1
|
Zhang DY, Zhang ZH, Liu WT, Zhou WM, Zhou PF, Wei JJ, Dai XJ, Zeng XL, Zhou YQ, Li HW, Zhang H, Shen AL, Cheng LS, Shen GD, He YF. A humanized anti-MSLN×4-1BB bispecific antibody exhibits potent antitumour activity through 4-1BB signaling activation and fc function without systemic toxicity. J Transl Med 2025; 23:53. [PMID: 39806351 PMCID: PMC11726934 DOI: 10.1186/s12967-025-06107-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Agonistic monoclonal antibodies targeting 4-1BB/CD137 have shown preclinical promise, but their clinical development has been limited by severe liver toxicity or limited efficacy. Therefore, a safe and efficient immunostimulatory molecule is urgently needed for cancer immunotherapy. METHODS A novel anti-MSLN×4-1BB bispecific antibody (bsAb) was generated via antibody engineering, and its affinity and activity were detected via enzyme-linked immunosorbent assay (ELISA), flow cytometry, and T-cell activation and luciferase reporter assays. In vivo antitumour activity was assessed by establishing humanized mice bearing human MSLN-expressing MC38 (MC38/hMSLN) or CT26 (CT26/hMSLN) cells, and safety was further evaluated in cynomolgus monkeys. RESULTS We generated two humanized anti-MSLN×4-1BB bsAbs (HK013-G1/G4) by fusing an anti-4-1BB scFv to the C-terminus of an anti-MSLN VHH with an intact Fc fragment from human IgG1 or IgG4. The two bsAbs were able to block the binding of CA125 to MSLN and stimulate 4-1BB signaling pathway, which was strictly dependent on MSLN expression. In particular, HK013-G1 retained Fc function and induced ADCC effect in tumour cells, whereas HK013-G4 did not. Strikingly, HK013-G1 showed superior antitumour activity to HK013-G4 both in vitro and in vivo and remained effective even in the presence of soluble MSLN. HK013-G1 enhanced antitumour immunity and induced durable antigen-specific immune memory to prevent rechallenged tumour growth, even at a dose as low as 1 mg/kg. Furthermore, HK013-G1 did not induce nonspecific production of proinflammatory cytokines and showed good tolerability up to the highest tested dose (30 mg/kg weekly) for 5 weeks, with no HK013-G1-related adverse effects observed in cynomolgus monkeys. In addition, the mean half-life of HK013-G1 was approximately 61 and 97 h at single doses of 3 and 30 mg/kg, respectively. CONCLUSION The optimal anti-MSLN×4-1BB bsAb HK013-G1 exhibited synergistic antitumour effects by inducing an ADCC effect (innate immunity) and stimulating the 4-1BB signaling pathway (adaptive immunity) upon cross-bridging with MSLN with no systemic toxicity, which may offer the promise of an improved therapeutic window relative to that of 4-1BB agonists.
Collapse
Affiliation(s)
- Da-Yan Zhang
- Department of Medical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, 230031, Anhui, China
- Hefei HankeMab Biotechnology Co., Ltd.; Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd, Hefei, 230031, Anhui, China
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China
| | - Zhi-Hua Zhang
- Department of Medical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, 230031, Anhui, China
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China
| | - Wen-Ting Liu
- Hefei HankeMab Biotechnology Co., Ltd.; Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd, Hefei, 230031, Anhui, China
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China
| | - Wei-Ming Zhou
- Hefei HankeMab Biotechnology Co., Ltd.; Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd, Hefei, 230031, Anhui, China
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China
| | - Peng-Fei Zhou
- Hefei HankeMab Biotechnology Co., Ltd.; Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd, Hefei, 230031, Anhui, China
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China
| | - Juan-Juan Wei
- Hefei HankeMab Biotechnology Co., Ltd.; Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd, Hefei, 230031, Anhui, China
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China
| | - Xue-Jing Dai
- Hefei HankeMab Biotechnology Co., Ltd.; Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd, Hefei, 230031, Anhui, China
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China
| | - Xiao-Li Zeng
- Hefei HankeMab Biotechnology Co., Ltd.; Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd, Hefei, 230031, Anhui, China
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China
| | - Yu-Qiong Zhou
- Hefei HankeMab Biotechnology Co., Ltd.; Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd, Hefei, 230031, Anhui, China
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China
| | - Han-Wang Li
- Hefei HankeMab Biotechnology Co., Ltd.; Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd, Hefei, 230031, Anhui, China
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China
| | - Heng Zhang
- Hefei HankeMab Biotechnology Co., Ltd.; Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd, Hefei, 230031, Anhui, China
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China
| | - Ao-Lin Shen
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China.
- Department of General Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Lian-Sheng Cheng
- Hefei HankeMab Biotechnology Co., Ltd.; Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd, Hefei, 230031, Anhui, China.
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China.
| | - Guo-Dong Shen
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China.
- Department of Geriatrics, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Yi-Fu He
- Department of Medical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, 230031, Anhui, China.
- Anhui Province Key Laboratory of Geriatric Immunotherapy and Nutrition Therapy, Gerontology Institute of Anhui Province, Hefei, 230001, Anhui, China.
| |
Collapse
|
2
|
Hangiu O, Navarro R, Frago S, Rubio-Pérez L, Tapia-Galisteo A, Díez-Alonso L, Gómez-Rosel M, Silva-Pilipich N, Vanrell L, Smerdou C, Howard KA, Sanz L, Álvarez-Vallina L, Compte M. Effective cancer immunotherapy combining mRNA-encoded bispecific antibodies that induce polyclonal T cell engagement and PD-L1-dependent 4-1BB costimulation. Front Immunol 2025; 15:1494206. [PMID: 39835115 PMCID: PMC11743637 DOI: 10.3389/fimmu.2024.1494206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
Background Immune checkpoint inhibitors have revolutionized cancer therapy, but many patients fail to respond or develop resistance, often due to reduced T cell activity. Costimulation via 4-1BB has emerged as a promising approach to enhance the effector function of antigen-primed T cells. Bispecific T cell-engaging (TCE) antibodies are an effective way to provide tumor-specific T cell receptor-mediated signaling to tumor-infiltrating lymphocytes. mRNA-based delivery of bispecific antibodies, offer a novel approach to enhance tumor-specific immune responses while minimizing adverse effects. Methods Two bispecific antibodies were generated: the EGFR x CD3 TCE antibody (LiTE) and the PD-L1 x 4-1BB costimulatory antibody (LiTCo), which was further fused to a high FcRn albumin variant (Albu-LiTCo). The mRNA encoding these bispecific antibodies contains an N1-methylpseudouridine modified nucleoside and regulatory sequences to ensure proper expression and stability. A series of in vitro assays and cell-based analyses were performed to characterize both antibodies. The in vivo efficacy of the mRNA-encoded bispecific antibodies was evaluated in xenograft tumor models expressing EGFR. Results We investigated the combined effect of two mRNA-encoded Fc-free bispecific antibodies with complementary mechanisms of action: an EGFR-targeting TCE and a half-life extended PD-L1 x 4-1BB costimulatory antibody. The mRNAs encoding both bispecific LiTERNA and Albu-LiTCoRNA, showed similar binding specificity and in vitro function to their protein analogues. Pharmacokinetic studies demonstrated sustained expression of both bispecific antibodies following intravenous administration of the mRNAs formulated using a polymer/lipid-based nanoparticle (LNP) but different pharmacokinetic profiles, shorter for the TCE and longer for the PD-L1 x 4-1BB. When administered as a mRNA-LNP combination (ComboRNA), the growth of EGFR-positive tumors in immunocompetent mice was significantly inhibited, resulting in tumor regression in 20% of cases with no associated toxicity. Histological analysis confirmed increased T cell infiltration in the tumors treated with LITERNA and ComboRNA. Repeated administration resulted in sustained production of bispecific antibodies with different exposure cycles and potent antitumor activity with a favorable safety profile. Conclusions These results highlight the potential of combining two mRNA-encoded bispecific antibodies with different mechanisms of action and programmable half-life for cancer immunotherapy.
Collapse
Affiliation(s)
- Oana Hangiu
- Department of Antibody Engineering, Leadartis SL, Tres Cantos, Madrid, Spain
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario12 de Octubre (H12O), Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Rocío Navarro
- Department of Antibody Engineering, Leadartis SL, Tres Cantos, Madrid, Spain
| | - Susana Frago
- Department of Antibody Engineering, Leadartis SL, Tres Cantos, Madrid, Spain
| | - Laura Rubio-Pérez
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario12 de Octubre (H12O), Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Antonio Tapia-Galisteo
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario12 de Octubre (H12O), Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Laura Díez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario12 de Octubre (H12O), Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marina Gómez-Rosel
- Department of Antibody Engineering, Leadartis SL, Tres Cantos, Madrid, Spain
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario12 de Octubre (H12O), Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Noelia Silva-Pilipich
- Division of DNA and RNA Medicine, CIMA Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA) and Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
| | | | - Cristian Smerdou
- Division of DNA and RNA Medicine, CIMA Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA) and Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
| | - Kenneth A. Howard
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute Hospital Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario12 de Octubre (H12O), Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marta Compte
- Department of Antibody Engineering, Leadartis SL, Tres Cantos, Madrid, Spain
| |
Collapse
|
3
|
Orhan A, Justesen TF, Raskov H, Qvortrup C, Gögenur I. Introducing Neoadjuvant Immunotherapy for Colorectal Cancer: Advancing the Frontier. Ann Surg 2025; 281:95-104. [PMID: 39005208 DOI: 10.1097/sla.0000000000006443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
OBJECTIVE To give surgeons a review of the current and future use of neoadjuvant immunotherapy in patients with localized colorectal cancer (CRC). BACKGROUND Immunotherapy has revolutionized the standard of care in oncology and improved survival outcomes in several cancers. However, the applicability of immunotherapy is still an ongoing challenge. Some cancer types are less responsive to immunotherapy, and the heterogeneity in responses within cancer types is poorly understood. Clinical characteristics of the patient, the timing of immunotherapy in relation to surgery, diversities in the immune responses, clonal heterogeneity, different features of the tumor microenvironment, and genetic alterations are some factors among many that may influence the efficacy of immunotherapy. RESULTS In this narrative review, we describe the major types of immunotherapy used to treat localized CRC. Furthermore, we discuss the prediction of response to immunotherapy in relation to biomarkers and radiologic assessment. Finally, we consider the future perspectives of clinical implications and response patterns, as well as the potential and challenges of neoadjuvant immunotherapy in localized CRC. CONCLUSIONS Establishing mismatch repair (MMR) status at the time of diagnosis is central to the potential use of neoadjuvant immunotherapy, in particular immune checkpoint inhibitors, in localized CRC. To date, efficacy is primarily seen in patients with deficient MMR status and polymerase epsilon mutations, although a small group of patients with proficient MMR does respond. In conclusion, neoadjuvant immunotherapy shows promising complete response rates, which may open a future avenue of an organ-sparing watch-and-wait approach for a group of patients.
Collapse
Affiliation(s)
- Adile Orhan
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Tobias F Justesen
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Hans Raskov
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Camilla Qvortrup
- Department of Clinical Oncology, Rigshospitalet, Copenhagen, Denmark
| | - Ismail Gögenur
- Department of Surgery, Center for Surgical Science, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Herrera M, Pretelli G, Desai J, Garralda E, Siu LL, Steiner TM, Au L. Bispecific antibodies: advancing precision oncology. Trends Cancer 2024; 10:893-919. [PMID: 39214782 DOI: 10.1016/j.trecan.2024.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/29/2024] [Accepted: 07/12/2024] [Indexed: 09/04/2024]
Abstract
Bispecific antibodies (bsAbs) are engineered molecules designed to target two different epitopes or antigens. The mechanism of action is determined by the bsAb molecular targets and structure (or format), which can be manipulated to create variable and novel functionalities, including linking immune cells with tumor cells, or dual signaling pathway blockade. Several bsAbs have already changed the treatment landscape of hematological malignancies and select solid cancers. However, the mechanisms of resistance to these agents are understudied and the management of toxicities remains challenging. Herein, we review the principles in bsAb engineering, current understanding of mechanisms of action and resistance, data for clinical application, and provide a perspective on ongoing challenges and future developments in this field.
Collapse
Affiliation(s)
- Mercedes Herrera
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Giulia Pretelli
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jayesh Desai
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Elena Garralda
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Thiago M Steiner
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Lewis Au
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
Fenis A, Demaria O, Gauthier L, Vivier E, Narni-Mancinelli E. New immune cell engagers for cancer immunotherapy. Nat Rev Immunol 2024; 24:471-486. [PMID: 38273127 DOI: 10.1038/s41577-023-00982-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 01/27/2024]
Abstract
There have been major advances in the immunotherapy of cancer in recent years, including the development of T cell engagers - antibodies engineered to redirect T cells to recognize and kill cancer cells - for the treatment of haematological malignancies. However, the field still faces several challenges to develop agents that are consistently effective in a majority of patients and cancer types, such as optimizing drug dose, overcoming treatment resistance and improving efficacy in solid tumours. A new generation of T cell-targeted molecules was developed to tackle these issues that are potentially more effective and safer. In addition, agents designed to engage the antitumour activities of other immune cells, including natural killer cells and myeloid cells, are showing promise and have the potential to treat a broader range of cancers.
Collapse
Affiliation(s)
- Aurore Fenis
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille Université, Centre National de la Recherche Scientifique, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Olivier Demaria
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Laurent Gauthier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille Université, Centre National de la Recherche Scientifique, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- Assistance Publique-Hôpitaux de Marseille, Hôpital de la Timone, Marseille Immunopôle, Marseille, France
| | - Emilie Narni-Mancinelli
- Aix Marseille Université, Centre National de la Recherche Scientifique, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
| |
Collapse
|
6
|
Rubio-Pérez L, Frago S, Compte M, Navarro R, Harwood SL, Lázaro-Gorines R, Gómez-Rosel M, Hangiu O, Silva-Pilipich N, Vanrell L, Smerdou C, Álvarez-Vallina L. Characterization of a Trispecific PD-L1 Blocking Antibody That Exhibits EGFR-Conditional 4-1BB Agonist Activity. Antibodies (Basel) 2024; 13:34. [PMID: 38804302 PMCID: PMC11130918 DOI: 10.3390/antib13020034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Immune checkpoint blockade has changed the treatment paradigm for advanced solid tumors, but the overall response rates are still limited. The combination of checkpoint blockade with anti-4-1BB antibodies to stimulate tumor-infiltrating T cells has shown anti-tumor activity in human trials. However, the further clinical development of these antibodies has been hampered by significant off-tumor toxicities. Here, we generated an anti-4-1BB/EGFR/PD-L1 trispecific antibody consisting of a triple-targeting tandem trimerbody (TT) fused to an engineered silent Fc region. This antibody (IgTT-4E1-S) was designed to combine the blockade of the PD-L1/PD-1 axis with conditional 4-1BB costimulation specifically confined to the tumor microenvironment (TME). The antibody demonstrated simultaneous binding to purified EGFR, PD-L1, and 4-1BB in solution, effective blockade of the PD-L1/PD1 interaction, and potent 4-1BB-mediated costimulation, but only in the presence of EGFR-expressing cells. These results demonstrate the feasibility of IgTT-4E1-S specifically blocking the PD-L1/PD-1 axis and inducing EGFR-conditional 4-1BB agonist activity.
Collapse
Affiliation(s)
- Laura Rubio-Pérez
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario12 de Octubre (H12O), 28041 Madrid, Spain; (L.R.-P.); (R.L.-G.); (M.G.-R.); (O.H.)
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), 28041 Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
- Chair for Immunology UFV/Merck, Universidad Francisco de Vitoria (UFV), Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Susana Frago
- Department of Antibody Engineering, Leadartis SL, QUBE Technology Park, Tres Cantos, 28760 Madrid, Spain; (S.F.); (M.C.); (R.N.)
| | - Marta Compte
- Department of Antibody Engineering, Leadartis SL, QUBE Technology Park, Tres Cantos, 28760 Madrid, Spain; (S.F.); (M.C.); (R.N.)
| | - Rocío Navarro
- Department of Antibody Engineering, Leadartis SL, QUBE Technology Park, Tres Cantos, 28760 Madrid, Spain; (S.F.); (M.C.); (R.N.)
| | - Seandean L. Harwood
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark;
| | - Rodrigo Lázaro-Gorines
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario12 de Octubre (H12O), 28041 Madrid, Spain; (L.R.-P.); (R.L.-G.); (M.G.-R.); (O.H.)
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), 28041 Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Marina Gómez-Rosel
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario12 de Octubre (H12O), 28041 Madrid, Spain; (L.R.-P.); (R.L.-G.); (M.G.-R.); (O.H.)
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), 28041 Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Oana Hangiu
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario12 de Octubre (H12O), 28041 Madrid, Spain; (L.R.-P.); (R.L.-G.); (M.G.-R.); (O.H.)
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Antibody Engineering, Leadartis SL, QUBE Technology Park, Tres Cantos, 28760 Madrid, Spain; (S.F.); (M.C.); (R.N.)
| | - Noelia Silva-Pilipich
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain; (N.S.-P.); (C.S.)
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| | - Lucía Vanrell
- Facultad de Ingeniería, Universidad ORT Uruguay, 11100 Montevideo, Uruguay;
- Nanogrow Biotech, Montevideo 11500, Uruguay
| | - Cristian Smerdou
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain; (N.S.-P.); (C.S.)
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario12 de Octubre (H12O), 28041 Madrid, Spain; (L.R.-P.); (R.L.-G.); (M.G.-R.); (O.H.)
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), 28041 Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
- Chair for Immunology UFV/Merck, Universidad Francisco de Vitoria (UFV), Pozuelo de Alarcón, 28223 Madrid, Spain
| |
Collapse
|
7
|
Tapia-Galisteo A, Sánchez-Rodríguez I, Narbona J, Iglesias-Hernández P, Aragón-García S, Jiménez-Reinoso A, Compte M, Khan S, Tsuda T, Chames P, Lacadena J, Álvarez-Vallina L, Sanz L. Combination of T cell-redirecting strategies with a bispecific antibody blocking TGF-β and PD-L1 enhances antitumor responses. Oncoimmunology 2024; 13:2338558. [PMID: 38623463 PMCID: PMC11018002 DOI: 10.1080/2162402x.2024.2338558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/30/2024] [Indexed: 04/17/2024] Open
Abstract
T cell-based immunotherapies for solid tumors have not achieved the clinical success observed in hematological malignancies, partially due to the immunosuppressive effect promoted by the tumor microenvironment, where PD-L1 and TGF-β play a pivotal role. However, durable responses to immune checkpoint inhibitors remain limited to a minority of patients, while TGF-β inhibitors have not reached the market yet. Here, we describe a bispecific antibody for dual blockade of PD-L1 and TFG-β, termed AxF (scFv)2, under the premise that combination with T cell redirecting strategies would improve clinical benefit. The AxF (scFv)2 antibody was well expressed in mammalian and yeast cells, bound both targets and inhibited dose-dependently the corresponding signaling pathways in luminescence-based cellular reporter systems. Moreover, combined treatment with trispecific T-cell engagers (TriTE) or CAR-T cells significantly boosted T cell activation status and cytotoxic response in breast, lung and colorectal (CRC) cancer models. Importantly, the combination of an EpCAMxCD3×EGFR TriTE with the AxF (scFv)2 delayed CRC tumor growth in vivo and significantly enhanced survival compared to monotherapy with the trispecific antibody. In summary, we demonstrated the feasibility of concomitant blockade of PD-L1 and TGF-β by a single molecule, as well as its therapeutic potential in combination with different T cell redirecting agents to overcome tumor microenvironment-mediated immunosuppression.
Collapse
Affiliation(s)
- Antonio Tapia-Galisteo
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Cancer Immunotherapy Unit (UNICA), Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-oncology and Immunotherapy Group, Biomedical Research Institute Hospital 12 de Octubre, Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Iñigo Sánchez-Rodríguez
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Javier Narbona
- Department of Biochemistry and Molecular Biology, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Patricia Iglesias-Hernández
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Saray Aragón-García
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Anaïs Jiménez-Reinoso
- Cancer Immunotherapy Unit (UNICA), Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-oncology and Immunotherapy Group, Biomedical Research Institute Hospital 12 de Octubre, Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marta Compte
- Department of Antibody Engineering, Leadartis SL, Madrid, Spain
| | - Shaukat Khan
- Nemours Children’s Health Delaware, Wilmington, Delaware, USA
| | - Takeshi Tsuda
- Nemours Children’s Health Delaware, Wilmington, Delaware, USA
| | - Patrick Chames
- Aix Marseille Univ, CNRS, INSERM, Institute Paoli-Calmettes, CRCM, Marseille, France
| | - Javier Lacadena
- Department of Biochemistry and Molecular Biology, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-oncology and Immunotherapy Group, Biomedical Research Institute Hospital 12 de Octubre, Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| |
Collapse
|
8
|
Madsen AV, Pedersen LE, Kristensen P, Goletz S. Design and engineering of bispecific antibodies: insights and practical considerations. Front Bioeng Biotechnol 2024; 12:1352014. [PMID: 38333084 PMCID: PMC10850309 DOI: 10.3389/fbioe.2024.1352014] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Bispecific antibodies (bsAbs) have attracted significant attention due to their dual binding activity, which permits simultaneous targeting of antigens and synergistic binding effects beyond what can be obtained even with combinations of conventional monospecific antibodies. Despite the tremendous therapeutic potential, the design and construction of bsAbs are often hampered by practical issues arising from the increased structural complexity as compared to conventional monospecific antibodies. The issues are diverse in nature, spanning from decreased biophysical stability from fusion of exogenous antigen-binding domains to antibody chain mispairing leading to formation of antibody-related impurities that are very difficult to remove. The added complexity requires judicious design considerations as well as extensive molecular engineering to ensure formation of high quality bsAbs with the intended mode of action and favorable drug-like qualities. In this review, we highlight and summarize some of the key considerations in design of bsAbs as well as state-of-the-art engineering principles that can be applied in efficient construction of bsAbs with diverse molecular formats.
Collapse
Affiliation(s)
- Andreas V. Madsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lasse E. Pedersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Peter Kristensen
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Steffen Goletz
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
9
|
Salek-Ardakani S, Zajonc DM, Croft M. Agonism of 4-1BB for immune therapy: a perspective on possibilities and complications. Front Immunol 2023; 14:1228486. [PMID: 37662949 PMCID: PMC10469789 DOI: 10.3389/fimmu.2023.1228486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/03/2023] [Indexed: 09/05/2023] Open
Abstract
Costimulatory receptors on immune cells represent attractive targets for immunotherapy given that these molecules can increase the frequency of individual protective immune cell populations and their longevity, as well as enhance various effector functions. 4-1BB, a member of the TNF receptor superfamily, also known as CD137 and TNFRSF9, is one such molecule that is inducible on several cell types, including T cells and NK cells. Preclinical studies in animal models have validated the notion that stimulating 4-1BB with agonist reagents or its natural ligand could be useful to augment conventional T cell and NK cell immunity to protect against tumor growth and against viral infection. Additionally, stimulating 4-1BB can enhance regulatory T cell function and might be useful in the right context for suppressing autoimmunity. Two human agonist antibodies to 4-1BB have been produced and tested in clinical trials for cancer, with variable results, leading to the production of a wealth of second-generation antibody constructs, including bi- and multi-specifics, with the hope of optimizing activity and selectivity. Here, we review the progress to date in agonism of 4-1BB, discuss the complications in targeting the immune system appropriately to elicit the desired activity, together with challenges in engineering agonists, and highlight the untapped potential of manipulating this molecule in infectious disease and autoimmunity.
Collapse
Affiliation(s)
| | - Dirk M. Zajonc
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Medicine, University of California (UC) San Diego, La Jolla, CA, United States
| |
Collapse
|
10
|
Tapia-Galisteo A, Compte M, Álvarez-Vallina L, Sanz L. When three is not a crowd: trispecific antibodies for enhanced cancer immunotherapy. Theranostics 2023; 13:1028-1041. [PMID: 36793863 PMCID: PMC9925307 DOI: 10.7150/thno.81494] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 12/31/2022] [Indexed: 02/04/2023] Open
Abstract
Despite the clinical success of the first bispecific antibody approved by the FDA against B cell malignancies (blinatumomab), many obstacles remain such as dosing, treatment resistance, and modest efficacy in solid tumors. To overcome these limitations, considerable efforts have been dedicated to the development of multispecific antibodies, opening up new avenues to address both the complex biology of cancer and the onset of anti-tumoral immune responses. Simultaneous targeting of two tumor-associated antigens is presumed to enhance cancer cell selectivity and reduce immune escape. Co-engagement of CD3, along with agonists of co-stimulatory molecules or antagonists of co-inhibitory immune checkpoint receptors in a single molecule, may revert T cell exhaustion. Similarly, targeting of two activating receptors in NK cells may improve their cytotoxic potency. And these are only examples of the potential of antibody-based molecular entities engaging three (or more) relevant targets. From the perspective of health care costs, multispecific antibodies are appealing, since a similar (or superior) therapeutic effect could be obtained with a single therapeutic agent as with a combination of different monoclonal antibodies. Despite challenges in production, multispecific antibodies are endowed with unprecedented properties, which may render them more potent biologics for cancer therapy.
Collapse
Affiliation(s)
- Antonio Tapia-Galisteo
- Immuno-oncology and Immunotherapy Group, Biomedical Research Institute Hospital 12 de Octubre, Madrid, Spain.,Cancer Immunotherapy Unit (UNICA), Hospital Universitario 12 de Octubre, Madrid, Spain.,H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marta Compte
- Department of Antibody Engineering, Leadartis S.L., Madrid, Spain
| | - Luis Álvarez-Vallina
- Immuno-oncology and Immunotherapy Group, Biomedical Research Institute Hospital 12 de Octubre, Madrid, Spain.,Cancer Immunotherapy Unit (UNICA), Hospital Universitario 12 de Octubre, Madrid, Spain.,H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| |
Collapse
|
11
|
Daniluk K, Lange A, Pruchniewski M, Małolepszy A, Sawosz E, Jaworski S. Delivery of Melittin as a Lytic Agent via Graphene Nanoparticles as Carriers to Breast Cancer Cells. J Funct Biomater 2022; 13:278. [PMID: 36547538 PMCID: PMC9787603 DOI: 10.3390/jfb13040278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/01/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Melittin, as an agent to lyse biological membranes, may be a promising therapeutic agent in the treatment of cancer. However, because of its nonspecific actions, there is a need to use a delivery method. The conducted research determined whether carbon nanoparticles, such as graphene and graphene oxide, could be carriers for melittin to breast cancer cells. The studies included the analysis of intracellular pH, the potential of cell membranes, the type of cellular transport, and the expression of receptor proteins. By measuring the particle size, zeta potential, and FT-IT analysis, we found that the investigated nanoparticles are connected by electrostatic interactions. The level of melittin encapsulation with graphene was 86%, while with graphene oxide it was 78%. A decrease in pHi was observed for all cell lines after administration of melittin and its complex with graphene. The decrease in membrane polarization was demonstrated for all lines treated with melittin and its complex with graphene and after exposure to the complex of melittin with graphene oxide for the MDA-MB-231 and HFFF2 lines. The results showed that the investigated melittin complexes and the melittin itself act differently on different cell lines (MDA-MB-231 and MCF-7). It has been shown that in MDA-MD-231 cells, melittin in a complex with graphene is transported to cells via caveolin-dependent endocytosis. On the other hand, the melittin-graphene oxide complex can reach breast cancer cells through various types of transport. Other differences in protein expression changes were also observed for tumor lines after exposure to melittin and complexes.
Collapse
Affiliation(s)
- Karolina Daniluk
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Agata Lange
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Michał Pruchniewski
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Artur Małolepszy
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, 00-654 Warsaw, Poland
| | - Ewa Sawosz
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Sławomir Jaworski
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| |
Collapse
|