl groups, because of an enhanced reactivity of its thiol ester
- Author
-
Harwood, Seandean Lykke, Nielsen, Nadia Sukusu, Jensen, Kathrine Tejlgård, Nielsen, Peter Kresten, Thøgersen, Ida B., and Enghild, Jan J.
- Published
- 2021
- Full Text
- View/download PDF
19. α2-macroglobulin:Investigation of its native conformation, bait region, and thiol ester using mass spectrometry and mutant characterization
- Author
-
Harwood, Seandean Lykke
- Subjects
protein, biochemistry, protease, protease inhibitor, cross-linking ,protein, biokemi, protease, krydsbinding - Abstract
α2-macrocroglobulin (A2M) er et protein, der findes i humant blod og besidder en unik mekanisme, hvorved det fanger og inhiberer protein-kløvende enzymer, kaldet proteaser. Mekanismen initieres ved, at en protease kløver i et bestemt område på A2M, der agerer som lokkemad. Denne kløvning får A2M til at lukke sig omkring proteasen, som derefter er indespærret og ikke længere kan kløve andre proteiner. Proteaser indvirker i kritiske biologiske processer såsom inflammation, blodkoagulation og omstrukturering af væv, og idet A2M kan inhibere de fleste humane proteaser formodes A2M også at spille en rolle i disse processer. Til trods for at der er blevet forsket i A2M siden 1950’erne, er der stadig uafklarede spørgsmål omkring dets struktur, mekanisme og biologisk funktion.Strukturen af A2M i sin native tilstand har ikke kunne løses ved traditionel røntgen krystallografi pga. dårlig spredning af røntgen stråler fra A2M krystaller. Derforhar jeg koordineret et samarbejdsstudie, hvor flere lav-opløsningsmetoder er blevetanvendt. På baggrund af studiet har vi konstrueret en model for A2M, som viser hvordan dets fire proteinkæder er sat sammen og hvordan det ser ud, når det møder proteaser, hvilket belyser den protease-hæmmende mekanisme. Modellen har en lav opløsningsgrad, men får opbakning fra flere validerende forsøg og stemmer overens med de kendte strukturer på lignende proteiner såsom komplementfaktoren C3.Jeg har igangsat produktion af rekombinant A2M og flere beslægtede proteiner i humane celler. Det har givet mulighed for at indføre ændringer i A2M for at undersøge detaljer i dets funktion. For eksempel har jeg vist, at A2Ms reaktive thiol ester gruppe kan erstattes med en disulfidbro, hvilket viser at thiol esteren ikke spiller en afgørende rolle i A2Ms mekanisme. På lignende vis kan A2Ms lokkemadsområde erstattes med enny sekvens, der ikke genkendes og kløves af humane proteaser, hvori nye kløvningssteder kan blive indsat. Denne nye sekvens blev brugt som udgangspunkt til at lave nye protease-hæmmere, der var selektive overfor bestemte proteaser.Derudover har jeg været en del af den tidlige karakterising af A2ML1, en protease-hæmmer tilhørende A2Ms proteinfamilie, som hovedsageligt er til at finde i detyderste lag af hud. Der er fundet en sammenhæng mellem genet for A2ML1 og flere sjældne sygdomme, heriblandt paraneoplastisk pemfigus, kronisk mellemørebetændelse og Noonan syndrom. Vi har opdaget at thiol esteren fra A2ML1 reagerer effektivt med hydroxyler, hvilket adskiller den fra A2M, men gør at den i i højere grad ligner komplementfaktorer som C3 og C4B. Thiol esterens hydroxyl reaktivitet viste sig at bidrage til A2ML1s hæmning af proteaser.Tilsammen har disse resultater fremmet forskningsfeltet indenfor A2M og lignende protease-hæmmere. Der er blevet grundlagt metoder og fremgange der vilgavne studier af lignende proteiner og fremtidig udvikling af A2M-baserede behandlingsformer.
- Published
- 2021
20. Substituting the Thiol Ester of Human A2M or C3 with a Disulfide Produces Native Proteins with Altered Proteolysis-Induced Conformational Changes
- Author
-
Harwood, Seandean Lykke, primary, Nielsen, Nadia Sukusu, additional, Pedersen, Henrik, additional, Kjøge, Katarzyna, additional, Nielsen, Peter Kresten, additional, Andersen, Gregers Rom, additional, and Enghild, Jan J., additional
- Published
- 2020
- Full Text
- View/download PDF
21. Carcinoembryonic Antigen (CEA)-Specific 4-1BB-Costimulation Induced by CEA-Targeted 4-1BB-Agonistic Trimerbodies
- Author
-
Mikkelsen, Kasper, primary, Harwood, Seandean Lykke, additional, Compte, Marta, additional, Merino, Nekane, additional, Mølgaard, Kasper, additional, Lykkemark, Simon, additional, Alvarez-Mendez, Ana, additional, Blanco, Francisco J., additional, and Álvarez-Vallina, Luis, additional
- Published
- 2019
- Full Text
- View/download PDF
22. 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, Alvarez-Vallina, Luis, Sub Cell Biology, Celbiologie, Sub Cell Biology, and Celbiologie
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,T cell ,CD3 ,medicine.medical_treatment ,Immunology ,lcsh:RC254-282 ,T cell redirection ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,medicine ,trimerbody ,Immunology and Allergy ,Original Research ,cancer immunotherapy ,Immunological synapse formation ,biology ,T-cell receptor ,t cell redirection ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Molecular biology ,3. Good health ,bispecific antibody ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,epithelial growth factor receptor ,Cancer research ,biology.protein ,Antibody ,Clone (B-cell biology) ,lcsh:RC581-607 - 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 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
23. ATTACK, a novel bispecific T cell-recruiting antibody with trivalent EGFR binding and monovalent CD3 binding for cancer immunotherapy
- Author
-
Sub Cell Biology, Celbiologie, 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, Alvarez-Vallina, Luis, Sub Cell Biology, Celbiologie, 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
- Published
- 2017
24. Intramolecular trimerization, a novel strategy for making multispecific antibodies with controlled orientation of the antigen binding domains
- Author
-
Alvarez-Cienfuegos, Ana, primary, Nuñez-Prado, Natalia, additional, Compte, Marta, additional, Cuesta, Angel M., additional, Blanco-Toribio, Ana, additional, Harwood, Seandean Lykke, additional, Villate, Maider, additional, Merino, Nekane, additional, Bonet, Jaume, additional, Navarro, Rocio, additional, Muñoz-Briones, Clara, additional, Sørensen, Karen Marie Juul, additional, Mølgaard, Kasper, additional, Oliva, Baldo, additional, Sanz, Laura, additional, Blanco, Francisco J., additional, and Alvarez-Vallina, Luis, additional
- Published
- 2016
- Full Text
- View/download PDF
25. Efficacy and toxicity management of CAR-T-cell immunotherapy: a matter of responsiveness control or tumour-specificity?
- Author
-
Alonso-Camino, Vanesa, primary, Harwood, Seandean Lykke, additional, Álvarez-Méndez, Ana, additional, and Alvarez-Vallina, Luis, additional
- Published
- 2016
- Full Text
- View/download PDF
26. 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
27. 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
- Full Text
- View/download PDF
28. 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
- Full Text
- View/download PDF
29. 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
- Full Text
- View/download PDF
30. Engineering New Protease Inhibitors Using α 2 -Macroglobulin.
- Author
-
Harwood SL and Enghild JJ
- Subjects
- Humans, Pregnancy, Female, Protease Inhibitors pharmacology, Enzyme Inhibitors, alpha-Macroglobulins chemistry, alpha-Macroglobulins metabolism, Endopeptidases metabolism, Peptide Hydrolases, Macroglobulins, Pregnancy-Associated alpha 2-Macroglobulins
- Abstract
Protease inhibitors of the alpha-macroglobulin family (αM) have a unique mechanism that allows them to trap proteases that is dependent not on the protease's class, but rather on its cleavage specificity. Proteases trigger a conformational change in the αM protein by cleaving within a "bait region," resulting in the sequestering of the protease inside the αM molecule. This nonspecific inhibitory mechanism appears to have arisen early in the αM family, and the broad protease-trapping capacity that it allows may play a role in pathogen defense.Human α
2 -macroglobulin (A2M) is a tetrameric αM whose bait region is permissive to cleavage by most proteases, making it a broad-spectrum protease inhibitor. Recent work has demonstrated that the inhibitory capacity of A2M derives directly from its bait region sequence: modifying the bait region sequence to introduce or remove protease cleavage sites will modify A2M's inhibition of the relevant proteases accordingly. Thus, changing the amino acid sequence of the bait region presents an effective avenue for protein engineering of new protease inhibitors if the substrate specificity of the target protease is known. The design of new A2M-based protease inhibitors with tailored inhibitory capacities has potential applications in basic research and the clinic. In this chapter, we describe the general approach and considerations for the bait region engineering of A2M., (© 2024. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.