28 results on '"Tapia‐Galisteo, Antonio"'
Search Results
2. Tumor targeted 4-1BB agonist antibody-albumin fusions with high affinity to FcRn induce anti-tumor immunity without toxicity
- Author
-
Hangiu, Oana, Compte, Marta, Dinesen, Anders, Navarro, Rocio, Tapia-Galisteo, Antonio, Mandrup, Ole A., Erce-Llamazares, Ainhoa, Lázaro-Gorines, Rodrigo, Nehme-Álvarez, Daniel, Domínguez-Alonso, Carmen, Harwood, Seandean L., Alfonso, Carlos, Blanco, Belen, Rubio-Pérez, Laura, Jiménez-Reinoso, Anaïs, Díez-Alonso, Laura, Blanco, Francisco J., Sanz, Laura, Howard, Kenneth A., and Álvarez-Vallina, Luis
- Published
- 2022
- Full Text
- View/download PDF
3. Combination of T cell-redirecting strategies with a bispecific antibody blocking TGF-β and PD-L1 enhances antitumor responses
- Author
-
Tapia-Galisteo, Antonio, primary, Sánchez-Rodríguez, Iñigo, additional, Narbona, Javier, additional, Iglesias-Hernández, Patricia, additional, Aragón-García, Saray, additional, Jiménez-Reinoso, Anaïs, additional, Compte, Marta, additional, Khan, Shaukat, additional, Tsuda, Takeshi, additional, Chames, Patrick, additional, Lacadena, Javier, additional, Álvarez-Vallina, Luis, additional, and Sanz, Laura, additional
- Published
- 2024
- Full Text
- View/download PDF
4. 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
5. Engineered T cells secreting anti-BCMA T cell engagers control multiple myeloma and promote immune memory in vivo.
- Author
-
Díez-Alonso, Laura, Falgas, Aïda, Arroyo-Ródenas, Javier, Romencín, Paola A., Martínez, Alba, Gómez-Rosel, Marina, Blanco, Belén, Jiménez-Reinoso, Anaïs, Mayado, Andrea, Pérez-Pons, Alba, Aguilar-Sopeña, Óscar, Ramírez-Fernández, Ángel, Segura-Tudela, Alejandro, Perez-Amill, Lorena, Tapia-Galisteo, Antonio, Domínguez-Alonso, Carmen, Rubio-Pérez, Laura, Jara, Maria, Solé, Francesc, and Hangiu, Oana
- Subjects
MULTIPLE myeloma ,T cells ,IMMUNOLOGIC memory ,BISPECIFIC antibodies ,HEMATOLOGIC malignancies - Abstract
Multiple myeloma is the second most common hematological malignancy in adults and remains an incurable disease. B cell maturation antigen (BCMA)–directed immunotherapy, including T cells bearing chimeric antigen receptors (CARs) and systemically injected bispecific T cell engagers (TCEs), has shown remarkable clinical activity, and several products have received market approval. However, despite promising results, most patients eventually become refractory and relapse, highlighting the need for alternative strategies. Engineered T cells secreting TCE antibodies (STAb) represent a promising strategy that combines the advantages of adoptive cell therapies and bispecific antibodies. Here, we undertook a comprehensive preclinical study comparing the therapeutic potential of T cells either expressing second-generation anti-BCMA CARs (CAR-T) or secreting BCMAxCD3 TCEs (STAb-T) in a T cell–limiting experimental setting mimicking the conditions found in patients with relapsed/refractory multiple myeloma. STAb-T cells recruited T cell activity at extremely low effector-to-target ratios and were resistant to inhibition mediated by soluble BCMA released from the cell surface, resulting in enhanced cytotoxic responses and prevention of immune escape of multiple myeloma cells in vitro. These advantages led to robust expansion and persistence of STAb-T cells in vivo, generating long-lived memory BCMA-specific responses that could control multiple myeloma progression in xenograft models, outperforming traditional CAR-T cells. These promising preclinical results encourage clinical testing of the BCMA-STAb-T cell approach in relapsed/refractory multiple myeloma. Editor's summary: Taking a STAb at cellular therapy. Multiple myeloma is a hematological cancer with limited treatment options, particularly after relapse. Chimeric antigen receptor (CAR) T cells are being explored for multiple myeloma, much like for other hematological malignancies, but patients still relapse. Here, Díez-Alonso et al. developed STAb-T cells, which, rather than express a CAR, secrete T cell–engaging antibodies targeting B cell maturation antigen (BCMA) and CD3. The authors compared STAb-T cells with traditional CAR-T cells in vitro and in vivo, showing that the STAb-T cells more effectively killed target cells at low frequencies and were resistant to inhibition mediated by soluble BCMA released from myeloma cells, preventing immune escape. This translated to improved in vivo efficacy in a murine multiple myeloma model, supporting further development of BCMA-specific STAb-T cells as a therapeutic for this and other hematological malignancies. —Courtney Malo [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. 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
7. A PD-L1/EGFR bispecific antibody combines immune checkpoint blockade and direct anti-cancer action for an enhanced anti-tumor response
- Author
-
Rubio-Pérez, Laura, primary, Lázaro-Gorines, Rodrigo, additional, Harwood, Seandean L., additional, Compte, Marta, additional, Navarro, Rocío, additional, Tapia-Galisteo, Antonio, additional, Bonet, Jaume, additional, Blanco, Belén, additional, Lykkemark, Simon, additional, Ramírez-Fernández, Ángel, additional, Ferreras-Gutiérrez, Mariola, additional, Domínguez-Alonso, Carmen, additional, Díez-Alonso, Laura, additional, Segura-Tudela, Alejandro, additional, Hangiu, Oana, additional, Erce-Llamazares, Ainhoa, additional, Blanco, Francisco J., additional, Santos, Cruz, additional, Rodríguez-Peralto, José L., additional, Sanz, Laura, additional, and Álvarez-Vallina, Luis, additional
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Overcoming CAR-Mediated CD19 Downmodulation and Leukemia Relapse with T Lymphocytes Secreting Anti-CD19 T-cell Engagers
- Author
-
Blanco, Belén, primary, Ramírez-Fernández, Ángel, primary, Bueno, Clara, primary, Argemí-Muntadas, Lidia, primary, Fuentes, Patricia, primary, Aguilar-Sopeña, Óscar, primary, Gutierrez-Agüera, Francisco, primary, Zanetti, Samanta Romina, primary, Tapia-Galisteo, Antonio, primary, Díez-Alonso, Laura, primary, Segura-Tudela, Alejandro, primary, Castellà, Maria, primary, Marzal, Berta, primary, Betriu, Sergi, primary, Harwood, Seandean L., primary, Compte, Marta, primary, Lykkemark, Simon, primary, Erce-Llamazares, Ainhoa, primary, Rubio-Pérez, Laura, primary, Jiménez-Reinoso, Anaïs, primary, Domínguez-Alonso, Carmen, primary, Neves, Maria, primary, Morales, Pablo, primary, Paz-Artal, Estela, primary, Guedan, Sonia, primary, Sanz, Laura, primary, Toribio, María L., primary, Roda-Navarro, Pedro, primary, Juan, Manel, primary, Menéndez, Pablo, primary, and Álvarez-Vallina, Luis, primary
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Data from Overcoming CAR-Mediated CD19 Downmodulation and Leukemia Relapse with T Lymphocytes Secreting Anti-CD19 T-cell Engagers
- Author
-
Blanco, Belén, primary, Ramírez-Fernández, Ángel, primary, Bueno, Clara, primary, Argemí-Muntadas, Lidia, primary, Fuentes, Patricia, primary, Aguilar-Sopeña, Óscar, primary, Gutierrez-Agüera, Francisco, primary, Zanetti, Samanta Romina, primary, Tapia-Galisteo, Antonio, primary, Díez-Alonso, Laura, primary, Segura-Tudela, Alejandro, primary, Castellà, Maria, primary, Marzal, Berta, primary, Betriu, Sergi, primary, Harwood, Seandean L., primary, Compte, Marta, primary, Lykkemark, Simon, primary, Erce-Llamazares, Ainhoa, primary, Rubio-Pérez, Laura, primary, Jiménez-Reinoso, Anaïs, primary, Domínguez-Alonso, Carmen, primary, Neves, Maria, primary, Morales, Pablo, primary, Paz-Artal, Estela, primary, Guedan, Sonia, primary, Sanz, Laura, primary, Toribio, María L., primary, Roda-Navarro, Pedro, primary, Juan, Manel, primary, Menéndez, Pablo, primary, and Álvarez-Vallina, Luis, primary
- Published
- 2023
- Full Text
- View/download PDF
10. Figure S1 from An Fc-free EGFR-specific 4-1BB-agonistic Trimerbody Displays Broad Antitumor Activity in Humanized Murine Cancer Models without Toxicity
- Author
-
Compte, Marta, primary, Harwood, Seandean L., primary, Erce-Llamazares, Ainhoa, primary, Tapia-Galisteo, Antonio, primary, Romero, Eduardo, primary, Ferrer, Irene, primary, Garrido-Martin, Eva M., primary, Enguita, Ana B., primary, Ochoa, Maria C., primary, Blanco, Belén, primary, Oteo, Marta, primary, Merino, Nekane, primary, Nehme-Álvarez, Daniel, primary, Hangiu, Oana, primary, Domínguez-Alonso, Carmen, primary, Zonca, Manuela, primary, Ramírez-Fernández, Angel, primary, Blanco, Francisco J., primary, Morcillo, Miguel A., primary, Muñoz, Ines G., primary, Melero, Ignacio, primary, Rodriguez-Peralto, José L., primary, Paz-Ares, Luis, primary, Sanz, Laura, primary, and Alvarez-Vallina, Luis, primary
- Published
- 2023
- Full Text
- View/download PDF
11. When three is not a crowd: trispecific antibodies for enhanced cancer immunotherapy
- Author
-
Tapia-Galisteo, Antonio, primary, Compte, Marta, additional, Álvarez-Vallina, Luis, additional, and Sanz, Laura, additional
- Published
- 2023
- Full Text
- View/download PDF
12. 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
13. Generación y caracterización de fragmentos de anticuerpo recombinantes multiespecíficos para inmunoterapia del cáncer
- Author
-
Tapia Galisteo, Antonio and Sanz Alcober, Laura
- Subjects
Fisiología - Abstract
En las últimas décadas, los anticuerpos biespecíficos (AcBs) han demostrado su potencial terapéutico en cáncer, culminando con la aprobación de blinatumomab para el tratamiento de la leucemia linfoblástica aguda (2014) y amivantamab para el del cáncer de pulmón no microcítico (2021). Sin embargo, a pesar del incremento exponencial del número de AcBs en desarrollo, su uso terapéutico ha mostrado limitaciones de eficacia y seguridad inherentes a su diseño. En este trabajo se describe y evalúa un nuevo formato de anticuerpo triespecífico (AcTs) que redirige la actividad de linfocitos T hacia células de cáncer colorrectal (CCR). El AcTs en formato TriTE (del inglés, Trispecific T-cell Engager) se encuentra formado por un scFv (del inglés, single-chain Fv) anti-CD3 flanqueado por dos dominios VHH frente a los antígenos asociados a tumor (AAT) EpCAM y EGFR. Este anticuerpo, eficientemente expresado en células de mamífero y en levaduras, es capaz de reconocer sus antígenos diana en la superficie celular, así como de desencadenar la actividad citotóxica de linfocitos T de forma específica hacia células de CCR que expresan EpCAM y/o EGFR, aunque en menor medida en las células que expresan solo uno de los AAT. In vitro, la lisis de células de CCR que expresan ambos AAT promovida por TriTE fue al menos 20 veces más potente que la de células solo EpCAM+ o EGFR+. Además, fue capaz de retrasar el crecimiento tumoral y aumentar de forma significativa la supervivencia in vivo. La ausencia de actividad citotóxica frente a células de CCR doble negativas y la actividad limitada frente a células que solo expresan uno de los AAT, apuntan hacia una mayor especificidad y un mejor perfil de seguridad de TriTE con respecto a los AcBs convencionales. Además, el reconocimiento dual de dos AAT distintos puede reducir el riesgo de escape tumoral asociado a la heterogeneidad antigénica intrínseca o a la pérdida de su expresión por la presión selectiva de las terapias basadas en AcBs...
- Published
- 2022
14. Generación y caracterización de fragmentos de anticuerpo recombinantes multiespecíficos para inmunoterapia del cáncer
- Author
-
Sanz Alcober, Laura, Tapia Galisteo, Antonio, Sanz Alcober, Laura, and Tapia Galisteo, Antonio
- Abstract
En las últimas décadas, los anticuerpos biespecíficos (AcBs) han demostrado su potencial terapéutico en cáncer, culminando con la aprobación de blinatumomab para el tratamiento de la leucemia linfoblástica aguda (2014) y amivantamab para el del cáncer de pulmón no microcítico (2021). Sin embargo, a pesar del incremento exponencial del número de AcBs en desarrollo, su uso terapéutico ha mostrado limitaciones de eficacia y seguridad inherentes a su diseño. En este trabajo se describe y evalúa un nuevo formato de anticuerpo triespecífico (AcTs) que redirige la actividad de linfocitos T hacia células de cáncer colorrectal (CCR). El AcTs en formato TriTE (del inglés, Trispecific T-cell Engager) se encuentra formado por un scFv (del inglés, single-chain Fv) anti-CD3 flanqueado por dos dominios VHH frente a los antígenos asociados a tumor (AAT) EpCAM y EGFR. Este anticuerpo, eficientemente expresado en células de mamífero y en levaduras, es capaz de reconocer sus antígenos diana en la superficie celular, así como de desencadenar la actividad citotóxica de linfocitos T de forma específica hacia células de CCR que expresan EpCAM y/o EGFR, aunque en menor medida en las células que expresan solo uno de los AAT. In vitro, la lisis de células de CCR que expresan ambos AAT promovida por TriTE fue al menos 20 veces más potente que la de células solo EpCAM+ o EGFR+. Además, fue capaz de retrasar el crecimiento tumoral y aumentar de forma significativa la supervivencia in vivo. La ausencia de actividad citotóxica frente a células de CCR doble negativas y la actividad limitada frente a células que solo expresan uno de los AAT, apuntan hacia una mayor especificidad y un mejor perfil de seguridad de TriTE con respecto a los AcBs convencionales. Además, el reconocimiento dual de dos AAT distintos puede reducir el riesgo de escape tumoral asociado a la heterogeneidad antigénica intrínseca o a la pérdida de su expresión por la presión selectiva de las terapias basadas en AcBs..., In the last decades, biespecific antibodies (BsAb) have shown a great therapeutic potential in cancer, leading to the approval of blinatumomab for acute lymphoblastic leukemia treatment (2014) and amivantamab for non-small cell lung cancer (2021). In spite of the increasing number of BsAb in development, their therapeutic use has revealed limitations related to efficacy and safety. Here, we describe and characterize a new trispecific antibody (TsAb) format, named TriTE (Trispecific T-cell Engager), which redirects T cell response toward colorectal cancer (CRC) cells. The TriTE antibody consists of two anti-EGFR and anti-EpCAM single-domain VHH antibodies fused to an anti-CD3 scFv (single-chain Fv) antibody fragment. This antibody, which is efficiently expressed by mammalian and yeast cells, is able to recognize its target antigens on the cell surface, as well as trigger specific cytotoxic T cell response against EGFR- and/or EpCAM-expressing CRC cells, although to a lesser extent in those which express a single tumor-associated antigen (TAA). In vitro, specific cytolysis of double-positive CRC cells induced by TriTE was at least 20-fold stronger compared to that of single-positive CRC cells. Moreover, TriTE was able to delay the tumor growth and significantly increased survival in vivo. A higher specificity and a better safety profile of TriTE regarding conventional BsAb are expected, since double-negative CRC cells were spared and the killing of single-positive cells was limited. In addition, the dual targeting of two different TAA can decrease the risk of tumor scape due to intratumor antigenic heterogeneity or antigen loss associated to selective pressure by BsAb-based therapies...
- Published
- 2022
15. 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
16. Overcoming CAR-Mediated CD19 Downmodulation and Leukemia Relapse with T Lymphocytes Secreting Anti-CD19 T-cell Engagers
- Author
-
Blanco, Belén, primary, Ramírez-Fernández, Ángel, additional, Bueno, Clara, additional, Argemí-Muntadas, Lidia, additional, Fuentes, Patricia, additional, Aguilar-Sopeña, Óscar, additional, Gutierrez-Agüera, Francisco, additional, Zanetti, Samanta Romina, additional, Tapia-Galisteo, Antonio, additional, Díez-Alonso, Laura, additional, Segura-Tudela, Alejandro, additional, Castellà, Maria, additional, Marzal, Berta, additional, Betriu, Sergi, additional, Harwood, Seandean L., additional, Compte, Marta, additional, Lykkemark, Simon, additional, Erce-Llamazares, Ainhoa, additional, Rubio-Pérez, Laura, additional, Jiménez-Reinoso, Anaïs, additional, Domínguez-Alonso, Carmen, additional, Neves, Maria, additional, Morales, Pablo, additional, Paz-Artal, Estela, additional, Guedan, Sonia, additional, Sanz, Laura, additional, Toribio, María L., additional, Roda-Navarro, Pedro, additional, Juan, Manel, additional, Menéndez, Pablo, additional, and Álvarez-Vallina, Luis, additional
- Published
- 2022
- Full Text
- View/download PDF
17. 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
18. TGF-β-induced IGFBP-3 is a key paracrine factor from activated pericytes that promotes colorectal cancer cell migration and invasion
- Author
-
Navarro, Rocío, Tapia-Galisteo, Antonio, Martín-García, Laura, Tarín, Carlos, Corbacho, Cesáreo, Sánchez-Tirado, Esther, Campuzano, Susana, González-Cortés, Araceli, Yáñez-Sedeño, Paloma, Compte, Marta, Álvarez-Vallina, Luis, Sanz, Laura, Tapia‐Galisteo, Antonio, Martín‐García, Laura, Gómez-López, Gonzalo, Sánchez‐Tirado, Esther, González‐Cortés, Araceli, Yáñez‐Sedeño, Paloma, Álvarez‐Vallina, Luis, European Research Council, Instituto de Salud Carlos III, Comunidad de Madrid (España), Ministerio de Economía y Competitividad (España), European Research Council (ERC), Instituto de Salud Carlos III - ISCIII, and Comunidad de Madrid
- Subjects
0301 basic medicine ,Cancer Research ,Carcinogenesis ,Metastasis ,0302 clinical medicine ,Cell Movement ,Transforming Growth Factor beta ,pericyte ,Research Articles ,Cell migration ,General Medicine ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.anatomical_structure ,Phenotype ,Oncology ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Molecular Medicine ,Female ,Pericyte ,Colorectal Neoplasms ,Research Article ,Signal Transduction ,TGF-β ,Mice, Nude ,colorectal cancer ,Biology ,lcsh:RC254-282 ,03 medical and health sciences ,Paracrine signalling ,Cell Line, Tumor ,Paracrine Communication ,Genetics ,medicine ,Animals ,Humans ,tumor microenvironment ,Neoplasm Invasiveness ,Autocrine signalling ,TGF‐β ,Cell Proliferation ,Tumor microenvironment ,IGFBP-3 ,medicine.disease ,IGFBP‐3 ,digestive system diseases ,030104 developmental biology ,Insulin-Like Growth Factor Binding Protein 3 ,Drug Resistance, Neoplasm ,Cancer cell ,Cancer research ,Cytokine secretion ,Pericytes ,Proto-Oncogene Proteins c-akt - Abstract
In this work, we show that TGF‐β produced by colorectal cancer cells activates pericytes that in turn promote their tumorigenic properties through the release of soluble factors, including IGFBP‐3, with a prominent role in cancer cell migration and invasion. Moreover, co‐implantation of colorectal cancer cells and pericytes in immunodeficient mice increased tumor growth., The crosstalk between cancer cells and the tumor microenvironment has been implicated in cancer progression and metastasis. Fibroblasts and immune cells are widely known to be attracted to and modified by cancer cells. However, the role of pericytes in the tumor microenvironment beyond endothelium stabilization is poorly understood. Here, we report that pericytes promoted colorectal cancer (CRC) cell proliferation, migration, invasion, stemness, and chemoresistance in vitro, as well as tumor growth in a xenograft CRC model. We demonstrate that coculture with human CRC cells induced broad transcriptomic changes in pericytes, mostly associated with TGF‐β receptor activation. The prognostic value of a TGF‐β response signature in pericytes was analyzed in CRC patient data sets. This signature was found to be a good predictor of CRC relapse. Moreover, in response to stimulation by CRC cells, pericytes expressed high levels of TGF‐β1, initiating an autocrine activation loop. Investigation of secreted mediators and underlying molecular mechanisms revealed that IGFBP‐3 is a key paracrine factor from activated pericytes affecting CRC cell migration and invasion. In summary, we demonstrate that the interplay between pericytes and CRC cells triggers a vicious cycle that stimulates pericyte cytokine secretion, in turn increasing CRC cell tumorigenic properties. Overall, we provide another example of how cancer cells can manipulate the tumor microenvironment.
- Published
- 2020
19. Case Report: An EGFR-Targeted 4-1BB-agonistic Trimerbody Does Not Induce Hepatotoxicity in Transgenic Mice With Liver Expression of Human EGFR
- Author
-
Palazon, Asis, Compte, Marta, Harwood, Seandean L., Martínez-Torrecuadrada, Jorge, Pérez-Chacón, Gema, González-García, Patricia, Tapia-Galisteo, Antonio, Bergen en Henegouwen, Paul M. P. Van, Sánchez Muñoz, Aranzazu, Fabregat, Isabel, Sanz, Laura, Zapata, Juan M., Álvarez-Vallina, Luis, Palazon, Asis, Compte, Marta, Harwood, Seandean L., Martínez-Torrecuadrada, Jorge, Pérez-Chacón, Gema, González-García, Patricia, Tapia-Galisteo, Antonio, Bergen en Henegouwen, Paul M. P. Van, Sánchez Muñoz, Aranzazu, Fabregat, Isabel, Sanz, Laura, Zapata, Juan M., and Álvarez-Vallina, Luis
- Abstract
Agonistic monoclonal antibodies (mAbs) targeting the co-stimulatory receptor 4-1BB are among the most effective immunotherapeutic agents across pre-clinical cancer models. However, clinical development of full-length 4-1BB agonistic mAbs, has been hampered by dose-limiting liver toxicity. We have previously developed an EGFR-targeted 4-1BBagonistic trimerbody (1D8N/CEGa1) that induces potent anti-tumor immunity without systemic toxicity, in immunocompetent mice bearing murine colorectal carcinoma cells expressing human EGFR. Here, we study the impact of human EGFR expression on mouse liver in the toxicity profile of 1D8N/CEGa1. Systemic administration of IgG-based anti-4-1BB agonist resulted in nonspecific immune stimulation and hepatotoxicity in a liver-specific human EGFR-transgenic immunocompetent mouse, whereas in 1D8N/CEGa1-treated mice no such immune-related adverse effects were observed. Collectively, these data support the role of FcgR interactions in the major offtumor toxicities associated with IgG-based 4-1BB agonists and further validate the safety profile of EGFR-targeted Fc-less 4-1BB-agonistic trimerbodies in systemic cancer immunotherapy protocols., Ministerio de Ciencia e Innovación, Ministerio de Economía y Competitividad, Instituto de Salud Carlos III, FEDER, Asociación Española contra el Cáncer, Depto. de Bioquímica y Biología Molecular, TRUE, pub
- Published
- 2021
20. Case Report: An EGFR-Targeted 4-1BB-agonistic Trimerbody Does Not Induce Hepatotoxicity in Transgenic Mice With Liver Expression of Human EGFR
- Author
-
Sub Cell Biology, Celbiologie, Compte, Marta, Harwood, Seandean L., Martínez-Torrecuadrada, Jorge, Perez-Chacon, Gema, González-García, Patricia, Tapia-Galisteo, Antonio, Van Bergen en Henegouwen, Paul M.P., Sánchez, Aránzazu, Fabregat, Isabel, Sanz, Laura, Zapata, Juan M., Alvarez-Vallina, Luis, Sub Cell Biology, Celbiologie, Compte, Marta, Harwood, Seandean L., Martínez-Torrecuadrada, Jorge, Perez-Chacon, Gema, González-García, Patricia, Tapia-Galisteo, Antonio, Van Bergen en Henegouwen, Paul M.P., Sánchez, Aránzazu, Fabregat, Isabel, Sanz, Laura, Zapata, Juan M., and Alvarez-Vallina, Luis
- Published
- 2021
21. An Fc-free EGFR-specific 4-1BB-agonistic Trimerbody Displays Broad Antitumor Activity in Humanized Murine Cancer Models without Toxicity
- Author
-
Compte, Marta, primary, Harwood, Seandean L., additional, Erce-Llamazares, Ainhoa, additional, Tapia-Galisteo, Antonio, additional, Romero, Eduardo, additional, Ferrer, Irene, additional, Garrido-Martin, Eva M., additional, Enguita, Ana B., additional, Ochoa, Maria C., additional, Blanco, Belén, additional, Oteo, Marta, additional, Merino, Nekane, additional, Nehme-Álvarez, Daniel, additional, Hangiu, Oana, additional, Domínguez-Alonso, Carmen, additional, Zonca, Manuela, additional, Ramírez-Fernández, Angel, additional, Blanco, Francisco J., additional, Morcillo, Miguel A., additional, Muñoz, Ines G., additional, Melero, Ignacio, additional, Rodriguez-Peralto, José L., additional, Paz-Ares, Luis, additional, Sanz, Laura, additional, and Alvarez-Vallina, Luis, additional
- Published
- 2021
- Full Text
- View/download PDF
22. Case Report: An EGFR-Targeted 4-1BB-agonistic Trimerbody Does Not Induce Hepatotoxicity in Transgenic Mice With Liver Expression of Human EGFR
- Author
-
Compte, Marta, primary, Harwood, Seandean L., additional, Martínez-Torrecuadrada, Jorge, additional, Perez-Chacon, Gema, additional, González-García, Patricia, additional, Tapia-Galisteo, Antonio, additional, Van Bergen en Henegouwen, Paul M. P., additional, Sánchez, Aránzazu, additional, Fabregat, Isabel, additional, Sanz, Laura, additional, Zapata, Juan M., additional, and Alvarez-Vallina, Luis, additional
- Published
- 2021
- Full Text
- View/download PDF
23. TGF‐β‐induced IGFBP‐3 is a key paracrine factor from activated pericytes that promotes colorectal cancer cell migration and invasion
- Author
-
Navarro, Rocío, primary, Tapia‐Galisteo, Antonio, additional, Martín‐García, Laura, additional, Tarín, Carlos, additional, Corbacho, Cesáreo, additional, Gómez‐López, Gonzalo, additional, Sánchez‐Tirado, Esther, additional, Campuzano, Susana, additional, González‐Cortés, Araceli, additional, Yáñez‐Sedeño, Paloma, additional, Compte, Marta, additional, Álvarez‐Vallina, Luis, additional, and Sanz, Laura, additional
- Published
- 2020
- Full Text
- View/download PDF
24. Anti-tumoral potential of a human granulysin-based, CEA-targeted cytolytic immunotoxin
- Author
-
Ibáñez-Pérez, Raquel, primary, Guerrero-Ochoa, Patricia, additional, Al-Wasaby, Sameer, additional, Navarro, Rocío, additional, Tapia-Galisteo, Antonio, additional, De Miguel, Diego, additional, Gonzalo, Oscar, additional, Conde, Blanca, additional, Martínez-Lostao, Luis, additional, Hurtado-Guerrero, Ramón, additional, Sanz, Laura, additional, and Anel, Alberto, additional
- Published
- 2019
- Full Text
- View/download PDF
25. 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
26. 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
27. 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
28. 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
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.