1
|
Wei X, Zhao L, Yang F, Yang Y, Zhang H, Du K, Tian X, Fan R, Si G, Wang K, Li Y, Wei Z, He M, Sui J. A CD25×TIGIT bispecific antibody induces anti-tumor activity through selective intratumoral Treg cell depletion. Mol Ther 2024; 32:4075-4094. [PMID: 39245938 DOI: 10.1016/j.ymthe.2024.09.010] [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: 12/13/2023] [Revised: 07/29/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024] Open
Abstract
Intratumoral regulatory T cells (Tregs) express high levels of CD25 and TIGIT, which are also recognized as markers of effector T cell (Teff) activation. Targeting these molecules each alone with monoclonal antibodies (mAbs) poses a risk of concurrently depleting both Teffs and peripheral Tregs, thereby compromising the effectiveness and selectivity of intratumoral Treg depletion. Here, leveraging the increased abundance of CD25+ TIGIT+ double-positive Tregs in the solid tumor microenvironment (but not in peripheral tissues), we explore the feasibility of using a CD25×TIGIT bispecific antibody (bsAb) to selectively deplete intratumoral Tregs. We initially constructed a bsAb co-targeting mouse CD25 and TIGIT, NSWm7210, and found that NSWm7210 conferred enhanced intratumoral Treg depletion, Teff activation, and tumor suppression as compared to the parental monotherapies in mouse models. We subsequently constructed a bsAb co-targeting human CD25 and TIGIT (NSWh7216), which preferentially eliminated CD25+ TIGIT+ double-positive cells over single-positive cells in vitro. NSWh7216 exhibited enhanced anti-tumor activity without toxicity of peripheral Tregs in CD25 humanized mice compared to the parental monotherapies. Our study illustrates the use of CD25×TIGIT bsAbs as effective agents against solid tumors based on selective depletion of intratumoral Tregs.
Collapse
Affiliation(s)
- Xin Wei
- School of Life Sciences, Beijing Normal University, Beijing 100875, China; National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China
| | - Linlin Zhao
- National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China
| | - Fang Yang
- National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China
| | - Yajing Yang
- National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China
| | - Huixiang Zhang
- National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China
| | - Kaixin Du
- National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China
| | - Xinxin Tian
- National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China
| | - Ruihua Fan
- National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China
| | - Guangxu Si
- National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China
| | - Kailun Wang
- National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China
| | - Yulu Li
- National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China
| | - Zhizhong Wei
- National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China
| | - Miaomiao He
- National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China
| | - Jianhua Sui
- National Institute of Biological Sciences, 7 Science Park Road, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China.
| |
Collapse
|
2
|
Shin JH, Oh S, Jang MH, Lee SY, Min C, Eu YJ, Begum H, Kim JC, Lee GR, Oh HB, Paul MJ, Ma JKC, Gwak HS, Youn H, Kim SR. Enhanced efficacy of glycoengineered rice cell-produced trastuzumab. PLANT BIOTECHNOLOGY JOURNAL 2024; 22:3068-3081. [PMID: 39016470 DOI: 10.1111/pbi.14429] [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: 01/24/2024] [Revised: 06/03/2024] [Accepted: 06/27/2024] [Indexed: 07/18/2024]
Abstract
For several decades, a plant-based expression system has been proposed as an alternative platform for the production of biopharmaceuticals including therapeutic monoclonal antibodies (mAbs), but the immunogenicity concerns associated with plant-specific N-glycans attached in plant-based biopharmaceuticals has not been completely solved. To eliminate all plant-specific N-glycan structure, eight genes involved in plant-specific N-glycosylation were mutated in rice (Oryza sativa) using the CRISPR/Cas9 system. The glycoengineered cell lines, PhytoRice®, contained a predominant GnGn (G0) glycoform. The gene for codon-optimized trastuzumab (TMab) was then introduced into PhytoRice® through Agrobacterium co-cultivation. Selected cell lines were suspension cultured, and TMab secreted from cells was purified from the cultured media. The amino acid sequence of the TMab produced by PhytoRice® (P-TMab) was identical to that of TMab. The inhibitory effect of P-TMab on the proliferation of the BT-474 cancer cell line was significantly enhanced at concentrations above 1 μg/mL (****P < 0.0001). P-TMab bound to a FcγRIIIa variant, FcγRIIIa-F158, more than 2.7 times more effectively than TMab. The ADCC efficacy of P-TMab against Jurkat cells was 2.6 times higher than that of TMab in an in vitro ADCC assay. Furthermore, P-TMab demonstrated efficient tumour uptake with less liver uptake compared to TMab in a xenograft assay using the BT-474 mouse model. These results suggest that the glycoengineered PhytoRice® could be an alternative platform for mAb production compared to current CHO cells, and P-TMab has a novel and enhanced efficacy compared to TMab.
Collapse
Affiliation(s)
- Jun-Hye Shin
- Department of Life Science, Sogang University, Seoul, South Korea
- PhytoMab Co. Ltd., Seoul, South Korea
| | - Sera Oh
- Department of Nuclear Medicine, Cancer Imaging Center, Seoul National University Hospital, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | | | - Seok-Yong Lee
- Department of Nuclear Medicine, Cancer Imaging Center, Seoul National University Hospital, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Chanhong Min
- Department of Chemistry, Sogang University, Seoul, South Korea
| | | | - Hilal Begum
- Department of Life Science, Sogang University, Seoul, South Korea
| | - Jong-Chan Kim
- Department of Life Science, Sogang University, Seoul, South Korea
| | - Gap Ryol Lee
- Department of Life Science, Sogang University, Seoul, South Korea
| | - Han-Bin Oh
- Department of Chemistry, Sogang University, Seoul, South Korea
| | - Matthew J Paul
- Hotung Molecular Immunology Unit, Institute for Infection and Immunity, St George's University of London, London, UK
| | - Julian K-C Ma
- Hotung Molecular Immunology Unit, Institute for Infection and Immunity, St George's University of London, London, UK
| | - Ho-Shin Gwak
- National Cancer Center Korea, Goyang-si, Kyunggi-do, South Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Cancer Imaging Center, Seoul National University Hospital, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Seong-Ryong Kim
- Department of Life Science, Sogang University, Seoul, South Korea
- PhytoMab Co. Ltd., Seoul, South Korea
| |
Collapse
|
3
|
Tirone B, Scarabosio A, Surico PL, Parodi PC, D’Esposito F, Avitabile A, Foti C, Gagliano C, Zeppieri M. Targeted Drug Delivery in Periorbital Non-Melanocytic Skin Malignancies. Bioengineering (Basel) 2024; 11:1029. [PMID: 39451404 PMCID: PMC11504966 DOI: 10.3390/bioengineering11101029] [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: 09/16/2024] [Revised: 10/05/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Targeted drug delivery has emerged as a transformative approach in the treatment of periorbital skin malignancies, offering the potential for enhanced efficacy and reduced side effects compared to traditional therapies. This review provides a comprehensive overview of targeted therapies in the context of periorbital malignancies, including basal cell carcinoma, squamous cell carcinoma, sebaceous gland carcinoma, and Merkel cell carcinoma. It explores the mechanisms of action for various targeted therapies, such as monoclonal antibodies, small molecule inhibitors, and immunotherapies, and their applications in treating these malignancies. Additionally, this review addresses the management of ocular and periocular side effects associated with these therapies, emphasizing the importance of a multidisciplinary approach to minimize impact and ensure patient adherence. By integrating current findings and discussing emerging trends, this review aims to highlight the advancements in targeted drug delivery and its potential to improve treatment outcomes and quality of life for patients with periorbital skin malignancies.
Collapse
Affiliation(s)
- Benedetta Tirone
- Dermatology and Venerology Section, Department of Precision and Regenerative Medicine and Ionan Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| | - Anna Scarabosio
- Clinic of Plastic and Reconstructive Surgery, Ospedale Santa Maria della Misericordia, 33100 Udine, Italy
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Pier Luigi Surico
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
- Department of Ophthalmology, Campus Bio-Medico University, 00128 Rome, Italy
| | - Pier Camillo Parodi
- Clinic of Plastic and Reconstructive Surgery, Ospedale Santa Maria della Misericordia, 33100 Udine, Italy
| | - Fabiana D’Esposito
- Imperial College Ophthalmic Research Group (ICORG) Unit, Imperial College, 153-173 Marylebone Rd, London NW15QH, UK
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Via Pansini 5, 80131 Napoli, Italy
| | - Alessandro Avitabile
- Eye Clinic Catania San Marco Hospital, Viale Carlo Azeglio Ciampi, 95121 Catania, Italy
| | - Caterina Foti
- Dermatology and Venerology Section, Department of Precision and Regenerative Medicine and Ionan Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| | - Caterina Gagliano
- Mediterranean Foundation “G.B. Morgagni”, 95125 Catania, Italy
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, p.le S. Maria della Misericordia 15, 33100 Udine, Italy
| |
Collapse
|
4
|
Robinson WH, Fiorentino D, Chung L, Moreland LW, Deodhar M, Harler MB, Saulsbery C, Kunder R. Cutting-edge approaches to B-cell depletion in autoimmune diseases. Front Immunol 2024; 15:1454747. [PMID: 39445025 PMCID: PMC11497632 DOI: 10.3389/fimmu.2024.1454747] [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: 06/25/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
B-cell depletion therapy (BCDT) has been employed to treat autoimmune disease for ~20 years. Immunoglobulin G1 (IgG1) monoclonal antibodies targeting CD20 and utilizing effector function (eg, antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, antibody-dependent cellular phagocytosis) to eliminate B cells have historically been the predominant therapeutic approaches. More recently, diverse BCDT approaches targeting a variety of B-cell surface antigens have been developed for use in hematologic malignancies, including effector-function-enhanced monoclonal antibodies, chimeric antigen receptor T-cell (CAR-T) treatment, and bispecific T-cell engagers (TCEs). The latter category of antibodies employs CD3 engagement to augment the killing of target cells. Given the improvement in B-cell depletion observed with CAR-T and TCEs compared with conventional monospecific antibodies for treatment of hematologic malignancies and the recent case reports demonstrating therapeutic benefit of CAR-T in autoimmune disease, there is potential for these mechanisms to be effective for B-cell-mediated autoimmune disease. In this review, we discuss the various BCDTs that are being developed in autoimmune diseases, describing the molecule designs, depletion mechanisms, and potential advantages and disadvantages of each approach as they pertain to safety, efficacy, and patient experience. Additionally, recent advances and strategies with TCEs are presented to help broaden understanding of the potential for bispecific antibodies to safely and effectively engage T cells for deep B-cell depletion in autoimmune diseases.
Collapse
Affiliation(s)
- William H. Robinson
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States
| | - David Fiorentino
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States
| | - Lorinda Chung
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States
- Palo Alto VA Health Care System, Palo Alto, CA, United States
| | - Larry W. Moreland
- Division of Rheumatology, School of Medicine, University of Colorado Anschutz, Aurora, CO, United States
| | | | | | | | | |
Collapse
|
5
|
Haryadi R, Chan KF, Lin PC, Tan YL, Wan C, Shahreel W, Tay SJ, Nguyen-Khuong T, Walsh I, Song Z. Generating and characterizing a comprehensive panel of CHO cells glycosylation mutants for advancing glycobiology and biotechnology research. Sci Rep 2024; 14:23068. [PMID: 39367021 PMCID: PMC11452509 DOI: 10.1038/s41598-024-73722-z] [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: 04/10/2024] [Accepted: 09/20/2024] [Indexed: 10/06/2024] Open
Abstract
This report describes the development and characterization of a comprehensive collection of CHO cell glycosylation mutants with significant potential for advancing glycobiology and biotechnology. EPO-Fc and trastuzumab, two model molecules, were produced using these mutants to assess the effects of mutated glycogenes, and LC-MS/MS analysis was employed to quantitatively analyse their N-glycans. EPO-Fc exhibited exclusively homogeneous Man9 glycans only when nearly all α-mannosidases in the genome were inactivated, except lysosomal MAN2B1. Some mutants lacking GnT-I activity produce mostly Man5 N-glycans, while their O-glycan and glycolipid profiles can differ due to other mutations in the cell. GnT-II deficiency prevents GnT-V from adding GlcNAc to the core N-glycan, resulting in branches attaching solely to the α1,3-linked mannose, leaving the α1,6-linked mannose free. The mutant-produced antibody's single-branched glycan contains more sialic acid than the dual-branched glycans produced in CHO-K1 cells. Trastuzumab produced in these mutants provided insights into how Fc N-glycans impact the antibody's interaction with FcγR1 and FcγR2a, FcγR3a, and their influence on antibody-dependent cellular cytotoxicity (ADCC). In the study of Fc glycans in Fc-FcγR1 and FcγR2a interactions, we observed a consistent glycan-related impact on binding to both receptors, indicating a common interaction mechanism between Fc glycans and both FcγRI and FcγRIIa. CHO mutants produced trimeric gp120 demonstrated distinct reactivity with multiple broadly neutralizing anti-HIV antibodies, confirming the involvement of gp120 glycans in interactions with specific broadly neutralizing antibodies. Finally, one of the mutants produced human β-glucocerebrosidase with uniform Man5 N-glycans, showcasing its potential for glycoengineered production and enhancement in therapeutic efficacy.
Collapse
Affiliation(s)
- Ryan Haryadi
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Kah Fai Chan
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Pao Chun Lin
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Yun Lei Tan
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Corrine Wan
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Wahyu Shahreel
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Shi Jie Tay
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Terry Nguyen-Khuong
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Ian Walsh
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01, Centros, 138668, Singapore
| | - Zhiwei Song
- Bioprocessing Technology Institute, A*STAR, 20 Biopolis Way, #06-01, Centros, 138668, Singapore.
| |
Collapse
|
6
|
Krištić J, Lauc G. The importance of IgG glycosylation-What did we learn after analyzing over 100,000 individuals. Immunol Rev 2024. [PMID: 39364834 DOI: 10.1111/imr.13407] [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] [Indexed: 10/05/2024]
Abstract
All four subclasses of immunoglobulin G (IgG) antibodies have glycan structures attached to the protein part of the IgG molecules. Glycans linked to the Fc portion of IgG are found in all IgG antibodies, while about one-fifth of IgG antibodies in plasma also have glycans attached to the Fab portion of IgG. The IgG3 subclass is characterized by more complex glycosylation compared to other IgG subclasses. In this review, we discuss the significant influence that glycans exert on the structural and functional properties of IgG. We provide a comprehensive overview of how the composition of these glycans can affect IgG's effector functions by modulating its interactions with Fcγ receptors and other molecules such as the C1q component of complement, which in turn influence various immune responses triggered by IgG, including antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). In addition, the importance of glycans for the efficacy of therapeutics like monoclonal antibodies and intravenous immunoglobulin (IVIg) therapy is discussed. Moreover, we offer insights into IgG glycosylation characteristics and roles derived from general population, disease-specific, and interventional studies. These studies indicate that IgG glycans are important biomarkers and functional effectors in health and disease.
Collapse
Affiliation(s)
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
7
|
Beyze A, Larroque C, Le Quintrec M. The role of antibody glycosylation in autoimmune and alloimmune kidney diseases. Nat Rev Nephrol 2024; 20:672-689. [PMID: 38961307 DOI: 10.1038/s41581-024-00850-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 07/05/2024]
Abstract
Immunoglobulin glycosylation is a pivotal mechanism that drives the diversification of antibody functions. The composition of the IgG glycome is influenced by environmental factors, genetic traits and inflammatory contexts. Differential IgG glycosylation has been shown to intricately modulate IgG effector functions and has a role in the initiation and progression of various diseases. Analysis of IgG glycosylation is therefore a promising tool for predicting disease severity. Several autoimmune and alloimmune disorders, including critical and potentially life-threatening conditions such as systemic lupus erythematosus, anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis and antibody-mediated kidney graft rejection, are driven by immunoglobulin. In certain IgG-driven kidney diseases, including primary membranous nephropathy, IgA nephropathy and lupus nephritis, particular glycome characteristics can enhance in situ complement activation and the recruitment of innate immune cells, resulting in more severe kidney damage. Hypofucosylation, hypogalactosylation and hyposialylation are the most common IgG glycosylation traits identified in these diseases. Modulating IgG glycosylation could therefore be a promising therapeutic strategy for regulating the immune mechanisms that underlie IgG-driven kidney diseases and potentially reduce the burden of immunosuppressive drugs in affected patients.
Collapse
Affiliation(s)
- Anaïs Beyze
- Institute of Regenerative Medicine and Biotherapy, IRMB U1183, Montpellier, France.
- Department of Nephrology, Dialysis and Transplantation, Montpellier University Hospital, Montpellier, France.
- University of Montpellier, Montpellier, France.
| | - Christian Larroque
- Institute of Regenerative Medicine and Biotherapy, IRMB U1183, Montpellier, France
- Department of Nephrology, Dialysis and Transplantation, Montpellier University Hospital, Montpellier, France
- University of Montpellier, Montpellier, France
| | - Moglie Le Quintrec
- Institute of Regenerative Medicine and Biotherapy, IRMB U1183, Montpellier, France.
- Department of Nephrology, Dialysis and Transplantation, Montpellier University Hospital, Montpellier, France.
- University of Montpellier, Montpellier, France.
| |
Collapse
|
8
|
McCraw AJ, Palhares LCGF, Hendel JL, Gardner RA, Santaolalla A, Crescioli S, McDonnell J, Van Hemelrijck M, Chenoweth A, Spencer DIR, Wagner GK, Karagiannis SN. IgE glycosylation and impact on structure and function: A systematic review. Allergy 2024; 79:2625-2661. [PMID: 39099223 DOI: 10.1111/all.16259] [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: 02/20/2024] [Revised: 06/20/2024] [Accepted: 07/21/2024] [Indexed: 08/06/2024]
Abstract
The impact of human IgE glycosylation on structure, function and disease mechanisms is not fully elucidated, and heterogeneity in different studies renders drawing conclusions challenging. Previous reviews discussed IgE glycosylation focusing on specific topics such as health versus disease, FcεR binding or impact on function. We present the first systematic review of human IgE glycosylation conducted utilizing the PRISMA guidelines. We sought to define the current consensus concerning the roles of glycosylation on structure, biology and disease. Despite diverse analytical methodologies, source, expression systems and the sparsity of data on IgE antibodies from non-allergic individuals, collectively evidence suggests differential glycosylation profiles, particularly in allergic diseases compared with healthy states, and indicates functional impact, and contributions to IgE-mediated hypersensitivities and atopic diseases. Beyond allergic diseases, dysregulated terminal glycan structures, including sialic acid, may regulate IgE metabolism. Glycan sites such as N394 may contribute to stabilizing IgE structure, with alterations in these glycans likely influencing both structure and IgE-FcεR interactions. This systematic review therefore highlights critical IgE glycosylation attributes in health and disease that may be exploitable for therapeutic intervention, and the need for novel analytics to explore pertinent research avenues.
Collapse
Affiliation(s)
- Alexandra J McCraw
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, Guy's Hospital, King's College London, London, UK
| | - Lais C G F Palhares
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, Guy's Hospital, King's College London, London, UK
| | - Jenifer L Hendel
- Department of Chemistry, Trent University, Peterborough, Ontario, Canada
| | | | - Aida Santaolalla
- Translational Oncology & Urology Research (TOUR), School of Cancer and Pharmaceutical Sciences, King's College, London, UK
| | - Silvia Crescioli
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, Guy's Hospital, King's College London, London, UK
| | - James McDonnell
- Randall Centre for Cell and Molecular Biophysics, School of Basic & Medical Biosciences, King's College London, London, UK
| | - Mieke Van Hemelrijck
- Translational Oncology & Urology Research (TOUR), School of Cancer and Pharmaceutical Sciences, King's College, London, UK
| | - Alicia Chenoweth
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, Guy's Hospital, King's College London, London, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, Guy's Cancer Centre, King's College London, London, UK
| | | | - Gerd K Wagner
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, Guy's Hospital, King's College London, London, UK
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, Belfast, UK
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, Guy's Hospital, King's College London, London, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, Guy's Cancer Centre, King's College London, London, UK
| |
Collapse
|
9
|
Mao C, Eberle K, Chen X, Zhou Y, Li J, Xin H, Gao W. FcRider: a recombinant Fc nanoparticle with endogenous adjuvant activities for hybrid immunization. Antib Ther 2024; 7:295-306. [PMID: 39381134 PMCID: PMC11456856 DOI: 10.1093/abt/tbae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/15/2024] [Accepted: 08/21/2024] [Indexed: 10/10/2024] Open
Abstract
Active immunization (vaccination) induces long-lasting immunity with memory, which takes weeks to months to develop. Passive immunization (transfer of neutralizing antibodies) provides immediate protection, yet with high cost and effects being comparatively short-lived. No currently approved adjuvants are compatible with formulations to combine active and passive immunizations, not to mention their huge disparities in administration routes and dosage. To solve this, we engineered the Fc fragment of human IgG1 into a hexamer nanoparticle and expressed its afucosylated form in Fut8-/- CHO cells, naming it "FcRider." FcRider is highly soluble with long-term stability, easily produced at high levels equivalent to those of therapeutic antibodies, and is amenable to conventional antibody purification schemes. Most importantly, FcRider possesses endogenous adjuvant activities. Using SWHEL B cell receptor (BCR) transgenic mice, we found that HEL-FcRider induced GL7+ germinal center B cells and HEL-specific IgG. Similarly, immunizing mice with UFO-BG-FcRider, a fusion containing the stabilized human immunodeficiency virus-1 (HIV-1) Env protein as immunogen, promoted somatic hypermutation and generation of long CDR3 of the IgG heavy chains. Intramuscular injection of (Fba + Met6)3-FcRider, a fusion with two peptide epitopes from Candida albicans cell surface, stimulated strong antigen-specific IgG titers. In three different models, we showed that afucosylated FcRider functions as a multivalent immunogen displayer and stimulates antigen-specific B cells without any exogenous adjuvant. As an antibody derivative, afucosylated FcRider could be a novel platform combining vaccines and therapeutic antibodies, integrating active and passive immunizations into single-modality "hybrid immunization" to provide complete and long-lasting protection against infections, and may open new avenues in cancer immunotherapy as well.
Collapse
Affiliation(s)
- Changchuin Mao
- Antagen Pharmaceuticals, Inc., Canton, MA 02021, United States
| | - Karen Eberle
- Department of Microbiology and Immunology, Louisiana State University Health Science Center, New Orleans, LA 70112, United States
| | - Xiaojie Chen
- Base&Byte Biotechnology Co., Ltd., Changping District, Beijing 102206, PR China
| | - Yiming Zhou
- Base&Byte Biotechnology Co., Ltd., Changping District, Beijing 102206, PR China
| | - Jun Li
- Department of Biological Sciences, Florida International University, Miami, FL 33199, United States
| | - Hong Xin
- Department of Microbiology and Immunology, Louisiana State University Health Science Center, New Orleans, LA 70112, United States
| | - Wenda Gao
- Antagen Pharmaceuticals, Inc., Canton, MA 02021, United States
| |
Collapse
|
10
|
Herrera M, Pretelli G, Desai J, Garralda E, Siu LL, Steiner TM, Au L. Bispecific antibodies: advancing precision oncology. Trends Cancer 2024; 10:893-919. [PMID: 39214782 DOI: 10.1016/j.trecan.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/29/2024] [Accepted: 07/12/2024] [Indexed: 09/04/2024]
Abstract
Bispecific antibodies (bsAbs) are engineered molecules designed to target two different epitopes or antigens. The mechanism of action is determined by the bsAb molecular targets and structure (or format), which can be manipulated to create variable and novel functionalities, including linking immune cells with tumor cells, or dual signaling pathway blockade. Several bsAbs have already changed the treatment landscape of hematological malignancies and select solid cancers. However, the mechanisms of resistance to these agents are understudied and the management of toxicities remains challenging. Herein, we review the principles in bsAb engineering, current understanding of mechanisms of action and resistance, data for clinical application, and provide a perspective on ongoing challenges and future developments in this field.
Collapse
Affiliation(s)
- Mercedes Herrera
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Giulia Pretelli
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jayesh Desai
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Elena Garralda
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Thiago M Steiner
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Lewis Au
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
| |
Collapse
|
11
|
Nagy Á, Ulmert D, Zedan W, Storey CM, Park J, Geres S, Lückerath K, Sjöström K, Westin H, Peekhaus N, Thorek DL, Karlström AE, Altai M. Impact of site-specific conjugation strategies on the pharmacokinetics of antibody conjugated radiotherapeutics. Eur J Med Chem 2024; 280:116927. [PMID: 39378827 DOI: 10.1016/j.ejmech.2024.116927] [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: 07/09/2024] [Revised: 09/21/2024] [Accepted: 09/29/2024] [Indexed: 10/10/2024]
Abstract
Antibody radionuclide conjugates are an emerging modality for targeted imaging and potent therapy of disseminated disease. Coupling of radionuclides to monoclonal antibodies (mAbs) is typically achieved by applying non-site-specific labelling techniques. With the ambition of reducing variability, increasing labelling efficacy and stability, several site-specific conjugation strategies have been developed in recent years for toxin- and fluorophore-mAb conjugates. In this study, we studied two site-specific labelling strategies for the conjugation of the macrocyclic chelating agent, DOTA, to the anti-Leucine Rich Repeat Containing 15 (LRRC15) mAb DUNP19. Specifically, one approach utilized a DOTA-bearing peptide (FcIII) with a strong affinity for the fragment crystallizable (Fc) domain of the human IgG1 of DUNP19 (DUNP19LF-FcIII-DOTASS), while the other leveraged a chemo-enzymatic technique to substitute the N-linked bi-antennary oligosaccharides in the human IgG1 Fc domain with DOTA (DUNP19LF-gly-DOTASS). To assess if these methods impact the antibody's binding properties and targeting efficacy, comparative in vitro and in vivo studies of the generated DUNP19-conjugates were performed. While the LRRC15 binding of both radioimmunoconjugates remained intact, the conjugation methods had different impacts on their abilities to interact with FcRn and FcγRs. In vitro assessments of DUNP19LF-FcIII-DOTASS and DUNP19LF-gly-DOTASS demonstrated markedly decreased affinity for FcRn and FcγRIIIa (CD16), respectively. DUNP19LF-FcIII-DOTASS demonstrated increased blood and tissue kinetics in vivo, confirming loss of FcRn binding. While the ablated FcγR interaction of DUNP19LF-gly-DOTASS had no immediate impact on in vivo biodistribution, reduced immunotherapeutic effect can be expected in future studies as a result of reduced NK-cells interaction. In conclusion, our findings underscore the necessity for meticulous consideration and evaluation of mAb labelling strategies, extending beyond mere conjugation efficiency and radiolabeling yields. Notably, site-specific labelling methods were found to significantly influence the immunological impact of Fc interactions. Therefore, it is of paramount importance to consider the intended diagnostic or therapeutic application of the construct and to adopt conjugation strategies that ensure the preservation of critical pharmacological properties and functionality of the antibody in use.
Collapse
Affiliation(s)
- Ábel Nagy
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| | - David Ulmert
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden
| | - Wahed Zedan
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Claire M Storey
- Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Julie Park
- Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Susanne Geres
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Katharina Lückerath
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, DKTK, Essen, Germany
| | | | | | - Norbert Peekhaus
- Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Daniel Lj Thorek
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA; Oncologic Imaging Program, Siteman Cancer Center, St. Louis, Missouri, USA
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm, Sweden
| | - Mohamed Altai
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden; Lund University Cancer Centre (LUCC), Lund University, Lund, Sweden.
| |
Collapse
|
12
|
Zhang Y, Zhang M, Wu H, Wu X, Zheng H, Feng J, Wang M, Wang J, Luo L, Xiao H, Qiao C, Li X, Zheng Y, Huang W, Wang Y, Wang Y, Feng J, Chen G. Afucosylated anti-EBOV antibody MIL77-3 engages sGP to elicit NK cytotoxicity. J Virol 2024; 98:e0068524. [PMID: 39162435 PMCID: PMC11406966 DOI: 10.1128/jvi.00685-24] [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: 06/02/2024] [Accepted: 06/24/2024] [Indexed: 08/21/2024] Open
Abstract
MIL77-3 is one component of antibody cocktail that is produced in our lab and represents an effective regimen for animals suffering from Zaire Ebolavirus (EBOV) infection. MIL77-3 is engineered to increase its affinity for the FcγRIIIa (CD16a) by deleting the fucose in the framework region. The potential effects of this modification on host immune responses, however, remain largely unknown. Herein, we demonstrated that MIL77-3 recognized secreted glycoproptein (sGP), produced by EBOV, and formed the immunocomplex to potently augment antibody-dependent cytotoxicity of human peripheral blood-derived natural killer cells (pNKs), including CD56dim and CD56bright subpopulations, in contrast to the counterparts (Mab114, rEBOV548, fucosylated MIL77-3). Intriguingly, this effect was not observed when NK92-CD16a cell line was utilized and restored by the addition of beads-coupled or membrane-anchored sGP in combination with MIL77-3. Furthermore, sGP bound to unrecognized receptors on T cells contaminated in pNKs rather than NK92-CD16a cells. Administration of beads-coupled sGP/MIL77-3 complex in mice elicited NK activation. Overall, this work reveals an immune-stimulating function of sGP/MIL77-3 complex by triggering cytotoxic activity of NK cells, highlighting the necessity to evaluate the potential impact of MIL77-3 on host immune reaction in clinical trials. IMPORTANCE Zaire Ebolavirus (EBOV) is highly lethal and causes sporadic outbreaks. The passive administration of monoclonal antibodies (mAbs) represents a promising treatment regimen against EBOV. Mounting evidence has shown that the efficacy of a subset of therapeutic mAbs in vivo is intimately associated with its capacity to trigger NK activity, supporting glycomodification of Fc region of anti-EBOV mAbs as a putative strategy to enhance Fc-mediated immune effector function as well as protection in vivo. Our work here uncovers the potential harmful influence of this modification on host immune responses, especially for mAbs with cross-reactivity to secreted glycoproptein (sGP) (e.g., MIL77-3), and highlights it is necessary to evaluate the NK-stimulating activity of a fucosylated mAb engaged with sGP when a new candidate is developed.
Collapse
Affiliation(s)
- Yuting Zhang
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Min Zhang
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Haiyan Wu
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiaonan Wu
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Hang Zheng
- Institute of Pharmacology and Toxicology, Beijing, China
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Junjuan Feng
- Institute of Pharmacology and Toxicology, Beijing, China
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Mianjing Wang
- Institute of Pharmacology and Toxicology, Beijing, China
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Jing Wang
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Longlong Luo
- Institute of Pharmacology and Toxicology, Beijing, China
| | - He Xiao
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Chunxia Qiao
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Xinying Li
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Yuanqiang Zheng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Yi Wang
- Department of Hematology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jiannan Feng
- Institute of Pharmacology and Toxicology, Beijing, China
| | - Guojiang Chen
- Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
13
|
Graham LV, Fisher JG, Doyle ADP, Sale B, Del Rio L, French AJE, Mayor NP, Turner TR, Marsh SGE, Cragg MS, Forconi F, Khakoo SI, Blunt MD. KIR2DS2+ NK cells in cancer patients demonstrate high activation in response to tumour-targeting antibodies. Front Oncol 2024; 14:1404051. [PMID: 39286025 PMCID: PMC11402612 DOI: 10.3389/fonc.2024.1404051] [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: 03/20/2024] [Accepted: 08/13/2024] [Indexed: 09/19/2024] Open
Abstract
Strategies to mobilise natural killer (NK) cells against cancer include tumour-targeting antibodies, NK cell engagers (NKCEs) and the adoptive transfer of ex vivo expanded healthy donor-derived NK cells. Genetic and functional studies have revealed that expression of the activating killer immunoglobulin-like receptor KIR2DS2 is associated with enhanced function in NK cells from healthy donors and improved outcome in several different malignancies. The optimal strategy to leverage KIR2DS2+ NK cells therapeutically is however currently unclear. In this study, we therefore evaluated the response of KIR2DS2-expressing NK cells to activation against cancer with clinically relevant tumour-targeting antibodies and following ex vivo expansion. We identified that KIR2DS2high NK cells from patients with chronic lymphocytic leukaemia and hepatocellular carcinoma had enhanced activation in response to tumour-targeting antibodies compared to KIR2DS2- NK cells. However, the superior function of healthy donor derived KIR2DS2high NK cells was lost following ex vivo expansion which is required for adoptive transfer-based therapeutic strategies. These data provide evidence that targeting KIR2DS2 directly in cancer patients may allow for the utilisation of their enhanced effector function, however such activity may be lost following their ex vivo expansion.
Collapse
Affiliation(s)
- Lara V Graham
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom
| | - Jack G Fisher
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom
| | - Amber D P Doyle
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom
| | - Ben Sale
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Luis Del Rio
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Albert J E French
- Anthony Nolan Research Institute, Royal Free Hospital, London, United Kingdom
| | - Neema P Mayor
- Anthony Nolan Research Institute, Royal Free Hospital, London, United Kingdom
- Department of Academic Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Thomas R Turner
- Anthony Nolan Research Institute, Royal Free Hospital, London, United Kingdom
- Department of Academic Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Steven G E Marsh
- Anthony Nolan Research Institute, Royal Free Hospital, London, United Kingdom
- Department of Academic Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Mark S Cragg
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
- Antibody and Vaccine Group, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Francesco Forconi
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
- Haematology Department, Cancer Care Directorate, University Hospital Southampton National Health Service (NHS) Trust, Southampton, United Kingdom
| | - Salim I Khakoo
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom
| | - Matthew D Blunt
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
14
|
Xu T, Tong L, Zhang Z, Zhou H, Zheng P. Glycosylation in Drosophila S2 cells. Biotechnol Bioeng 2024. [PMID: 39140464 DOI: 10.1002/bit.28827] [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: 03/08/2024] [Revised: 07/12/2024] [Accepted: 08/04/2024] [Indexed: 08/15/2024]
Abstract
In recent years, there has been a remarkable surge in the approval of therapeutic protein drugs, particularly recombinant glycoproteins. Drosophila melanogaster S2 cells have become an appealing platform for the production of recombinant proteins due to their simplicity and low cost in cell culture. However, a significant limitation associated with using the S2 cell expression system is its propensity to introduce simple paucimannosidic glycosylation structures, which differs from that in the mammalian expression system. It is well established that the glycosylation patterns of glycoproteins have a profound impact on the physicochemical properties, bioactivity, and immunogenicity. Therefore, understanding the mechanisms behind these glycosylation modifications and implementing measures to address it has become a subject of considerable interest. This review aims to comprehensively summarize recent advancements in glycosylation modification in S2 cells, with a particular focus on comparing the glycosylation patterns among S2, other insect cells, and mammalian cells, as well as developing strategies for altering the glycosylation patterns of recombinant glycoproteins.
Collapse
Affiliation(s)
- Tingting Xu
- Department of General Medicine, People's Hospital of Longhua, Shenzhen, China
| | - Lixiang Tong
- Department of General Medicine, People's Hospital of Longhua, Shenzhen, China
| | - Zhifu Zhang
- Department of General Medicine, People's Hospital of Longhua, Shenzhen, China
| | - Hairong Zhou
- Department of General Medicine, People's Hospital of Longhua, Shenzhen, China
| | - Peilin Zheng
- Department of General Medicine, People's Hospital of Longhua, Shenzhen, China
| |
Collapse
|
15
|
Saporiti S, Bianchi D, Ben Mariem O, Rossi M, Guerrini U, Eberini I, Centola F. In silico evaluation of the role of Fab glycosylation in cetuximab antibody dynamics. Front Immunol 2024; 15:1429600. [PMID: 39185413 PMCID: PMC11342397 DOI: 10.3389/fimmu.2024.1429600] [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: 05/08/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
Introduction N-glycosylation is a post-translational modification that is highly important for the development of monoclonal antibodies (mAbs), as it regulates their biological activity, particularly in terms of immune effector functions. While typically added at the Fc level, approximately 15-25% of circulating antibodies exhibit glycosylation in the Fab domains as well. To the best of our knowledge, cetuximab (Erbitux®) is the only therapeutic antibody presenting Fab glycosylation approved world-wide targeting the epidermal growth factor receptor for the treatment of metastatic-colorectal and head and neck cancers. Additionally, it can trigger antibody-dependent cell cytotoxicity (ADCC), a response that typically is influenced by N-glycosylation at Fc level. However, the role of Fab glycosylation in cetuximab remains poorly understood. Hence, this study aims to investigate the structural role of Fab glycosylation on the conformational behavior of cetuximab. Methods The study was performed in silico via accelerated molecular dynamics simulations. The commercial cetuximab was compared to its form without Fab glycosylation and structural descriptors were evaluated to establish conformational differences. Results The results clearly show a correlation between the Fab glycosylation and structural descriptors that may modulate the conformational freedom of the antibody, potentially affecting Fc effector functions, and suggesting a negative role of Fab glycosylation on the interaction with FcγRIIIa. Conclusion Fab glycosylation of cetuximab is the most critical challenge for biosimilar development, but the differences highlighted in this work with respect to its aglycosylated form can improve the knowledge and represent also a great opportunity to develop novel strategies of biotherapeutics.
Collapse
Affiliation(s)
- Simona Saporiti
- Analytical Excellence and Program Management, Merck Serono S.p.A., Rome, Italy
| | - Davide Bianchi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Omar Ben Mariem
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Mara Rossi
- Analytical Excellence and Program Management, Merck Serono S.p.A., Rome, Italy
| | - Uliano Guerrini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Ivano Eberini
- Dipartimento di Scienze Farmacologiche e Biomolecolari & Data Science Research Center (DSRC), Università degli Studi di Milano, Milan, Italy
| | - Fabio Centola
- Analytical Excellence and Program Management, Merck Serono S.p.A., Rome, Italy
| |
Collapse
|
16
|
Shi Y, Sun Y, Seki A, Rutz S, Koerber JT, Wang J. A real-time antibody-dependent cellular phagocytosis assay by live cell imaging. J Immunol Methods 2024; 531:113715. [PMID: 38936465 DOI: 10.1016/j.jim.2024.113715] [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: 02/27/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Antibody-dependent cellular phagocytosis (ADCP) is a cellular process by which antibody-opsonized targets (pathogens or cells) activate the Fc receptors on the surface of phagocytes to induce phagocytosis, resulting in internalization and degradation of pathogens or target cells through phagosome acidification. Besides NK cells-mediated antibody-dependent cellular cytotoxicity (ADCC), tumor-infiltrated monocytes and macrophages can directly kill tumor cells in the presence of tumor antigen-specific antibodies through ADCP, representing another attractive strategy for cancer immunotherapy. Even though several methods have been developed to measure ADCP, an automated and high-throughput quantitative assay should offer highly desirable advantages for drug discovery. In this study we established a new ADCP assay to identify therapeutical monoclonal antibodies (mAbs) that facilitate macrophages phagocytosis of live target cells. We used Incucyte, an imaging system for live cell analysis. By labeling the live target cells with a pH sensitive dye (pHrodo), we successfully monitored the ADCP in real time. We demonstrated that our image-based assay is robust and quantitative, suitable for screening and characterization of therapeutical mAbs that directly kill target cells through ADCP. Furthermore, we found different subtypes of macrophages have distinct ADCP activities using both mouse and human primary macrophages differentiated in vitro. By studying various mAbs with mutations in their Fc regions using our assay, we showed that the variants with increased binding to Fc gamma receptors (FcγRs) have enhanced ADCP activities.
Collapse
Affiliation(s)
- Yongchang Shi
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc, South San Francisco, CA 94080, USA
| | - Yonglian Sun
- Department of Antibody Engineering, Genentech, Inc, South San Francisco, CA 94080, USA
| | - Akiko Seki
- Department of Cancer Immunology, Genentech Inc, South San Francisco, CA 94080, USA
| | - Sascha Rutz
- Department of Cancer Immunology, Genentech Inc, South San Francisco, CA 94080, USA
| | - James T Koerber
- Department of Antibody Engineering, Genentech, Inc, South San Francisco, CA 94080, USA
| | - Jianyong Wang
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc, South San Francisco, CA 94080, USA.
| |
Collapse
|
17
|
Nadat F, Clark B. Forming a new perspective: Post-structural approaches to determination of donor compatibility and post-transplant assessment of allograft health. Int J Immunogenet 2024; 51:195-205. [PMID: 38711186 DOI: 10.1111/iji.12675] [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: 03/13/2024] [Accepted: 03/25/2024] [Indexed: 05/08/2024]
Abstract
The purpose of this review is to encourage a new perspective on the question of donor-recipient compatibility and post-transplant assessment of graft health based on functional measures. The premise is that we should be better sighted on what (and how) the immune system responds toward rather than what is merely there. Continuance of the pursuit of further and better definition of antigens and antibodies is not however discouraged but seen as necessary to improved understanding of the structural correlates of functional immunity. There currently exists, in the opinion of the authors, an opportunity for histocompatibility and immunogenetics laboratories to develop and widen their scope of involvement into these new areas of laboratory activity in support and to the benefit of the transplant programmes they serve.
Collapse
Affiliation(s)
- Fatima Nadat
- Functional Assessment of Transplant Immunology Group, St James's University Hospital, Leeds, UK
| | - Brendan Clark
- Functional Assessment of Transplant Immunology Group, St James's University Hospital, Leeds, UK
| |
Collapse
|
18
|
Palma JA, Bunyatov MI, Hulbert SW, Jewett MC, DeLisa MP. Bacterial glycoengineering: Cell-based and cell-free routes for producing biopharmaceuticals with customized glycosylation. Curr Opin Chem Biol 2024; 81:102500. [PMID: 38991462 DOI: 10.1016/j.cbpa.2024.102500] [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: 04/08/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024]
Abstract
Glycosylation plays a pivotal role in tuning the folding and function of proteins. Because most human therapeutic proteins are glycosylated, understanding and controlling glycosylation is important for the design, optimization, and manufacture of biopharmaceuticals. Unfortunately, natural eukaryotic glycosylation pathways are complex and often produce heterogeneous glycan patterns, making the production of glycoproteins with chemically precise and homogeneous glycan structures difficult. To overcome these limitations, bacterial glycoengineering has emerged as a simple, cost-effective, and scalable approach to produce designer glycoprotein therapeutics and vaccines in which the glycan structures are engineered to reduce heterogeneity and improve biological and biophysical attributes of the protein. Here, we discuss recent advances in bacterial cell-based and cell-free glycoengineering that have enabled the production of biopharmaceutical glycoproteins with customized glycan structures.
Collapse
Affiliation(s)
- Jaymee A Palma
- Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA
| | - Mehman I Bunyatov
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Sophia W Hulbert
- Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA
| | - Michael C Jewett
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Matthew P DeLisa
- Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA; Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; Cornell Institute of Biotechnology, Cornell University, Biotechnology Building, Ithaca, NY 14853, USA.
| |
Collapse
|
19
|
Suzuki H, Ohishi T, Tanaka T, Kaneko MK, Kato Y. Anti-HER2 Cancer-Specific mAb, H 2Mab-250-hG 1, Possesses Higher Complement-Dependent Cytotoxicity than Trastuzumab. Int J Mol Sci 2024; 25:8386. [PMID: 39125956 PMCID: PMC11313270 DOI: 10.3390/ijms25158386] [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: 04/22/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Cancer-specific monoclonal antibodies (CasMabs) that recognize cancer-specific antigens with in vivo antitumor efficacy are innovative therapeutic strategies for minimizing adverse effects. We previously established a cancer-specific anti-human epidermal growth factor receptor 2 (HER2) monoclonal antibody (mAb), H2Mab-250/H2CasMab-2. In flow cytometry and immunohistochemistry, H2Mab-250 reacted with HER2-positive breast cancer cells but did not show reactivity to normal epithelial cells. In contrast, a clinically approved anti-HER2 mAb, trastuzumab, strongly recognizes both breast cancer and normal epithelial cells in flow cytometry. The human IgG1 version of H2Mab-250 (H2Mab-250-hG1) possesses compatible in vivo antitumor effects against breast cancer xenografts to trastuzumab despite the lower affinity and effector activation than trastuzumab in vitro. This study compared the antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cellular cytotoxicity (CDC) between H2Mab-250-hG1 and trastuzumab. Both H2Mab-250-hG1 and trastuzumab showed ADCC activity against HER2-overexpressed Chinese hamster ovary -K1 and breast cancer cell lines (BT-474 and SK-BR-3) in the presence of human natural killer cells. Some tendency was observed where trastuzumab showed a more significant ADCC effect compared to H2Mab-250-hG1. Importantly, H2Mab-250-hG1 exhibited superior CDC activity in these cells compared to trastuzumab. Similar results were obtained in the mouse IgG2a types of both H2Mab-250 and trastuzumab. These results suggest the different contributions of ADCC and CDC activities to the antitumor effects of H2Mab-250-hG1 and trastuzumab, and indicate a future direction for the clinical development of H2Mab-250-hG1 against HER2-positive tumors.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (H.S.); (T.T.); (M.K.K.)
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, 18-24 Miyamoto, Numazu-shi, Shizuoka 410-0301, Japan;
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (H.S.); (T.T.); (M.K.K.)
| | - Mika K. Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (H.S.); (T.T.); (M.K.K.)
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (H.S.); (T.T.); (M.K.K.)
| |
Collapse
|
20
|
Moquist PN, Zhang X, Leiske CI, Eng-Duncan NML, Zeng W, Bindman NA, Wo SW, Wong A, Henderson CM, Crowder K, Lyon R, Doronina SO, Senter PD, Neff-LaFord HD, Sussman D, Gardai SJ, Levengood MR. Reversible Chemical Modification of Antibody Effector Function Mitigates Unwanted Systemic Immune Activation. Bioconjug Chem 2024; 35:855-866. [PMID: 38789102 PMCID: PMC11191404 DOI: 10.1021/acs.bioconjchem.4c00212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Antibody effector functions including antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) are mediated through the interaction of the antibody Fc region with Fcγ receptors present on immune cells. Several approaches have been used to modulate antibody Fc-Fcγ interactions with the goal of driving an effective antitumor immune response, including Fc point mutations and glycan modifications. However, robust antibody-Fcγ engagement and immune cell binding of Fc-enhanced antibodies in the periphery can lead to the unwanted induction of systemic cytokine release and other dose-limiting infusion-related reactions. Creating a balance between effective engagement of Fcγ receptors that can induce antitumor activity without incurring systemic immune activation is an ongoing challenge in the field of antibody and immuno-oncology therapeutics. Herein, we describe a method for the reversible chemical modulation of antibody-Fcγ interactions using simple poly(ethylene glycol) (PEG) linkers conjugated to antibody interchain disulfides with maleimide attachments. This method enables dosing of a therapeutic with muted Fcγ engagement that is restored in vivo in a time-dependent manner. The technology was applied to an effector function enhanced agonist CD40 antibody, SEA-CD40, and experiments demonstrate significant reductions in Fc-induced immune activation in vitro and in mice and nonhuman primates despite showing retained efficacy and improved pharmacokinetics compared to the parent antibody. We foresee that this simple, modular system can be rapidly applied to antibodies that suffer from systemic immune activation due to peripheral FcγR binding immediately upon infusion.
Collapse
Affiliation(s)
- Philip N. Moquist
- ADC
Chemistry, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United states
| | - Xinqun Zhang
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Chris I. Leiske
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | | | - Weiping Zeng
- ADC
In Vivo Pharmacology, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Noah A. Bindman
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Serena W. Wo
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Abbie Wong
- ADC
Translational Sciences, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Clark M. Henderson
- ADC
Translational Sciences, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Karalyne Crowder
- Non-Clinical
Sciences, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Robert Lyon
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Svetlana O. Doronina
- ADC
Chemistry, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United states
| | - Peter D. Senter
- ADC
Chemistry, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United states
| | - Haley D. Neff-LaFord
- Non-Clinical
Sciences, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Django Sussman
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| | - Shyra J. Gardai
- Immunology, Pfizer,
Inc., 21823 30th Dr.
SE, Bothell, Washington 98021, United States
| | - Matthew R. Levengood
- ADC
Antibody Engineering, Pfizer, Inc., 21823 30th Dr. SE, Bothell, Washington 98021, United States
| |
Collapse
|
21
|
Mahomed S. Broadly neutralizing antibodies for HIV prevention: a comprehensive review and future perspectives. Clin Microbiol Rev 2024; 37:e0015222. [PMID: 38687039 PMCID: PMC11324036 DOI: 10.1128/cmr.00152-22] [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: 05/02/2024] Open
Abstract
SUMMARYThe human immunodeficiency virus (HIV) epidemic remains a formidable global health concern, with 39 million people living with the virus and 1.3 million new infections reported in 2022. Despite anti-retroviral therapy's effectiveness in pre-exposure prophylaxis, its global adoption is limited. Broadly neutralizing antibodies (bNAbs) offer an alternative strategy for HIV prevention through passive immunization. Historically, passive immunization has been efficacious in the treatment of various diseases ranging from oncology to infectious diseases. Early clinical trials suggest bNAbs are safe, tolerable, and capable of reducing HIV RNA levels. Although challenges such as bNAb resistance have been noted in phase I trials, ongoing research aims to assess the additive or synergistic benefits of combining multiple bNAbs. Researchers are exploring bispecific and trispecific antibodies, and fragment crystallizable region modifications to augment antibody efficacy and half-life. Moreover, the potential of other antibody isotypes like IgG3 and IgA is under investigation. While promising, the application of bNAbs faces economic and logistical barriers. High manufacturing costs, particularly in resource-limited settings, and logistical challenges like cold-chain requirements pose obstacles. Preliminary studies suggest cost-effectiveness, although this is contingent on various factors like efficacy and distribution. Technological advancements and strategic partnerships may mitigate some challenges, but issues like molecular aggregation remain. The World Health Organization has provided preferred product characteristics for bNAbs, focusing on optimizing their efficacy, safety, and accessibility. The integration of bNAbs in HIV prophylaxis necessitates a multi-faceted approach, considering economic, logistical, and scientific variables. This review comprehensively covers the historical context, current advancements, and future avenues of bNAbs in HIV prevention.
Collapse
Affiliation(s)
- Sharana Mahomed
- Centre for the AIDS
Programme of Research in South Africa (CAPRISA), Doris Duke Medical
Research Institute, Nelson R Mandela School of Medicine, University of
KwaZulu-Natal, Durban,
South Africa
| |
Collapse
|
22
|
Roider HG, Hoff S, Tseng SY, Berndt S, Trautwein M, Filarsky K, Gritzan U, Camps J, Nadler WM, Grudzinska-Goebel J, Ellinger P, Pesch T, Soon CF, Geyer M, Gluske K, Stelte-Ludwig B, Gorjánácz M. Selective depletion of tumor-infiltrating regulatory T cells with BAY 3375968, a novel Fc-optimized anti-CCR8 antibody. Clin Exp Med 2024; 24:122. [PMID: 38856863 PMCID: PMC11164760 DOI: 10.1007/s10238-024-01362-8] [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: 03/15/2024] [Accepted: 04/28/2024] [Indexed: 06/11/2024]
Abstract
Regulatory T cells (Tregs) are known to facilitate tumor progression by suppressing CD8+ T cells within the tumor microenvironment (TME), thereby also hampering the effectiveness of immune checkpoint inhibitors (ICIs). While systemic depletion of Tregs can enhance antitumor immunity, it also triggers undesirable autoimmune responses. Therefore, there is a need for therapeutic agents that selectively target Tregs within the TME without affecting systemic Tregs. In this study, as shown also by others, the chemokine (C-C motif) receptor 8 (CCR8) was found to be predominantly expressed on Tregs within the TME of both humans and mice, representing a unique target for selective depletion of tumor-residing Tregs. Based on this, we developed BAY 3375968, a novel anti-human CCR8 antibody, along with respective surrogate anti-mouse CCR8 antibodies, and demonstrated their in vitro mode-of-action through induction of potent antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) activities. In vivo, anti-mouse CCR8 antibodies effectively depleted Tregs within the TME primarily via ADCP, leading to increased CD8+ T cell infiltration and subsequent tumor growth inhibition across various cancer models. This monotherapeutic efficacy was significantly enhanced in combination with ICIs. Collectively, these findings suggest that CCR8 targeting represents a promising strategy for Treg depletion in cancer therapies. BAY 3375968 is currently under investigation in a Phase I clinical trial (NCT05537740).
Collapse
MESH Headings
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/drug effects
- Receptors, CCR8/immunology
- Receptors, CCR8/antagonists & inhibitors
- Animals
- Mice
- Humans
- Tumor Microenvironment/immunology
- Tumor Microenvironment/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Female
- Antibody-Dependent Cell Cytotoxicity
- Lymphocyte Depletion
- Cell Line, Tumor
- Phagocytosis/drug effects
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
Collapse
Affiliation(s)
| | | | - Su-Yi Tseng
- Bayer AG, Pharmaceuticals, San Francisco, USA
| | | | | | - Katharina Filarsky
- Bayer AG, Pharmaceuticals, Wuppertal, Germany
- Current address: Roche Diagnostics GmbH, Penzberg, Germany
| | - Uwe Gritzan
- Bayer AG, Pharmaceuticals, Wuppertal, Germany
- Current address: Memorial Sloan Kettering Cancer Center, New York, USA
| | | | | | | | | | | | | | | | | | - Beatrix Stelte-Ludwig
- Bayer AG, Pharmaceuticals, Wuppertal, Germany
- Current address: Vincerx Pharma, Monheim am Rhein, Germany
| | | |
Collapse
|
23
|
Krishna S, Jung ST, Lee EY. Escherichia coli and Pichia pastoris: microbial cell-factory platform for -full-length IgG production. Crit Rev Biotechnol 2024:1-23. [PMID: 38797692 DOI: 10.1080/07388551.2024.2342969] [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: 12/13/2023] [Accepted: 03/18/2024] [Indexed: 05/29/2024]
Abstract
Owing to the unmet demand, the pharmaceutical industry is investigating an alternative host to mammalian cells to produce antibodies for a variety of therapeutic and research applications. Regardless of some disadvantages, Escherichia coli and Pichia pastoris are the preferred microbial hosts for antibody production. Despite the fact that the production of full-length antibodies has been successfully demonstrated in E. coli, which has mostly been used to produce antibody fragments, such as: antigen-binding fragments (Fab), single-chain fragment variable (scFv), and nanobodies. In contrast, Pichia, a eukaryotic microbial host, is mostly used to produce glycosylated full-length antibodies, though hypermannosylated glycan is a major challenge. Advanced strategies, such as the introduction of human-like glycosylation in endotoxin-edited E. coli and cell-free system-based glycosylation, are making progress in creating human-like glycosylation profiles of antibodies in these microbes. This review begins by explaining the structural and functional requirements of antibodies and continues by describing and analyzing the potential of E. coli and P. pastoris as hosts for providing a favorable environment to create a fully functional antibody. In addition, authors compare these microbes on certain features and predict their future in antibody production. Briefly, this review analyzes, compares, and highlights E. coli and P. pastoris as potential hosts for antibody production.
Collapse
Affiliation(s)
- Shyam Krishna
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Sang Taek Jung
- BK21 Graduate Program, Department of Biomedical Sciences, Graduate School, Korea University, Seoul, Republic of Korea
| | - Eun Yeol Lee
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
24
|
Xu P, Ou YC, Smith M, Paulson J, Schmidt MA, Kandari L, Parsons R, Khetan A. Application of fucosylation inhibitors for production of afucosylated antibody. Biotechnol Prog 2024; 40:e3438. [PMID: 38415431 DOI: 10.1002/btpr.3438] [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: 07/07/2023] [Revised: 12/21/2023] [Accepted: 01/24/2024] [Indexed: 02/29/2024]
Abstract
Fucosylation is an important quality attribute for therapeutic antibodies. Afucosylated antibodies exhibit higher therapeutic efficacies than their fucosylated counterparts through antibody-dependent cellular cytotoxicity (ADCC) mechanism. Since higher potency is beneficial in reducing dose or duration of the treatment, afucosylated antibodies have attracted a great deal of interest in biotherapeutics development. In this study, novel small molecules GDP-D-Rhamnose and its derivatives (Ac-GDP-D-Rhamnose and rhamnose sodium phosphate) were synthesized to inhibit the enzyme in the GDP-fucose synthesis pathway. Addition of these compounds into cell culture increased antibody afucosylation levels in a dose-dependent manner and had no significant impact on other protein quality attributes. A novel and effective mechanism to generate afucosylated antibody is demonstrated for biologics discovery, analytical method development, process development, and other applications.
Collapse
Affiliation(s)
- Ping Xu
- Biologics Development, Global Product Development & Supply, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Yu Chuan Ou
- Biologics Development, Global Product Development & Supply, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Michael Smith
- Chemical Process Development, Global Product Development & Supply, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Jim Paulson
- Chemical Process Development, Global Product Development & Supply, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Michael A Schmidt
- Chemical Process Development, Global Product Development & Supply, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Lakshmi Kandari
- Biologics Development, Global Product Development & Supply, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Rodney Parsons
- Chemical Process Development, Global Product Development & Supply, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| | - Anurag Khetan
- Biologics Development, Global Product Development & Supply, Bristol Myers Squibb, New Brunswick, New Jersey, USA
| |
Collapse
|
25
|
Wainberg ZA, Kang YK, Lee KW, Qin S, Yamaguchi K, Kim IH, Saeed A, Oh SC, Li J, Turk HM, Teixeira A, Hitre E, Udrea AA, Cardellino GG, Sanchez RG, Zahlten-Kümeli A, Taylor K, Enzinger PC. Bemarituzumab as first-line treatment for locally advanced or metastatic gastric/gastroesophageal junction adenocarcinoma: final analysis of the randomized phase 2 FIGHT trial. Gastric Cancer 2024; 27:558-570. [PMID: 38308771 PMCID: PMC11016503 DOI: 10.1007/s10120-024-01466-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/04/2024] [Indexed: 02/05/2024]
Abstract
BACKGROUND We report the final results of the randomized phase 2 FIGHT trial that evaluated bemarituzumab, a humanized monoclonal antibody selective for fibroblast growth factor receptor 2b (FGFR2b), plus mFOLFOX6 in patients with FGFR2b-positive (2 + /3 + membranous staining by immunohistochemistry), HER-2-negative gastric or gastroesophageal junction cancer (GC). METHODS Patients received bemarituzumab (15 mg/kg) or placebo once every 2 weeks with an additional bemarituzumab (7.5 mg/kg) or placebo dose on cycle 1 day 8. All patients received mFOLFOX6. The primary endpoint was investigator-assessed progression-free survival (PFS). Secondary endpoints included overall survival (OS), objective response rate, and safety. Efficacy was evaluated after a minimum follow-up of 24 months. RESULTS In the bemarituzumab-mFOLFOX6 (N = 77) and placebo-mFOLFOX6 (N = 78) arms, respectively, 59.7% and 66.7% of patients were FGFR2b-positive in ≥ 10% of tumor cells. The median PFS (95% confidence interval [CI]) was 9.5 months (7.3-13.7) with bemarituzumab-mFOLFOX6 and 7.4 months (5.7-8.4) with placebo-mFOLFOX6 (hazard ratio [HR], 0.72; 95% CI 0.49-1.08); median OS (95% CI) was 19.2 (13.6-24.2) and 13.5 (9.3-15.9) months, respectively (HR 0.77; 95% CI 0.52-1.14). Observed efficacy in FGFR2b-positive GC in ≥ 10% of tumor cells was: PFS: HR 0.43 (95% CI 0.26-0.73); OS: HR 0.52 (95% CI 0.31-0.85). No new safety findings were reported. CONCLUSIONS In FGFR2b-positive advanced GC, the combination of bemarituzumab-mFOLFOX6 led to numerically longer median PFS and OS compared with mFOLFOX6 alone. Efficacy was more pronounced with FGFR2b overexpression in ≥ 10% of tumor cells. Confirmatory phase 3 trials are ongoing (NCT05052801, NCT05111626). CLINICAL TRIAL REGISTRATION NCT03694522.
Collapse
Affiliation(s)
- Zev A Wainberg
- Division of Hematology-Oncology, Department of Medicine, University of California Los Angeles Medical Center, David Geffen School of Medicine, 2825 Santa Monica Blvd., Suite 200, Santa Monica, Los Angeles, CA, 90404-2429, USA.
| | - Yoon-Koo Kang
- Asan Medical Centre, University of Ulsan College of Medicine, Seoul, South Korea
| | - Keun-Wook Lee
- Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Shukui Qin
- Nanjing Tianyinshan Hospital, The 1st Affiliated Hospital of China Pharmaceutical University, Nanjing, China
| | - Kensei Yamaguchi
- Gastroenterological Chemotherapy Department, The Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - In-Ho Kim
- Department of Oncology, The Catholic University of Korea, Seoul St Mary's Hospital, Seoul, South Korea
| | - Anwaar Saeed
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sang Cheul Oh
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, South Korea
| | - Jin Li
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haci Mehmet Turk
- Department of Medical Oncology, Bezmialem Vakif University, Istanbul, Turkey
| | - Alexandra Teixeira
- Gastroenterology Division, Hospital da Senhora da Oliveira, Guimarães, Portugal
| | - Erika Hitre
- Department of Medical Oncology and Clinical Pharmacology "B", National Institute of Oncology, Budapest, Hungary
| | - Adrian A Udrea
- Medical Oncology, Medisprof Cancer Center, Cluj-Napoca, Romania
| | | | | | | | | | - Peter C Enzinger
- Department of Medicine, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
26
|
Sudol ASL, Crispin M, Tews I. The IgG-specific endoglycosidases EndoS and EndoS2 are distinguished by conformation and antibody recognition. J Biol Chem 2024; 300:107245. [PMID: 38569940 PMCID: PMC11063906 DOI: 10.1016/j.jbc.2024.107245] [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: 12/01/2023] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/05/2024] Open
Abstract
The IgG-specific endoglycosidases EndoS and EndoS2 from Streptococcus pyogenes can remove conserved N-linked glycans present on the Fc region of host antibodies to inhibit Fc-mediated effector functions. These enzymes are therefore being investigated as therapeutics for suppressing unwanted immune activation, and have additional application as tools for antibody glycan remodeling. EndoS and EndoS2 differ in Fc glycan substrate specificity due to structural differences within their catalytic glycosyl hydrolase domains. However, a chimeric EndoS enzyme with a substituted glycosyl hydrolase from EndoS2 loses catalytic activity, despite high structural homology between the two enzymes, indicating either mechanistic divergence of EndoS and EndoS2, or improperly-formed domain interfaces in the chimeric enzyme. Here, we present the crystal structure of the EndoS2-IgG1 Fc complex determined to 3.0 Å resolution. Comparison of complexed and unliganded EndoS2 reveals relative reorientation of the glycosyl hydrolase, leucine-rich repeat and hybrid immunoglobulin domains. The conformation of the complexed EndoS2 enzyme is also different when compared to the earlier EndoS-IgG1 Fc complex, and results in distinct contact surfaces between the two enzymes and their Fc substrate. These findings indicate mechanistic divergence of EndoS2 and EndoS. It will be important to consider these differences in the design of IgG-specific enzymes, developed to enable customizable antibody glycosylation.
Collapse
Affiliation(s)
- Abigail S L Sudol
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton, UK.
| | - Ivo Tews
- School of Biological Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
27
|
Yang J, Ostafe R, Bruening ML. In-Membrane Enrichment and Peptic Digestion to Facilitate Analysis of Monoclonal Antibody Glycosylation. Anal Chem 2024; 96:6347-6355. [PMID: 38607313 PMCID: PMC11283323 DOI: 10.1021/acs.analchem.4c00030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
The number of therapeutic monoclonal antibodies (mAbs) is growing rapidly due to their widespread use for treating various diseases and health conditions. Assessing the glycosylation profile of mAbs during production is essential to ensuring their safety and efficacy. This research aims to rapidly isolate and digest mAbs for liquid chromatography-tandem mass spectrometry (LC-MS/MS) identification of glycans and monitoring of glycosylation patterns, potentially during manufacturing. Immobilization of an Fc region-specific ligand, oFc20, in a porous membrane enables the enrichment of mAbs from cell culture supernatant and efficient elution with an acidic solution. Subsequent digestion of the mAb eluate occurred in a pepsin-modified membrane within 5 min. The procedure does not require alkylation and desalting, greatly shortening the sample preparation time. Subsequent LC-MS/MS analysis identified 11 major mAb N-glycan proteoforms and assessed the relative peak areas of the glycosylated peptides. This approach is suitable for the glycosylation profiling of various human IgG mAbs, including biosimilars and different IgG subclasses. The total time required for this workflow is less than 2 h, whereas the conventional enzymatic release and labeling of glycans can take much longer. Thus, the integrated membranes are suitable for facilitating the analysis of mAb glycosylation patterns.
Collapse
Affiliation(s)
- Junyan Yang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Raluca Ostafe
- Molecular Evolution, Protein Engineering and Production Facility; Purdue Institute for Inflammation, Immunology and Infection Diseases, Purdue University, West Lafayette, IN 47907, United States
| | - Merlin L. Bruening
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| |
Collapse
|
28
|
Delgado SR, Faissner S, Linker RA, Rammohan K. Key characteristics of anti-CD20 monoclonal antibodies and clinical implications for multiple sclerosis treatment. J Neurol 2024; 271:1515-1535. [PMID: 37906325 PMCID: PMC10973056 DOI: 10.1007/s00415-023-12007-3] [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: 05/23/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 11/02/2023]
Abstract
The recent success of anti-CD20 monoclonal antibody therapies in the treatment of multiple sclerosis (MS) has highlighted the role of B cells in the pathogenesis of MS. In people with MS, the inflammatory characteristics of B-cell activity are elevated, leading to increased pro-inflammatory cytokine release, diminished anti-inflammatory cytokine production and an accumulation of pathogenic B cells in the cerebrospinal fluid. Rituximab, ocrelizumab, ofatumumab, ublituximab and BCD-132 are anti-CD20 therapies that are either undergoing clinical development, or have been approved, for the treatment of MS. Despite CD20 being a common target for these therapies, differences have been reported in their mechanistic, pharmacological and clinical characteristics, which may have substantial clinical implications. This narrative review explores key characteristics of these therapies. By using clinical trial data and real-world evidence, we discuss their mechanisms of action, routes of administration, efficacy (in relation to B-cell kinetics), safety, tolerability and convenience of use. Clinicians, alongside patients and their families, should consider the aspects discussed in this review as part of shared decision-making discussions to improve outcomes and health-related quality of life for people living with MS.
Collapse
Affiliation(s)
- Silvia R Delgado
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Simon Faissner
- Department of Neurology, Ruhr-University Bochum, St Josef-Hospital, Bochum, Germany
| | - Ralf A Linker
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Kottil Rammohan
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
29
|
Pinkeova A, Kosutova N, Jane E, Lorencova L, Bertokova A, Bertok T, Tkac J. Medical Relevance, State-of-the-Art and Perspectives of "Sweet Metacode" in Liquid Biopsy Approaches. Diagnostics (Basel) 2024; 14:713. [PMID: 38611626 PMCID: PMC11011756 DOI: 10.3390/diagnostics14070713] [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: 02/07/2024] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
This review briefly introduces readers to an area where glycomics meets modern oncodiagnostics with a focus on the analysis of sialic acid (Neu5Ac)-terminated structures. We present the biochemical perspective of aberrant sialylation during tumourigenesis and its significance, as well as an analytical perspective on the detection of these structures using different approaches for diagnostic and therapeutic purposes. We also provide a comparison to other established liquid biopsy approaches, and we mathematically define an early-stage cancer based on the overall prognosis and effect of these approaches on the patient's quality of life. Finally, some barriers including regulations and quality of clinical validations data are discussed, and a perspective and major challenges in this area are summarised.
Collapse
Affiliation(s)
- Andrea Pinkeova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia; (A.P.); (N.K.); (E.J.); (L.L.)
- Glycanostics, Ltd., Kudlakova 7, 841 08 Bratislava, Slovakia;
| | - Natalia Kosutova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia; (A.P.); (N.K.); (E.J.); (L.L.)
| | - Eduard Jane
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia; (A.P.); (N.K.); (E.J.); (L.L.)
| | - Lenka Lorencova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia; (A.P.); (N.K.); (E.J.); (L.L.)
| | - Aniko Bertokova
- Glycanostics, Ltd., Kudlakova 7, 841 08 Bratislava, Slovakia;
| | - Tomas Bertok
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia; (A.P.); (N.K.); (E.J.); (L.L.)
| | - Jan Tkac
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 845 38 Bratislava, Slovakia; (A.P.); (N.K.); (E.J.); (L.L.)
- Glycanostics, Ltd., Kudlakova 7, 841 08 Bratislava, Slovakia;
| |
Collapse
|
30
|
Zedan HT, Smatti MK, Al-Sadeq DW, Al Khatib HA, Nicolai E, Pieri M, Bernardini S, Hssain AA, Taleb S, Qotba H, Issa K, Abu Raddad LJ, Althani AA, Nasrallah GK, Yassine HM. SARS-CoV-2 infection triggers more potent antibody-dependent cellular cytotoxicity (ADCC) responses than mRNA-, vector-, and inactivated virus-based COVID-19 vaccines. J Med Virol 2024; 96:e29527. [PMID: 38511514 DOI: 10.1002/jmv.29527] [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: 05/15/2023] [Revised: 02/08/2024] [Accepted: 03/01/2024] [Indexed: 03/22/2024]
Abstract
Neutralizing antibodies (NAbs) are elicited after infection and vaccination and have been well studied. However, their antibody-dependent cellular cytotoxicity (ADCC) functionality is still poorly characterized. Here, we investigated ADCC activity in convalescent sera from infected patients with wild-type (WT) severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) or omicron variant compared with three coronavirus disease 2019 (COVID-19) vaccine platforms and postvaccination breakthrough infection (BTI). We analyzed ADCC activity targeting SARS-CoV-2 spike (S) and nucleocapsid (N) proteins in convalescent sera following WT SARS-CoV-2-infection (n = 91), including symptomatic and asymptomatic infections, omicron-infection (n = 8), COVID-19 vaccination with messenger RNA- (mRNA)- (BNT162b2 or mRNA-1273, n = 77), adenovirus vector- (n = 41), and inactivated virus- (n = 46) based vaccines, as well as post-mRNA vaccination BTI caused by omicron (n = 28). Correlations between ADCC, binding, and NAb titers were reported. ADCC was elicited within the first month postinfection and -vaccination and remained detectable for ≥3 months. WT-infected symptomatic patients had higher S-specific ADCC levels than asymptomatic and vaccinated individuals. Also, no difference in N-specific ADCC activity was seen between symptomatic and asymptomatic patients, but the levels were higher than the inactivated vaccine. Notably, omicron infection showed reduced overall ADCC activity compared to WT SARS-CoV-2 infection. Although post-mRNA vaccination BTI elicited high levels of binding and NAbs, ADCC activity was significantly reduced. Also, there was no difference in ADCC levels across the four vaccines, although NAbs and binding antibody titers were significantly higher in mRNA-vaccinated individuals. All evaluated vaccine platforms are inferior in inducing ADCC compared to natural infection with WT SARS-CoV-2. The inactivated virus-based vaccine can induce N-specific ADCC activity, but its relevance to clinical outcomes requires further investigation. Our data suggest that ADCC could be used to estimate the extra-neutralization level against COVID-19 and provides evidence that vaccination should focus on other Fc-effector functions besides NAbs. Also, the decreased susceptibility of the omicron variant to ADCC offers valuable guidance for forthcoming efforts to identify the specific targets of antibodies facilitating ADCC.
Collapse
Affiliation(s)
- Hadeel T Zedan
- Infectious Diseases Department, Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, Doha, Qatar
| | - Maria K Smatti
- Infectious Diseases Department, Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
| | - Duaa W Al-Sadeq
- College of Medicine, Member of QU Health, Qatar University, Doha, Qatar
| | - Hebah A Al Khatib
- Infectious Diseases Department, Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
| | - Eleonora Nicolai
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Massimo Pieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ali Ait Hssain
- Medical Intensive Care Unit, Hamad Medical Corporation, Doha, Qatar
| | - Sara Taleb
- Department of Research, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Hamda Qotba
- Department of Clinical Research, Primary Health Care Centers, Doha, Qatar
- Department of Pathology, Sidra Medicine, Doha, Qatar
| | - Khodr Issa
- Proteomics, Inflammatory Response, and Mass Spectrometry (PRISM) Laboratory, INSERM U-1192, University of Lille, Lille, France
| | - Laith J Abu Raddad
- Department of Population Health Sciences, Infectious Disease Epidemiology Group, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Asmaa A Althani
- Infectious Diseases Department, Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, Doha, Qatar
| | - Gheyath K Nasrallah
- Infectious Diseases Department, Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, Doha, Qatar
| | - Hadi M Yassine
- Infectious Diseases Department, Biomedical Research Center, Research Complex, Qatar University, Doha, Qatar
| |
Collapse
|
31
|
Qi F, Su H, Wang B, Qian L, Wang Y, Wang C, Hou Y, Chen P, Zhang Q, Li D, Tang H, Jiang J, Bian H, Chen Z, Zhang S. Hypoxia-activated ADCC-enhanced humanized anti-CD147 antibody for liver cancer imaging and targeted therapy with improved selectivity. MedComm (Beijing) 2024; 5:e512. [PMID: 38469549 PMCID: PMC10927247 DOI: 10.1002/mco2.512] [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: 11/26/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/13/2024] Open
Abstract
Therapeutic antibodies (Abs) improve the clinical outcome of cancer patients. However, on-target off-tumor toxicity limits Ab-based therapeutics. Cluster of differentiation 147 (CD147) is a tumor-associated membrane antigen overexpressed in cancer cells. Ab-based drugs targeting CD147 have achieved inadequate clinical benefits for liver cancer due to side effects. Here, by using glycoengineering and hypoxia-activation strategies, we developed a conditional Ab-dependent cellular cytotoxicity (ADCC)-enhanced humanized anti-CD147 Ab, HcHAb18-azo-PEG5000 (HAP18). Afucosylated ADCC-enhanced HcHAb18 Ab was produced by a fed-batch cell culture system. Azobenzene (Azo)-linked PEG5000 conjugation endowed HAP18 Ab with features of hypoxia-responsive delivery and selective targeting. HAP18 Ab potently inhibits the migration, invasion, and matrix metalloproteinase secretion, triggers the cytotoxicity and apoptosis of cancer cells, and induces ADCC, complement-dependent cytotoxicity, and Ab-dependent cellular phagocytosis under hypoxia. In xenograft mouse models, HAP18 Ab selectively targets hypoxic liver cancer tissues but not normal organs or tissues, and has potent tumor-inhibiting effects. HAP18 Ab caused negligible side effects and exhibited superior pharmacokinetics compared to those of parent HcHAb18 Ab. The hypoxia-activated ADCC-enhanced humanized HAP18 Ab safely confers therapeutic efficacy against liver cancer with improved selectivity. This study highlights that hypoxia activation is a promising strategy for improving the tumor targeting potential of anti-CD147 Ab drugs.
Collapse
Affiliation(s)
- Fang‐Zheng Qi
- Department of Cell Biology, School of MedicineNankai UniversityTianjinChina
| | - Hui‐Shan Su
- Department of Cell Biology, School of MedicineNankai UniversityTianjinChina
| | - Bo Wang
- Department of Cell Biology, School of MedicineNankai UniversityTianjinChina
| | - Luo‐Meng Qian
- Department of Cell Biology, School of MedicineNankai UniversityTianjinChina
| | - Yang Wang
- Department of Cell Biology, School of MedicineNankai UniversityTianjinChina
| | - Chen‐Hui Wang
- Department of Cell Biology, School of MedicineNankai UniversityTianjinChina
| | - Ya‐Xin Hou
- Department of Cell Biology, School of MedicineNankai UniversityTianjinChina
| | - Ping Chen
- National Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Qing Zhang
- National Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Dong‐Mei Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug ResearchNankai UniversityTianjinChina
| | - Hao Tang
- National Translational Science Center for Molecular Medicine, Department of Cell BiologyState Key Laboratory of Cancer BiologyAir Force Medical UniversityXi'anChina
| | - Jian‐Li Jiang
- National Translational Science Center for Molecular Medicine, Department of Cell BiologyState Key Laboratory of Cancer BiologyAir Force Medical UniversityXi'anChina
| | - Hui‐Jie Bian
- National Translational Science Center for Molecular Medicine, Department of Cell BiologyState Key Laboratory of Cancer BiologyAir Force Medical UniversityXi'anChina
| | - Zhi‐Nan Chen
- National Translational Science Center for Molecular Medicine, Department of Cell BiologyState Key Laboratory of Cancer BiologyAir Force Medical UniversityXi'anChina
| | - Si‐He Zhang
- Department of Cell Biology, School of MedicineNankai UniversityTianjinChina
| |
Collapse
|
32
|
Zhao Y, Raidas S, Mao Y, Li N. High-Throughput Glycan Profiling of Human Serum IgG Subclasses Using Parallel Reaction Monitoring Peptide Bond Fragmentation of Glycopeptides and Microflow LC-MS. J Proteome Res 2024; 23:585-595. [PMID: 38231888 DOI: 10.1021/acs.jproteome.3c00311] [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] [Indexed: 01/19/2024]
Abstract
LC-MS-based N-glycosylation profiling in four human serum IgG subclasses (IgG1, IgG2, IgG3, and IgG4) often requires additional affinity-based enrichment of specific IgG subclasses, owing to the high amino acid sequence similarity of Fc glycopeptides among subclasses. Notably, for IgG4 and the major allotype of IgG3, the glycopeptide precursors share identical retention time and mass and therefore cannot be distinguished based on precursor or glycan fragmentation. Here, we developed a parallel reaction monitoring (PRM)-based method for quantifying Fc glycopeptides through combined transitions generated from both glycosidic and peptide bond fragmentation. The latter enables the subpopulation of IgG3 and IgG4 to be directly distinguished according to mass differences without requiring further enrichment of specific IgG subclasses. In addition, a multinozzle electrospray emitter coupled to a capillary flow liquid chromatograph was used to increase the robustness and detection sensitivity of the method for low-yield peptide backbone fragment ions. The gradient was optimized to decrease the overall run time and make the method compatible with high-throughput analysis. We demonstrated that this method can be used to effectively monitor the relative levels of 13 representative glycoforms, with a good limit of detection for individual IgG subclasses.
Collapse
Affiliation(s)
- Yunlong Zhao
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, United States
| | - Shivkumar Raidas
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, United States
| | - Yuan Mao
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, United States
| | - Ning Li
- Analytical Chemistry, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, United States
| |
Collapse
|
33
|
Ma Y, Xiang Y, Li X, Zhang D, Chen Q. Recombinant streptococcal protein G-modified metal-organic framework ZIF-8 for the highly selective purification of immunoglobulin G from human serum. Anal Chim Acta 2024; 1288:342175. [PMID: 38220305 DOI: 10.1016/j.aca.2023.342175] [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: 10/07/2023] [Revised: 11/28/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024]
Abstract
A novel solid phase extractant His-rSPG@ZIF-8 was prepared by covalently coupling recombinant streptococcal protein G (His-rSPG) with ZIF-8. The His-rSPG@ZIF-8 composite was characterized by Fourier transform infrared spectroscopy (FT-IR), Raman spectroscopy (Raman), X-ray photoelectron spectroscopy (XPS), scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Due to the specific binding between the immunoglobulin binding region of His-rSPG and the Fc region of immunoglobulin G (IgG), the His-rSPG@ZIF-8 composite demonstrated exceptional selectivity in adsorbing IgG. In Britton-Robinson buffer (BR buffer) with a salt concentration of 500 mmol L-1 (0.04 mol L-1, pH 8.0), the His-rSPG@ZIF-8 composite exhibited a remarkable adsorption efficiency of 99.8 % for 0.05 mg of the composite on 200 μL of IgG solution (100 μg mL-1). The adsorption behavior of the His-rSPG@ZIF-8 composite aligns with the Langmuir adsorption model, and the theoretical maximum adsorption capacity is 1428.6 mg g-1. The adsorbed IgG molecules were successfully eluted using a SDS solution (0.5 %, m/m), resulting in a recovery rate of 91.2 %. Indeed, the His-rSPG@ZIF-8 composite was successfully utilized for the isolation and purification of IgG from human serum samples. The obtained IgG exhibited high purity, as confirmed by SDS-PAGE analysis. Additionally, LC-MS/MS analysis was employed to identify the human serum proteins following the adsorption and elution process using the His-rSPG@ZIF-8 composite material. The results revealed that the recovered solution contained an impressive content of immunoglobulin, accounting for 62.4 % of the total protein content. Furthermore, this process also led to the significant enrichment of low abundance proteins such as Serpin B4 and Cofilin-1. Consequently, the His-rSPG@ZIF-8 composite holds great promise for applications such as IgG purification and immunoassays. At the same time, it expands the application of metal-organic frameworks in the field of proteomics.
Collapse
Affiliation(s)
- Yufei Ma
- School of Pharmacy, Shenyang Medical College, Shenyang, 110034, People's Republic of China
| | - Yuhan Xiang
- School of Pharmacy, Shenyang Medical College, Shenyang, 110034, People's Republic of China
| | - Xin Li
- Department of Science and Technology, Shenyang Medical College, Shenyang, 110034, People's Republic of China.
| | - Dandan Zhang
- School of Public Health, Shenyang Medical College, Shenyang, 110034, People's Republic of China.
| | - Qing Chen
- School of Pharmacy, Shenyang Medical College, Shenyang, 110034, People's Republic of China.
| |
Collapse
|
34
|
Zhang M, Zhang Y, Wu H, Wang X, Zheng H, Feng J, Wang J, Luo L, Xiao H, Qiao C, Li X, Zheng Y, Huang W, Wang Y, Wang Y, Shi Y, Feng J, Chen G. Functional characterization of AF-04, an afucosylated anti-MARV GP antibody. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166964. [PMID: 37995774 DOI: 10.1016/j.bbadis.2023.166964] [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: 11/11/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
Marburg virus (MARV), one member of the Filoviridae family, cause sporadic outbreaks of hemorrhagic fever with high mortality rates. No countermeasures are currently available for the prevention or treatment of MARV infection. Monoclonal antibodies (mAbs) are promising candidates to display high neutralizing activity against MARV infection in vitro and in vivo. Recently, growing evidence has shown that immune effector function including antibody-dependent cell-mediated cytotoxicity (ADCC) is also required for in vivo efficacy of a panel of antibodies. Glyco-engineered methods are widely utilized to augment ADCC function of mAbs. In this study, we generated a fucose-knockout MARV GP-specific mAb named AF-04 and showed that afucosylation dramatically increased its binding affinity to polymorphic FcγRIIIa (F176/V176) compared with the parental AF-03. Accordingly, AF-04-mediated NK cell activation and NFAT expression downstream of FcγRIIIa in effector cells were also augmented. In conclusion, this work demonstrates that AF-04 represents a novel avenue for the treatment of MARV-caused disease.
Collapse
Affiliation(s)
- Min Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - Yuting Zhang
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010110, China
| | - Haiyan Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - Xinwei Wang
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010110, China
| | - Hang Zheng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010110, China
| | - Junjuan Feng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010110, China
| | - Jing Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - Longlong Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - He Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - Chunxia Qiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - Xinying Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - Yuanqiang Zheng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010110, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 102600, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 102600, China
| | - Yi Wang
- Department of Hematology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing 100071, China.
| | - Yanchun Shi
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010110, China.
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China.
| | - Guojiang Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China.
| |
Collapse
|
35
|
Morse JW, Gui X, Deng M, Huang R, Ye X, Zhao P, Fan X, Xiong W, Zhang C, Zhang N, An Z. Fc gamma receptors promote antibody-induced LILRB4 internalization and immune regulation of monocytic AML. Antib Ther 2024; 7:13-27. [PMID: 38235377 PMCID: PMC10791040 DOI: 10.1093/abt/tbad025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/16/2023] [Accepted: 10/29/2023] [Indexed: 01/19/2024] Open
Abstract
The immune checkpoint leukocyte immunoglobulin-like receptor B4 (LILRB4) is found specifically on the cell surface of acute monocytic leukemia (monocytic AML), an aggressive and common subtype of AML. We have developed a humanized monoclonal IgG1 LILRB4-blocking antibody (h128-3), which improved immune regulation but reduced cell surface expression of LILRB4 in monocytic AML models by 40-60%. Interestingly, most of this effect was neutralized by mutation of the Fc region of the antibody (h128-3/N297A), which prevents interaction with Fc gamma receptors (FcγRs). This suggested that there is FcγR-dependent antigenic modulation underlying h128-3's effects, a mechanism known to alter the function of antibodies targeting B-cell malignancies. We disrupted the Fc-FcγR interaction pharmacologically and with stable CRISPR-Cas9-mediated genetic knockout of FcγRs in monocytic AML cell lines to investigate the role of FcγR-dependent antigenic modulation in the regulation of LILRB4 by h128-3. When FcγRI is inhibited or removed from the surface of monocytic AML cells, h128-3 cannot optimally perform its blocking function, resulting in activation of the LILRB4 inhibitory receptor and leading to a 15-25% decrease in T-cell-mediated cytotoxicity in vitro. In the absence of FcγRI, scaffolding by FcγRIIa allows h128-3 to maintain LILRB4-blocking function. Here we define a FcγR-dependent antigenic modulation mechanism underlying the function of an immunoreceptor blocking antibody for the first time in myeloid malignancy. This research will facilitate the development of safe, precision-targeted antibody therapeutics in myeloid malignancies with greater potency and efficacy.
Collapse
Affiliation(s)
- Joshua W Morse
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, TX 77030, USA
| | - Xun Gui
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, TX 77030, USA
| | - Mi Deng
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ryan Huang
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Xiaohua Ye
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, TX 77030, USA
| | - Peng Zhao
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, TX 77030, USA
| | - Xuejun Fan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, TX 77030, USA
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, TX 77030, USA
| | - Chengcheng Zhang
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, TX 77030, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, TX 77030, USA
| |
Collapse
|
36
|
Barlan K, Bhide GP, White DR, Lake MR, Lu C, Rieder SE, Fan L, Hsieh CL. Genome-scale functional genomics screening highlights genes impacting protein fucosylation in Chinese hamster ovary cells. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:52-58. [PMID: 37844762 DOI: 10.1016/j.slasd.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 10/18/2023]
Abstract
N-linked glycosylation is a common post-translational modification that has various effects on multiple types of proteins. The extent to which an N-linked glycoprotein is modified and the identity of glycans species involved is of great interest to the biopharmaceutical industry, since glycosylation can impact the efficacy and safety of therapeutic monoclonal antibodies (mAbs). mAbs lacking core fucose, for example, display enhanced clinical efficacy through increased antibody-dependent cellular cytotoxicity. We performed a genome-wide CRISPR knockout screen in Chinese hamster ovary (CHO) cells, the workhorse cell culture system for industrial production of mAbs, aimed at identifying novel regulators of protein fucosylation. Using a lectin binding assay, we identified 224 gene perturbations that significantly alter protein fucosylation, including well-known glycosylation genes. This functional genomics framework could readily be extended and applied to study the genetic pathways involved in regulation of other glycoforms. We hope this resource will provide useful guidance toward the development of next generation CHO cell lines and mAb therapeutics.
Collapse
Affiliation(s)
- Kari Barlan
- Genomics Research Center, North Chicago, IL, United States
| | - Gaurang P Bhide
- Biologics Analytical Research and Development, Worcester, MA, United States
| | - Derek R White
- Biologics Production, AbbVie, North Chicago, IL, United States
| | - Marc R Lake
- Biologics Production, AbbVie, North Chicago, IL, United States
| | - Charles Lu
- Genomics Research Center, North Chicago, IL, United States
| | - Stephanie E Rieder
- Biologics Science and Technology, AbbVie Bioresearch Center, Worcester, MA, United States
| | - Lianchun Fan
- Biologics Science and Technology, AbbVie Bioresearch Center, Worcester, MA, United States
| | - Chen-Lin Hsieh
- Genomics Research Center, North Chicago, IL, United States.
| |
Collapse
|
37
|
Hale RC, Morais D, Chou J, Stowell SR. The role of glycosylation in clinical allergy and immunology. J Allergy Clin Immunol 2024; 153:55-66. [PMID: 37717626 PMCID: PMC10872775 DOI: 10.1016/j.jaci.2023.09.003] [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: 02/08/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/19/2023]
Abstract
While glycans are among the most abundant macromolecules on the cell with widespread functions, their role in immunity has historically been challenging to study. This is in part due to difficulties assimilating glycan analysis into routine approaches used to interrogate immune cell function. Despite this, recent developments have illuminated fundamental roles for glycans in host immunity. The growing field of glycoimmunology continues to leverage new tools and approaches to uncover the function of glycans and glycan-binding proteins in immunity. Here we utilize clinical vignettes to examine key roles of glycosylation in allergy, inborn errors of immunity, and autoimmunity. We will discuss the diverse functions of glycans as epitopes, as modulators of antibody function, and as regulators of immune cell function. Finally, we will highlight immune modulatory therapies that harness the critical role of glycans in the immune system.
Collapse
Affiliation(s)
- Rebecca C Hale
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass; Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Dominique Morais
- Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass.
| | - Sean R Stowell
- Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass; Harvard Glycomics Center, Harvard Medical School, Boston, Mass.
| |
Collapse
|
38
|
Kim HJ, Aktas O, Patterson KR, Korff S, Kunchok A, Bennett JL, Weinshenker BG, Paul F, Hartung H, Cimbora D, Smith MA, Mittereder N, Rees WA, She D, Cree BAC. Inebilizumab reduces neuromyelitis optica spectrum disorder risk independent of FCGR3A polymorphism. Ann Clin Transl Neurol 2023; 10:2413-2420. [PMID: 37804003 PMCID: PMC10723240 DOI: 10.1002/acn3.51911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/24/2023] [Accepted: 09/13/2023] [Indexed: 10/08/2023] Open
Abstract
Inebilizumab, a humanized, glycoengineered, IgG1 monoclonal antibody that depletes CD19+ B-cells, is approved to treat aquaporin 4 (AQP4) IgG-seropositive neuromyelitis optica spectrum disorder (NMOSD). Inebilizumab is afucosylated and engineered for enhanced affinity to Fc receptor III-A (FCGR3A) receptors on natural killer cells to maximize antibody-dependent cellular cytotoxicity. Previously, the F allele polymorphism at amino acid 158 of the FCGR3A gene (F158) was shown to decrease IgG-binding affinity and reduce rituximab (anti-CD20) efficacy for NMOSD attack prevention. In contrast, our current findings from inebilizumab-treated NMOSD patients indicate similar clinical outcomes between those with F158 and V158 allele genotypes.
Collapse
Affiliation(s)
- Ho Jin Kim
- Department of NeurologyResearch Institute and Hospital of National Cancer CenterGoyangSouth Korea
| | - Orhan Aktas
- Medical FacultyHeinrich Heine University DüsseldorfDüsseldorfGermany
| | | | | | - Amy Kunchok
- Department of NeurologyMellen Center for Multiple Sclerosis, Cleveland ClinicOhioClevelandUSA
| | - Jeffrey L. Bennett
- Department of Neurology, Programs in Neuroscience and ImmunologyUniversity of Colorado School of Medicine, Anschutz Medical CampusColoradoAuroraUSA
| | | | - Friedemann Paul
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and CharitéUniversitätsmedizin Berlin, Corporate Member of Freie Universitat Berlin and Humboldt‐Universitat zu BerlinBerlinGermany
| | - Hans‐Peter Hartung
- Medical FacultyHeinrich Heine University DüsseldorfDüsseldorfGermany
- Brain and Mind CentreUniversity of SydneyNew South WalesSydneyAustralia
- Department of NeurologyMedical University ViennaViennaAustria
- Department of NeurologyPalacky University in OlomoucOlomoucCzech Republic
| | | | | | | | | | - Dewei She
- Horizon TherapeuticsIllinoisDeerfieldUSA
| | - Bruce A. C. Cree
- Department of Neurology, UCSF Weill Institute for NeurosciencesUniversity of California San FranciscoCaliforniaSan FranciscoUSA
| |
Collapse
|
39
|
Gong YT, Zhang LJ, Liu YC, Tang M, Lin JY, Chen XY, Chen YX, Yan Y, Zhang WD, Jin JM, Luan X. Neutrophils as potential therapeutic targets for breast cancer. Pharmacol Res 2023; 198:106996. [PMID: 37972723 DOI: 10.1016/j.phrs.2023.106996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Breast cancer (BC) remains the foremost cause of cancer mortality globally, with neutrophils playing a critical role in its pathogenesis. As an essential tumor microenvironment (TME) component, neutrophils are emerging as pivotal factors in BC progression. Growing evidence has proved that neutrophils play a Janus- role in BC by polarizing into the anti-tumor (N1) or pro-tumor (N2) phenotype. Clinical trials are evaluating neutrophil-targeted therapies, including Reparixin (NCT02370238) and Tigatuzumab (NCT01307891); however, their clinical efficacy remains suboptimal. This review summarizes the evidence regarding the close relationship between neutrophils and BC, emphasizing the critical roles of neutrophils in regulating metabolic and immune pathways. Additionally, we summarize the existing therapeutic approaches that target neutrophils, highlighting the challenges, and affirming the rationale for continuing to explore neutrophils as a viable therapeutic target in BC management.
Collapse
Affiliation(s)
- Yi-Ting Gong
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Li-Jun Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi-Chen Liu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Min Tang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jia-Yi Lin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xin-Yi Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi-Xu Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yue Yan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wei-Dong Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Second Military Medical University, Shanghai 201203, China; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China.
| | - Jin-Mei Jin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
40
|
Guo Y, Hao Y, Shen L, Du Y, Wang X, Gao L, Feng X, Zhai Y, Liu Z, Xu E, Yang Y, Xi Y, Yang B, Zhang L. TSTA3 overexpression promotes malignant characteristics in LUSC by regulating LAMP2-mediated autophagy and tumor microenvironment. Cancer Cell Int 2023; 23:285. [PMID: 37986192 PMCID: PMC10662648 DOI: 10.1186/s12935-023-03109-z] [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: 09/11/2023] [Accepted: 10/25/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND TSTA3 gene encoding GDP-L-fucose synthase has recently been proved to be closely related to the prognosis of patients with various tumors. However, its role in lung cancer is still unclear. The purpose of this study is to explore the expression level, prognostic effect, potential function and mechanism of TSTA3 in lung cancer. METHODS Based on TCGA database, Kaplan-Meier and COX regression was used to analyze the relationship between TSTA3 expression and prognosis of lung cancer patients. Immunohistochemistry was used to determine the TSTA3 protein expression in lung cancer and normal tissues. The function of TSTA3 in lung squamous cell carcinoma (LUSC) cell was determined by CCK8, colony formation, transwell assay in vitro and subcutaneous xenografts in vivo. Transcriptome analysis, Lyso-Tracker Red staining and rescue experiment were used to explore the possible underlying mechanism. RESULTS The expression of TSTA3 was significantly increased in lung cancer, especially in LUSC, and was significantly correlated with the malignant characteristics of LUSC. COX regression analysis showed that the high expression of TSTA3 was an independent prognostic factor in LUSC patients. This was also confirmed by immunohistochemical staining. Compared with the control group, the proliferation, colony formation, invasion and migration ability of LUSC cells with TSTA3 overexpression was enhanced. Similarly, the ability of cell proliferation, colony formation, invasion and migration were weakened after transient knockdown of TSTA3. In vivo experiment showed that compared with control group, TSTA3 overexpression significantly promoted the growth of tumor and shortened survival time. In addition, transcriptome sequencing analysis showed that the differentially expressed genes between TSTA3 overexpression and control group was mainly concentrated in the lysosome pathway. Further study found that TSTA3 might affect the proliferation, invasion and migration of LUSC by regulating the expression of lysosome-associated membrane protein 2 (LAMP2) in LUSC. CONCLUSION The expression level of TSTA3 in LUSC is significantly higher than that in normal tissues. High expression of TSTA3 is associated with poor prognosis of LUSC patients. TSTA3 may affect the proliferation, invasion and migration of LUSC by regulating LAMP2.
Collapse
Affiliation(s)
- Yanlin Guo
- Basic Medical Sciences Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Yanlong Hao
- Basic Medical Sciences Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Liuyi Shen
- Basic Medical Sciences Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Yu Du
- Basic Medical Sciences Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Xiaohui Wang
- Basic Medical Sciences Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Lvye Gao
- Basic Medical Sciences Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Xuefei Feng
- Basic Medical Sciences Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Yuanfang Zhai
- Basic Medical Sciences Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Zhifei Liu
- Fifth Middle School of Taiyuan, Taiyuan, Shanxi, China
| | - Enwei Xu
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Yue Yang
- Department of Thoracic Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Yanfeng Xi
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Bin Yang
- Department of Thoracic Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, 030001, People's Republic of China.
| | - Ling Zhang
- Basic Medical Sciences Center of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China.
| |
Collapse
|
41
|
van Pul L, Maurer I, Boeser-Nunnink BD, Harskamp AM, van Dort KA, Kootstra NA. A genetic variation in fucosyltransferase 8 accelerates HIV-1 disease progression indicating a role for N-glycan fucosylation. AIDS 2023; 37:1959-1969. [PMID: 37598360 PMCID: PMC10552802 DOI: 10.1097/qad.0000000000003689] [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: 04/12/2023] [Revised: 07/25/2023] [Accepted: 08/07/2023] [Indexed: 08/22/2023]
Abstract
OBJECTIVES Core fucosylation by fucosyltransferase 8 (FUT8) is an important posttranslational modification that impacts components of the immune system. Genetic variations in FUT8 can alter its function and could, therefore, play a role in the antiviral immune response and pathogenesis of HIV-1. This study analysed the effect of a single nucleotide polymorphism (SNP) in FUT8 on the clinical course of HIV-1 infection. DESIGN/METHODS The effect of SNPs in FUT8 on untreated HIV-1 disease outcome were analysed in a cohort of 304 people with HIV-1 (PWH) using survival analysis. Flow-cytometry was used to determine the effect of SNP on T-cell activation, differentiation and exhaustion/senescence. T-cell function was determined by proliferation assay and by measuring intracellular cytokine production. The effect of the SNP on HIV-1 replication was determined by in-vitro HIV-1 infections. Sensitivity of HIV-1 produced in PBMC with or without the SNP to broadly neutralizing antibodies was determined using a TZM-bl based neutralization assay. RESULTS Presence of the minor allele of SNP rs4131564 was associated with accelerated disease progression. The SNP had no effect on T-cell activation and T-cell differentiation in PWH. Additionally, no differences in T-cell functionality as determined by proliferation and cytokine production was observed. HIV-1 replication and neutralization sensitivity was also unaffected by the SNP in FUT8. CONCLUSION SNP rs4131564 in FUT8 showed a major impact on HIV-1 disease course underscoring a role for N-glycan fucosylation even though no clear effect on the immune system or HIV-1 could be determined in vitro .
Collapse
Affiliation(s)
- Lisa van Pul
- Amsterdam Institute for Infection and Immunity
- Department of Experimental Immunology, Amsterdam UMC, location University of Amsterdam, Amsterdam, The Netherlands
| | - Irma Maurer
- Amsterdam Institute for Infection and Immunity
- Department of Experimental Immunology, Amsterdam UMC, location University of Amsterdam, Amsterdam, The Netherlands
| | - Brigitte D.M. Boeser-Nunnink
- Amsterdam Institute for Infection and Immunity
- Department of Experimental Immunology, Amsterdam UMC, location University of Amsterdam, Amsterdam, The Netherlands
| | - Agnes M. Harskamp
- Amsterdam Institute for Infection and Immunity
- Department of Experimental Immunology, Amsterdam UMC, location University of Amsterdam, Amsterdam, The Netherlands
| | - Karel A. van Dort
- Amsterdam Institute for Infection and Immunity
- Department of Experimental Immunology, Amsterdam UMC, location University of Amsterdam, Amsterdam, The Netherlands
| | - Neeltje A. Kootstra
- Amsterdam Institute for Infection and Immunity
- Department of Experimental Immunology, Amsterdam UMC, location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
42
|
Finotti G, Pietronigro E, Balanzin C, Lonardi S, Constantin G, Chao MP, Tecchio C, Vermi W, Cassatella MA. slan+ Monocytes Kill Cancer Cells Coated in Therapeutic Antibody by Trogoptosis. Cancer Immunol Res 2023; 11:1538-1552. [PMID: 37695535 DOI: 10.1158/2326-6066.cir-23-0239] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/04/2023] [Accepted: 09/01/2023] [Indexed: 09/12/2023]
Abstract
Monocytes positive for 6-Sulfo LacNAc (slan) are a major subset of nonclassical CD14dimCD16+ monocytes in humans. We have shown that slan+ cells infiltrate lymphomas and elicit an antibody-dependent cellular cytotoxicity (ADCC) of neoplastic B cells mediated by the anti-CD20 therapeutic rituximab. Herein, by performing blocking experiments and flow cytometry analyses, as well as confocal microscopy and live-cell imaging assays, we extended the findings to other humanized antibodies and deciphered the underlying effector mechanism(s). Specifically, we show that, after coculture with target cells coated with anti-CD20 or anti-CD38, slan+ monocytes mediate trogocytosis, a cell-cell contact dependent, antibody-mediated process that triggers an active, mechanic disruption of target cell membranes. Trogocytosis by slan+ monocytes leads to a necrotic type of target cell death known as trogoptosis, which, once initiated, was partially sustained by endogenous TNFα. We also found that slan+ monocytes, unlike natural killer (NK) cells, mediate a direct ADCC with all types of anti-CD47 analyzed, and this was independent of their IgG isotype. The latter findings unveil a potentially relevant contribution by slan+ monocytes in mediating the therapeutic efficacy of anti-CD47 in clinical practice, which could be particularly important when NK cells are exhausted or deficient in number. Overall, our observations shed new light on the cytotoxic mechanisms exerted by slan+ monocytes in antibody-dependent tumor cell targeting and advance our knowledge on how to expand our therapeutic arsenal for cancer therapy.
Collapse
Affiliation(s)
- Giulia Finotti
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Enrica Pietronigro
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Camillo Balanzin
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Silvia Lonardi
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gabriela Constantin
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Mark P Chao
- Division of Hematology, Stanford University, Stanford, California
| | - Cristina Tecchio
- Section of Hematology and Bone Marrow Transplant Unit, Department of Medicine, University of Verona, Verona, Italy
| | - William Vermi
- Section of Pathology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marco A Cassatella
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
43
|
Abramson HN. Immunotherapy of Multiple Myeloma: Current Status as Prologue to the Future. Int J Mol Sci 2023; 24:15674. [PMID: 37958658 PMCID: PMC10649824 DOI: 10.3390/ijms242115674] [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: 09/13/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
The landscape of therapeutic measures to treat multiple myeloma has undergone a seismic shift since the dawn of the current century. This has been driven largely by the introduction of new classes of small molecules, such as proteasome blockers (e.g., bortezomib) and immunomodulators (e.g., lenalidomide), as well as by immunotherapeutic agents starting with the anti-CD38 monoclonal antibody daratumumab in 2015. Recently, other immunotherapies have been added to the armamentarium of drugs available to fight this malignancy. These include the bispecifics teclistamab, talquetamab, and elranatamab, and the chimeric antigen receptor (CAR) T-cell products idecabtagene vicleucel (ide-cel) and ciltacabtagene autoleucel (cilta-cel). While the accumulated benefits of these newer agents have resulted in a more than doubling of the disease's five-year survival rate to nearly 60% and improved quality of life, the disease remains incurable, as patients become refractory to the drugs and experience relapse. This review covers the current scope of antimyeloma immunotherapeutic agents, both those in clinical use and in development. Included in the discussion are additional monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), bi- and multitargeted mAbs, and CAR T-cells and emerging natural killer (NK) cells, including products intended for "off-the-shelf" (allogeneic) applications. Emphasis is placed on the benefits of each along with the challenges that need to be surmounted if MM is to be cured.
Collapse
Affiliation(s)
- Hanley N Abramson
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
44
|
Rocamora F, Peralta AG, Shin S, Sorrentino J, Wu MYM, Toth EA, Fuerst TR, Lewis NE. Glycosylation shapes the efficacy and safety of diverse protein, gene and cell therapies. Biotechnol Adv 2023; 67:108206. [PMID: 37354999 PMCID: PMC11168894 DOI: 10.1016/j.biotechadv.2023.108206] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/26/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023]
Abstract
Over recent decades, therapeutic proteins have had widespread success in treating a myriad of diseases. Glycosylation, a near universal feature of this class of drugs, is a critical quality attribute that significantly influences the physical properties, safety profile and biological activity of therapeutic proteins. Optimizing protein glycosylation, therefore, offers an important avenue to developing more efficacious therapies. In this review, we discuss specific examples of how variations in glycan structure and glycoengineering impacts the stability, safety, and clinical efficacy of protein-based drugs that are already in the market as well as those that are still in preclinical development. We also highlight the impact of glycosylation on next generation biologics such as T cell-based cancer therapy and gene therapy.
Collapse
Affiliation(s)
- Frances Rocamora
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Angelo G Peralta
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Seunghyeon Shin
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - James Sorrentino
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mina Ying Min Wu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eric A Toth
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA
| | - Thomas R Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
45
|
García-Alija M, van Moer B, Sastre DE, Azzam T, Du JJ, Trastoy B, Callewaert N, Sundberg EJ, Guerin ME. Modulating antibody effector functions by Fc glycoengineering. Biotechnol Adv 2023; 67:108201. [PMID: 37336296 PMCID: PMC11027751 DOI: 10.1016/j.biotechadv.2023.108201] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Antibody based drugs, including IgG monoclonal antibodies, are an expanding class of therapeutics widely employed to treat cancer, autoimmune and infectious diseases. IgG antibodies have a conserved N-glycosylation site at Asn297 that bears complex type N-glycans which, along with other less conserved N- and O-glycosylation sites, fine-tune effector functions, complement activation, and half-life of antibodies. Fucosylation, galactosylation, sialylation, bisection and mannosylation all generate glycoforms that interact in a specific manner with different cellular antibody receptors and are linked to a distinct functional profile. Antibodies, including those employed in clinical settings, are generated with a mixture of glycoforms attached to them, which has an impact on their efficacy, stability and effector functions. It is therefore of great interest to produce antibodies containing only tailored glycoforms with specific effects associated with them. To this end, several antibody engineering strategies have been developed, including the usage of engineered mammalian cell lines, in vitro and in vivo glycoengineering.
Collapse
Affiliation(s)
- Mikel García-Alija
- Structural Glycobiology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia 48903, Spain
| | - Berre van Moer
- VIB Center for Medical Biotechnology, VIB, Zwijnaarde, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium
| | - Diego E Sastre
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tala Azzam
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jonathan J Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Beatriz Trastoy
- Structural Glycoimmunology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia, 48903, Spain; Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain.
| | - Nico Callewaert
- VIB Center for Medical Biotechnology, VIB, Zwijnaarde, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium.
| | - Eric J Sundberg
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Marcelo E Guerin
- Structural Glycobiology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia 48903, Spain; Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain.
| |
Collapse
|
46
|
Shivatare VS, Huang HW, Tseng TH, Chuang PK, Zeng YF, Wong CH. Probing the Internalization and Efficacy of Antibody-Drug Conjugate via Site-Specific Fc-Glycan Labelling of a Homogeneous Antibody Targeting SSEA-4 Bearing Tumors. Isr J Chem 2023; 63:e202300042. [PMID: 38348405 PMCID: PMC10861153 DOI: 10.1002/ijch.202300042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Indexed: 02/15/2024]
Abstract
Antibody drug conjugates (ADC) are an emerging class of pharmaceuticals consisting of cytotoxic agents covalently attached to an antibody designed to target a specific cancer cell surface molecule followed by internalization and intracellular release of payload to exhibit its anticancer activity. Targeted delivery of cytotoxic payload to a variety of specific cells has been demonstrated to have significant enhancement in clinical efficacy and dramatic reduction in off-target toxicity. Site-specific conjugation of payload to the antibody is highly desirable for development of ADC with well-defined antibody-to-drug ratio, enhanced internalization, reduced toxicity, improved stability, desired pharmacological profile and optimal therapeutic index. Here, we reported a site-specific conjugation strategy for evaluation of antibody internalization and efficacy of ADC designed to target SSEA4 on solid tumors. This strategy stems from the azido-fucose tag of a homogeneous antibody Fc-glycan generated via in vitro glycoengineering approach for site-specific conjugation and optimization of antibody-drug ratio to exhibit optimal efficacy. The ADC consisting of a chimeric anti-SSEA4 antibody chMC813-70, conjugated to the antineo-plastic agent monomethyl auristatin E via both cleavable and non-cleavable linkers showed excellent cytotoxicity profile towards SSEA4-bearing cancer cells. A clear distinction in cytotoxicity was observed among cancer cells with different SSEA4 expression levels.
Collapse
Affiliation(s)
- Vidya S Shivatare
- The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, USA
| | - Han-Wen Huang
- The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, USA
| | - Tzu-Hao Tseng
- The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, USA
| | - Po-Kai Chuang
- The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, USA
| | - Yi-Fang Zeng
- The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, USA
| | - Chi-Huey Wong
- The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, USA
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
47
|
Nguyen NTB, Leung HW, Pang KT, Tay SJ, Walsh I, Choo ABH, Yang Y. Optimizing effector functions of monoclonal antibodies via tailored N-glycan engineering using a dual landing pad CHO targeted integration platform. Sci Rep 2023; 13:15620. [PMID: 37731040 PMCID: PMC10511539 DOI: 10.1038/s41598-023-42925-1] [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: 03/10/2023] [Accepted: 09/16/2023] [Indexed: 09/22/2023] Open
Abstract
Monoclonal antibodies (mAbs) eliminate cancer cells via various effector mechanisms including antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), which are influenced by the N-glycan structures on the Fc region of mAbs. Manipulating these glycan structures on mAbs allows for optimization of therapeutic benefits associated with effector functions. Traditional approaches such as gene deletion or overexpression often lead to only all-or-nothing changes in gene expression and fail to modulate the expression of multiple genes at defined ratios and levels. In this work, we have developed a CHO cell engineering platform enabling modulation of multiple gene expression to tailor the N-glycan profiles of mAbs for enhanced effector functions. Our platform involves a CHO targeted integration platform with two independent landing pads, allowing expression of multiple genes at two pre-determined genomic sites. By combining with internal ribosome entry site (IRES)-based polycistronic vectors, we simultaneously modulated the expression of α-mannosidase II (MANII) and chimeric β-1,4-N-acetylglucosaminyl-transferase III (cGNTIII) genes in CHO cells. This strategy enabled the production of mAbs carrying N-glycans with various levels of bisecting and non-fucosylated structures. Importantly, these engineered mAbs exhibited different degrees of effector cell activation and CDC, facilitating the identification of mAbs with optimal effector functions. This platform was demonstrated as a powerful tool for producing antibody therapeutics with tailored effector functions via precise engineering of N-glycan profiles. It holds promise for advancing the field of metabolic engineering in mammalian cells.
Collapse
Affiliation(s)
- Ngan T B Nguyen
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Hau Wan Leung
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Kuin Tian Pang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Shi Jie Tay
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Ian Walsh
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Andre B H Choo
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore
| | - Yuansheng Yang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore, 138668, Singapore.
| |
Collapse
|
48
|
Juric V, Mayes E, Binnewies M, Lee T, Canaday P, Pollack JL, Rudolph J, Du X, Liu VM, Dash S, Palmer R, Jahchan NS, Ramoth ÅJ, Lacayo S, Mankikar S, Norng M, Brassell C, Pal A, Chan C, Lu E, Sriram V, Streuli M, Krummel MF, Baker KP, Liang L. TREM1 activation of myeloid cells promotes antitumor immunity. Sci Transl Med 2023; 15:eadd9990. [PMID: 37647386 DOI: 10.1126/scitranslmed.add9990] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/02/2023] [Indexed: 09/01/2023]
Abstract
Myeloid cells in the tumor microenvironment (TME) can exist in immunosuppressive and immunostimulatory states that impede or promote antitumor immunity, respectively. Blocking suppressive myeloid cells or increasing stimulatory cells to enhance antitumor immune responses is an area of interest for therapeutic intervention. Triggering receptor expressed on myeloid cells-1 (TREM1) is a proinflammatory receptor that amplifies immune responses. TREM1 is expressed on neutrophils, subsets of monocytes and tissue macrophages, and suppressive myeloid populations in the TME, including tumor-associated neutrophils, monocytes, and tumor-associated macrophages. Depletion or inhibition of immunosuppressive myeloid cells, or stimulation by TREM1-mediated inflammatory signaling, could be used to promote an immunostimulatory TME. We developed PY159, an afucosylated humanized anti-TREM1 monoclonal antibody with enhanced FcγR binding. PY159 is a TREM1 agonist that induces signaling, leading to up-regulation of costimulatory molecules on monocytes and macrophages, production of proinflammatory cytokines and chemokines, and enhancement of T cell activation in vitro. An antibody against mouse TREM1, PY159m, promoted antitumor efficacy in syngeneic mouse tumor models. These results suggest that PY159-mediated agonism of TREM1 on tumoral myeloid cells can promote a proinflammatory TME and offer a promising strategy for immunotherapy.
Collapse
Affiliation(s)
- Vladislava Juric
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Erin Mayes
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Mikhail Binnewies
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Tian Lee
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Pamela Canaday
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Joshua L Pollack
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Joshua Rudolph
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Xiaoyan Du
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Victoria M Liu
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Subhadra Dash
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Rachael Palmer
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Nadine S Jahchan
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Åsa Johanna Ramoth
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Sergio Lacayo
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Shilpa Mankikar
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Manith Norng
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Chris Brassell
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Aritra Pal
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Christopher Chan
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Erick Lu
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Venkataraman Sriram
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Michel Streuli
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kevin P Baker
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Linda Liang
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| |
Collapse
|
49
|
Smith FD, Pierce RH, Thisted T, van der Horst EH. Conditionally Active, pH-Sensitive Immunoregulatory Antibodies Targeting VISTA and CTLA-4 Lead an Emerging Class of Cancer Therapeutics. Antibodies (Basel) 2023; 12:55. [PMID: 37753969 PMCID: PMC10525963 DOI: 10.3390/antib12030055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Immune checkpoints and other immunoregulatory targets can be difficult to precisely target due to expression on non-tumor immune cells critical to maintaining immune homeostasis in healthy tissues. On-target/off-tumor binding of therapeutics results in significant pharmacokinetic and pharmacodynamic problems. Target-mediated drug disposition (TMDD) significantly limits effective intratumoral drug levels and adversely affects anti-tumor efficacy. Target engagement outside the tumor environment may lead to severe immune-related adverse events (irAEs), resulting in a narrowing of the therapeutic window, sub-optimal dosing, or cessation of drug development altogether. Overcoming these challenges has become tractable through recent advances in antibody engineering and screening approaches. Here, we review the discovery and development of conditionally active antibodies with minimal binding to target at physiologic pH but high-affinity target binding at the low pH of the tumor microenvironment by focusing on the discovery and improved properties of pH-dependent mAbs targeting two T cell checkpoints, VISTA and CTLA-4.
Collapse
Affiliation(s)
- F. Donelson Smith
- Sensei Biotherapeutics, Inc., 1405 Research Blvd., Suite 125, Rockville, MD 20850, USA;
| | | | - Thomas Thisted
- Sensei Biotherapeutics, Inc., 1405 Research Blvd., Suite 125, Rockville, MD 20850, USA;
| | | |
Collapse
|
50
|
Zhang M, Lam KP, Xu S. Natural Killer Cell Engagers (NKCEs): a new frontier in cancer immunotherapy. Front Immunol 2023; 14:1207276. [PMID: 37638058 PMCID: PMC10450036 DOI: 10.3389/fimmu.2023.1207276] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/03/2023] [Indexed: 08/29/2023] Open
Abstract
Natural Killer (NK) cells are a type of innate lymphoid cells that play a crucial role in immunity by killing virally infected or tumor cells and secreting cytokines and chemokines. NK cell-mediated immunotherapy has emerged as a promising approach for cancer treatment due to its safety and effectiveness. NK cell engagers (NKCEs), such as BiKE (bispecific killer cell engager) or TriKE (trispecific killer cell engager), are a novel class of antibody-based therapeutics that exhibit several advantages over other cancer immunotherapies harnessing NK cells. By bridging NK and tumor cells, NKCEs activate NK cells and lead to tumor cell lysis. A growing number of NKCEs are currently undergoing development, with some already in clinical trials. However, there is a need for more comprehensive studies to determine how the molecular design of NKCEs affects their functionality and manufacturability, which are crucial for their development as off-the-shelf drugs for cancer treatment. In this review, we summarize current knowledge on NKCE development and discuss critical factors required for the production of effective NKCEs.
Collapse
Affiliation(s)
- Minchuan Zhang
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Kong-Peng Lam
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore, Singapore
| | - Shengli Xu
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|