1
|
Kemper K, Gielen E, Boross P, Houtkamp M, Plantinga TS, de Poot SAH, Burm SM, Janmaat ML, Koopman LA, van den Brink EN, Rademaker R, Verzijl D, Engelberts PJ, Satijn D, Sasser AK, Breij ECW. Mechanistic and pharmacodynamic studies of DuoBody-CD3x5T4 in preclinical tumor models. Life Sci Alliance 2022; 5:5/11/e202201481. [PMID: 36271507 PMCID: PMC9458754 DOI: 10.26508/lsa.202201481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
CD3 bispecific antibodies (bsAbs) show great promise as anticancer therapeutics. Here, we show in-depth mechanistic studies of a CD3 bsAb in solid cancer, using DuoBody-CD3x5T4. Cross-linking T cells with tumor cells expressing the oncofetal antigen 5T4 was required to induce cytotoxicity. Naive and memory CD4+ and CD8+ T cells were equally effective at mediating cytotoxicity, and DuoBody-CD3x5T4 induced partial differentiation of naive T-cell subsets into memory-like cells. Tumor cell kill was associated with T-cell activation, proliferation, and production of cytokines, granzyme B, and perforin. Genetic knockout of FAS or IFNGR1 in 5T4+ tumor cells abrogated tumor cell kill. In the presence of 5T4+ tumor cells, bystander kill of 5T4− but not of 5T4−IFNGR1− tumor cells was observed. In humanized xenograft models, DuoBody-CD3x5T4 antitumor activity was associated with intratumoral and peripheral blood T-cell activation. Lastly, in dissociated patient-derived tumor samples, DuoBody-CD3x5T4 activated tumor-infiltrating lymphocytes and induced tumor-cell cytotoxicity, even when most tumor-infiltrating lymphocytes expressed PD-1. These data provide an in-depth view on the mechanism of action of a CD3 bsAb in preclinical models of solid cancer.
Collapse
|
2
|
Hagiyama M, Mimae T, Wada A, Takeuchi F, Yoneshige A, Inoue T, Kotoku N, Hamada H, Sekido Y, Okada M, Ito A. Possible Therapeutic Utility of anti-Cell Adhesion Molecule 1 Antibodies for Malignant Pleural Mesothelioma. Front Cell Dev Biol 2022; 10:945007. [PMID: 35903548 PMCID: PMC9315061 DOI: 10.3389/fcell.2022.945007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/23/2022] [Indexed: 11/15/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a highly aggressive malignant tumor, and the effective therapeutic drugs are limited. Thus, the establishment of novel therapeutic method is desired. Considerable proportion of MPMs are shown to express cell adhesion molecule 1 (CADM1), and to use CADM1 to bind to and proliferate on the pleural mesothelial surface, suggesting that CADM1 is a possible therapeutic target. Here, anti-CADM1 ectodomain chicken monoclonal antibodies, 3E1 and 9D2, were examined for their possible therapeutic utility. The full-length form of CADM1 was expressed in eight out of twelve human MPM cell lines. MPM cell lines were cultured on a confluent monolayer of mesothelial MeT-5A cells in the presence of 9D2, the neutralizing antibody. 9D2 suppressed the cell growth of CADM1-positive MPM cells with the loss and aggregation of CADM1 molecules on the MPM cell membrane, but not of CADM1-negative MPM cells. Co-addition of 3E1, lacking the neutralizing action, enhanced the growth-suppressive effect of 9D2. The two antibodies were tested as drug delivery vectors. 3E1 was converted into a humanized antibody (h3E1) and conjugated with monomethyl auristatin E (MMAE), a tubulin polymerization inhibitor. When the resulting h3E1–MMAE antibody-drug conjugate (ADC) was added to the standard cultures of CADM1-positive MPM cells, it suppressed the cell growth in a dose-dependent manner. Co-addition of 9D2 enhanced the growth-suppressive effect of h3E1–MMAE ADC. Anti-CADM1 ectodomain antibodies were suggested to serve as both antibody drugs and drug vectors in the treatment of MPM.
Collapse
Affiliation(s)
- Man Hagiyama
- Department of Pathology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Takahiro Mimae
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Akihiro Wada
- Department of Pathology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Fuka Takeuchi
- Division of Molecular Pathology, Graduate School of Medical Science, Kindai University, Osaka, Japan
| | - Azusa Yoneshige
- Department of Pathology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Takao Inoue
- Department of Pathology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Naoyuki Kotoku
- College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| | - Hironobu Hamada
- Department of Physical Analysis and Therapeutic Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yoshitaka Sekido
- Division of Cancer Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Morihito Okada
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Akihiko Ito
- Department of Pathology, Kindai University Faculty of Medicine, Osaka, Japan
- Division of Molecular Pathology, Graduate School of Medical Science, Kindai University, Osaka, Japan
- *Correspondence: Akihiko Ito,
| |
Collapse
|
3
|
Wu Y, Li Q, Kong Y, Wang Z, Lei C, Li J, Ding L, Wang C, Cheng Y, Wei Y, Song Y, Yang Z, Tu C, Ding Y, Ying T. A highly stable human single-domain antibody-drug conjugate exhibits superior penetration and treatment of solid tumors. Mol Ther 2022; 30:2785-2799. [PMID: 35462042 PMCID: PMC9372316 DOI: 10.1016/j.ymthe.2022.04.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/04/2022] [Accepted: 04/18/2022] [Indexed: 10/18/2022] Open
Abstract
The inefficient tumor penetration of therapeutic antibodies has hampered their effective use in treating solid tumors. Here, we report the identification of a fully human single-domain antibody (UdAb), designated as n501, targeting the oncofetal antigen 5T4. The high-resolution crystal structure indicates that n501 adopts a compact structure very similar to that of camelid nanobodies, and binds tightly to all eight leucine-rich repeats of 5T4. Furthermore, the UdAb n501 exhibits exceptionally high stability, with no apparent activity changes over 4 weeks of storage at various temperatures. Importantly, the UdAb-based antibody-drug conjugate (n501-SN38) showed much deeper tumor penetration, significantly higher tumor uptake, and faster accumulation at tumor sites than conventional IgG1-based antibody-drug conjugate (m603-SN38), resulting in improved tumor inhibition. These results highlight the potential of UdAb-based antibody-drug conjugates as a potential class of antitumor therapeutics with characteristics of high stability and strong tumor penetration for the effective treatment of solid tumors.
Collapse
|
4
|
Obacz J, Yung H, Shamseddin M, Linnane E, Liu X, Azad AA, Rassl DM, Fairen-Jimenez D, Rintoul RC, Nikolić MZ, Marciniak SJ. Biological basis for novel mesothelioma therapies. Br J Cancer 2021; 125:1039-1055. [PMID: 34226685 PMCID: PMC8505556 DOI: 10.1038/s41416-021-01462-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/13/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Mesothelioma is an aggressive cancer that is associated with exposure to asbestos. Although asbestos is banned in several countries, including the UK, an epidemic of mesothelioma is predicted to affect middle-income countries during this century owing to their heavy consumption of asbestos. The prognosis for patients with mesothelioma is poor, reflecting a failure of conventional chemotherapy that has ultimately resulted from an inadequate understanding of its biology. However, recent work has revolutionised the study of mesothelioma, identifying genetic and pathophysiological vulnerabilities, including the loss of tumour suppressors, epigenetic dysregulation and susceptibility to nutrient stress. We discuss how this knowledge, combined with advances in immunotherapy, is enabling the development of novel targeted therapies.
Collapse
Affiliation(s)
- Joanna Obacz
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Henry Yung
- UCL Respiratory, Division of Medicine Rayne Institute, University College London, London, UK
| | - Marie Shamseddin
- Wellcome Sanger Institute, Wellcome Trust Genome Campus, Saffron Walden, UK
| | - Emily Linnane
- Adsorption & Advanced Materials Laboratory, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Xiewen Liu
- Adsorption & Advanced Materials Laboratory, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Arsalan A Azad
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Doris M Rassl
- Department of Histopathology, Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - David Fairen-Jimenez
- Adsorption & Advanced Materials Laboratory, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Robert C Rintoul
- Department of Oncology, University of Cambridge, Cambridge, UK
- Department of Thoracic Oncology, Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - Marko Z Nikolić
- UCL Respiratory, Division of Medicine Rayne Institute, University College London, London, UK
| | - Stefan J Marciniak
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
5
|
High Prevalence of 5T4/Trophoblast Glycoprotein in Soft Tissue Sarcomas. Cancers (Basel) 2021; 13:cancers13194841. [PMID: 34638324 PMCID: PMC8508483 DOI: 10.3390/cancers13194841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
The expression of 5T4/trophoblast glycoprotein was evaluated in several histological subtypes of soft tissue sarcoma (STS) to determine whether the prevalence and level of expression of this membrane-associated glycoprotein is sufficient for use in targeted therapies. Tumor tissue microarrays containing cores from different histological subtypes of STS were stained using a standardized immunohistochemical staining method to detect 5T4; the level of staining was assessed using a semi-quantitative scoring method. No 5T4 staining was seen in the angiosarcomas and liposarcomas investigated in this study. 5T4 staining in the other STS subtypes was seen in more than 50% of cases, warranting further investigation into whether this antigen could evoke an anti-tumor immune response or can be used as target for the delivery of more potent toxins through antibody drug conjugates.
Collapse
|