1
|
Lodge J, Kajtar L, Duxbury R, Hall D, Burley GA, Cordy J, Yates JW, Rattray Z. Quantifying antibody binding: techniques and therapeutic implications. MAbs 2025; 17:2459795. [PMID: 39957177 PMCID: PMC11834528 DOI: 10.1080/19420862.2025.2459795] [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: 12/13/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/18/2025] Open
Abstract
The binding kinetics of an antibody for its target antigen represent key determinants of its biological function and success as a novel biotherapeutic. Defining these interactions and kinetics is critical for understanding the pharmacological and pharmacodynamic profiles of antibodies in therapeutic applications, with line of sight to clinical translation. In this review, we discuss the latest developments in approaches to measure and modulate antibody-antigen interactions, including antibody engineering, novel antibody formats, current, and emerging technologies for measuring antibody-antigen binding interactions, and emerging perspectives within the field. We also explore how emerging computational methods are set to become powerful tools for modeling antibody-binding interactions under physiologically relevant conditions. Finally, we consider the therapeutic implications of modulating target engagement in terms of pharmacodynamics and pharmacokinetics.
Collapse
Affiliation(s)
- James Lodge
- Large Molecule Discovery, GSK, Stevenage, UK
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Lewis Kajtar
- Large Molecule Discovery, GSK, Stevenage, UK
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Rachel Duxbury
- Large Molecule Discovery, GSK, Stevenage, UK
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - David Hall
- Large Molecule Discovery, GSK, Stevenage, UK
| | - Glenn A. Burley
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | | | | | - Zahra Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
2
|
Bielski P, Barczyński J, Mikitiuk M, Myrcha M, Rykała K, Boon L, Gąsior W, Hec-Gałązka A, Holak TA, Sitar T. The bispecific antibody targeting VISTA and PD-L1 shows enhanced tumor inhibitory activity in pancreatic, endometrial and breast cancers compared to mono- and combination immune checkpoint blockade. Front Immunol 2025; 16:1486799. [PMID: 40416959 PMCID: PMC12099300 DOI: 10.3389/fimmu.2025.1486799] [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: 08/26/2024] [Accepted: 03/25/2025] [Indexed: 05/27/2025] Open
Abstract
Background The introduction of checkpoint immunotherapeutic agents in the last decade has revolutionized cancer treatment. Although anti-PD-1, anti-PD-L1 and anti-CTLA4 are promising therapies, many patients fail to respond or relapse due to drug resistance potentially due to redundancy of immune checkpoints. One of the ways to improve the efficacy of this cancer treatment is to target two or even three immune checkpoints. To date, the benefit of combined anti-VISTA/anti-PD-L1 therapy has been confirmed, but no one has investigated the efficacy of blocking these negative immune checkpoints with a bispecific anti-VISTA/anti-PD-L1 antibody. Methods In this study, the bispecific antibodies (bsAbs) were produced in three formats: symmetric (IgG-HC-scFv), asymmetric (Fab-scFv-Fc(KIH)) and 2 x scFv. The binding and blocking properties of these bispecific antibodies (bsAbs) and their efficacy compared to monotherapy and combination therapy were then determined using endometrial (RL95-2), pancreatic (PANC-1) and breast (BT-20) cancer cell lines. Results The bsAbs generated in this study showed weaker binding properties to PD-1 and VISTA in ELISA (EC50) than the parent antibodies (atezolizumab and onvatilimab). Blockade of VISTA/VSIG-3 binding was also weaker with bsAbs compared to onvatilimab, but the ability to block the PD-1/PD-L1 pathway was slightly better than with atezolizumab. The Fc-based bsAbs showed statistically significant higher levels of lysis of endometrial, breast and pancreatic cancer cells. The symmetric bsAbs (IgG-HC-scFv) showed the most promising therapeutic potential. Higher levels of cancer cell lysis were associated with higher levels of pro-inflammatory cytokines. Both the asymmetric and symmetric bsAbs resulted in higher secretion levels of IFN-γ, TNFα and Granzyme B than anti-VISTA, anti-PD-L1 monotherapy and anti-VISTA/anti-PD-L1 combination therapy. Conclusion The high level of tumor cell lysis and increased expression of pro-inflammatory cytokines induced by the Fc-based bsAbs suggest a novel approach for the treatment of pancreatic, endometrial and breast cancer.
Collapse
Affiliation(s)
- Przemysław Bielski
- Recepton Sp. z o.o., Gdansk, Poland
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Krakow, Poland
- JJP Biologics Sp. z o.o., Warsaw, Poland
| | - Jan Barczyński
- Recepton Sp. z o.o., Gdansk, Poland
- JJP Biologics Sp. z o.o., Warsaw, Poland
| | - Michał Mikitiuk
- Recepton Sp. z o.o., Gdansk, Poland
- JJP Biologics Sp. z o.o., Warsaw, Poland
| | | | | | - Louis Boon
- JJP Biologics Sp. z o.o., Warsaw, Poland
| | | | | | | | - Tomasz Sitar
- Recepton Sp. z o.o., Gdansk, Poland
- JJP Biologics Sp. z o.o., Warsaw, Poland
| |
Collapse
|
3
|
Thakor AS. The Third Pillar of Precision Medicine - Precision Delivery. MedComm (Beijing) 2025; 6:e70200. [PMID: 40297244 PMCID: PMC12035764 DOI: 10.1002/mco2.70200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
Precision Medicine is thought of as having two main pillars: Precision Diagnosis and Precision Therapy. However, for Precision Medicine to reach its full potential, a third pillar is needed that we propose to call Precision Delivery. In the laboratory, many therapies show great efficacy when tested directly with target cells. However, upon clinical translation, they are often given via intravenous or oral administration, resulting in their systemic distribution. To ensure therapies reach target sites at the correct therapeutic levels, they are often given at higher concentrations. However, this can be associated with off-target effects, side-effects, and unwanted interactions. Delivery strategies can help mitigate this by "spatially re-coupling" therapies in vivo with target cells. This review explains the concept of Precision Delivery, which can be thought of as three interconnected, but independent, modules: targeted delivery, microenvironment modulation, and cellular interactions. While locoregional approaches directly deliver therapies into target tissues through endovascular, endoluminal, percutaneous, and implantation techniques, microenvironment modulation technologies facilitate the movement of therapies across biological barriers and through tissue matrices, so optimized therapies can reach and interact with target cells. We highlight new innovations driving advances in Precision Delivery, while also discussing the considerations and challenges that Precision Delivery faces as it becomes increasingly integrated into treatment workflows.
Collapse
Affiliation(s)
- Avnesh S. Thakor
- Department of RadiologyCenter for Interventional Radiology Innovation at Stanford (IRIS)School of MedicineStanford UniversityPalo AltoCaliforniaUSA
| |
Collapse
|
4
|
Saini S, Kumar Y. Structural and functional analysis of engineered antibodies for cancer immunotherapy: insights into protein compactness and solvent accessibility. J Biomol Struct Dyn 2025; 43:3859-3872. [PMID: 38173178 DOI: 10.1080/07391102.2023.2300129] [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: 08/14/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024]
Abstract
Antibodies are crucial tools in various biomedical applications, including immunotherapy. In this study, we focused on designing and engineering antibodies to enhance their structural dynamics and functional properties. By employing advanced computational techniques and experimental validation, we gained crucial insights into the impact of specific mutations on the engineered antibodies. This study investigates the design and engineering of antibodies to improve their structural dynamics and functional properties. Structural attributes, such as protein compactness and solvent accessibility, were assessed, revealing interesting trends in anti-CD3 and anti-HER2 antibodies. Mutations in CD3 antibodies resulted in a more stable conformation, while mutant HER2 antibodies exhibited altered interaction with the target. Analysis of secondary structure assignments demonstrated significant changes in the folding and stability of the mutant antibodies compared to the wild-type counterparts. The conformational landscape of the engineered antibodies was explored, providing insights into folding pathways and binding mechanisms. Overall, the current study highlights the significance of antibody design and engineering in modulating structural dynamics and functional properties. The findings contribute to developing improved immunotherapeutic strategies by optimising antibody-based therapeutics for targeted diseases with enhanced efficacy and precision.
Collapse
Affiliation(s)
- Samvedna Saini
- Department of Biological Sciences and Engineering (BSE), Netaji Subhas University of Technology (NSUT), New Delhi, India
| | - Yatender Kumar
- Department of Biological Sciences and Engineering (BSE), Netaji Subhas University of Technology (NSUT), New Delhi, India
| |
Collapse
|
5
|
Lizama-Muñoz A, Plaza-Diaz J. Bispecific Antibodies, Nanobodies and Extracellular Vesicles: Present and Future to Cancer Target Therapy. Biomolecules 2025; 15:639. [PMID: 40427532 PMCID: PMC12109199 DOI: 10.3390/biom15050639] [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: 04/01/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Cancer remains one of the leading causes of mortality worldwide, with a growing need for precise and effective treatments. Traditional therapies such as chemotherapy and radiotherapy have limitations, including off-target effects and drug resistance. In recent years, targeted therapies have emerged as promising alternatives, aiming to improve treatment specificity and reduce systemic toxicity. Among the most innovative approaches, bispecific antibodies, nanobodies, and extracellular vesicles offer distinct and complementary mechanisms for cancer therapy. Bispecific antibodies enhance immune responses and enable dual-targeting of cancer cells, nanobodies provide superior tumor penetration due to their small size, and extracellular vesicles present a novel platform for drug and RNA delivery. This work aims to review and analyze these three approaches, assessing their current applications, advantages, challenges, and future perspectives.
Collapse
Affiliation(s)
- Asier Lizama-Muñoz
- Department of Biochemistry, Molecular Biology and Immunology III, Faculty of Medicine, University of Granada, 18016 Granada, Spain;
- Clinical Analysis and Immunology Department, University Hospital Virgen de las Nieves, 18014 Granada, Spain
| | - Julio Plaza-Diaz
- ANUT-DSM (Alimentaciò, Nutrició Desenvolupament i Salut Mental), Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, 43201 Reus, Spain
- School of Health Sciences, Universidad Internacional de La Rioja, Avenida de la Paz 137, 26006 Logroño, Spain
| |
Collapse
|
6
|
Yan Y, Yuan J, Peng Y, Zhou C, Liu X, Sun L, Song Q. Bispecific antibodies combined with chemotherapy in solid tumor treatment, the path forward? Front Immunol 2025; 16:1568724. [PMID: 40352940 PMCID: PMC12061958 DOI: 10.3389/fimmu.2025.1568724] [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: 01/30/2025] [Accepted: 04/03/2025] [Indexed: 05/14/2025] Open
Abstract
Background Bispecific antibodies (bsAbs) introduced a novel strategy in anticancer therapy when chemotherapy alone could not meet life expectancy. Nonetheless, the efficacy of monotherapy was limited, and the safety profile of bsAbs combined with chemotherapy remained uncertain. Methods Literature retrieval was carried out through PubMed, Embase, and Cochrane from inception to January, 2025. Progression-free survival (PFS), overall survival (OS), and overall response rate (ORR), along with adverse effects (AEs), were utilized to assess the efficacy and safety. Publication bias was calculated using Funnel plots and Egger's test. Heterogeneity was examined through subgroup and sensitivity analyses. The protocol was preregistered in the International Prospective Register of Systematic Reviews (CRD42025633628). Results A total of 8 eligible clinical studies with 2,495 patients were included. Compared with chemotherapy alone, bsAb+chemotherapy exhibited positive outcomes in PFS (hazard ratio (HR): 0.52; 95% confidence interval (CI): 0.44-0.60; p<0.01), OS (HR: 0.67, 95% CI: 0.57-0.77; p<0.01), and ORR (HR: 0.31, 95% CI: 0.16-0.47; p<0.01). Subgroup analysis revealed that female patients, Asian patients, those under 65 years of age, and patients treated with IgG-like bsAb were more likely to benefit from the survival advantages of bsAb+chemotherapy. Despite the occurrence of leukopenia, metabolism-related, and skin-related AEs, RR of AEs in other systems showed no statistical significance. Conclusion BsAb+chemotherapy was superior to chemotherapy alone, especially in female patients, Asian patients, those under 65 years of age, and patients receiving IgG-like bsAb. Additionally, while the AEs associated with bsAb+chemotherapy are generally manageable, there is still room for improvement. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42025633628.
Collapse
Affiliation(s)
- Yici Yan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Jing Yuan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yanyang Peng
- School of Humanities and Management, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenxi Zhou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Xinbo Liu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Leitao Sun
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiaoling Song
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
7
|
Puttkammer JR, Barreto JN, Jensen CJ, Nedved AN, Wilson-Miller JL, Cole KC, Holmes LM, Kosobud AR, Kapoor P, Gertz MA, Dingli D, Gonsalves WI, Kumar SK, Hayman SR, Kourelis TV, Warsame R, Binder M, Cook J, Lin Y, Sandahl TB. Outpatient Management of Bispecific Related Toxicities: An Observational Study of Safety Outcomes and Resource Utilization. JCO Oncol Pract 2025:OP2400930. [PMID: 40233295 DOI: 10.1200/op-24-00930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/21/2025] [Accepted: 03/11/2025] [Indexed: 04/17/2025] Open
Abstract
PURPOSE Cytokine release syndrome (CRS) of any grade occurs in 56%-80% of bispecific antibodies (BsAbs) used in multiple myeloma (MM). Risk mitigation strategies are required to expedite escalation of care if toxicities develop. The rarity of grade 3 or 4 CRS compels protocolized outpatient management of BsAbs as outpatient practice may avoid hospital admissions and costly resource utilization without compromising safety. MATERIALS AND METHODS Patients with MM who received BsAb step-up dosing (SUD) from August 23, 2023, to March 29, 2024, were enrolled. Baseline demographics, patient outcomes, and CRS management were reviewed to assess the safety of outpatient practice. Resource utilization was also analyzed. RESULTS In this study, 34 patients received outpatient SUD of BsAbs (teclistamab n = 17, talequetamab n = 17) with 16 remaining outpatient throughout the entire SUD period. CRS occurred in seven patients who did not require hospitalization, demonstrating the safety of outpatient management of BsAb toxicity, regardless of low-grade CRS. CRS was observed in 24 patients (maximum grade 2) and immune effector cell-associated neurotoxicity syndrome in four patients (maximum grade 3). All patients with CRS received steroids, including 12 patients at home when instructed. Tocilizumab was given to 13 patients, accounting for 18 doses. If all doses of BsAbs and tocilizumab were given outpatient, the medication margin would be $115,004 in US dollars (USD). CONCLUSION An outpatient-based practice for BsAb administration demonstrated safety and cost savings. The description of our practice and results of this study provide valuable insights into the safety, feasibility, and resource stewardship of outpatient management of BsAbs. Future research should attempt to predict and stratify CRS risk to deliver a tailored supportive strategy and continued increase of outpatient management.
Collapse
Affiliation(s)
| | | | | | | | | | - Kristin C Cole
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Lucy M Holmes
- Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| | - Allison R Kosobud
- Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| | - Prashant Kapoor
- Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| | - Morie A Gertz
- Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| | - David Dingli
- Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| | - Wilson I Gonsalves
- Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| | - Shaji K Kumar
- Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| | - Suzanne R Hayman
- Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| | | | - Rahma Warsame
- Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| | - Moritz Binder
- Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| | - Joselle Cook
- Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| | - Yi Lin
- Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
8
|
Cao X, Li J, Liu S, Liu A, Zhang L, Chen F, Li Y, Ma H, Sun W, Ouyang S, Dai L, Liu J. Plasma IgG and IgM autoantibodies to COPT1 as potential biomarkers for detection of non-small cell lung cancer. Front Immunol 2025; 16:1455095. [PMID: 40292291 PMCID: PMC12021867 DOI: 10.3389/fimmu.2025.1455095] [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: 06/26/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Background Early diagnosis of lung cancer is crucial for improving patient outcomes. Autoantibodies against tumor-associated antigens (TAAs) found in the plasma can serve as biomarkers for lung cancer detection. Copper transporter 1 (COPT1) is abnormally expressed in several cancers including lung cancer. The purpose of this study is to explore the significance of anti-COPT1 autoantibodies in the clinical diagnosis of non-small cell lung cancer (NSCLC). Methods The expression level of COPT1 in NSCLC and normal tissues was analyzed based on TCGA and the Human Protein Atlas (HPA) database. Through enzyme-linked immunosorbent assay (ELISA), the expression levels of anti-COPT1 autoantibodies in plasma samples from normal controls (NC), patients with benign pulmonary nodules (BPN), and patients with NSCLC were detected in the discovery (89 NC and 89 NSCLC) and verification (321 NC, 321 BPN and 321 NSCLC) groups. The ELISA results were verified by western blotting and indirect immunofluorescence experiments. Results Based on HPA and TCGA databases, the mRNA and protein levels of COPT1 were higher in NSCLC tissues than in normal tissues. The levels of anti-COPT1-IgG and anti-COPT1-IgM autoantibodies were significantly higher in patients with NSCLC (P<0.05). Anti-COPT1-IgG and anti-COPT1-IgM could discriminate NSCLC from NC with area under the curve (AUC) values of 0.733 (95% CI: 0.694-0.771) and 0.679 (95% CI: 0.638-0.720), respectively. Additionally, the combination of anti-COPT1-IgG, anti-COPT1-IgM, and carcinoembryonic antigen (CEA) could enhance the efficacy of NSCLC diagnosis from BPN with increased AUC values. Conclusions Our study indicated the potential significance of anti-COPT1-IgG and anti-COPT1-IgM autoantibodies as novel biomarkers for the detection of NSCLC. Furthermore, the combination of anti-COPT1-IgG and anti-COPT1-IgM improved the diagnostic value.
Collapse
Affiliation(s)
- Xiaobin Cao
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
- Beijing Genomics Institution (BGI) College, Zhengzhou University, Zhengzhou, China
| | - Jing Li
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Siyu Liu
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
- Beijing Genomics Institution (BGI) College, Zhengzhou University, Zhengzhou, China
| | - Aichen Liu
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Lulu Zhang
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Fengqi Chen
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Yutong Li
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
- Beijing Genomics Institution (BGI) College, Zhengzhou University, Zhengzhou, China
| | - Hanke Ma
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Wenke Sun
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Songyun Ouyang
- Department of Respiratory and Sleep Medicine in the First Affiliated hospital, Zhengzhou University, Zhengzhou, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| | - Jingjing Liu
- Henan Institute of Medical and Pharmaceutical Sciences & Henan Key Medical Laboratory of Tumor Molecular Biomarkers, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Gao B, Sabnis R, Kotnis S, Feliciano S, Poling K, Mei T, Feng M, Das JK, Song J, Sun Q. Modular Platform for Efficient Assembly of Multifunctional Antibodies Using Orthogonal Protein-Protein Interactions. ACS APPLIED MATERIALS & INTERFACES 2025; 17:20685-20692. [PMID: 40159649 PMCID: PMC11986891 DOI: 10.1021/acsami.4c21958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025]
Abstract
Multifunctional antibodies, capable of simultaneously engaging multiple targets, are a unique class of antibodies that have sparked growing interest. Current approaches for making multifunctional antibodies, including chemical conjugation or genetic modifications, suffer from low product yield, complex structure design, and complicated manufacturing processes. In this study, we report a modular post-translational platform with highly specific protein-protein interactions for multifunctional antibody assembly and an elastin-like polypeptide (ELP) for easy purification. We generated and purified multifunctional antibodies with over 90% assembled scaffold and overall product purity. Additionally, we assembled antibodies with diverse applications, including detecting cancer, inhibiting cancer cell growth, and directing T cells to cancer cells for enhanced therapeutic efficacy. This platform offers high assembly efficiency, easy purification, and modularity for the redesign of antibody functions.
Collapse
Affiliation(s)
- Baizhen Gao
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Rushant Sabnis
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Siddhi Kotnis
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Sofia Feliciano
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Kyge Poling
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Tracy Mei
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Min Feng
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Jugal Kishore Das
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas 77807, United States
| | - Jianxun Song
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas 77807, United States
| | - Qing Sun
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
- Interdisciplinary
Graduate Program in Genetics and Genomics, Texas A&M University, College
Station, Texas 77843, United States
| |
Collapse
|
10
|
Rezazadeh‐Gavgani E, Majidazar R, Lotfinejad P, Kazemi T, Shamekh A. Immune Checkpoint Molecules: A Review on Pathways and Immunotherapy Implications. Immun Inflamm Dis 2025; 13:e70196. [PMID: 40243372 PMCID: PMC12004596 DOI: 10.1002/iid3.70196] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Today, treating cancer patients with monoclonal antibodies (mAbs), by targeting immune checkpoints, is one of the most outstanding immunotherapeutic methods. Immune checkpoints are special molecules having regulatory role in immune system responses. Once these molecules are presented on cancer cells, these cells will be capable of evading the immune system through their own specific pathways. This Evasion can be prevented by counterbalancing immune system responses with immune checkpoints related antibodies. AIMS The current study aimed to highlight immunotherapy and its methods, describe the immune checkpoints pathways, outline the immune checkpoint inhibitors (ICIs), and recent advances in this field, and sketch an outlook on the best treatment options for the most prevalent cancers. MATERIALS & METHODS This research implemented a narrative review method. A comprehensive literature review on the history, molecular and cellular biology, and the clinical aspects of immune checkpoint molecules was performed to illustrate the pathways involved in various cancers. Also, currently-available and future potential immunotherapies targeting these pathways were extracted from the searched studies. RESULTS The immune checkpoint family consists of many molecules, including CTLA-4, PD-1, PD-L1, LAG-3, TIM-3, and TIGIT. Attempts to modify these molecules in cancer treatment led to the development of therapeutic monoclonal antibodies. Most of these antibodies have entered clinical studies and some of them have been approved by the Food and Drug Administration (FDA) to be used in cancer patients' treatment plans. DISCUSSION With these novel treatments and the combination therapies they offer, there is also hope for better treatment outcomes for the previously untreatable metastatic cancers. In spite of the beneficial aspects of immune checkpoint therapy, similar to other treatments, they may cause side effects in some patients. Therefore, more studies are needed to reduce the probable side effects and uncover their underlying mechanism. CONCLUSION Based on the data shown in this review, there is still a lack of knowledge about the complete properties of ICIs and the possible combination therapies that we may be able to implement to achieve a better treatment response in cancer patients.
Collapse
Affiliation(s)
| | - Reza Majidazar
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Parisa Lotfinejad
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Tohid Kazemi
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Ali Shamekh
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Aging Research InstituteTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
11
|
Roussot N, Kaderbhai C, Ghiringhelli F. Targeting Immune Checkpoint Inhibitors for Non-Small-Cell Lung Cancer: Beyond PD-1/PD-L1 Monoclonal Antibodies. Cancers (Basel) 2025; 17:906. [PMID: 40075753 PMCID: PMC11898530 DOI: 10.3390/cancers17050906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Non-small-cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. Immunotherapy targeting the PD-1/PD-L1 axis has revolutionized treatment, providing durable responses in a subset of patients. However, with fewer than 50% of patients achieving significant benefits, there is a critical need to expand therapeutic strategies. This review explores emerging targets in immune checkpoint inhibition beyond PD-1/PD-L1, including CTLA-4, TIGIT, LAG-3, TIM-3, NKG2A, and CD39/CD73. We highlight the biological basis of CD8 T cell exhaustion in shaping the antitumor immune response. Novel therapeutic approaches targeting additional inhibitory receptors (IR) are discussed, with a focus on their distinct mechanisms of action and combinatory potential with existing therapies. Despite significant advancements, challenges remain in overcoming resistance mechanisms and optimizing patient selection. This review underscores the importance of dual checkpoint blockade and innovative bispecific antibody engineering to maximize therapeutic outcomes for NSCLC patients.
Collapse
Affiliation(s)
- Nicolas Roussot
- Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France; (C.K.); (F.G.)
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc, 21000 Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Team TIRECs: Therapies and Immune REsponse in CancerS, 21000 Dijon, France
- UFR Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
| | - Courèche Kaderbhai
- Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France; (C.K.); (F.G.)
| | - François Ghiringhelli
- Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France; (C.K.); (F.G.)
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc, 21000 Dijon, France
- Centre de Recherche INSERM LNC-UMR1231, Team TIRECs: Therapies and Immune REsponse in CancerS, 21000 Dijon, France
- UFR Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
| |
Collapse
|
12
|
Naatz LC, Dong S, Evavold B, Ye X, Chen M. Bispecific killer engager for targeted depletion of PD-1 positive lymphocytes: A new avenue for autoimmune disease treatment. Acta Pharm Sin B 2025; 15:1230-1241. [PMID: 40370541 PMCID: PMC12069113 DOI: 10.1016/j.apsb.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 05/16/2025] Open
Abstract
Bispecific killer cell engagers (BiKEs) are a powerful tool to incite the killing power of natural killer (NK) cells. Here, we posited that the BiKE technology could be utilized to deplete activated immune cells expressing programmed death-1 (PD-1+ cells), and hence treat autoimmune diseases since these cells drive the disorders. We designed and generated PD-1 BiKE that targets an activating NK cell receptor, CD16, and PD-1. PD-1 BiKE showed specific binding to PD-1+ cells and engaged CD16 simultaneously. PD-1 BiKE enhanced NK cell-mediated apoptosis and depletion of PD-1+ Raji cells, but not PD-1- Raji cells. Further, PD-1 BiKE induced apoptosis of primary PD-1+ T lymphocytes that are highly relevant to autoimmune disease progression. The BiKE depleted 42% of primary T cells that were stimulated in vitro. Importantly, those ablated primary T cells were activated cells. Meanwhile, naive T cells were spared by the BiKE treatment, supporting the crucial selectivity of PD-1 BiKE-directed cell depletion. Lastly, PD-1 BiKE is more effective than a conventional depleting antibody in the depletion of PD-1+ cells. The current work supports PD-1 BiKE is a selective, potent, and safe tool to deplete PD-1+ cells.
Collapse
Affiliation(s)
- Lauren C. Naatz
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Shuyun Dong
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Brian Evavold
- Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Xiangyang Ye
- Department of Pharmacotherapy, University of Utah, Salt Lake City, UT 84112, USA
| | - Mingnan Chen
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
13
|
Murphy MB, Vitale L, O'Neill T, Maurer DM, Malenchek L, Crocker A, Patterson C, Mills-Chen L, Saley V, Antczak NM, Boyer JM, McManus KM, Montanari NR, Hammond RA, Goldstein J, Thomas LJ, Keler T, Alvarado D. Dual Inhibition of Mast Cells and Thymic Stromal Lymphopoietin Using a Novel Bispecific Antibody, CDX-622. Allergy 2025. [PMID: 39976188 DOI: 10.1111/all.16509] [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: 09/26/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Mast cells (MCs) respond to an array of allergens that drive allergic and inflammatory diseases. Stem cell factor (SCF), the ligand for the receptor KIT, is required for MC survival and function. Thymic stromal lymphopoietin (TSLP) is an alarmin that promotes Type 2 inflammation in asthma and other inflammatory diseases. We describe CDX-622, a bispecific antibody (bsAb), that targets both SCF and TSLP to neutralize these distinct cytokines. METHODS The bsAb CDX-622 was developed from novel antagonist monoclonal antibodies (mAbs) to SCF (SCF-12) and TSLP (1D10). CDX-622 encodes the full-length 1D10 mAb and the single-chain variable fragment of SCF-12, linked to the C-terminus of the 1D10 heavy chain. CDX-622 was modified to prevent Fcγ receptor interactions and enhance FcRn binding. CDX-622 was tested using in vitro assays of MC and dendritic cell (DC) activation, an ex vivo human skin model, and in vivo studies in nonhuman primates. RESULTS Novel SCF and TSLP mAbs with neutralizing activity were generated. The bsAb CDX-622 potently inhibited SCF-driven MC degranulation and TSLP-mediated CCL17 release by DCs. In human skin samples treated with SCF and TSLP, CDX-622 markedly reduced proinflammatory, MC, and DC-related RNA signatures. Additionally, CDX-622 and SCF-12 mAb administered to cynomolgus macaques (Macaca fascicularis) had a profound effect on MCs without any observed toxicity. CONCLUSIONS CDX-622 is a potent inhibitor of MCs through the neutralization of SCF and effectively blocks Type 2 inflammatory responses driven by TSLP. Dual inhibition of these cytokines may lead to improved clinical outcomes in certain inflammatory disorders.
Collapse
Affiliation(s)
| | - Laura Vitale
- Celldex Therapeutics, Inc., Hampton, New Jersey, USA
| | | | | | | | | | | | | | | | | | - James M Boyer
- Celldex Therapeutics, Inc., Hampton, New Jersey, USA
| | | | | | | | | | | | - Tibor Keler
- Celldex Therapeutics, Inc., Hampton, New Jersey, USA
| | | |
Collapse
|
14
|
Marischen L, Fritsch J, Ilic J, Wahl L, Bertsch T, Knop S, Bold A. Two Are Better than One: The Bi-Specific Antibody Mosunetuzumab Reveals an Improved Immune Response of Vγ9Vδ2 T Cells Targeting CD20 in Malignant B Cells in Comparison to the Mono-Specific Antibody Obinutuzumab. Int J Mol Sci 2025; 26:1262. [PMID: 39941030 PMCID: PMC11818642 DOI: 10.3390/ijms26031262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
In treating cancer, immunotherapy has been established as a later-line treatment option in clinical practice. That includes stem cell transplantation, modified or activated immune cells, and antibodies directed against aberrant cells. As an unconventional immune cell subgroup, γδ T cells have been shown to provide effects against malignant cells. They exhibit an MHC-independent activation process, which could diminish graft-versus-host disease after an adoptive transfer of allogeneic cells. Over the last years, the efficacy of therapeutic antibodies has been improved. As a bi-specific antibody, mosunetuzumab binds to both CD3 and CD20, thereby providing close proximity between effector and target cells. Here, we set out to analyze the efficiency of γδ T cells' anti-tumor effects in combination with mosunetuzumab vs. the monoclonal anti-CD20 antibody obinutuzumab. Mosunetuzumab revealed improved responses of γδ T cells regarding their expression of IFN-γ and CD107a and their cytotoxicity towards malignant B cells from lymphoma B cell lines. In comparison to obinutuzumab, mosunetuzumab led to an equivalent or enhanced cytotoxicity against B cell lymphoma cell lines and primary patient samples, where this effect was even more prominent. In summary, we consider the combination of stimulated γδ T cells and mosunetuzumab to be a promising therapeutic approach for future clinical trials.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/pharmacology
- Antigens, CD20/immunology
- Cell Line, Tumor
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/pathology
- Interferon-gamma/metabolism
- Antibodies, Bispecific/pharmacology
- Antibodies, Bispecific/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/drug effects
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Lothar Marischen
- Department of Hematology and Medical Oncology, Paracelsus Medical University, 90419 Nuremberg, Germany
| | - Jürgen Fritsch
- Department of Infection Prevention and Infectious Diseases, University Hospital of Regensburg, 93053 Regensburg, Germany
| | - Jovana Ilic
- Department of Hematology and Medical Oncology, Paracelsus Medical University, 90419 Nuremberg, Germany
| | - Laura Wahl
- Department of Hematology and Medical Oncology, Paracelsus Medical University, 90419 Nuremberg, Germany
| | - Thomas Bertsch
- Institute for Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Paracelsus Medical University, 90419 Nuremberg, Germany
| | - Stefan Knop
- Department of Hematology and Medical Oncology, Paracelsus Medical University, 90419 Nuremberg, Germany
| | - Anna Bold
- Department of Hematology and Medical Oncology, Paracelsus Medical University, 90419 Nuremberg, Germany
| |
Collapse
|
15
|
Hushmandi K, Einollahi B, Lee EHC, Sakaizawa R, Glaviano A, Reiter RJ, Saadat SH, Farani MR, Huh YS, Aref AR, Salimimoghadam S, Kumar AP. Bispecific antibodies as powerful immunotherapeutic agents for urological cancers: Recent innovations based on preclinical and clinical evidence. Int J Biol Sci 2025; 21:1410-1435. [PMID: 39990653 PMCID: PMC11844292 DOI: 10.7150/ijbs.96155] [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: 03/10/2024] [Accepted: 08/25/2024] [Indexed: 02/25/2025] Open
Abstract
Conventional immunotherapy has emerged as a key option for cancer treatment. However, its efficacy has been limited in urological cancers, especially prostate cancer, because of the immunosuppressive tumor microenvironment (TME), difficulty in drug delivery, aberrant immune response, and damage to normal cells. Bispecific antibodies (BsAbs) are engineered proteins with two different antigen-binding domains, designed using different technologies and in various formats. BsAb-based tumor immunotherapy has yielded optimistic results in preclinical and clinical investigations of many tumor types, including urological cancers. However, a series of challenges, including tumor heterogeneity, TME, Ab immunogenicity, adverse effects, serum half-life, low response rates, and drug resistance, hamper the application of BsAbs. In this review, we provide insights into the most common BsAb platforms with different mechanisms of action, which are under preclinical and clinical research, along with ways to overcome the challenges in BsAb administration for treating urological cancer.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reo Sakaizawa
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, Texas USA
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon 22212, Republic of Korea
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon 22212, Republic of Korea
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX Inc., Boston, MA, USA
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
16
|
Kim HU, Kim YK. Bispecific antibodies and CLEM: an analytical approach to advanced cell imaging for therapeutic strategies. Appl Microsc 2025; 55:1. [PMID: 39828773 PMCID: PMC11743405 DOI: 10.1186/s42649-024-00106-y] [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: 10/30/2024] [Accepted: 12/15/2024] [Indexed: 01/22/2025] Open
Abstract
The development of bispecific antibodies (BsAbs) represents a significant advancement in therapeutic antibody design, enabling the simultaneous targeting of two different antigens. This dual-targeting capability enhances therapeutic efficacy, particularly in complex diseases like cancer, where tumor heterogeneity presents a significant challenge for traditional treatments. By bridging two distinct pathways, BsAbs can improve specificity and minimize off-target effects, making them invaluable in therapeutic contexts. Integrating advanced imaging techniques, particularly Correlative Light and Electron Microscopy (CLEM), offers a unique opportunity to visualize the dynamic interactions of BsAbs within cellular environments. CLEM combines the strengths of optical and electron microscopy, allowing researchers to observe real-time antibody-antigen interactions at nanoscale resolution. This synergy not only deepens our understanding of BsAbs' mechanisms of action but also provides critical insights into their spatial distribution, binding kinetics, and functional dynamics in live cells. In this review, the integration of BsAbs and CLEM paves the way for targeted therapeutic strategies, fostering the development of more effective treatments that can adapt to the complexities of disease pathology.
Collapse
Affiliation(s)
- Han-Ul Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Kangwon Center for Systems Imaging, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Young Kwan Kim
- Kangwon Center for Systems Imaging, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, Republic of Korea.
| |
Collapse
|
17
|
Wang M, Yu F, Zhang Y. Present and future of cancer nano-immunotherapy: opportunities, obstacles and challenges. Mol Cancer 2025; 24:26. [PMID: 39827147 PMCID: PMC11748575 DOI: 10.1186/s12943-024-02214-5] [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: 09/24/2024] [Accepted: 12/25/2024] [Indexed: 01/22/2025] Open
Abstract
Clinically, multimodal therapies are adopted worldwide for the management of cancer, which continues to be a leading cause of death. In recent years, immunotherapy has firmly established itself as a new paradigm in cancer care that activates the body's immune defense to cope with cancer. Immunotherapy has resulted in significant breakthroughs in the treatment of stubborn tumors, dramatically improving the clinical outcome of cancer patients. Multiple forms of cancer immunotherapy, including immune checkpoint inhibitors (ICIs), adoptive cell therapy and cancer vaccines, have become widely available. However, the effectiveness of these immunotherapies is not much satisfying. Many cancer patients do not respond to immunotherapy, and disease recurrence appears to be unavoidable because of the rapidly evolving resistance. Moreover, immunotherapies can give rise to severe off-target immune-related adverse events. Strategies to remove these hindrances mainly focus on the development of combinatorial therapies or the exploitation of novel immunotherapeutic mediations. Nanomaterials carrying anticancer agents to the target site are considered as practical approaches for cancer treatment. Nanomedicine combined with immunotherapies offers the possibility to potentiate systemic antitumor immunity and to facilitate selective cytotoxicity against cancer cells in an effective and safe manner. A myriad of nano-enabled cancer immunotherapies are currently under clinical investigation. Owing to gaps between preclinical and clinical studies, nano-immunotherapy faces multiple challenges, including the biosafety of nanomaterials and clinical trial design. In this review, we provide an overview of cancer immunotherapy and summarize the evidence indicating how nanomedicine-based approaches increase the efficacy of immunotherapies. We also discuss the key challenges that have emerged in the era of nanotechnology-based cancer immunotherapy. Taken together, combination nano-immunotherapy is drawing increasing attention, and it is anticipated that the combined treatment will achieve the desired success in clinical cancer therapy.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China.
| | - Fei Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China
| | - Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China
| |
Collapse
|
18
|
Bangolo A, Amoozgar B, Mansour C, Zhang L, Gill S, Ip A, Cho C. Comprehensive Review of Early and Late Toxicities in CAR T-Cell Therapy and Bispecific Antibody Treatments for Hematologic Malignancies. Cancers (Basel) 2025; 17:282. [DOI: 1.bangolo a, amoozgar b, mansour c, zhang l, gill s, ip a, cho c.comprehensive review of early and late toxicities in car t-cell therapy and bispecific antibody treatments for hematologic malignancies.cancers (basel).2025 jan 17;17(2):282.doi: 10.3390/cancers17020282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2025] Open
Abstract
Chimeric antigen receptor T-cell (or CAR-T) therapy and bispecific antibodies (BsAbs) have revolutionized the treatment of hematologic malignancies, offering new options for relapsed or refractory cases. However, these therapies carry risks of early complications, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), and delayed issues like graft-versus-host disease (GVHD), infections, and secondary cancers. Effective management requires early diagnosis using advanced biomarkers and imaging, along with prompt interventions involving immunosuppressants, corticosteroids, and cytokine inhibitors. A multidisciplinary approach is essential, integrating hematologists, oncologists, and infectious disease specialists, with emerging strategies like targeted biologics and personalized medicine showing promise in balancing efficacy with toxicity management. Ongoing research is critical to refine diagnostics and treatments, ensuring that these therapies not only extend survival but also improve patients’ quality of life. This review provides critical insights for healthcare professionals to quickly recognize and treat complications of CAR-T and BsAbs therapies. By focusing on early detection through biomarkers and imaging and outlining timely therapeutic interventions, it aims to equip the multidisciplinary care team with the knowledge necessary to manage the challenges of these advanced treatments effectively, ultimately optimizing patient outcomes.
Collapse
Affiliation(s)
- Ayrton Bangolo
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Behzad Amoozgar
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | | | - Lili Zhang
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Sarvarinder Gill
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Andrew Ip
- Division of Lymphoma, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Christina Cho
- Division of Stem Cell Transplant and Cellular Therapy, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| |
Collapse
|
19
|
Huang RR, Spliedt M, Kaufman T, Gorlatov S, Barat B, Shah K, Gill J, Stahl K, DiChiara J, Wang Q, Li JC, Alderson R, Moore PA, Brown JG, Tamura J, Zhang X, Bonvini E, Diedrich G. A Strategy for Simultaneous Engineering of Interspecies Cross-Reactivity, Thermostability, and Expression of a Bispecific 5T4 x CD3 DART ® Molecule for Treatment of Solid Tumors. Antibodies (Basel) 2025; 14:7. [PMID: 39846615 PMCID: PMC11755548 DOI: 10.3390/antib14010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/11/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025] Open
Abstract
Background: Bispecific antibodies represent a promising class of biologics for cancer treatment. However, their dual specificity and complex structure pose challenges in the engineering process, often resulting in molecules with good functional but poor physicochemical properties. Method: To overcome limitations in the properties of an anti-5T4 x anti-CD3 (α5T4 x αCD3) DART molecule, a phage-display method was developed, which succeeded in simultaneously engineering cross-reactivity to the cynomolgus 5T4 ortholog, improving thermostability and the elevating expression level. Results: This approach generated multiple DART molecules that exhibited significant improvements in all three properties. The lead DART molecule demonstrated potent in vitro and in vivo anti-tumor activity. Although its clearance in human FcRn-transgenic mice was comparable to that of the parental molecule, faster clearance was observed in cynomolgus monkeys. The lead α5T4 x αCD3 DART molecule displayed no evidence of off-target binding or polyspecificity, suggesting that the increased affinity for the target may account for its accelerated clearance in cynomolgus monkeys. Conclusions: This may reflect target-mediated drug disposition (TMDD), a potential limitation of targeting 5T4, despite its limited expression in healthy tissues.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Gundo Diedrich
- MacroGenics Inc., Rockville, MD 20850, USA; (M.S.); (T.K.); (S.G.); (B.B.); (K.S.); (J.G.); (K.S.); (J.D.); (Q.W.); (J.C.L.); (R.A.); (P.A.M.); (J.G.B.); (J.T.); (X.Z.); (E.B.)
| |
Collapse
|
20
|
Bangolo A, Amoozgar B, Mansour C, Zhang L, Gill S, Ip A, Cho C. Comprehensive Review of Early and Late Toxicities in CAR T-Cell Therapy and Bispecific Antibody Treatments for Hematologic Malignancies. Cancers (Basel) 2025; 17:282. [PMID: 39858064 PMCID: PMC11764151 DOI: 10.3390/cancers17020282] [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: 12/24/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Chimeric antigen receptor T-cell (or CAR-T) therapy and bispecific antibodies (BsAbs) have revolutionized the treatment of hematologic malignancies, offering new options for relapsed or refractory cases. However, these therapies carry risks of early complications, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), and delayed issues like graft-versus-host disease (GVHD), infections, and secondary cancers. Effective management requires early diagnosis using advanced biomarkers and imaging, along with prompt interventions involving immunosuppressants, corticosteroids, and cytokine inhibitors. A multidisciplinary approach is essential, integrating hematologists, oncologists, and infectious disease specialists, with emerging strategies like targeted biologics and personalized medicine showing promise in balancing efficacy with toxicity management. Ongoing research is critical to refine diagnostics and treatments, ensuring that these therapies not only extend survival but also improve patients' quality of life. This review provides critical insights for healthcare professionals to quickly recognize and treat complications of CAR-T and BsAbs therapies. By focusing on early detection through biomarkers and imaging and outlining timely therapeutic interventions, it aims to equip the multidisciplinary care team with the knowledge necessary to manage the challenges of these advanced treatments effectively, ultimately optimizing patient outcomes.
Collapse
Affiliation(s)
- Ayrton Bangolo
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA; (B.A.); (L.Z.); (S.G.)
| | - Behzad Amoozgar
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA; (B.A.); (L.Z.); (S.G.)
| | | | - Lili Zhang
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA; (B.A.); (L.Z.); (S.G.)
| | - Sarvarinder Gill
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA; (B.A.); (L.Z.); (S.G.)
| | - Andrew Ip
- Division of Lymphoma, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA;
| | - Christina Cho
- Division of Stem Cell Transplant and Cellular Therapy, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA;
| |
Collapse
|
21
|
Morabito F, Martino EA, Nizzoli ME, Talami A, Pozzi S, Martino M, Neri A, Gentile M. Comparative Analysis of Bispecific Antibodies and CAR T-Cell Therapy in Follicular Lymphoma. Eur J Haematol 2025; 114:4-16. [PMID: 39462177 PMCID: PMC11613673 DOI: 10.1111/ejh.14335] [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: 08/15/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024]
Abstract
The treatment landscape for relapsed/refractory follicular lymphoma (RR-FL) is marked by a pivotal debate between chimeric antigen receptor T-cell (CAR-T) therapy and bispecific antibodies (BsAbs). While both CAR-T therapy and BsAbs target similar immunobiology and molecular markers, their efficacy comparisons are hindered by the lack of direct clinical trial comparisons. Key trials, such as the ZUMA-5 study, underscore axicabtagene ciloleucel (axi-cel)'s efficacy in treating RR-FL, achieving a 79% complete response rate with a median duration of response exceeding 3 years. Similarly, lisocabtagene maraleucel (liso-cel) in the TRANSCEND FL study reports a 94% complete response rate, emphasizing robust outcomes in heavily pretreated patients. Among BsAbs, mosunetuzumab showed promise in the GO29781 trial, with a 62% overall response rate in heavily pretreated RR-FL patients. Thus, CAR-T therapy offers potential curative benefits with a single infusion. However, its efficacy is tempered by significant adverse events such as cytokine release syndrome (CRS), neurotoxicity, and cytopenias, requiring specialized management and patient monitoring. In contrast, BsAbs provide a more tolerable treatment option counterbalancing by lower response rates and frequent dosing requirements. Personalized treatment strategies are crucial because of these distinct efficacy and safety profiles. When considering cost-effectiveness, both therapies need to be evaluated in the context of their clinical outcomes and quality of life improvements. Cost-effectiveness considerations are essential; while CAR-T therapies incur higher initial costs, their potential for long-term remission may mitigate expenses associated with repeated treatments or hospitalizations. Future research into resistance mechanisms and optimal therapeutic sequencing will further refine RR-FL management strategies.
Collapse
Affiliation(s)
| | | | | | - Annalisa Talami
- Hematology UnitAzienda USL‐IRCSS di Reggio EmiliaReggio EmiliaItaly
| | - Stefano Pozzi
- Hematology UnitAzienda USL‐IRCSS di Reggio EmiliaReggio EmiliaItaly
| | - Massimo Martino
- Department of Hemato‐Oncology and Radiotherapy, Hematology and Stem Cell Transplantation and Cellular Therapies Unit (CTMO)Grande Ospedale Metropolitano “Bianchi‐Melacrino‐Morelli”Reggio CalabriaItaly
| | - Antonino Neri
- Scientific DirectorateAzienda USL‐IRCCS di Reggio EmiliaReggio EmiliaItaly
| | - Massimo Gentile
- Hematology Unit, Department of Onco‐HematologyAO of CosenzaCosenzaItaly
- Department of Pharmacy, Health and Nutritional ScienceUniversity of CalabriaRendeItaly
| |
Collapse
|
22
|
Fitriana W, Sakai T, Duan L, Hengphasatporn K, Shigeta Y, Mashima T, Uda T, Hifumi E, Hirota S. Experimental and Computational Studies on Domain-Swapped Structure Stabilization of an Antibody Light Chain by Disulfide Bond Introduction. J Med Chem 2024; 67:22313-22321. [PMID: 39656517 DOI: 10.1021/acs.jmedchem.4c02570] [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: 12/28/2024]
Abstract
Development of different platforms would be useful for designing functional antibodies to improve the efficiency of antibody-based drugs. Three-dimensional domain swapping (3D-DS) may occur in the variable region of antibody light chain #4C214A, and a pair of domain-swapped dimers may interact with each other to form a tetramer. In this study, to stabilize the 3D-DS dimer structure in #4C214A, Val2 in strand A (swapping region) and Thr97 in strand G were replaced with Cys residues, generating #4 V2C/T97C/C214A with a Cys2-Cys97 disulfide bond that cross-links strands A and G of different protomers. The #4 V2C/T97C/C214A tetramer did not dissociate into monomers at low protein concentration (6 μM); however, some of the tetramers were converted to monomers by disulfide bond reduction. Two-dimensional free energy profile analysis for the tetramerization of two 3D-DS dimers was performed by molecular dynamics simulation. These results show that disulfide bond introduction is useful for controlling the dimerization/dissociation of the variable region through 3D-DS.
Collapse
Affiliation(s)
- Wahyu Fitriana
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Takahiro Sakai
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Lian Duan
- Center for Computational Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
| | - Kowit Hengphasatporn
- Center for Computational Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Yasuteru Shigeta
- Center for Computational Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Tsuyoshi Mashima
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
- Medilux Research Center, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Taizo Uda
- Nanotechnology Laboratory, Institute of Systems, Information Technologies and Nanotechnologies (ISIT), 4-1 Kyudai-Shinmachi, Fukuoka 879-5593, Japan
| | - Emi Hifumi
- Institute for Research Management, Oita University, 700 Dannoharu, Oita-Shi, Oita 870-1192, Japan
| | - Shun Hirota
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| |
Collapse
|
23
|
Jureczek J, Kałwak K, Dzięgiel P. Antibody-Based Immunotherapies for the Treatment of Hematologic Malignancies. Cancers (Basel) 2024; 16:4181. [PMID: 39766080 PMCID: PMC11674729 DOI: 10.3390/cancers16244181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/05/2025] Open
Abstract
Despite the great advancements in treatment strategies for hematological malignancies (HMs) over the years, their effective treatment remains challenging. Conventional treatment strategies are burdened with several serious drawbacks limiting their effectiveness and safety. Improved understanding of tumor immunobiology has provided novel anti-cancer strategies targeting selected immune response components. Currently, immunotherapy is counted as the fourth pillar of oncological treatment (together with surgery, chemo- and radiotherapy) and is becoming standard in the treatment regimen, alone or in combination therapy. Several categories of immunotherapies have been developed and are currently being assessed in clinical trials for the treatment of blood cancers, including immune checkpoint inhibitors, antigen-targeted antibodies, antibody-drug conjugates, tumor vaccines, and adoptive cell therapies. However, monoclonal antibodies (mAbs) and their derivatives have achieved the most notable clinical outcome so far. Since the approval of rituximab for treating B-cell malignancies, the availability of mAbs against tumor-specific surface molecules for clinical use has flourished. Antibody-based therapy has become one of the most successful strategies for immunotherapeutic cancer treatment in the last few decades, and many mAbs have already been introduced into standard treatment protocols for some hematologic malignancies. To further increase the efficacy of mAbs, they can be conjugated to radioisotopes or cytostatic drugs, so-called antibody-drug conjugates. Moreover, with the growing recognition of T-cell immunity's role in cancer development, strategies aimed at enhancing T cell activation and inhibiting mechanisms that suppress T cell function are actively being developed. This review provides a comprehensive overview of the current status of immunotherapeutic strategies based on monoclonal antibodies and their derivatives, including antibody-drug conjugates, bispecific T-cell engagers, and checkpoint inhibitors, approved for the treatment of various HMs.
Collapse
Affiliation(s)
- Justyna Jureczek
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Krzysztof Kałwak
- Clinical Department of Paediatric Bone Marrow Transplantation, Oncology and Haematology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| |
Collapse
|
24
|
Mohamed A, Elhawi M, Trybula M, Elshawy M, Chakrabarti S, Selfridge E, Asa SL. Role of Bispecific Antibodies in Gastroenteropancreatic Neuroendocrine Neoplasms (GEP-NENs): Review of Literature. Clin Med Insights Oncol 2024; 18:11795549241285213. [PMID: 39651421 PMCID: PMC11624570 DOI: 10.1177/11795549241285213] [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: 09/25/2023] [Accepted: 09/01/2024] [Indexed: 12/11/2024] Open
Abstract
Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) are a heterogeneous group of neoplasms with an increasing incidence in the last few decades. Despite therapeutic advances in the management of GEP-NENs, resistance to many of these treatments has made their management a great challenge. One of the most recent advances in oncologic therapy is targeting multiple receptors simultaneously and engaging immune cells in the tumor microenvironment through bispecific antibodies (BsAbs). Since the FDA approval of the anti-CD3 × anti CD19 BsAb blinatumomab, for management of B-cell acute lymphoblastic leukemia, around a hundred different BsAbs have been developed and tested in various clinical trials. In this article, we review the current development of BsAbs developed or being currently tested for the management of GEP-NENs, their mechanism of action, current results from ongoing trials, toxicities, and upcoming trials.
Collapse
Affiliation(s)
- Amr Mohamed
- Division of Medical Oncology, Department of Medicine, University Hospitals, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Mai Elhawi
- Department of Clinical Oncology, Ain Shams University Hospital, Cairo, Egypt
| | - Marcus Trybula
- Division of Medical Oncology, Department of Medicine, University Hospitals, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Mohamed Elshawy
- Department of Surgery, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Sakti Chakrabarti
- Division of Medical Oncology, Department of Medicine, University Hospitals, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Eva Selfridge
- Division of Medical Oncology, Department of Medicine, University Hospitals, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Sylvia L Asa
- Department of Pathology, University Hospitals, Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
25
|
Föller S, Regett N, Lataster L, Radziwill G, Takors R. Optimum blue light exposure: a means to increase cell-specific productivity in Chinese hamster ovary cells. Appl Microbiol Biotechnol 2024; 108:530. [PMID: 39636393 PMCID: PMC11621146 DOI: 10.1007/s00253-024-13363-4] [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: 08/22/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
Research for biopharmaceutical production processes with mammalian cells steadily aims to enhance the cell-specific productivity as a means for optimizing total productivities of bioreactors. Whereas current technologies such as pH, temperature, and osmolality shift require modifications of the cultivation medium, the use of optogenetic switches in recombinant producer cells might be a promising contact-free alternative. However, the proper application of optogenetically engineered cells requires a detailed understanding of basic cellular responses of cells that do not yet contain the optogenetic switches. The knowhow of ideal light exposure to enable the optimum use of related approaches is missing so far. Consequently, the current study set out to find optimum conditions for IgG1 producing Chinese hamster ovary (CHO) cells which were exposed to blue LED light. Growth characteristics, cell-specific productivity using enzyme-linked immunosorbent assay, as well as cell cycle distribution using flow cytometry were analyzed. Whereas too harsh light exposure causes detrimental growth effects that could be compensated with antioxidants, a surprising boost of cell-specific productivity by 57% occurred at optimum high light doses. The increase coincided with an increased number of cells in the G1 phase of the cell cycle after 72 h of illumination. The results present a promising new approach to boost biopharmaceutical productivity of mammalian cells simply by proper light exposure without any further optogenetic engineering. KEY POINTS: • Blue LED light hinders growth in CHO DP-12 cells • Antioxidants protect to a certain degree from blue light effects • Illumination with blue LED light raises cell-specific productivity.
Collapse
Affiliation(s)
- Stefanie Föller
- Institute of Biochemical Engineering, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany.
| | - Niklas Regett
- Institute of Biochemical Engineering, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Levin Lataster
- Institute of Biology II, University of Freiburg, 79098, Freiburg, Germany
| | - Gerald Radziwill
- Institute of Biology II, University of Freiburg, 79098, Freiburg, Germany
| | - Ralf Takors
- Institute of Biochemical Engineering, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany.
| |
Collapse
|
26
|
Li CL, Ma XY, Yi P. Bispecific Antibodies, Immune Checkpoint Inhibitors, and Antibody-Drug Conjugates Directing Antitumor Immune Responses: Challenges and Prospects. Cell Biochem Funct 2024; 42:e70011. [PMID: 39463028 DOI: 10.1002/cbf.70011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/27/2024] [Accepted: 10/13/2024] [Indexed: 10/29/2024]
Abstract
Tumor immunotherapy includes bispecific antibodies (BsAbs), immune checkpoint inhibitors (ICIs), vaccines, and adoptive cell immunotherapy. BsAbs belong to the family of antibodies that can specifically target two or more different antigens and are a promising option for tumor immunotherapy. Immune checkpoints are antibodies targeting PD-1, PD-L1, and CTLA4 and have demonstrated remarkable therapeutic efficacy in the treatment of hematological and solid tumors, whose combination therapies have been shown to synergistically enhance the antitumor effects of BsAbs. In addition, the clinical efficacy of existing monoclonal antibodies targeting PD-1 (e.g., ipilimumab, nivolumab, pembrolizumab, and cemiplimab) and PD-L1 (e.g., atezolizumab, avelumab, and durvalumab) could also be enhanced by conjugation to small drugs as antibody-drug conjugates (ADCs). The development of truly effective therapies for patients with treatment-resistant cancers can be achieved by optimizing the various components of ADCs.
Collapse
Affiliation(s)
- Chen Lu Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Yuan Ma
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yi
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Ruan DY, Wei XL, Liu FR, Hu XC, Zhang J, Ji DM, Huang DZ, Zhao YQ, Pan HM, Liao WJ, Yang KY, Xu N, Lu XX, Chen YL, Zhang W, Zhou H, Zhao HY, Xu RH. The first-in-class bispecific antibody IBI318 (LY3434172) targeting PD-1 and PD-L1 in patients with advanced tumors: a phase Ia/Ib study. J Hematol Oncol 2024; 17:118. [PMID: 39614368 PMCID: PMC11606118 DOI: 10.1186/s13045-024-01644-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND There is an unmet clinical need to enhance the response rate and safety of anti-PD-1/PD-L1-based cancer immunotherapy (IO). Herein, we presented the clinical study of IBI318 (LY3434172), a first-in-class bispecific antibody (bsAb) targeting PD-1 and PD-L1, in patients with advanced tumors. METHODS In this open-label, multicenter Phase Ia/Ib study of IBI318, the Phase Ia involved dose escalation and a safety dose expansion, while the Phase Ib focused on preliminary safety and efficacy evaluation in non-small cell lung cancer (NSCLC) and nasopharyngeal carcinoma (NPC). In Phase Ia, patients with advanced tumors received IBI318 doses ranging from 0.3 to 1200 mg every two weeks (Q2W) to determine the recommended Phase 2 dose (RP2D). In Phase Ib, NSCLC or NPC patients from five cohorts with varying treatment histories received IBI318 at the RP2D. The primary endpoint was safety and the secondary endpoints included efficacy assessed by investigators according to RECIST v1.1, pharmacokinetics, immunogenicity, and pharmacodynamics. RESULTS From February 11, 2019, to January 25, 2022, a total of 103 eligible patients were enrolled (Phase Ia, n = 55; Phase Ib, n = 48). The median follow-up was 10.1 months (range 0.7-28.6). The RP2D was determined to be 300 mg Q2W. Treatment-related adverse events (TRAEs) of any grades occurred in 88 patients (85.4%), while 10 patients (9.7%) experienced grade ≥ 3 TRAEs. The objective response rate (ORR) was 15.5% and the disease control rate (DCR) was 49.5% in all patients. In Phase Ib, the confirmed ORR was 45.5% in treatment-naïve NSCLC patients and 30.0% in IO-naïve NPC patients who had failed or were intolerant to platinum-based treatments. CONCLUSIONS IBI318 demonstrated a favorable safety profile and preliminary efficacy in treatment-naïve NSCLC and IO-naïve NPC patients. Further clinical studies are needed to assess the full therapeutic potential of PD-1/PD-L1 dual inhibition with bsAbs.
Collapse
Affiliation(s)
- Dan-Yun Ruan
- Department of Clinical Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China
| | - Xiao-Li Wei
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China
| | - Fu-Rong Liu
- Department of Clinical Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China
| | - Xi-Chun Hu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Jian Zhang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Dong-Mei Ji
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Ding-Zhi Huang
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China
| | - Yan-Qiu Zhao
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, The Academy of Medical Science, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Hong-Min Pan
- Medical Oncology, Sir Run Run Shaw Hospital (SRRSH), affiliated with the Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Wang-Jun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Kun-Yu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiao-Xiao Lu
- Innovent Biologics (Suzhou) Co. Ltd, Suzhou, People's Republic of China
| | - Yu-Ling Chen
- Innovent Biologics (Suzhou) Co. Ltd, Suzhou, People's Republic of China
| | - Wen Zhang
- Innovent Biologics (Suzhou) Co. Ltd, Suzhou, People's Republic of China
| | - Hui Zhou
- Innovent Biologics (Suzhou) Co. Ltd, Suzhou, People's Republic of China
| | - Hong-Yun Zhao
- Department of Clinical Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China.
| | - Rui-Hua Xu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
28
|
Mussa A, Ismail NH, Hamid M, Al-Hatamleh MAI, Bragoli A, Hajissa K, Mokhtar NF, Mohamud R, Uskoković V, Hassan R. Understanding the role of TNFR2 signaling in the tumor microenvironment of breast cancer. J Exp Clin Cancer Res 2024; 43:312. [PMID: 39609700 PMCID: PMC11603874 DOI: 10.1186/s13046-024-03218-1] [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: 08/21/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed malignancy among women. It is characterized by a high level of heterogeneity that emerges from the interaction of several cellular and soluble components in the tumor microenvironment (TME), such as cytokines, tumor cells and tumor-associated immune cells. Tumor necrosis factor (TNF) receptor 2 (TNFR2) appears to play a significant role in microenvironmental regulation, tumor progression, immune evasion, drug resistance, and metastasis of many types of cancer, including BC. However, the significance of TNFR2 in BC biology is not fully understood. This review provides an overview of TNFR2 biology, detailing its activation and its interactions with important signaling pathways in the TME (e.g., NF-κB, MAPK, and PI3K/Akt pathways). We discuss potential therapeutic strategies targeting TNFR2, with the aim of enhancing the antitumor immune response to BC. This review provides insights into role of TNFR2 as a major immune checkpoint for the future treatment of patients with BC.
Collapse
Affiliation(s)
- Ali Mussa
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
- Department of Biology, Faculty of Education, Omdurman Islamic University, P.O. Box 382, Omdurman, Sudan
| | - Nor Hayati Ismail
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
| | - Mahasin Hamid
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Hunan Province, Changsha, 410013, China
- Department of Zoology, Faculty of Sciences and Information Technology, University of Nyala, Nyala, 63311, Sudan
| | - Mohammad A I Al-Hatamleh
- Division of Hematology and Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Anthony Bragoli
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Khalid Hajissa
- Department of Zoology, Faculty of Science and Technology, Omdurman Islamic University, P.O. Box 382, Omdurman, Sudan
| | - Noor Fatmawati Mokhtar
- Institute for Research in Molecular Medicine (iNFORMM), Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia.
| | - Vuk Uskoković
- TardigradeNano LLC, Irvine, CA, 92604, USA
- Division of Natural Sciences, Fullerton College, Fullerton, CA, 92832, USA
| | - Rosline Hassan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia.
| |
Collapse
|
29
|
Whitehead CA, Wines BD, Davies AM, McDonnell JM, Trist HM, Esparon SE, Hogarth PM. Stellabody: A novel hexamer-promoting mutation for improved IgG potency. Immunol Rev 2024; 328:438-455. [PMID: 39364646 PMCID: PMC11659935 DOI: 10.1111/imr.13400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
Advances in antibody engineering are being directed at the development of next generation immunotherapeutics with improved potency. Hexamerisation of IgG is a normal physiological aspect of IgG biology and recently described mutations that facilitate this process have a substantial impact upon monoclonal antibody behavior resulting in the elicitation of dramatically enhanced complement-dependent cytotoxicity, Fc receptor function, and enhanced antigen binding effects, such as targeted receptor agonism or microbe neutralization. Whereas the discovery of IgG hexamerisation enhancing mutations has largely focused on residues with exposure at the surface of the Fc-Fc and CH2-CH3 interfaces, our unique approach is the engineering of the mostly buried residue H429 in the CH3 domain. Selective substitution at position 429 forms the basis of Stellabody technology, where the choice of amino acid results in distinct hexamerisation outcomes. H429F results in monomeric IgG that hexamerises after target binding, so called "on-target" hexamerisation, while the H429Y mutant forms pH-sensitive hexamers in-solution prior to antigen binding. Moreover, Stellabody technologies are broadly applicable across the family of antibody-based biologic therapeutics, including conventional mAbs, bispecific mAbs, and Ig-like biologics such as Fc-fusions, with applications in diverse diseases.
Collapse
Affiliation(s)
- Clarissa A. Whitehead
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Bruce D. Wines
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Anna M. Davies
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's HouseLondonUK
| | - James M. McDonnell
- Randall Centre for Cell and Molecular Biophysics, King's College London, New Hunt's HouseLondonUK
| | - Halina M. Trist
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
| | | | - P. Mark Hogarth
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of Immunology, School of Translational MedicineMonash UniversityMelbourneVictoriaAustralia
- Department of Clinical PathologyThe University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
30
|
Choi SM, Lee JH, Ko S, Hong SS, Jin HE. Mechanism of Action and Pharmacokinetics of Approved Bispecific Antibodies. Biomol Ther (Seoul) 2024; 32:708-722. [PMID: 39448393 PMCID: PMC11535297 DOI: 10.4062/biomolther.2024.146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Bispecific antibodies represent a significant advancement in therapeutic antibody engineering, offering the ability to simultaneously target two distinct antigens. This dual-targeting capability enhances therapeutic efficacy, especially in complex diseases, such as cancer and autoimmune disorders, where drug resistance and incomplete target coverage are prevalent challenges. Bispecific antibodies facilitate immune cell engagement and disrupt multiple signaling pathways, providing a more comprehensive treatment approach than traditional monoclonal antibodies. However, the intricate structure of bispecific antibodies introduces unique pharmacokinetic challenges, including issues related to their absorption, distribution, metabolism, and excretion, which can significantly affect their efficacy and safety. This review provides an in-depth analysis of the structural design, mechanisms of action, and pharmacokinetics of the currently approved bispecific antibodies. It also highlights the engineering innovations that have been implemented to overcome these challenges, such as Fc modifications and advanced dimerization techniques, which enhance the stability and half-life of bispecific antibodies. Significant progress has been made in bispecific antibody technology; however, further research is necessary to broaden their clinical applications, enhance their safety profiles, and optimize their incorporation into combination therapies. Continuous advancements in this field are expected to enable bispecific antibodies to provide more precise and effective therapeutic strategies for a range of complex diseases, ultimately improving patient outcomes and advancing precision medicine.
Collapse
Affiliation(s)
- Seong Min Choi
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Ju-Hee Lee
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Soyeon Ko
- Department of Biomedical Sciences, College of Medicine & Program in Biomedicals Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine & Program in Biomedicals Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Hyo-Eon Jin
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
31
|
Yoshida J, Kato Y, Isogawa A, Tanaka Y, Kumagai I, Asano R, Nakanishi T, Makabe K. Construction of bispecific antibodies by specific pairing between the heavy chain and the light chain using removable SpyCatcher/SnoopCatcher units. J Biol Eng 2024; 18:57. [PMID: 39402666 PMCID: PMC11476941 DOI: 10.1186/s13036-024-00454-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
During the production of bispecific antibodies (bsAbs), nonspecific pairing results in low yields of target bsAb molecules, an issue known as the "mispairing problem." Several antibody engineering techniques have been developed to overcome mispairing issues. Here, we introduce "bsAb by external pairing and excision" (BAPE), a novel chain pairing method that induces specific chain pairing by fusing external SpyCatcher/Tag and SnoopCatcher/Tag units. These tags are then removed via protease cleavage. In this study, we applied this method to force the correct pairings of heavy and light chains while the heavy-chain pairing was achieved by the Knobs-into-Holes mutation. We then confirmed the formation of interchain bridges with covalent isopeptide bonds. Both anti-CD3/anti-Her2 and anti-CD3/anti-EGFR bsAbs displayed satisfactory target binding activities and in vitro cell-killing activity with activated T-cells.
Collapse
Affiliation(s)
- Jyunna Yoshida
- Graduate School of Science and Engineering, Yamagata University, 4-3-16 Jyonan, Yonezawa, Yamagata, 992-8510, Japan
| | - Yuki Kato
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Ai Isogawa
- Department of Chemistry and Bioengineering, Division of Science and Engineering for Materials, Chemistry and Biology, Graduate School of Engineering, Osaka Metropolitan University, Sugimoto 3-3-138, Sumiyoshi-ku, Osaka, 558-8585, Japan
| | - Yoshikazu Tanaka
- Graduate School of Life Sciences, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan
| | - Izumi Kumagai
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Ryutaro Asano
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Takeshi Nakanishi
- Department of Chemistry and Bioengineering, Division of Science and Engineering for Materials, Chemistry and Biology, Graduate School of Engineering, Osaka Metropolitan University, Sugimoto 3-3-138, Sumiyoshi-ku, Osaka, 558-8585, Japan
| | - Koki Makabe
- Graduate School of Science and Engineering, Yamagata University, 4-3-16 Jyonan, Yonezawa, Yamagata, 992-8510, Japan.
| |
Collapse
|
32
|
Toledo-Stuardo K, Ribeiro CH, González-Herrera F, Matthies DJ, Le Roy MS, Dietz-Vargas C, Latorre Y, Campos I, Guerra Y, Tello S, Vásquez-Sáez V, Novoa P, Fehring N, González M, Rodríguez-Siza J, Vásquez G, Méndez P, Altamirano C, Molina MC. Therapeutic antibodies in oncology: an immunopharmacological overview. Cancer Immunol Immunother 2024; 73:242. [PMID: 39358613 PMCID: PMC11448508 DOI: 10.1007/s00262-024-03814-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/16/2024] [Indexed: 10/04/2024]
Abstract
The biotechnological development of monoclonal antibodies and their immunotherapeutic use in oncology have grown exponentially in the last decade, becoming the first-line therapy for some types of cancer. Their mechanism of action is based on the ability to regulate the immune system or by interacting with targets that are either overexpressed in tumor cells, released into the extracellular milieu or involved in processes that favor tumor growth. In addition, the intrinsic characteristics of each subclass of antibodies provide specific effector functions against the tumor by activating antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, and antibody-dependent cellular phagocytosis, among other mechanisms. The rational design and engineering of monoclonal antibodies have improved their pharmacokinetic and pharmacodynamic features, thus optimizing the therapeutic regimens administered to cancer patients and improving their clinical outcomes. The selection of the immunoglobulin G subclass, modifications to its crystallizable region (Fc), and conjugation of radioactive substances or antineoplastic drugs may all improve the antitumor effects of therapeutic antibodies. This review aims to provide insights into the immunological and pharmacological aspects of therapeutic antibodies used in oncology, with a rational approach at molecular modifications that can be introduced into these biological tools, improving their efficacy in the treatment of cancer.
Collapse
Affiliation(s)
- Karen Toledo-Stuardo
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Carolina H Ribeiro
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Fabiola González-Herrera
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Douglas J Matthies
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - María Soledad Le Roy
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Claudio Dietz-Vargas
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Yesenia Latorre
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Ivo Campos
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Yuneisy Guerra
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Samantha Tello
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Valeria Vásquez-Sáez
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Pedro Novoa
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Nicolás Fehring
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Mauricio González
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Jose Rodríguez-Siza
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Gonzalo Vásquez
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Pamela Méndez
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile
| | - Claudia Altamirano
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
- Centro Regional de Estudio en Alimentos Saludables, Valparaíso, Chile
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy (IMPACT), Santiago, Chile
| | - María Carmen Molina
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Avda. Independencia 1027, Block I, 3er piso, Santiago, Chile.
| |
Collapse
|
33
|
Ho KW, Liu YL, Liao TY, Liu ES, Cheng TL. Strategies for Non-Covalent Attachment of Antibodies to PEGylated Nanoparticles for Targeted Drug Delivery. Int J Nanomedicine 2024; 19:10045-10064. [PMID: 39371476 PMCID: PMC11453133 DOI: 10.2147/ijn.s479270] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/05/2024] [Indexed: 10/08/2024] Open
Abstract
Polyethylene glycol (PEG)-modified nanoparticles (NPs) often struggle with reduced effectiveness against metastasis and liquid tumors due to limited tumor cell uptake and therapeutic efficacy. To address this, actively targeted liposomes with enhanced tumor selectivity and internalization are being developed to improve uptake and treatment outcomes. Using bi-functional proteins to functionalize PEGylated NPs and enhance targeted drug delivery through non-covalent attachment methods has emerged as a promising approach. Among these, the one-step and two-step targeting strategies stand out for their simplicity, efficiency, and versatility. The one-step strategy integrates streptavidin-tagged antibodies or bispecific antibodies (bsAbs: PEG/DIG × marker) directly into PEGylated NPs. This method uses the natural interactions between antibodies and PEG for stable, specific binding, allowing the modification of biotin/Fc-binding molecules like protein A, G, or anti-Fc peptide. Simply mixing bsAbs with PEGylated NPs improves tumor targeting and internalization. The two-step strategy involves first accumulating bsAbs (PEG/biotin × tumor marker) on the tumor cell surface, triggering an initial attack via antibody-dependent and complement-dependent cytotoxicity. These bsAbs then capture PEGylated NPs, initiating a second wave of internalization and cytotoxicity. Both strategies aim to enhance the targeting capabilities of PEGylated NPs by enabling specific recognition and binding to disease-specific markers or receptors. This review provides potential pathways for accelerating clinical translation in the development of targeted nanomedicine.
Collapse
Affiliation(s)
- Kai-Wen Ho
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yen-Ling Liu
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tzu-Yi Liao
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - En-Shuo Liu
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tian-Lu Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
34
|
Yoon C, Lee E, Kim D, Joung S, Kim Y, Jung H, Kim Y, Lee GM. SiMPl-GS: Advancing Cell Line Development via Synthetic Selection Marker for Next-Generation Biopharmaceutical Production. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405593. [PMID: 39105414 PMCID: PMC11481413 DOI: 10.1002/advs.202405593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/24/2024] [Indexed: 08/07/2024]
Abstract
Rapid and efficient cell line development (CLD) process is essential to expedite therapeutic protein development. However, the performance of widely used glutamine-based selection systems is limited by low selection efficiency, stringency, and the inability to select multiple genes. Therefore, an AND-gate synthetic selection system is rationally designed using split intein-mediated protein ligation of glutamine synthetase (GS) (SiMPl-GS). Split sites of the GS are selected using a computational approach and validated with GS-knockout Chinese hamster ovary cells for their potential to enable cell survival in a glutamine-free medium. In CLD, SiMPl-GS outperforms the wild-type GS by selectively enriching high producers. Unlike wild-type GS, SiMPl-GS results in cell pools in which most cells produce high levels of therapeutic proteins. Harnessing orthogonal split intein pairs further enables the selection of four plasmids with a single selection, streamlining multispecific antibody-producing CLD. Taken together, SiMPl-GS is a simple yet effective means to expedite CLD for therapeutic protein production.
Collapse
Affiliation(s)
- Chansik Yoon
- Department of Biological SciencesKAISTDaejeon34141Republic of Korea
| | - Eun‐ji Lee
- Biotherapeutics Translational Research CenterKRIBBDaejeon34113Republic of Korea
- Department of Bioprocess Engineering, KRIBB School of BiotechnologyUSTDaejeon34141Republic of Korea
| | - Dongil Kim
- Department of Biological SciencesKAISTDaejeon34141Republic of Korea
| | - Siyun Joung
- Department of Biological SciencesKAISTDaejeon34141Republic of Korea
| | - Yujin Kim
- Department of Biological SciencesKAISTDaejeon34141Republic of Korea
| | - Heungchae Jung
- Department of Bioprocess Engineering, KRIBB School of BiotechnologyUSTDaejeon34141Republic of Korea
- BIO CenterDaejeon TechnoparkDaejeon34054Republic of Korea
| | - Yeon‐Gu Kim
- Biotherapeutics Translational Research CenterKRIBBDaejeon34113Republic of Korea
- Department of Bioprocess Engineering, KRIBB School of BiotechnologyUSTDaejeon34141Republic of Korea
| | - Gyun Min Lee
- Department of Biological SciencesKAISTDaejeon34141Republic of Korea
| |
Collapse
|
35
|
Bartoszewska E, Tota M, Kisielewska M, Skowron I, Sebastianka K, Stefaniak O, Molik K, Rubin J, Kraska K, Choromańska A. Overcoming Antigen Escape and T-Cell Exhaustion in CAR-T Therapy for Leukemia. Cells 2024; 13:1596. [PMID: 39329777 PMCID: PMC11430486 DOI: 10.3390/cells13181596] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Leukemia is a prevalent pediatric cancer with significant challenges, particularly in relapsed or refractory cases. Chimeric antigen receptor T-cell (CAR-T) therapy has emerged as a personalized cancer treatment, modifying patients' T cells to target and destroy resistant cancer cells. This study reviews the current therapeutic options of CAR-T therapy for leukemia, addressing the primary obstacles such as antigen escape and T-cell exhaustion. We explore dual-targeting strategies and their potential to improve treatment outcomes by preventing the loss of target antigens. Additionally, we examine the mechanisms of T-cell exhaustion and strategies to enhance CAR-T persistence and effectiveness. Despite remarkable clinical successes, CAR-T therapy poses risks such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Our findings highlight the need for ongoing research to optimize CAR-T applications, reduce toxicities, and extend this innovative therapy to a broader range of hematologic malignancies. This comprehensive review aims to provide valuable insights for improving leukemia treatment and advancing the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Elżbieta Bartoszewska
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Maciej Tota
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Monika Kisielewska
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Izabela Skowron
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Kamil Sebastianka
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Oliwia Stefaniak
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Klaudia Molik
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Jakub Rubin
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Karolina Kraska
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland (M.K.); (I.S.); (K.S.); (O.S.); (K.M.); (J.R.); (K.K.)
- Student Research Group No K148, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
36
|
Tan X, Huang Z, Pei H, Jia Z, Zheng J. Molecular glue-mediated targeted protein degradation: A novel strategy in small-molecule drug development. iScience 2024; 27:110712. [PMID: 39297173 PMCID: PMC11409024 DOI: 10.1016/j.isci.2024.110712] [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] [Indexed: 09/21/2024] Open
Abstract
Small-molecule drugs are effective and thus most widely used. However, their applications are limited by their reliance on active high-affinity binding sites, restricting their target options. A breakthrough approach involves molecular glues, a novel class of small-molecule compounds capable of inducing protein-protein interactions (PPIs). This opens avenues to target conventionally undruggable proteins, overcoming limitations seen in conventional small-molecule drugs. Molecular glues play a key role in targeted protein degradation (TPD) techniques, including ubiquitin-proteasome system-based approaches such as proteolysis targeting chimeras (PROTACs) and molecular glue degraders and recently emergent lysosome system-based techniques like molecular degraders of extracellular proteins through the asialoglycoprotein receptors (MoDE-As) and macroautophagy degradation targeting chimeras (MADTACs). These techniques enable an innovative targeted degradation strategy for prolonged inhibition of pathology-associated proteins. This review provides an overview of them, emphasizing the clinical potential of molecular glues and guiding the development of molecular-glue-mediated TPD techniques.
Collapse
Affiliation(s)
- Xueqiang Tan
- Department of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Zuyi Huang
- Department of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Hairun Pei
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, Beijing Technology & Business University, Beijing 100048, China
| | - Zongchao Jia
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Jimin Zheng
- Department of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
37
|
Mandalawatta HP, Rajendra K, Fairfax K, Hewitt AW. Emerging trends in virus and virus-like particle gene therapy delivery to the brain. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102280. [PMID: 39206077 PMCID: PMC11350507 DOI: 10.1016/j.omtn.2024.102280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Recent advances in gene therapy and gene-editing techniques offer the very real potential for successful treatment of neurological diseases. However, drug delivery constraints continue to impede viable therapeutic interventions targeting the brain due to its anatomical complexity and highly restrictive microvasculature that is impervious to many molecules. Realizing the therapeutic potential of gene-based therapies requires robust encapsulation and safe and efficient delivery to the target cells. Although viral vectors have been widely used for targeted delivery of gene-based therapies, drawbacks such as host genome integration, prolonged expression, undesired off-target mutations, and immunogenicity have led to the development of alternative strategies. Engineered virus-like particles (eVLPs) are an emerging, promising platform that can be engineered to achieve neurotropism through pseudotyping. This review outlines strategies to improve eVLP neurotropism for therapeutic brain delivery of gene-editing agents.
Collapse
Affiliation(s)
| | - K.C. Rajendra
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Kirsten Fairfax
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Alex W. Hewitt
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
38
|
Dong W, Wang W, Cao C. The Evolution of Antibody-Drug Conjugates: Toward Accurate DAR and Multi-specificity. ChemMedChem 2024; 19:e202400109. [PMID: 38758596 DOI: 10.1002/cmdc.202400109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/18/2024]
Abstract
Antibody-drug conjugates (ADCs) consist of antibodies, linkers and payloads. They offer targeted delivery of potent cytotoxic drugs to tumor cells, minimizing off-target effects. However, the therapeutic efficacy of ADCs is compromised by heterogeneity in the drug-to-antibody ratio (DAR), which impacts both cytotoxicity and pharmacokinetics (PK). Additionally, the emergence of drug resistance poses significant challenges to the clinical advancement of ADCs. To overcome these limitations, a variety of strategies have been developed, including the design of multi-specific drugs with accurate DAR. This review critically summarizes the current challenges faced by ADCs, categorizing key issues and evaluating various innovative solutions. We provide an in-depth analysis of the latest methodologies for achieving homogeneous DAR and explore design strategies for multi-specific drugs aimed at combating drug resistance. Our discussion offers a current perspective on the advancements made in refining ADC technologies, with an emphasis on enhancing therapeutic outcomes.
Collapse
Affiliation(s)
- Wenge Dong
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Wanqi Wang
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Chan Cao
- State Key Laboratory of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
39
|
Raja A, Kasana A, Verma V. Next-Generation Therapeutic Antibodies for Cancer Treatment: Advancements, Applications, and Challenges. Mol Biotechnol 2024:10.1007/s12033-024-01270-y. [PMID: 39222285 DOI: 10.1007/s12033-024-01270-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
The field of cancer treatment has evolved significantly over the last decade with the emergence of next-generation therapeutic antibodies. Conventional treatments like chemotherapy pose significant challenges, including adverse side effects. Monoclonal antibodies have paved the way for more targeted and effective interventions. The evolution from chimeric to humanized and fully human antibodies has led to a reduction in immunogenicity and enhanced tolerance in vivo. The advent of next-generation antibodies, including bispecific antibodies, nanobodies, antibody-drug conjugates, glyco-engineered antibodies, and antibody fragments, represents a leap forward in cancer therapy. These innovations offer increased potency, adaptability, and reduced drug resistance. Challenges such as target validation, immunogenicity, and high production costs exist. However, technological advancements in antibody engineering techniques provide optimism for addressing these issues. The future promises a paradigm shift, where ongoing research will propel these powerful antibodies to the forefront, revolutionizing the fight against cancer and creating new preventive and curative treatments. This review provides an overview of three next-generation antibody-based molecules, namely bispecific antibodies, antibody-drug conjugates, and nanobodies that have shown promising results in cancer treatment. It discusses the evolution of antibodies from conventional forms to next-generation molecules, along with their applications in cancer treatment, production methods, and associated challenges. The review aims to offer researchers insights into the evolving landscape of next-generation antibody-based cancer therapeutics and their potential to revolutionize treatment strategies.
Collapse
Affiliation(s)
- Abhavya Raja
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, 201310, Uttar Pradesh, India
| | - Abhishek Kasana
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, 201310, Uttar Pradesh, India
| | - Vaishali Verma
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, 201310, Uttar Pradesh, India.
| |
Collapse
|
40
|
Arslan M, Asim M, Sattar H, Khan A, Thoppil Ali F, Zehra M, Talluri K. Role of Radiology in the Diagnosis and Treatment of Breast Cancer in Women: A Comprehensive Review. Cureus 2024; 16:e70097. [PMID: 39449897 PMCID: PMC11500669 DOI: 10.7759/cureus.70097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Breast cancer remains a leading cause of morbidity and mortality among women worldwide. Early detection and precise diagnosis are critical for effective treatment and improved patient outcomes. This review explores the evolving role of radiology in the diagnosis and treatment of breast cancer, highlighting advancements in imaging technologies and the integration of artificial intelligence (AI). Traditional imaging modalities such as mammography, ultrasound, and magnetic resonance imaging have been the cornerstone of breast cancer diagnostics, with each modality offering unique advantages. The advent of radiomics, which involves extracting quantitative data from medical images, has further augmented the diagnostic capabilities of these modalities. AI, particularly deep learning algorithms, has shown potential in improving diagnostic accuracy and reducing observer variability across imaging modalities. AI-driven tools are increasingly being integrated into clinical workflows to assist in image interpretation, lesion classification, and treatment planning. Additionally, radiology plays a crucial role in guiding treatment decisions, particularly in the context of image-guided radiotherapy and monitoring response to neoadjuvant chemotherapy. The review also discusses the emerging field of theranostics, where diagnostic imaging is combined with therapeutic interventions to provide personalized cancer care. Despite these advancements, challenges such as the need for large annotated datasets and the integration of AI into clinical practice remain. The review concludes that while the role of radiology in breast cancer management is rapidly evolving, further research is required to fully realize the potential of these technologies in improving patient outcomes.
Collapse
Affiliation(s)
| | - Muhammad Asim
- Emergency Medicine, Royal Free Hospital, London, GBR
| | - Hina Sattar
- Medicine, Dow University of Health Sciences, Karachi, PAK
| | - Anita Khan
- Medicine, Khyber Girls Medical College, Peshawar, PAK
| | | | - Muneeza Zehra
- Internal Medicine, Karachi Medical and Dental College, Karachi, PAK
| | - Keerthi Talluri
- General Medicine, GSL (Ganni Subba Lakshmi garu) Medical College, Rajahmundry, IND
| |
Collapse
|
41
|
Son A, Park J, Kim W, Lee W, Yoon Y, Ji J, Kim H. Integrating Computational Design and Experimental Approaches for Next-Generation Biologics. Biomolecules 2024; 14:1073. [PMID: 39334841 PMCID: PMC11430650 DOI: 10.3390/biom14091073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/13/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Therapeutic protein engineering has revolutionized medicine by enabling the development of highly specific and potent treatments for a wide range of diseases. This review examines recent advances in computational and experimental approaches for engineering improved protein therapeutics. Key areas of focus include antibody engineering, enzyme replacement therapies, and cytokine-based drugs. Computational methods like structure-based design, machine learning integration, and protein language models have dramatically enhanced our ability to predict protein properties and guide engineering efforts. Experimental techniques such as directed evolution and rational design approaches continue to evolve, with high-throughput methods accelerating the discovery process. Applications of these methods have led to breakthroughs in affinity maturation, bispecific antibodies, enzyme stability enhancement, and the development of conditionally active cytokines. Emerging approaches like intracellular protein delivery, stimulus-responsive proteins, and de novo designed therapeutic proteins offer exciting new possibilities. However, challenges remain in predicting in vivo behavior, scalable manufacturing, immunogenicity mitigation, and targeted delivery. Addressing these challenges will require continued integration of computational and experimental methods, as well as a deeper understanding of protein behavior in complex physiological environments. As the field advances, we can anticipate increasingly sophisticated and effective protein therapeutics for treating human diseases.
Collapse
Affiliation(s)
- Ahrum Son
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA;
| | - Jongham Park
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (W.L.); (Y.Y.)
| | - Woojin Kim
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (W.L.); (Y.Y.)
| | - Wonseok Lee
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (W.L.); (Y.Y.)
| | - Yoonki Yoon
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (W.L.); (Y.Y.)
| | - Jaeho Ji
- Department of Convergent Bioscience and Informatics, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea;
| | - Hyunsoo Kim
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; (J.P.); (W.K.); (W.L.); (Y.Y.)
- Department of Convergent Bioscience and Informatics, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea;
- Protein AI Design Institute, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
- SCICS (Sciences for Panomics), 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| |
Collapse
|
42
|
Qiang M, Liu H, Yang L, Wang H, Guo R. Immunotherapy for small cell lung cancer: the current state and future trajectories. Discov Oncol 2024; 15:355. [PMID: 39152301 PMCID: PMC11329494 DOI: 10.1007/s12672-024-01119-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/21/2024] [Indexed: 08/19/2024] Open
Abstract
Small cell lung cancer (SCLC) constitutes approximately 10% to 15% of all lung cancer diagnoses and represents a pressing global public health challenge due to its high mortality rates. The efficacy of conventional treatments for SCLC is suboptimal, characterized by limited anti-tumoral effects and frequent relapses. In this context, emerging research has pivoted towards immunotherapy combined with chemotherapy, a rapidly advancing field that has shown promise in ameliorating the clinical outcomes of SCLC patients. Through originally developed for non-small cell lung cancer (NSCLC), these therapies have extended new treatment avenues for SCLC. Currently, a nexus of emerging hot-spot treatments has demonstrated significant therapeutic efficacy. Based on the amalgamation of chemotherapy and immunotherapy, and the development of new immunotherapy agents, the treatment of SCLC has seen the hoping future. Progress has been achieved in enhancing the tumor immune microenvironment through the concomitant use of chemotherapy, immunotherapy, and tyrosine kinase inhibitors (TKI), as evinced by emerging clinical trial data. Moreover, a tripartite approach involving immunotherapy, targeted therapy, and chemotherapy appears auspicious for future clinical applications. Overcoming resistance to post-immunotherapy regimens remains an urgent area of exploration. Finally, bispecific antibodies, adoptive cell transfer (ACT), oncolytic virus, monotherapy, including Delta-like ligand 3 (DLL3) and T cell immunoreceptor with Ig and ITIM domains (TIGIT), as well as precision medicine, may present a prospective route towards achieving curative outcomes in SCLC. This review aims to synthesize extant literature and highlight future directions in SCLC treatment, acknowledging the persistent challenges in the field. Furthermore, the continual development of novel therapeutic agents and technologies renders the future of SCLC treatment increasingly optimistic.
Collapse
Affiliation(s)
- Min Qiang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hongyang Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lei Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Jilin University, Changchun, China.
| |
Collapse
|
43
|
Shao C, Tang B, Chu JCH, Lau KM, Wong WT, Che CM, Tai WCS, Wong WT, Wong CTT. Macrophage-engaging peptidic bispecific antibodies (pBsAbs) for immunotherapy via a facile bioconjugation strategy. Chem Sci 2024; 15:11272-11278. [PMID: 39055004 PMCID: PMC11268508 DOI: 10.1039/d4sc00851k] [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: 02/06/2024] [Accepted: 04/29/2024] [Indexed: 07/27/2024] Open
Abstract
Bispecific antibodies are artificial molecules that fuse two different antigen-binding sites of monoclonal antibodies into one single entity. They have emerged as a promising next-generation anticancer treatment. Despite the fascinating applications of bispecific antibodies, the design and production of bispecific antibodies remain tedious and challenging, leading to a long R&D process and high production costs. We herein report an unprecedented strategy to cyclise and conjugate tumour-targeting peptides on the surface of a monoclonal antibody to form a novel type of bispecific antibody, namely the peptidic bispecific antibody (pBsAb). Such design combines the merits of highly specific monoclonal antibodies and serum-stable cyclic peptides that endows an additional tumour-targeting ability to the monoclonal antibody for binding with two different antigens. Our results show that the novel pBsAb, which comprises EGFR-binding cyclic peptides and an anti-SIRP-α monoclonal antibody, could serve as a macrophage-engaging bispecific antibody to initiate enhanced macrophage-cancer cell interaction and block the "don't eat me" signal between CD47-SIRP-α, as well as promoting antibody-dependent cellular phagocytosis and 3D cell spheroid infiltration. These findings give rise to a new type of bispecific antibody and a new platform for the rapid generation of new bispecific antibodies for research and potential therapeutic uses.
Collapse
Affiliation(s)
- Chihao Shao
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| | - Bo Tang
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| | - Jacky C H Chu
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F, Building 17W, Hong Kong Science Park New Territories Hong Kong China
| | - Kwai Man Lau
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| | - Wai-Ting Wong
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| | - Chi-Ming Che
- Laboratory for Synthetic Chemistry and Chemical Biology Limited Units 1503-1511, 15/F, Building 17W, Hong Kong Science Park New Territories Hong Kong China
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, The University of Hong Kong Pokfulam Road Hong Kong China
| | - William C S Tai
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| | - Wing-Tak Wong
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| | - Clarence T T Wong
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University Kowloon Hong Kong China
| |
Collapse
|
44
|
Cai A, Chen Y, Wang LS, Cusick JK, Shi Y. Depicting Biomarkers for HER2-Inhibitor Resistance: Implication for Therapy in HER2-Positive Breast Cancer. Cancers (Basel) 2024; 16:2635. [PMID: 39123362 PMCID: PMC11311605 DOI: 10.3390/cancers16152635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
HER2 (human epidermal growth factor receptor 2) is highly expressed in a variety of cancers, including breast, lung, gastric, and pancreatic cancers. Its amplification is linked to poor clinical outcomes. At the genetic level, HER2 is encoded by the ERBB2 gene (v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2), which is frequently mutated or amplified in cancers, thus spurring extensive research into HER2 modulation and inhibition as viable anti-cancer strategies. An impressive body of FDA-approved drugs, including anti-HER2 monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), and HER2-tyrosine kinase inhibitors (TKIs), have demonstrated success in enhancing overall survival (OS) and disease progression-free survival (PFS). Yet, drug resistance remains a persistent challenge and raises the risks of metastatic potential and tumor relapse. Research into alternative therapeutic options for HER2+ breast cancer therefore proves critical for adapting to this ever-evolving landscape. This review highlights current HER2-targeted therapies, discusses predictive biomarkers for drug resistance, and introduces promising emergent therapies-especially combination therapies-that are aimed at overcoming drug resistance in the context of HER2+ breast cancer.
Collapse
Affiliation(s)
- Alvan Cai
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
| | - Yuan Chen
- Section Pathology of the Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany;
| | - Lily S. Wang
- University of California, Berkeley, CA 94720, USA;
| | - John K. Cusick
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
| | - Yihui Shi
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA; (A.C.); (J.K.C.)
- California Pacific Medical Center Research Institute, Sutter Bay Hospitals, San Francisco, CA 94107, USA
| |
Collapse
|
45
|
High P, Guernsey C, Subramanian S, Jacob J, Carmon KS. The Evolving Paradigm of Antibody-Drug Conjugates Targeting the ErbB/HER Family of Receptor Tyrosine Kinases. Pharmaceutics 2024; 16:890. [PMID: 39065587 PMCID: PMC11279420 DOI: 10.3390/pharmaceutics16070890] [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: 06/14/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024] Open
Abstract
Current therapies targeting the human epidermal growth factor receptor (HER) family, including monoclonal antibodies (mAbs) and tyrosine kinase inhibitors (TKIs), are limited by drug resistance and systemic toxicities. Antibody-drug conjugates (ADCs) are one of the most rapidly expanding classes of anti-cancer therapeutics with 13 presently approved by the FDA. Importantly, ADCs represent a promising therapeutic option with the potential to overcome traditional HER-targeted therapy resistance by delivering highly potent cytotoxins specifically to HER-overexpressing cancer cells and exerting both mAb- and payload-mediated antitumor efficacy. The clinical utility of HER-targeted ADCs is exemplified by the immense success of HER2-targeted ADCs including trastuzumab emtansine and trastuzumab deruxtecan. Still, strategies to improve upon existing HER2-targeted ADCs as well as the development of ADCs against other HER family members, particularly EGFR and HER3, are of great interest. To date, no HER4-targeting ADCs have been reported. In this review, we extensively detail clinical-stage EGFR-, HER2-, and HER3-targeting monospecific ADCs as well as novel clinical and pre-clinical bispecific ADCs (bsADCs) directed against this receptor family. We close by discussing nascent trends in the development of HER-targeting ADCs, including novel ADC payloads and HER ligand-targeted ADCs.
Collapse
Affiliation(s)
- Peyton High
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
| | - Cara Guernsey
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
| | - Shraddha Subramanian
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
| | - Joan Jacob
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
| | - Kendra S. Carmon
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (P.H.); (C.G.); (S.S.); (J.J.)
| |
Collapse
|
46
|
Saleh K, Khoury R, Khalife N, Chahine C, Ibrahim R, Tikriti Z, Le Cesne A. The Evolving Role of Bispecific Antibodies in Diffuse Large B-Cell Lymphoma. J Pers Med 2024; 14:666. [PMID: 39063920 PMCID: PMC11278258 DOI: 10.3390/jpm14070666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
The advent of targeted therapies such as monoclonal antibodies, adoptive T-cell therapies, and antibody-drug conjugates (ADCs) dramatically changed the treatment landscape of diffuse large B-cell lymphoma (DLBCL) over the last two decades. Rituximab was the first one approved. Chimeric antigen receptor T-cells are currently approved as second-line treatment in patients with DLBCL refractory to first-line chemo-immunotherapy. Polatuzumab, a CD79b-targeting ADC, is approved as first-line treatment in high-risk patients in combination with chemo-immunotherapy. Bispecific antibodies (BsAbs) are a novel category of drugs that are also changing the treatment paradigm of patients with DLBCL. They are engineered to bind to two different targets at the same time. To date, two BsAbs (glofitamab and epcoritamab) are approved as monotherapy in third-line treatment in DLBCL. Combination strategies with chemotherapy, immunotherapy, and ADCs are currently under investigation with encouraging results in first-line or subsequent lines of treatment. In the following review, we focus on the structure of BsAbs, the mechanism of action, clinical efficacy, and the mechanisms of resistance to BsAbs.
Collapse
Affiliation(s)
- Khalil Saleh
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (A.L.C.)
| | - Rita Khoury
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (A.L.C.)
| | - Nadine Khalife
- Department of Head and Neck Oncology, Gustave Roussy Cancer Campus, 94800 Villejuif, France;
| | - Claude Chahine
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (A.L.C.)
| | - Rebecca Ibrahim
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (A.L.C.)
| | - Zamzam Tikriti
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (A.L.C.)
| | - Axel Le Cesne
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France; (R.K.); (C.C.); (R.I.); (Z.T.); (A.L.C.)
| |
Collapse
|
47
|
Wang L, Jiang H, Yin X, Liang T, Li G, Ding C, Yang M, Zhang L, Liu J, Xu Y. PHE1-based IgG-like antibody platform provides a novel strategy for enhanced T-cell immunotherapy. Front Immunol 2024; 15:1415834. [PMID: 38933272 PMCID: PMC11201533 DOI: 10.3389/fimmu.2024.1415834] [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: 04/11/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Introduction Bispecific antibodies (BsAbs) can simultaneously target two epitopes of different antigenic targets, bringing possibilities for diversity in antibody drug design and are promising tools for the treatment of cancers and other diseases. T-cell engaging bsAb is an important application of the bispecific antibody, which could promote T cell-mediated tumor cell killing by targeting tumor-associated antigen (TAA) and CD3 at the same time. Methods This study comprised antibodies purification, Elisa assay for antigen binding, cytotoxicity assays, T cell activation by flow cytometry in vitro and xenogenic tumor model in vivo. Results We present a novel bsAb platform named PHE-Ig technique to promote cognate heavy chain (HC)-light chain (LC) pairing by replacing the CH1/CL regions of different monoclonal antibodies (mAbs) with the natural A and B chains of PHE1 fragment of Integrin β2 based on the knob-in-hole (KIH) technology. We had also verified that PHE-Ig technology can be effectively used as a platform to synthesize different desired bsAbs for T-cell immunotherapy. Especially, BCMA×CD3 PHE-Ig bsAbs exhibited robust anti-multiple myeloma (MM) activity in vitro and in vivo. Discussion Moreover, PHE1 domain was further shortened with D14G and R41S mutations, named PHE-S, and the PHE-S-based BCMA×CD3 bsAbs also showed anti BCMA+ tumor effect in vitro and in vivo, bringing more possibilities for the development and optimization of different bsAbs. To sum up, PHE1-based IgG-like antibody platform for bsAb construction provides a novel strategy for enhanced T-cell immunotherapy.
Collapse
Affiliation(s)
- Lingbin Wang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haojie Jiang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuying Yin
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Liang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoming Li
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Ding
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mina Yang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Zhang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Synvida Biotechnology Co., Ltd, Shanghai, China
| | - Yanyan Xu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
48
|
Lin CHT, Tariq MJ, Ullah F, Sannareddy A, Khalid F, Abbas H, Bader A, Samaras C, Valent J, Khouri J, Anwer F, Raza S, Dima D. Current Novel Targeted Therapeutic Strategies in Multiple Myeloma. Int J Mol Sci 2024; 25:6192. [PMID: 38892379 PMCID: PMC11172591 DOI: 10.3390/ijms25116192] [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: 04/27/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Multiple myeloma (MM) is a hematologic malignancy caused by the clonal expansion of immunoglobulin-producing plasma cells in the bone marrow and/or extramedullary sites. Common manifestations of MM include anemia, renal dysfunction, infection, bone pain, hypercalcemia, and fatigue. Despite numerous recent advancements in the MM treatment paradigm, current therapies demonstrate limited long-term effectiveness and eventual disease relapse remains exceedingly common. Myeloma cells often develop drug resistance through clonal evolution and alterations of cellular signaling pathways. Therefore, continued research of new targets in MM is crucial to circumvent cumulative drug resistance, overcome treatment-limiting toxicities, and improve outcomes in this incurable disease. This article provides a comprehensive overview of the landscape of novel treatments and emerging therapies for MM grouped by molecular target. Molecular targets outlined include BCMA, GPRC5D, FcRH5, CD38, SLAMF7, BCL-2, kinesin spindle protein, protein disulfide isomerase 1, peptidylprolyl isomerase A, Sec61 translocon, and cyclin-dependent kinase 6. Immunomodulatory drugs, NK cell therapy, and proteolysis-targeting chimera are described as well.
Collapse
Affiliation(s)
- Cindy Hsin-Ti Lin
- Department of Internal Medicine, Case Western Reserve University, MetroHealth Campus, Cleveland, OH 44109, USA
| | - Muhammad Junaid Tariq
- Department of Hematology-Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA;
| | - Fauzia Ullah
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | | | - Farhan Khalid
- Department of Internal Medicine, Monmouth Medical Center, Long Branch, NJ 07740, USA;
| | - Hasan Abbas
- Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Abbas Bader
- School of Medicine, University of Missouri–Kansas City, Kansas City, MO 64110, USA;
| | - Christy Samaras
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Jason Valent
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Jack Khouri
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Faiz Anwer
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Shahzad Raza
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Danai Dima
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
49
|
Braun A, Gouni S, Pulles A, Strati P, Minnema MC, Budde LE. Bispecific Antibody Use in Patients With Lymphoma and Multiple Myeloma. Am Soc Clin Oncol Educ Book 2024; 44:e433516. [PMID: 38935881 DOI: 10.1200/edbk_433516] [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: 06/29/2024]
Abstract
This article endeavors to navigate the clinical journey of bispecific antibodies (BsAbs), from elucidating common toxicities and management strategies to examining novel agents and broadening access in community health care. These drugs, commonly through T-cell activation, result in shared adverse events such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. Variations in target antigens and designs, however, might introduce unique toxicities for different BsAbs, warranting specific management approaches. Recent US Food and Drug Administration approvals of BsAbs targeting CD3+ T cells linked to CD20 for non-Hodgkin lymphoma and to B-cell maturation antigen or GPRC5D for multiple myeloma have transformed the treatment landscape for hematologic malignancies. Emerging new agents promise further enhancement and safety, exploring novel antigen targets, innovative structures such as trispecific antibodies, and the engagement of diverse immune cells. Simultaneously, the expansion of BsAbs into community practices is underway, demanding a multifaceted strategy that encompasses educational initiatives, operational adaptations, and collaborative frameworks. This ensures comprehensive treatment access, allowing every patient, irrespective of geographical or socioeconomic status, to benefit from these advancements in cancer therapy.
Collapse
Affiliation(s)
- Adam Braun
- City of Hope National Medical Center, Duarte, CA
| | | | - Astrid Pulles
- Department of Rheumatology & Clinical Immunology, Utrecht University, Utrecht, the Netherlands
| | - Paolo Strati
- MD Anderson Comprehensive Cancer Center, Houston, TX
| | - Monique C Minnema
- Department of Hematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | | |
Collapse
|
50
|
Asano R, Takeuchi M, Nakakido M, Ito S, Aikawa C, Yokoyama T, Senoo A, Ueno G, Nagatoishi S, Tanaka Y, Nakagawa I, Tsumoto K. Characterization of a novel format scFv×VHH single-chain biparatopic antibody against metal binding protein MtsA. Protein Sci 2024; 33:e5017. [PMID: 38747382 PMCID: PMC11094767 DOI: 10.1002/pro.5017] [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: 03/09/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/19/2024]
Abstract
Biparatopic antibodies (bpAbs) are engineered antibodies that bind to multiple different epitopes within the same antigens. bpAbs comprise diverse formats, including fragment-based formats, and choosing the appropriate molecular format for a desired function against a target molecule is a challenging task. Moreover, optimizing the design of constructs requires selecting appropriate antibody modalities and adjusting linker length for individual bpAbs. Therefore, it is crucial to understand the characteristics of bpAbs at the molecular level. In this study, we first obtained single-chain variable fragments and camelid heavy-chain variable domains targeting distinct epitopes of the metal binding protein MtsA and then developed a novel format single-chain bpAb connecting these fragment antibodies with various linkers. The physicochemical properties, binding activities, complex formation states with antigen, and functions of the bpAb were analyzed using multiple approaches. Notably, we found that the assembly state of the complexes was controlled by a linker and that longer linkers tended to form more compact complexes. These observations provide detailed molecular information that should be considered in the design of bpAbs.
Collapse
Affiliation(s)
- Risa Asano
- Department of BioengineeringSchool of Engineering, The University of TokyoTokyoJapan
| | - Miyu Takeuchi
- Department of BioengineeringSchool of Engineering, The University of TokyoTokyoJapan
| | - Makoto Nakakido
- Department of BioengineeringSchool of Engineering, The University of TokyoTokyoJapan
- Department of Chemistry and BiotechnologySchool of Engineering, The University of TokyoTokyoJapan
| | - Sho Ito
- Rigaku Corporation ROD Single Crystal Analysis Group Application LaboratoriesTokyoJapan
| | - Chihiro Aikawa
- Section of Applied Veterinary Sciences, Division of Veterinary Sciences, Department of Veterinary MedicineObihiro University of Agriculture and Veterinary MedicineHokkaidoJapan
| | - Takeshi Yokoyama
- Graduate School of Life Sciences, Tohoku UniversityMiyagiJapan
- The advanced center for innovations in next‐generation medicine (INGEM)Tohoku UniversityMiyagiJapan
| | - Akinobu Senoo
- Department of Chemistry and BiotechnologySchool of Engineering, The University of TokyoTokyoJapan
| | - Go Ueno
- RIKEN SPring‐8 CenterHyogoJapan
| | - Satoru Nagatoishi
- Medical Device Development and Regulation Research CenterSchool of Engineering, The University of TokyoTokyoJapan
| | - Yoshikazu Tanaka
- Graduate School of Life Sciences, Tohoku UniversityMiyagiJapan
- The advanced center for innovations in next‐generation medicine (INGEM)Tohoku UniversityMiyagiJapan
| | - Ichiro Nakagawa
- Department of MicrobiologyGraduate School of Medicine, Kyoto UniversityKyotoJapan
| | - Kouhei Tsumoto
- Department of BioengineeringSchool of Engineering, The University of TokyoTokyoJapan
- Department of Chemistry and BiotechnologySchool of Engineering, The University of TokyoTokyoJapan
- Medical Device Development and Regulation Research CenterSchool of Engineering, The University of TokyoTokyoJapan
- The Institute of Medical Science, The University of TokyoTokyoJapan
| |
Collapse
|