1
|
Lin SY, Schmidt EN, Takahashi-Yamashiro K, Macauley MS. Roles for Siglec-glycan interactions in regulating immune cells. Semin Immunol 2024; 77:101925. [PMID: 39706106 DOI: 10.1016/j.smim.2024.101925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
Cell surface complex carbohydrates, known as glycans, are positioned to be the first point of contact between two cells. Indeed, interactions between glycans with glycan-binding can modulate cell-cell interactions. This concept is particularly relevant for immune cells, which use an array of glycan-binding proteins to help in the process of differentiating 'self' from 'non-self'. This is exemplified by the sialic acid-binding immunoglobulin-type lectins (Siglecs), which recognize sialic acid. Given that sialic acid is relatively unique to vertebrates, immune cells leverage Siglecs to recognize sialic acid as a marker of 'self'. Siglecs serve many biological roles, with most of these functions regulated through interactions with their sialoglycan ligands. In this review, we provide a comprehensive update on the ligands of Siglecs and how Siglec-sialoglycan interactions help regulate immune cells in the adaptive and innate immune system.
Collapse
Affiliation(s)
- Sung-Yao Lin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Edward N Schmidt
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | | | - Matthew S Macauley
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada; Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada; Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
2
|
Tu H, Yuan L, Ni B, Lin Y, Wang K. Siglecs-mediated immune regulation in neurological disorders. Pharmacol Res 2024; 210:107531. [PMID: 39615617 DOI: 10.1016/j.phrs.2024.107531] [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: 07/25/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024]
Abstract
The surfaces of various immune cells are rich in glycan chains, including the sialic-acid-binding immunoglobulin-like lectins (Siglecs) family. As an emerging glyco-immune checkpoint, Siglecs have the ability to bind and interact with various glycoproteins, thereby eliciting a series of downstream reactions to modulate the immune response. The impact of Siglecs has been extensively studied in tumor immunotherapy. However, research in neurological disorders and neurological diseases is very limited, and therapeutic options involving Siglecs need further exploration. Siglecs play a crucial role in the development, homeostasis, and repair processes of the nervous system, especially in degenerative diseases. This review summarizes studies on the immunomodulatory role mediated by Siglecs expressed on different immune cells in various neurological disorders, elucidates how dysregulated sialic acid contributes to several psychiatric disorders, and discusses the progress and limitations of research on the treatment of neurological disorders.
Collapse
Affiliation(s)
- Huifang Tu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Limei Yuan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Bo Ni
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yufeng Lin
- Department of Neurology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300190, China.
| | - Kaiyuan Wang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| |
Collapse
|
3
|
Murphy PV, Dhara A, Fitzgerald LS, Hever E, Konda S, Mandal K. Small lectin ligands as a basis for applications in glycoscience and glycomedicine. Chem Soc Rev 2024; 53:9428-9445. [PMID: 39162695 DOI: 10.1039/d4cs00642a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Glycan recognition by lectins mediates important biological events. This Tutorial Review aims to introduce lectin-ligand interactions and show how these molecular recognition events inspire innovations such as: (i) glycomimetic ligands; (ii) multivalent ligand agonists/antagonists; (iii) ligands for precision delivery of therapies to cells, where therapies include vaccines, siRNA and LYTACs (iv) development of diagnostics. A small number of case studies are selected to demonstrate principles for development of new ligands for applications inspired by knowledge of natural glycan ligand structure and function.
Collapse
Affiliation(s)
- Paul V Murphy
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
- SSPC, SFI Research Centre for Pharmaceuticals, Galway, H91TK33, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, University Road, Galway, H91TK33, Ireland
| | - Ashis Dhara
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
- SSPC, SFI Research Centre for Pharmaceuticals, Galway, H91TK33, Ireland
| | - Liam S Fitzgerald
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
- SSPC, SFI Research Centre for Pharmaceuticals, Galway, H91TK33, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, University Road, Galway, H91TK33, Ireland
| | - Eoin Hever
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
| | - Saidulu Konda
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
| | - Kishan Mandal
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
| |
Collapse
|
4
|
Irons EE, Sajina GC, Lau JT. Sialic acid in the regulation of blood cell production, differentiation and turnover. Immunology 2024; 172:517-532. [PMID: 38503445 PMCID: PMC11223974 DOI: 10.1111/imm.13780] [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: 11/27/2023] [Accepted: 03/04/2024] [Indexed: 03/21/2024] Open
Abstract
Sialic acid is a unique sugar moiety that resides in the distal and most accessible position of the glycans on mammalian cell surface and extracellular glycoproteins and glycolipids. The potential for sialic acid to obscure underlying structures has long been postulated, but the means by which such structural changes directly affect biological processes continues to be elucidated. Here, we appraise the growing body of literature detailing the importance of sialic acid for the generation, differentiation, function and death of haematopoietic cells. We conclude that sialylation is a critical post-translational modification utilized in haematopoiesis to meet the dynamic needs of the organism by enforcing rapid changes in availability of lineage-specific cell types. Though long thought to be generated only cell-autonomously within the intracellular ER-Golgi secretory apparatus, emerging data also demonstrate previously unexpected diversity in the mechanisms of sialylation. Emphasis is afforded to the mechanism of extrinsic sialylation, whereby extracellular enzymes remodel cell surface and extracellular glycans, supported by charged sugar donor molecules from activated platelets.
Collapse
Affiliation(s)
| | | | - Joseph T.Y. Lau
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203 USA
| |
Collapse
|
5
|
Rajagopal D, MacLeod E, Corogeanu D, Vessillier S. Immune-related adverse events of antibody-based biological medicines in cancer therapy. J Cell Mol Med 2024; 28:e18470. [PMID: 38963257 PMCID: PMC11223167 DOI: 10.1111/jcmm.18470] [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: 01/19/2024] [Revised: 05/03/2024] [Accepted: 05/22/2024] [Indexed: 07/05/2024] Open
Abstract
Recombinant antibodies (Abs) are an integral modality for the treatment of multiple tumour malignancies. Since the Food and Drug Administration (FDA) approval of rituximab as the first monoclonal antibody (mAb) for cancer treatment, several mAbs and antibody (Ab)-based therapies have been approved for the treatment of solid tumour malignancies and other cancers. These Abs function by either blocking oncogenic pathways or angiogenesis, modulating immune response, or by delivering a conjugated drug. The use of Ab-based therapy in cancer patients who could benefit from the treatment, however, is still limited by associated toxicity profiles which may stem from biological features and processes related to target binding, alongside biochemical and/or biophysical characteristics of the therapeutic Ab. A significant immune-related adverse event (irAE) associated with Ab-based therapies is cytokine release syndrome (CRS), characterized by the development of fever, rash and even marked, life-threatening hypotension, and acute inflammation with secondary to systemic uncontrolled increase in a range of pro-inflammatory cytokines. Here, we review irAEs associated with specific classes of approved, Ab-based novel cancer immunotherapeutics, namely immune checkpoint (IC)-targeting Abs, bispecific Abs (BsAbs) and Ab-drug-conjugates (ADCs), highlighting the significance of harmonization in preclinical assay development for safety assessment of Ab-based biotherapeutics as an approach to support and refine clinical translation.
Collapse
Affiliation(s)
- Deepa Rajagopal
- Immunotherapy, Biotherapeutics and Advanced Therapies Division, Science, Research, and Innovation Group, Medicines and Healthcare products Regulatory Agency (MHRA)HertfordshireUK
| | - Elliot MacLeod
- Immunotherapy, Biotherapeutics and Advanced Therapies Division, Science, Research, and Innovation Group, Medicines and Healthcare products Regulatory Agency (MHRA)HertfordshireUK
- Present address:
Gilead Sciences, Winchester HouseOxfordUK
| | - Diana Corogeanu
- Immunotherapy, Biotherapeutics and Advanced Therapies Division, Science, Research, and Innovation Group, Medicines and Healthcare products Regulatory Agency (MHRA)HertfordshireUK
- Present address:
East Sussex Healthcare NHS Trust, Conquest HospitalEast SussexUK
| | - Sandrine Vessillier
- Immunotherapy, Biotherapeutics and Advanced Therapies Division, Science, Research, and Innovation Group, Medicines and Healthcare products Regulatory Agency (MHRA)HertfordshireUK
| |
Collapse
|
6
|
Vaughan HJ, Est-Witte S, Dockery LT, Urello MA, Boyd J, Keyser BD, Zhuang L, Marelli M, Christie RJ. A high-throughput lysosome trafficking assay guides ligand selection and elucidates differences in CD22-targeted nanodelivery. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2351791. [PMID: 38817250 PMCID: PMC11138227 DOI: 10.1080/14686996.2024.2351791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/30/2024] [Indexed: 06/01/2024]
Abstract
Targeted nanoparticles offer potential to selectively deliver therapeutics to cells; however, their subcellular fate following endocytosis must be understood to properly design mechanisms of drug release. Here we describe a nanoparticle platform and associated cell-based assay to observe lysosome trafficking of targeted nanoparticles in live cells. The nanoparticle platform utilizes two fluorescent dyes loaded onto PEG-poly(glutamic acid) and PEG-poly(Lysine) block co-polymers that also comprise azide reactive handles on PEG termini to attach antibody-based targeting ligands. Fluorophores were selected to be pH-sensitive (pHrodo Red) or pH-insensitive (Alexafluor 488) to report when nanoparticles enter low pH lysosomes. Dye-labelled block co-polymers were further assembled into polyion complex micelle nanoparticles and crosslinked through amide bond formation to form stable nano-scaffolds for ligand attachment. Cell binding and lysosome trafficking was determined in live cells by fluorescence imaging in 96-well plates and quantification of red- and green-fluorescence signals over time. The platform and assay was validated for selection of optimal antibody-derived targeting ligands directed towards CD22 for nanoparticle delivery. Kinetic analysis of uptake and lysosome trafficking indicated differences between ligand types and the ligand with the highest lysosome trafficking efficiency translated into effective DNA delivery with nanoparticles bearing the optimal ligand.
Collapse
Affiliation(s)
- Hannah J. Vaughan
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Lance T. Dockery
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Morgan A. Urello
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Jonathan Boyd
- Discovery Sciences, BioPharma R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Li Zhuang
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Marcello Marelli
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - R. James Christie
- Biologics Engineering, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
7
|
Lim HJ, McWilliam HEG. Quantitative Measurement of Plasma Membrane Protein Internalisation and Recycling in Heterogenous Cellular Samples by Flow Cytometry. Bio Protoc 2024; 14:e4986. [PMID: 38737503 PMCID: PMC11082785 DOI: 10.21769/bioprotoc.4986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
Plasma membrane proteins mediate important aspects of physiology, including nutrient acquisition, cell-cell interactions, and monitoring homeostasis. The trafficking of these proteins, involving internalisation from and/or recycling back to the cell surface, is often critical to their functions. These processes can vary among different proteins and cell types and states and are still being elucidated. Current strategies to measure surface protein internalisation and recycling are typically microscopy or biochemical assays; these are accurate but generally limited to analysing a homogenous cell population and are often low throughput. Here, we present flow cytometry-based methods involving probe-conjugated antibodies that enable quantification of internalisation or recycling rates at the single-cell level in complex samples. To measure internalisation, we detail an assay where the protein of interest is labelled with a specific antibody conjugated to a fluorescent oligonucleotide-labelled probe. To measure recycling, a specific antibody conjugated to a cleavable biotin group is employed. These probes permit the differentiation of molecules that have been internalised or recycled from those that have not. When combined with cell-specific marker panels, these methods allow the quantitative study of plasma membrane protein trafficking dynamics in a heterogenous cell mixture at the single-cell level. Key features • These assays allow sensitive quantification of internalised or recycled surface molecules using oligonucleotide or cleavable biotin-conjugated probes, respectively, and detected by flow cytometry. • They can be adapted to any membrane protein that transits via the cell surface and for which a specific purified antibody is available. • The dynamics of a cell surface protein can be measured in heterogenous cell populations simultaneously, including various cellular activation states. • The internalisation assay builds upon the method developed by Liu et al. [1,2] and extends its application to heterogenous human peripheral blood mononuclear cells. • These assays have been extensively used on suspension cells but have not been tested on adherent cells.
Collapse
Affiliation(s)
- Hui Jing Lim
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hamish E. G. McWilliam
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
8
|
Wang Y, Xu Z, Wu KL, Yu L, Wang C, Ding H, Gao Y, Sun H, Wu YH, Xia M, Chen Y, Xiao H. Siglec-15/sialic acid axis as a central glyco-immune checkpoint in breast cancer bone metastasis. Proc Natl Acad Sci U S A 2024; 121:e2312929121. [PMID: 38252825 PMCID: PMC10835054 DOI: 10.1073/pnas.2312929121] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024] Open
Abstract
Immunotherapy is a promising approach for treating metastatic breast cancer (MBC), offering new possibilities for therapy. While checkpoint inhibitors have shown great progress in the treatment of metastatic breast cancer, their effectiveness in patients with bone metastases has been disappointing. This lack of efficacy seems to be specific to the bone environment, which exhibits immunosuppressive features. In this study, we elucidate the multiple roles of the sialic acid-binding Ig-like lectin (Siglec)-15/sialic acid glyco-immune checkpoint axis in the bone metastatic niche and explore potential therapeutic strategies targeting this glyco-immune checkpoint. Our research reveals that elevated levels of Siglec-15 in the bone metastatic niche can promote tumor-induced osteoclastogenesis as well as suppress antigen-specific T cell responses. Next, we demonstrate that antibody blockade of the Siglec-15/sialic acid glyco-immune checkpoint axis can act as a potential treatment for breast cancer bone metastasis. By targeting this pathway, we not only aim to treat bone metastasis but also inhibit the spread of metastatic cancer cells from bone lesions to other organs.
Collapse
Affiliation(s)
- Yixian Wang
- Department of Chemistry, Rice University, Houston, TX77005
| | - Zhan Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX77030
| | - Kuan-Lin Wu
- Department of Chemistry, Rice University, Houston, TX77005
| | - Liqun Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX77030
| | - Chenhang Wang
- Department of Chemistry, Rice University, Houston, TX77005
| | - Haoxue Ding
- Department of Chemistry, Rice University, Houston, TX77005
| | - Yang Gao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX77030
| | - Han Sun
- Department of Chemistry, Rice University, Houston, TX77005
| | - Yi-Hsuan Wu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX77030
| | - Meng Xia
- Department of Chemistry, Rice University, Houston, TX77005
| | - Yuda Chen
- Department of Chemistry, Rice University, Houston, TX77005
| | - Han Xiao
- Department of Chemistry, Rice University, Houston, TX77005
- Department of Biosciences, Rice University, Houston, TX77005
- Department of Bioengineering, Rice University, Houston, TX77005
| |
Collapse
|
9
|
Clemente B, Denis M, Silveira CP, Schiavetti F, Brazzoli M, Stranges D. Straight to the point: targeted mRNA-delivery to immune cells for improved vaccine design. Front Immunol 2023; 14:1294929. [PMID: 38090568 PMCID: PMC10711611 DOI: 10.3389/fimmu.2023.1294929] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
With the deepening of our understanding of adaptive immunity at the cellular and molecular level, targeting antigens directly to immune cells has proven to be a successful strategy to develop innovative and potent vaccines. Indeed, it offers the potential to increase vaccine potency and/or modulate immune response quality while reducing off-target effects. With mRNA-vaccines establishing themselves as a versatile technology for future applications, in the last years several approaches have been explored to target nanoparticles-enabled mRNA-delivery systems to immune cells, with a focus on dendritic cells. Dendritic cells (DCs) are the most potent antigen presenting cells and key mediators of B- and T-cell immunity, and therefore considered as an ideal target for cell-specific antigen delivery. Indeed, improved potency of DC-targeted vaccines has been proved in vitro and in vivo. This review discusses the potential specific targets for immune system-directed mRNA delivery, as well as the different targeting ligand classes and delivery systems used for this purpose.
Collapse
|
10
|
Hulbert SW, Desai P, Jewett MC, DeLisa MP, Williams AJ. Glycovaccinology: The design and engineering of carbohydrate-based vaccine components. Biotechnol Adv 2023; 68:108234. [PMID: 37558188 DOI: 10.1016/j.biotechadv.2023.108234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/12/2023] [Accepted: 08/05/2023] [Indexed: 08/11/2023]
Abstract
Vaccines remain one of the most important pillars in preventative medicine, providing protection against a wide array of diseases by inducing humoral and/or cellular immunity. Of the many possible candidate antigens for subunit vaccine development, carbohydrates are particularly appealing because of their ubiquitous presence on the surface of all living cells, viruses, and parasites as well as their known interactions with both innate and adaptive immune cells. Indeed, several licensed vaccines leverage bacterial cell-surface carbohydrates as antigens for inducing antigen-specific plasma cells secreting protective antibodies and the development of memory T and B cells. Carbohydrates have also garnered attention in other aspects of vaccine development, for example, as adjuvants that enhance the immune response by either activating innate immune responses or targeting specific immune cells. Additionally, carbohydrates can function as immunomodulators that dampen undesired humoral immune responses to entire protein antigens or specific, conserved regions on antigenic proteins. In this review, we highlight how the interplay between carbohydrates and the adaptive and innate arms of the immune response is guiding the development of glycans as vaccine components that act as antigens, adjuvants, and immunomodulators. We also discuss how advances in the field of synthetic glycobiology are enabling the design, engineering, and production of this new generation of carbohydrate-containing vaccine formulations with the potential to prevent infectious diseases, malignancies, and complex immune disorders.
Collapse
Affiliation(s)
- Sophia W Hulbert
- Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA
| | - Primit Desai
- 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 F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; Cornell Institute of Biotechnology, Cornell University, Ithaca, NY 14853, USA.
| | - Asher J Williams
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; Department of Chemical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
11
|
O'Sullivan JA, Youngblood BA, Schleimer RP, Bochner BS. Siglecs as potential targets of therapy in human mast cell- and/or eosinophil-associated diseases. Semin Immunol 2023; 69:101799. [PMID: 37413923 PMCID: PMC10528103 DOI: 10.1016/j.smim.2023.101799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Siglecs (sialic acid-binding immunoglobulin-like lectins) are a family of vertebrate glycan-binding cell-surface proteins. The majority mediate cellular inhibitory activity once engaged by specific ligands or ligand-mimicking molecules. As a result, Siglec engagement is now of interest as a strategy to therapeutically dampen unwanted cellular responses. When considering allergic inflammation, human eosinophils and mast cells express overlapping but distinct patterns of Siglecs. For example, Siglec-6 is selectively and prominently expressed on mast cells while Siglec-8 is highly specific for both eosinophils and mast cells. This review will focus on a subset of Siglecs and their various endogenous or synthetic sialoside ligands that regulate eosinophil and mast cell function and survival. It will also summarize how certain Siglecs have become the focus of novel therapies for allergic and other eosinophil- and mast cell-related diseases.
Collapse
Affiliation(s)
- Jeremy A O'Sullivan
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Robert P Schleimer
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bruce S Bochner
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
12
|
Moles E, Howard CB, Huda P, Karsa M, McCalmont H, Kimpton K, Duly A, Chen Y, Huang Y, Tursky ML, Ma D, Bustamante S, Pickford R, Connerty P, Omari S, Jolly CJ, Joshi S, Shen S, Pimanda JE, Dolnikov A, Cheung LC, Kotecha RS, Norris MD, Haber M, de Bock CE, Somers K, Lock RB, Thurecht KJ, Kavallaris M. Delivery of PEGylated liposomal doxorubicin by bispecific antibodies improves treatment in models of high-risk childhood leukemia. Sci Transl Med 2023; 15:eabm1262. [PMID: 37196067 DOI: 10.1126/scitranslmed.abm1262] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 04/13/2023] [Indexed: 05/19/2023]
Abstract
High-risk childhood leukemia has a poor prognosis because of treatment failure and toxic side effects of therapy. Drug encapsulation into liposomal nanocarriers has shown clinical success at improving biodistribution and tolerability of chemotherapy. However, enhancements in drug efficacy have been limited because of a lack of selectivity of the liposomal formulations for the cancer cells. Here, we report on the generation of bispecific antibodies (BsAbs) with dual binding to a leukemic cell receptor, such as CD19, CD20, CD22, or CD38, and methoxy polyethylene glycol (PEG) for the targeted delivery of PEGylated liposomal drugs to leukemia cells. This liposome targeting system follows a "mix-and-match" principle where BsAbs were selected on the specific receptors expressed on leukemia cells. BsAbs improved the targeting and cytotoxic activity of a clinically approved and low-toxic PEGylated liposomal formulation of doxorubicin (Caelyx) toward leukemia cell lines and patient-derived samples that are immunophenotypically heterogeneous and representative of high-risk subtypes of childhood leukemia. BsAb-assisted improvements in leukemia cell targeting and cytotoxic potency of Caelyx correlated with receptor expression and were minimally detrimental in vitro and in vivo toward expansion and functionality of normal peripheral blood mononuclear cells and hematopoietic progenitors. Targeted delivery of Caelyx using BsAbs further enhanced leukemia suppression while reducing drug accumulation in the heart and kidneys and extended overall survival in patient-derived xenograft models of high-risk childhood leukemia. Our methodology using BsAbs therefore represents an attractive targeting platform to potentiate the therapeutic efficacy and safety of liposomal drugs for improved treatment of high-risk leukemia.
Collapse
Affiliation(s)
- Ernest Moles
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- Australian Centre for Nanomedicine, Faculty of Engineering, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Christopher B Howard
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia 4072, Australia
| | - Pie Huda
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia 4072, Australia
| | - Mawar Karsa
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Hannah McCalmont
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Kathleen Kimpton
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Alastair Duly
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Yongjuan Chen
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Yizhou Huang
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Melinda L Tursky
- Department of Haematology and Bone Marrow Transplant, St Vincent's Hospital Sydney, Sydney 2010, Australia
- St Vincent's Centre for Applied Medical Research (AMR), Sydney 2010, Australia
- St Vincent Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - David Ma
- Department of Haematology and Bone Marrow Transplant, St Vincent's Hospital Sydney, Sydney 2010, Australia
- St Vincent's Centre for Applied Medical Research (AMR), Sydney 2010, Australia
- St Vincent Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Sonia Bustamante
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, UNSW Sydney, Sydney 2052, Australia
| | - Russell Pickford
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, UNSW Sydney, Sydney 2052, Australia
| | - Patrick Connerty
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Sofia Omari
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Christopher J Jolly
- School of Biomedical Sciences, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
| | - Swapna Joshi
- School of Biomedical Sciences, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
| | - Sylvie Shen
- School of Biomedical Sciences, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
| | - John E Pimanda
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
- School of Biomedical Sciences, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- Department of Haematology, Prince of Wales Hospital, Sydney 2031, Australia
| | - Alla Dolnikov
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Laurence C Cheung
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, Western Australia 6009, Australia
- Curtin Medical School, Curtin University, Perth, Western Australia 6102, Australia
| | - Rishi S Kotecha
- Leukaemia Translational Research Laboratory, Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, Western Australia 6009, Australia
- Curtin Medical School, Curtin University, Perth, Western Australia 6102, Australia
- Department of Clinical Haematology, Oncology, Blood and Marrow Transplantation, Perth Children's Hospital, Perth, Western Australia 6009, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Murray D Norris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
- University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Sydney 2052, Australia
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Charles E de Bock
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Klaartje Somers
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Kristofer J Thurecht
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia 4072, Australia
- Centre for Advanced Imaging, ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, St Lucia 4072, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- Australian Centre for Nanomedicine, Faculty of Engineering, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| |
Collapse
|
13
|
CD33 isoforms in microglia and Alzheimer's disease: Friend and foe. Mol Aspects Med 2023; 90:101111. [PMID: 35940942 DOI: 10.1016/j.mam.2022.101111] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/18/2022] [Accepted: 07/27/2022] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is the most common form of neurodegenerative disease and is considered the main cause of dementia worldwide. Genome-wide association studies combined with integrated analysis of functional datasets support a critical role for microglia in AD pathogenesis, identifying them as important potential therapeutic targets. The ability of immunomodulatory receptors on microglia to control the response to pathogenic amyloid-β aggregates has gained significant interest. Siglec-3, also known as CD33, is one of these immunomodulatory receptors expressed on microglia that has been identified as an AD susceptibility factor. Here, we review recent advances made in understanding the multifaceted roles that CD33 plays in microglia with emphasis on two human-specific CD33 isoforms that differentially correlate with AD susceptibility. We also describe several different therapeutic approaches for targeting CD33 that have been advanced for the purpose of skewing microglial cell responses.
Collapse
|
14
|
Esapa B, Jiang J, Cheung A, Chenoweth A, Thurston DE, Karagiannis SN. Target Antigen Attributes and Their Contributions to Clinically Approved Antibody-Drug Conjugates (ADCs) in Haematopoietic and Solid Cancers. Cancers (Basel) 2023; 15:1845. [PMID: 36980732 PMCID: PMC10046624 DOI: 10.3390/cancers15061845] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
Antibody drug conjugates (ADCs) are powerful anti-cancer therapies comprising an antibody joined to a cytotoxic payload through a chemical linker. ADCs exploit the specificity of antibodies for their target antigens, combined with the potency of cytotoxic drugs, to selectively kill target antigen-expressing tumour cells. The recent rapid advancement of the ADC field has so far yielded twelve and eight ADCs approved by the US and EU regulatory bodies, respectively. These serve as effective targeted treatments for several haematological and solid tumour types. In the development of an ADC, the judicious choice of an antibody target antigen with high expression on malignant cells but restricted expression on normal tissues and immune cells is considered crucial to achieve selectivity and potency while minimising on-target off-tumour toxicities. Aside from this paradigm, the selection of an antigen for an ADC requires consideration of several factors relating to the expression pattern and biological features of the target antigen. In this review, we discuss the attributes of antigens selected as targets for antibodies used in clinically approved ADCs for the treatment of haematological and solid malignancies. We discuss target expression, functions, and cellular kinetics, and we consider how these factors might contribute to ADC efficacy.
Collapse
Affiliation(s)
- Benjamina Esapa
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Jiexuan Jiang
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Anthony Cheung
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London SE1 9RT, UK
| | - Alicia Chenoweth
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London SE1 9RT, UK
| | - David E. Thurston
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, London SE1 9NH, UK
| | - Sophia N. Karagiannis
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences, King’s College London, Guy’s Hospital, London SE1 9RT, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London SE1 9RT, UK
| |
Collapse
|
15
|
Shaw JL, Pablo JL, Greka A. Mechanisms of Protein Trafficking and Quality Control in the Kidney and Beyond. Annu Rev Physiol 2023; 85:407-423. [PMID: 36763970 DOI: 10.1146/annurev-physiol-031522-100639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Numerous trafficking and quality control pathways evolved to handle the diversity of proteins made by eukaryotic cells. However, at every step along the biosynthetic pathway, there is the potential for quality control system failure. This review focuses on the mechanisms of disrupted proteostasis. Inspired by diseases caused by misfolded proteins in the kidney (mucin 1 and uromodulin), we outline the general principles of protein biosynthesis, delineate the recognition and degradation pathways targeting misfolded proteins, and discuss the role of cargo receptors in protein trafficking and lipid homeostasis. We also discuss technical approaches including live-cell fluorescent microscopy, chemical screens to elucidate trafficking mechanisms, multiplexed single-cell CRISPR screening platforms to systematically delineate mechanisms of proteostasis, and the advancement of novel tools to degrade secretory and membrane-associated proteins. By focusing on components of trafficking that go awry, we highlight ongoing efforts to understand fundamental mechanisms of disrupted proteostasis and implications for the treatment of human proteinopathies.
Collapse
Affiliation(s)
- Jillian L Shaw
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; .,Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Juan Lorenzo Pablo
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; .,Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Anna Greka
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; .,Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Abstract
Vesicles mediate the trafficking of membranes/proteins in the endocytic and secretory pathways. These pathways are regulated by small GTPases of the Rab family. Rab proteins belong to the Ras superfamily of GTPases, which are significantly involved in various intracellular trafficking and signaling processes in the nervous system. Rab11 is known to play a key role especially in recycling many proteins, including receptors important for signal transduction and preservation of functional activities of nerve cells. Rab11 activity is controlled by GEFs (guanine exchange factors) and GAPs (GTPase activating proteins), which regulate its function through modulating GTP/GDP exchange and the intrinsic GTPase activity, respectively. Rab11 is involved in the transport of several growth factor molecules important for the development and repair of neurons. Overexpression of Rab11 has been shown to significantly enhance vesicle trafficking. On the other hand, a reduced expression of Rab11 was observed in several neurodegenerative diseases. Current evidence appears to support the notion that Rab11 and its cognate proteins may be potential targets for therapeutic intervention. In this review, we briefly discuss the function of Rab11 and its related interaction partners in intracellular pathways that may be involved in neurodegenerative processes.
Collapse
Affiliation(s)
| | - Jiri Novotny
- Jiri Novotny, Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic.
| |
Collapse
|
17
|
Marlin A, Le Pape F, Le Goff J, Hamon N, Troadec T, Tripier R, Berthou C, Patinec V. New Triazacycloalkane Derivatives as Cytotoxic Agents for CLL Treatment: From Proof of Concept to the Targeting Biomolecule. Bioconjug Chem 2022; 33:1377-1392. [PMID: 35709513 DOI: 10.1021/acs.bioconjchem.2c00227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The 1,4,7-tris-(2-pyridinylmethyl)-1,4,7-triazacyclononane ligand (no3py) and its bifunctional analogue no3pyCOOK were synthesized to investigate their action toward zinc(II) depletion related to the apoptosis phenomenon in chronic lymphocytic leukemia (CLL) cells. no3py was used as the "free" ligand, while its "graftable" derivative was conjugated on a newly synthesized bifunctional sialoglycan, 6'-SL-NH2, selected to specifically bind CD22 biomarker expressed on the B-CLL cell surface. Both compounds were produced with good yields thanks to a Sonogashira coupling reaction and an orthoester function, respectively, for the chelator and the targeting moiety. The newly reported bioconjugate 6'-SL-no3py was then obtained through a peptidic coupling reaction. Biological in vitro studies of no3py and 6'-SL-no3py consisting of real-time detection of cell health (cytotoxicity and proliferation) and caspases 3/7 activation (crucial enzymes whose activation triggers cell death signaling pathways) have been investigated. First, Ramos, Daudi, and Raji B-cell lines, which present different sensitivity to zinc(II) content variation, were incubated with no3py and 6'-SL-no3py. Then, a videomicroscope allowed the real-time monitoring of the morphological changes leading to cell death from the detection of the cytotoxicity, the antiproliferative effect, and the caspasic activity. In terms of mechanism, the Zn2+ chelator cytotoxic effect of no3py has been evidenced by a culture medium ion supplementation study and by the decrease of intracellular fluorescence of Zn-specific fluorophore zinquin in the presence of no3py and 6'-SL-no3py chelators. Finally, flow cytometry analysis with classical Annexin V staining was conducted to detect no3py- and 6'-SL-no3py-induced apoptotic cell death in B-CLL cells. Time-course analysis, using the Incucyte Live-Cell Analysis System, demonstrated that no3py induced cell death in a time- and dose-dependent manner with variability across cell lines. 6'-SL-no3py exhibited the same dose-dependent trend as no3py, showing the efficiency of the targeting moiety. In both cases, the chelators depicted proliferation curves that were inversely correlated with kinetic death. Morphological changes specific to apoptosis and caspase 3/7 activation were observed for the three cell lines treated with no3py and 6'-SL-no3py, highlighting their role as apoptotic agents. A higher concentration of 6'-SL-no3py is needed to reach 50% of the B-CLL mortality, confirming a targeting of the chelator to the cell membrane. Overall, our results proved that the biological properties of the triazamacrocyclic chelator still remain even after addition of the targeting moiety. The free chelator as well as the bioconjugate constitute promising cytotoxic agents for CLL therapy through apoptosis induction.
Collapse
Affiliation(s)
- Axia Marlin
- Univ. Brest, UMR-CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29238 Brest, France
| | - Fiona Le Pape
- Univ. Brest, UMR-INSERM 1227 LBAI, 5 Avenue Foch, CHU Morvan, BP 824, 29609 Brest, France
| | - Jocelyn Le Goff
- Univ. Brest, UMR-INSERM 1227 LBAI, 5 Avenue Foch, CHU Morvan, BP 824, 29609 Brest, France
| | - Nadège Hamon
- Univ. Brest, UMR-CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29238 Brest, France
| | - Thibault Troadec
- Univ. Brest, UMR-CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29238 Brest, France
| | - Raphaël Tripier
- Univ. Brest, UMR-CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29238 Brest, France
| | - Christian Berthou
- Univ. Brest, UMR-INSERM 1227 LBAI, 5 Avenue Foch, CHU Morvan, BP 824, 29609 Brest, France
| | - Véronique Patinec
- Univ. Brest, UMR-CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29238 Brest, France
| |
Collapse
|
18
|
Wong KL, Li Z, Ma F, Wang D, Song N, Chong CH, Luk KK, Leung SO. SM03, an Anti-CD22 Antibody, Converts Cis-to- Trans Ligand Binding of CD22 against α2,6-Linked Sialic Acid Glycans and Immunomodulates Systemic Autoimmune Diseases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2726-2737. [PMID: 35688465 DOI: 10.4049/jimmunol.2100820] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 03/08/2022] [Indexed: 06/15/2023]
Abstract
SM03, an anti-CD22 recombinant IgG1 mAb, is currently in a phase III clinical trial for the treatment of rheumatoid arthritis (NCT04312815). SM03 showed good safety and efficacy in phase I systemic lupus erythematosus and phase II moderate to severe rheumatoid arthritis clinical trials. We propose the success of SM03 as a therapeutic to systemic autoimmune diseases is through the utilization of a novel mechanism of action unique to SM03. CD22, an inhibitory coreceptor of the BCR, is a potential immunotherapeutic target against autoimmune diseases. SM03 could disturb the CD22 homomultimeric configuration through disrupting cis binding to α2,6-linked sialic acids, induce rapid internalization of CD22 from the cell surface of human B cells, and facilitate trans binding between CD22 to human autologous cells. This in turn increased the activity of the downstream immunomodulatory molecule Src homology region 2 domain-containing phosphatase 1 (SHP-1) and decreased BCR-induced NF-κB activation in human B cells and B cell proliferation. This mechanism of action gives rationale to support the significant amelioration of disease and good safety profile in clinical trials, as by enabling the "self" recognition mechanism of CD22 via trans binding to α2,6 sialic acid ligands on autologous cells, SM03 specifically restores immune tolerance of B cells to host tissues without affecting the normal B cell immune response to pathogens.
Collapse
Affiliation(s)
- Kin L Wong
- SinoMab BioScience Ltd., Hong Kong Special Administrative Region, China
| | - Zhengdong Li
- SinoMab BioScience Ltd., Hong Kong Special Administrative Region, China
| | - Felix Ma
- SinoMab BioScience Ltd., Hong Kong Special Administrative Region, China
| | - Dong Wang
- SinoMab BioScience Ltd., Hong Kong Special Administrative Region, China
| | - Nan Song
- SinoMab BioScience Ltd., Hong Kong Special Administrative Region, China
| | - Chi H Chong
- SinoMab BioScience Ltd., Hong Kong Special Administrative Region, China
| | - Ka K Luk
- SinoMab BioScience Ltd., Hong Kong Special Administrative Region, China
| | - Shui O Leung
- SinoMab BioScience Ltd., Hong Kong Special Administrative Region, China
| |
Collapse
|
19
|
Babulic JL, Capicciotti CJ. Exo-Enzymatic Cell-Surface Glycan Labeling for Capturing Glycan–Protein Interactions through Photo-Cross-Linking. Bioconjug Chem 2022; 33:773-780. [DOI: 10.1021/acs.bioconjchem.2c00043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Jonathan L. Babulic
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario K7L 3N6, Canada
| | - Chantelle J. Capicciotti
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario K7L 3N6, Canada
- Department of Chemistry, and Department of Surgery, Queen’s University, Kingston, Ontario K7L 3N6, Canada
| |
Collapse
|
20
|
Ammon A, Mellenthin L, Emmerich C, Naschberger E, Stürzl M, Mackensen A, Müller F. Reduced cytotoxicity by mutation of Lysine 590 of Pseudomonas exotoxin can be restored in an optimized, Lysine-free immunotoxin. IMMUNOTHERAPY ADVANCES 2022; 2:ltac007. [PMID: 35919491 PMCID: PMC9327129 DOI: 10.1093/immadv/ltac007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/17/2022] [Indexed: 12/02/2022] Open
Abstract
Immunotoxins, which are fusion proteins of an antibody fragment and a fragment of a bacterial or a plant toxin, induce apoptosis in target cells by inhibition of protein synthesis. ADP-ribosylating toxins often have few lysine residues in their catalytic domain. As they are the target for ubiquitination, the low number of lysines possibly prevents ubiquitin-dependent degradation of the toxin in the cytosol. To reduce this potential degradation, we aimed to generate a lysine-free (noK), Pseudomonas exotoxin (PE)-based immunotoxin. The new generation 24 kDa PE, which lacks all but the furin-cleavage site of domain II, was mutated at lysine 590 (K590) and at K606 in a CD22-targeting immunotoxin and activity was determined against various B cell malignancies in vitro and in vivo. On average, K590 mutated to arginine (R) reduced cytotoxicity by 1.3-fold and K606R enhanced cytotoxicity by 1.3-fold compared to wild type (wt). Mutating K590 to histidine or deleting K590 did not prevent this loss in cytotoxicity. Neither stability nor internalization rate of K590R could explain reduced cytotoxicity. These results highlight the relevance of lysine 590 for PE intoxication. In line with in vitro results, the K606R mutant was more than 1.8-fold more active than the other variants in vivo suggesting that this single mutation may be beneficial when targeting CD22-positive malignancies. Finally, reduced cytotoxicity by K590R was compensated for by K606R and the resulting lysine-free variant achieved wt-like activity in vitro and in vivo. Thus, PE24-noK may represent a promising candidate for down-stream applications that would interfere with lysines.
Collapse
Affiliation(s)
- Anna Ammon
- Department of Internal Medicine 5, Haematology and Oncology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Lisa Mellenthin
- Department of Internal Medicine 5, Haematology and Oncology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Charlotte Emmerich
- Department of Internal Medicine 5, Haematology and Oncology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Elisabeth Naschberger
- Division of Molecular and Experimental Surgery, Translational Research Center, Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Michael Stürzl
- Division of Molecular and Experimental Surgery, Translational Research Center, Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Haematology and Oncology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Fabian Müller
- Department of Internal Medicine 5, Haematology and Oncology, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
21
|
Harumoto T, Iwai H, Tanigawa M, Kubo T, Atsumi T, Tsutsumi K, Takashima M, Destito G, Soloff R, Tomizuka K, Nycholat C, Paulson J, Uehara K. Enhancement of Gene Knockdown on CD22-Expressing Cells by Chemically Modified Glycan Ligand-siRNA Conjugates. ACS Chem Biol 2022; 17:292-298. [PMID: 35020348 DOI: 10.1021/acschembio.1c00652] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Extrahepatic targeted delivery of oligonucleotides, such as small interfering RNA (siRNA) and antisense oligonucleotides (ASOs), is an attractive technology for the development of nucleic acid-based medicines. To target CD22-expressing B cells, several drug platforms have shown promise, including antibodies, antibody-drug conjugates, and nanoparticles, but to date CD22-targeted delivery of oligonucleotide therapeutics has not been reported. Here we report the uptake and enhancement of siRNA gene expression knockdown in CD22-expressing B cells using a chemically stabilized and modified CD22 glycan ligand-conjugated siRNA. This finding has the potential to broaden the use of siRNA technology, opening up novel therapeutic opportunities, and presents an innovative approach for targeted delivery of siRNAs to B cell lymphomas.
Collapse
Affiliation(s)
- Toshimasa Harumoto
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Hiroto Iwai
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Mari Tanigawa
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Toshiko Kubo
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Toshiyuki Atsumi
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Kyoko Tsutsumi
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Michio Takashima
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Giuseppe Destito
- Kyowa Kirin Inc., 9420 Athena Circle, La Jolla, California 92037, United States
| | - Rachel Soloff
- Kyowa Kirin Inc., 9420 Athena Circle, La Jolla, California 92037, United States
| | - Kazuma Tomizuka
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Corwin Nycholat
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - James Paulson
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Keiji Uehara
- Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Otemachi Financial City Grand Cube, 1-9-2 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
22
|
Wang J, Yang J, Kopeček J. Nanomedicines in B cell-targeting therapies. Acta Biomater 2022; 137:1-19. [PMID: 34687954 PMCID: PMC8678319 DOI: 10.1016/j.actbio.2021.10.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/29/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023]
Abstract
B cells play multiple roles in immune responses related to autoimmune diseases as well as different types of cancers. As such, strategies focused on B cell targeting attracted wide interest and developed intensively. There are several common mechanisms various B cell targeting therapies have relied on, including direct B cell depletion, modulation of B cell antigen receptor (BCR) signaling, targeting B cell survival factors, targeting the B cell and T cell costimulation, and immune checkpoint blockade. Nanocarriers, used as drug delivery vehicles, possess numerous advantages to low molecular weight drugs, reducing drug toxicity, enhancing blood circulation time, as well as augmenting targeting efficacy and improving therapeutic effect. Herein, we review the commonly used targets involved in B cell targeting approaches and the utilization of various nanocarriers as B cell-targeted delivery vehicles. STATEMENT OF SIGNIFICANCE: As B cells are engaged significantly in the development of many kinds of diseases, utilization of nanomedicines in B cell depletion therapies have been rapidly developed. Although numerous studies focused on B cell targeting have already been done, there are still various potential receptors awaiting further investigation. This review summarizes the most relevant studies that utilized nanotechnologies associated with different B cell depletion approaches, providing a useful tool for selection of receptors, agents and/or nanocarriers matching specific diseases. Along with uncovering new targets in the function map of B cells, there will be a growing number of candidates that can benefit from nanoscale drug delivery.
Collapse
Affiliation(s)
- Jiawei Wang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
23
|
Lim J, Sari-Ak D, Bagga T. Siglecs as Therapeutic Targets in Cancer. BIOLOGY 2021; 10:1178. [PMID: 34827170 PMCID: PMC8615218 DOI: 10.3390/biology10111178] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023]
Abstract
Hypersialylation is a common post-translational modification of protein and lipids found on cancer cell surfaces, which participate in cell-cell interactions and in the regulation of immune responses. Sialic acids are a family of nine-carbon α-keto acids found at the outermost ends of glycans attached to cell surfaces. Given their locations on cell surfaces, tumor cells aberrantly overexpress sialic acids, which are recognized by Siglec receptors found on immune cells to mediate broad immunomodulatory signaling. Enhanced sialylation exposed on cancer cell surfaces is exemplified as "self-associated molecular pattern" (SAMP), which tricks Siglec receptors found on leukocytes to greatly down-regulate immune responsiveness, leading to tumor growth. In this review, we focused on all 15 human Siglecs (including Siglec XII), many of which still remain understudied. We also highlighted strategies that disrupt the course of Siglec-sialic acid interactions, such as antibody-based therapies and sialic acid mimetics leading to tumor cell depletion. Herein, we introduced the central roles of Siglecs in mediating pro-tumor immunity and discussed strategies that target these receptors, which could benefit improved cancer immunotherapy.
Collapse
Affiliation(s)
- Jackwee Lim
- Singapore Immunology Network, A*STAR, 8a Biomedical Grove, Singapore 138648, Singapore;
| | - Duygu Sari-Ak
- Department of Medical Biology, School of Medicine, University of Health Sciences, Istanbul 34668, Turkey;
| | - Tanaya Bagga
- Singapore Immunology Network, A*STAR, 8a Biomedical Grove, Singapore 138648, Singapore;
| |
Collapse
|
24
|
Mohamed SMA, Wohlmann A, Schofield P, Sia KCS, McCalmont H, Savvides SN, Verstraete K, Kavallaris M, Christ D, Friedrich KH, Bayat N, Lock RB. A recombinant antibody fragment directed to the thymic stromal lymphopoietin receptor (CRLF2) efficiently targets pediatric Philadelphia chromosome-like acute lymphoblastic leukemia. Int J Biol Macromol 2021; 190:214-223. [PMID: 34481852 DOI: 10.1016/j.ijbiomac.2021.08.194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/21/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022]
Abstract
Antibody fragments are promising building blocks for developing targeted therapeutics, thus improving treatment efficacy while minimising off-target toxicity. Despite recent advances in targeted therapeutics, patients with Philadelphia-like acute lymphoblastic leukemia (Ph-like ALL), a high-risk malignancy, lack specific and effective targeted treatments. Cytokine receptor-like factor 2 (CRLF2) is overexpressed in 50% of Ph-like ALL cases, conferring the survival of leukemia blasts through activation of the JAK/STAT signalling pathway. Targeting such a vital cell-surface protein could result in potent anti-leukaemic efficacy and reduce the likelihood of relapse associated with antigen loss. Herein, we developed a novel single-chain variable fragment (scFv) against CRLF2 based on a monoclonal antibody raised against the recombinant extracellular domain of human TSLPRα chain. The scFv fragment demonstrated excellent binding affinity with CRLF2 protein in the nanomolar range. Cellular association studies in vitro using an inducible CRLF2 knockdown cell line and ex vivo using patient-derived xenografts revealed the selective association of the scFv with CRLF2. The fragment exhibited significant receptor antagonistic effects on STAT5 signalling, suggesting possible therapeutic implications in vivo. This study is the first to describe the potential use of a novel scFv for targeting Ph-like ALL.
Collapse
Affiliation(s)
- Sara M A Mohamed
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.; University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Andreas Wohlmann
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany
| | - Peter Schofield
- Garvan Institute of Medical Research, Sydney, NSW, Australia; St.Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW, Australia
| | - Keith C S Sia
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.; University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia
| | - Hannah McCalmont
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.; University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia
| | | | | | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.; University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia; Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, UNSW Sydney, Sydney, NSW, Australia
| | - Daniel Christ
- Garvan Institute of Medical Research, Sydney, NSW, Australia; St.Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW, Australia
| | | | - Narges Bayat
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.; University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.; University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia.
| |
Collapse
|
25
|
Rentzsch M, Wawrzinek R, Zelle-Rieser C, Strandt H, Bellmann L, Fuchsberger FF, Schulze J, Busmann J, Rademacher J, Sigl S, Del Frari B, Stoitzner P, Rademacher C. Specific Protein Antigen Delivery to Human Langerhans Cells in Intact Skin. Front Immunol 2021; 12:732298. [PMID: 34745102 PMCID: PMC8566742 DOI: 10.3389/fimmu.2021.732298] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/30/2021] [Indexed: 11/13/2022] Open
Abstract
Immune modulating therapies and vaccines are in high demand, not least to the recent global spread of SARS-CoV2. To achieve efficient activation of the immune system, professional antigen presenting cells have proven to be key coordinators of such responses. Especially targeted approaches, actively directing antigens to specialized dendritic cells, promise to be more effective and accompanied by reduced payload due to less off-target effects. Although antibody and glycan-based targeting of receptors on dendritic cells have been employed, these are often expensive and time-consuming to manufacture or lack sufficient specificity. Thus, we applied a small-molecule ligand that specifically binds Langerin, a hallmark receptor on Langerhans cells, conjugated to a model protein antigen. Via microneedle injection, this construct was intradermally administered into intact human skin explants, selectively loading Langerhans cells in the epidermis. The ligand-mediated cellular uptake outpaces protein degradation resulting in intact antigen delivery. Due to the pivotal role of Langerhans cells in induction of immune responses, this approach of antigen-targeting of tissue-resident immune cells offers a novel way to deliver highly effective vaccines with minimally invasive administration.
Collapse
Affiliation(s)
- Mareike Rentzsch
- Department of Biomolecular Systems, Max-Planck-Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Robert Wawrzinek
- Department of Biomolecular Systems, Max-Planck-Institute of Colloids and Interfaces, Potsdam, Germany
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Claudia Zelle-Rieser
- Langerhans Cell Research Lab, Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Helen Strandt
- Langerhans Cell Research Lab, Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lydia Bellmann
- Langerhans Cell Research Lab, Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix F. Fuchsberger
- Department of Biomolecular Systems, Max-Planck-Institute of Colloids and Interfaces, Potsdam, Germany
| | - Jessica Schulze
- Department of Biomolecular Systems, Max-Planck-Institute of Colloids and Interfaces, Potsdam, Germany
| | - Jil Busmann
- Department of Biomolecular Systems, Max-Planck-Institute of Colloids and Interfaces, Potsdam, Germany
| | - Juliane Rademacher
- Department of Biomolecular Systems, Max-Planck-Institute of Colloids and Interfaces, Potsdam, Germany
| | - Stephan Sigl
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Del Frari
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Patrizia Stoitzner
- Langerhans Cell Research Lab, Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Rademacher
- Department of Biomolecular Systems, Max-Planck-Institute of Colloids and Interfaces, Potsdam, Germany
| |
Collapse
|
26
|
Upregulation of CD22 by Chidamide promotes CAR T cells functionality. Sci Rep 2021; 11:20637. [PMID: 34667217 PMCID: PMC8526578 DOI: 10.1038/s41598-021-00227-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Treatment failure or relapse due to tumor escape caused by reduction in target antigen expression has become a challenge in the field of CART therapy. Target antigen density is closely related to the effectiveness of CART therapy, and reduced or lost target antigen expression limits the efficacy of CART therapy and hinders the durability of CAR T cells. Epigenetic drugs can regulate histones for molecular modifications to regulate the transcriptional, translational and post-translational modification processes of target agents, and we demonstrated for the first time the role in regulating CD22 expression and its effect on the efficacy of CD22 CART. In this paper, we found that Chidamide promoted the expression of CD22 on the surface of B-cell tumor cells in vitro and in vivo, and enhanced the function of CD22 CART. As for mechanisms, we demonstrated that Chidamide did not affect CD22 mRNA transcription, but significantly increased the expression of total CD22 protein, indicating that Chidamide may upregulate cell surface CD22 expression by affecting the distribution of CD22 protein. In summary, our results suggest that Chidamide may enhance the efficacy of CD22 CART by inhibiting histone deacetylases to regulate post-transcriptional modifications that affect protein distribution to increase the expression of CD22 on the cell surface.
Collapse
|
27
|
Glycans in autophagy, endocytosis and lysosomal functions. Glycoconj J 2021; 38:625-647. [PMID: 34390447 PMCID: PMC8497297 DOI: 10.1007/s10719-021-10007-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/14/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022]
Abstract
Glycans have been shown to function as versatile molecular signals in cells. This prompted us to look at their roles in endocytosis, endolysosomal system and autophagy. We start by introducing the cell biological aspects of these pathways, the concept of the sugar code, and provide an overview on the role of glycans in the targeting of lysosomal proteins and in lysosomal functions. Moreover, we review evidence on the regulation of endocytosis and autophagy by glycans. Finally, we discuss the emerging concept that cytosolic exposure of luminal glycans, and their detection by endogenous lectins, provides a mechanism for the surveillance of the integrity of the endolysosomal compartments, and serves their eventual repair or disposal.
Collapse
|
28
|
Hammood M, Craig AW, Leyton JV. Impact of Endocytosis Mechanisms for the Receptors Targeted by the Currently Approved Antibody-Drug Conjugates (ADCs)-A Necessity for Future ADC Research and Development. Pharmaceuticals (Basel) 2021; 14:ph14070674. [PMID: 34358100 PMCID: PMC8308841 DOI: 10.3390/ph14070674] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 12/17/2022] Open
Abstract
Biologically-based therapies increasingly rely on the endocytic cycle of internalization and exocytosis of target receptors for cancer therapies. However, receptor trafficking pathways (endosomal sorting (recycling, lysosome localization) and lateral membrane movement) are often dysfunctional in cancer. Antibody-drug conjugates (ADCs) have revitalized the concept of targeted chemotherapy by coupling inhibitory antibodies to cytotoxic payloads. Significant advances in ADC technology and format, and target biology have hastened the FDA approval of nine ADCs (four since 2019). Although the links between aberrant endocytic machinery and cancer are emerging, the impact of dysregulated internalization processes of ADC targets and response rates or resistance have not been well studied. This is despite the reliance on ADC uptake and trafficking to lysosomes for linker cleavage and payload release. In this review, we describe what is known about all the target antigens for the currently approved ADCs. Specifically, internalization efficiency and relevant intracellular sorting activities are described for each receptor under normal processes, and when complexed to an ADC. In addition, we discuss aberrant endocytic processes that have been directly linked to preclinical ADC resistance mechanisms. The implications of endocytosis in regard to therapeutic effectiveness in the clinic are also described. Unexpectedly, information on endocytosis is scarce (absent for two receptors). Moreover, much of what is known about endocytosis is not in the context of receptor-ADC/antibody complexes. This review provides a deeper understanding of the pertinent principles of receptor endocytosis for the currently approved ADCs.
Collapse
Affiliation(s)
- Manar Hammood
- Departément de Medécine Nucléaire et Radiobiologie, Faculté de Medécine et des Sciences de la Santé, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Andrew W. Craig
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Jeffrey V. Leyton
- Departément de Medécine Nucléaire et Radiobiologie, Faculté de Medécine et des Sciences de la Santé, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
- Centre d’Imagerie Moleculaire, Centre de Recherche, CHUS, Sherbrooke, QC J1H 5N4, Canada
- Correspondence: ; Tel.: +1-819-346-1110
| |
Collapse
|
29
|
Lin J, Jin J, Shen Y, Zhang L, Gong G, Bian H, Chen H, Nagle DG, Wu Y, Zhang W, Luan X. Emerging protein degradation strategies: expanding the scope to extracellular and membrane proteins. Theranostics 2021; 11:8337-8349. [PMID: 34373745 PMCID: PMC8344007 DOI: 10.7150/thno.62686] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/03/2021] [Indexed: 12/23/2022] Open
Abstract
Classic small molecule inhibitors that directly target pathogenic proteins typically rely on the accessible binding sites to achieve prolonged occupancy and influence protein functions. The emerging targeted protein degradation (TPD) strategies exemplified by PROteolysis TArgeting Chimeras (PROTACs) are revolutionizing conventional drug discovery modality to target proteins of interest (POIs) that were categorized as "undruggable" before, however, these strategies are limited within intracellular POIs. The novel new degrader technologies such as LYsosome-TArgeting Chimaeras (LYTACs) and Antibody-based PROTACs (AbTACs) have been successfully developed to expand the scope of TPD to extracellular and membrane proteins, fulfilling huge unmet medical needs. Here, we systematically review the currently viable protein degradation strategies, emphasize that LYTACs and AbTACs turn a new avenue for the development of TPD, and highlight the potential challenges and directions in this vibrant field.
Collapse
Affiliation(s)
- Jiayi Lin
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jinmei Jin
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yiwen Shen
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lijun Zhang
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Gang Gong
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huiting Bian
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hongzhuan Chen
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dale G. Nagle
- Department of Biomolecular Sciences and Research of Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677-1848, USA
| | - Ye Wu
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- School of Pharmacy, Second Military Medical University, Shanghai, 201203, China
| | - Xin Luan
- Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
30
|
Chu Y, Zhou X, Wang X. Antibody-drug conjugates for the treatment of lymphoma: clinical advances and latest progress. J Hematol Oncol 2021; 14:88. [PMID: 34090506 PMCID: PMC8180036 DOI: 10.1186/s13045-021-01097-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are a promising class of immunotherapies with the potential to specifically target tumor cells and ameliorate the therapeutic index of cytotoxic drugs. ADCs comprise monoclonal antibodies, cytotoxic payloads with inherent antitumor activity, and specialized linkers connecting the two. In recent years, three ADCs, brentuximab vedotin, polatuzumab vedotin, and loncastuximab tesirine, have been approved and are already establishing their place in lymphoma treatment. As the efficacy and safety of ADCs have moved in synchrony with advances in their design, a plethora of novel ADCs have garnered growing interest as treatments. In this review, we provide an overview of the essential elements of ADC strategies in lymphoma and elucidate the up-to-date progress, current challenges, and novel targets of ADCs in this rapidly evolving field.
Collapse
Affiliation(s)
- Yurou Chu
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, No.324, Jingwu Road, Jinan, 250021, Shandong, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, No.324, Jingwu Road, Jinan, 250021, Shandong, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- School of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, No.324, Jingwu Road, Jinan, 250021, Shandong, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- School of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
31
|
LYTACs that engage the asialoglycoprotein receptor for targeted protein degradation. Nat Chem Biol 2021; 17:937-946. [PMID: 33767387 PMCID: PMC8387313 DOI: 10.1038/s41589-021-00770-1] [Citation(s) in RCA: 258] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 02/10/2021] [Indexed: 02/06/2023]
Abstract
Selective protein degradation platforms have afforded new development opportunities for therapeutics and tools for biological inquiry. The first lysosome targeting chimeras (LYTACs) targeted extracellular and membrane proteins for degradation by bridging a target protein to the cation-independent mannose-6-phosphate receptor (CI-M6PR). Here, we developed LYTACs that engage the asialoglycoprotein receptor (ASGPR), a liver-specific lysosomal targeting receptor, to degrade extracellular proteins in a cell type-specific manner. We conjugated binders to a tri-GalNAc motif that engages ASGPR to drive downregulation of proteins. Degradation of EGFR by GalNAc-LYTAC attenuated EGFR signaling compared to inhibition with an antibody. Furthermore, we demonstrated that a LYTAC comprising a 3.4 kDa peptide binder linked to a tri-GalNAc ligand degrades integrins and reduces cancer cell proliferation. Degradation with a single tri-GalNAc ligand prompted site-specific conjugation on antibody scaffolds, which improved the pharmacokinetic profile of GalNAc-LYTACs in vivo. GalNAc-LYTACs thus represent an avenue for cell-type restricted protein degradation.
Collapse
|
32
|
Smith BAH, Bertozzi CR. The clinical impact of glycobiology: targeting selectins, Siglecs and mammalian glycans. Nat Rev Drug Discov 2021; 20:217-243. [PMID: 33462432 PMCID: PMC7812346 DOI: 10.1038/s41573-020-00093-1] [Citation(s) in RCA: 256] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 01/31/2023]
Abstract
Carbohydrates - namely glycans - decorate every cell in the human body and most secreted proteins. Advances in genomics, glycoproteomics and tools from chemical biology have made glycobiology more tractable and understandable. Dysregulated glycosylation plays a major role in disease processes from immune evasion to cognition, sparking research that aims to target glycans for therapeutic benefit. The field is now poised for a boom in drug development. As a harbinger of this activity, glycobiology has already produced several drugs that have improved human health or are currently being translated to the clinic. Focusing on three areas - selectins, Siglecs and glycan-targeted antibodies - this Review aims to tell the stories behind therapies inspired by glycans and to outline how the lessons learned from these approaches are paving the way for future glycobiology-focused therapeutics.
Collapse
Affiliation(s)
- Benjamin A H Smith
- Department of Chemical & Systems Biology and ChEM-H, Stanford School of Medicine, Stanford, CA, USA
| | - Carolyn R Bertozzi
- Department of Chemical & Systems Biology and ChEM-H, Stanford School of Medicine, Stanford, CA, USA.
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
33
|
Current Status on Therapeutic Molecules Targeting Siglec Receptors. Cells 2020; 9:cells9122691. [PMID: 33333862 PMCID: PMC7765293 DOI: 10.3390/cells9122691] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/09/2020] [Accepted: 12/12/2020] [Indexed: 12/15/2022] Open
Abstract
The sialic acid-binding immunoglobulin-type of lectins (Siglecs) are receptors that recognize sialic acid-containing glycans. In the majority of the cases, Siglecs are expressed on immune cells and play a critical role in regulating immune cell signaling. Over the years, it has been shown that the sialic acid-Siglec axis participates in immunological homeostasis, and that any imbalance can trigger different pathologies, such as autoimmune diseases or cancer. For all this, different therapeutics have been developed that bind to Siglecs, either based on antibodies or being smaller molecules. In this review, we briefly introduce the Siglec family and we compile a description of glycan-based molecules and antibody-based therapies (including CAR-T and bispecific antibodies) that have been designed to therapeutically targeting Siglecs.
Collapse
|
34
|
Valverde P, Martínez JD, Cañada FJ, Ardá A, Jiménez-Barbero J. Molecular Recognition in C-Type Lectins: The Cases of DC-SIGN, Langerin, MGL, and L-Sectin. Chembiochem 2020; 21:2999-3025. [PMID: 32426893 PMCID: PMC7276794 DOI: 10.1002/cbic.202000238] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/19/2020] [Indexed: 12/16/2022]
Abstract
Carbohydrates play a pivotal role in intercellular communication processes. In particular, glycan antigens are key for sustaining homeostasis, helping leukocytes to distinguish damaged tissues and invading pathogens from healthy tissues. From a structural perspective, this cross-talk is fairly complex, and multiple membrane proteins guide these recognition processes, including lectins and Toll-like receptors. Since the beginning of this century, lectins have become potential targets for therapeutics for controlling and/or avoiding the progression of pathologies derived from an incorrect immune outcome, including infectious processes, cancer, or autoimmune diseases. Therefore, a detailed knowledge of these receptors is mandatory for the development of specific treatments. In this review, we summarize the current knowledge about four key C-type lectins whose importance has been steadily growing in recent years, focusing in particular on how glycan recognition takes place at the molecular level, but also looking at recent progresses in the quest for therapeutics.
Collapse
Affiliation(s)
- Pablo Valverde
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
| | - J Daniel Martínez
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
| | - F Javier Cañada
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Avda Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Ana Ardá
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
| | - Jesús Jiménez-Barbero
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
- Ikerbasque, Basque Foundation for Science, 48009, Bilbao, Spain
- Department of Organic Chemistry II, Faculty of Science and Technology, UPV-EHU, 48940, Leioa, Spain
| |
Collapse
|
35
|
Rueda-Gensini L, Cifuentes J, Castellanos MC, Puentes PR, Serna JA, Muñoz-Camargo C, Cruz JC. Tailoring Iron Oxide Nanoparticles for Efficient Cellular Internalization and Endosomal Escape. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1816. [PMID: 32932957 PMCID: PMC7559083 DOI: 10.3390/nano10091816] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022]
Abstract
Iron oxide nanoparticles (IONs) have been widely explored for biomedical applications due to their high biocompatibility, surface-coating versatility, and superparamagnetic properties. Upon exposure to an external magnetic field, IONs can be precisely directed to a region of interest and serve as exceptional delivery vehicles and cellular markers. However, the design of nanocarriers that achieve an efficient endocytic uptake, escape lysosomal degradation, and perform precise intracellular functions is still a challenge for their application in translational medicine. This review highlights several aspects that mediate the activation of the endosomal pathways, as well as the different properties that govern endosomal escape and nuclear transfection of magnetic IONs. In particular, we review a variety of ION surface modification alternatives that have emerged for facilitating their endocytic uptake and their timely escape from endosomes, with special emphasis on how these can be manipulated for the rational design of cell-penetrating vehicles. Moreover, additional modifications for enhancing nuclear transfection are also included in the design of therapeutic vehicles that must overcome this barrier. Understanding these mechanisms opens new perspectives in the strategic development of vehicles for cell tracking, cell imaging and the targeted intracellular delivery of drugs and gene therapy sequences and vectors.
Collapse
Affiliation(s)
- Laura Rueda-Gensini
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Javier Cifuentes
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Maria Claudia Castellanos
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Paola Ruiz Puentes
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Julian A. Serna
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Carolina Muñoz-Camargo
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
| | - Juan C. Cruz
- Department of Biomedical Engineering, School of Engineering, Universidad de Los Andes, Carrera 1 No. 18A-12, 111711 Bogotá, Colombia; (L.R.-G.); (J.C.); (M.C.C.); (P.R.P.); (J.A.S.)
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide 5005, Australia
| |
Collapse
|
36
|
Meyer SJ, Böser A, Korn MA, Koller C, Bertocci B, Reimann L, Warscheid B, Nitschke L. Cullin 3 Is Crucial for Pro-B Cell Proliferation, Interacts with CD22, and Controls CD22 Internalization on B Cells. THE JOURNAL OF IMMUNOLOGY 2020; 204:3360-3374. [DOI: 10.4049/jimmunol.1900925] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 04/03/2020] [Indexed: 12/15/2022]
|
37
|
Etienne F, Berthaud M, Nguyen F, Bernardeau K, Maurel C, Bodet-Milin C, Diab M, Abadie J, Gouilleux-Gruart V, Vidal A, Bourgeois M, Chouin N, Ibisch C, Davodeau F. SPECT-CT Imaging of Dog Spontaneous Diffuse Large B-Cell Lymphoma Targeting CD22 for the Implementation of a Relevant Preclinical Model for Human. Front Oncol 2020; 10:20. [PMID: 32117707 PMCID: PMC7018706 DOI: 10.3389/fonc.2020.00020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 01/08/2020] [Indexed: 11/13/2022] Open
Abstract
Antibodies directed against CD22 have been used in radioimmunotherapy (RIT) clinical trials to treat patients with diffuse large B-cell lymphoma (DLBCL) with promising results. However, relevant preclinical models are needed to facilitate the evaluation and optimization of new protocols. Spontaneous DLBCL in dogs is a tumor model that may help accelerate the development of new methodologies and therapeutic strategies for RIT targeting CD22. Seven murine monoclonal antibodies specific for canine CD22 were produced by the hybridoma method and characterized. The antibodies' affinity and epitopic maps, their internalization capability and usefulness for diagnosis in immunohistochemistry were determined. Biodistribution and PET imaging on a mouse xenogeneic model of dog DLBCL was used to choose the most promising antibody for our purposes. PET-CT results confirmed biodistribution study observations and allowed tumor localization. The selected antibody, 10C6, was successfully used on a dog with spontaneous DLBCL for SPECT-CT imaging in the context of disease staging, validating its efficacy for diagnosis and the feasibility of future RIT assays. This first attempt at phenotypic imaging on dogs paves the way to implementing quantitative imaging methodologies that would be transposable to humans in a theranostic approach. Taking into account the feedback of existing human radioimmunotherapy clinical trials targeting CD22, animal trials are planned to investigate protocol improvements that are difficult to consider in humans due to ethical concerns.
Collapse
Affiliation(s)
- Floriane Etienne
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France.,AMaROC, Oniris (Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering), Nantes, France
| | - Maxime Berthaud
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | - Frédérique Nguyen
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France.,AMaROC, Oniris (Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering), Nantes, France
| | - Karine Bernardeau
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France.,P2R "Production de Protéines Recombinantes", CRCINA, SFR-Santé, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Catherine Maurel
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | - Caroline Bodet-Milin
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France.,Nuclear Medicine, University Hospital, Nantes, France
| | - Maya Diab
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | - Jérôme Abadie
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France.,AMaROC, Oniris (Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering), Nantes, France
| | | | - Aurélien Vidal
- Groupement d'Intérêt Public ARRONAX Cyclotron, Saint-Herblain, France
| | - Mickaël Bourgeois
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France.,Groupement d'Intérêt Public ARRONAX Cyclotron, Saint-Herblain, France
| | - Nicolas Chouin
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France.,AMaROC, Oniris (Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering), Nantes, France
| | - Catherine Ibisch
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France.,AMaROC, Oniris (Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering), Nantes, France
| | - François Davodeau
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| |
Collapse
|
38
|
Birrer MJ, Moore KN, Betella I, Bates RC. Antibody-Drug Conjugate-Based Therapeutics: State of the Science. J Natl Cancer Inst 2020; 111:538-549. [PMID: 30859213 DOI: 10.1093/jnci/djz035] [Citation(s) in RCA: 257] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/21/2019] [Accepted: 03/08/2019] [Indexed: 02/06/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are complex engineered therapeutics consisting of monoclonal antibodies, directed toward tumor-associated antigens, to which highly potent cytotoxic agents are attached using chemical linkers. This targeted drug delivery strategy couples the precision of the antibody targeting moiety with the cytocidal activity of the payload, which is generally too toxic on its own to be systemically administered. In this manner, ADCs confer a means to reduce off-target toxicities in patients by limiting the exposure of normal tissues to the payload, thus broadening the potential therapeutic window compared with traditional chemotherapy. The pace of ADC development is accelerating, with the number of investigational agents in human trials having more than tripled over the past 5 years, underscoring the enthusiasm for this transformative approach to cancer treatment. Here, we review the key structural elements of ADC design (antibody, linker, and payload), highlighting critical aspects and technological advances that have affected the clinical effectiveness of this class of biopharmaceuticals. The ADC field continues to evolve, including ongoing efforts aimed at improving target selection, developing payloads with varied mechanisms of action and increased potency, designing innovative bioconjugation strategies, as well as maximizing efficacy and tolerability in patients. An overview of the current clinical trial landscape is provided, with emphasis on the clinical experience of the four ADCs to have received regulatory approval to date, as well as additional promising candidates currently in late-stage clinical development in both solid tumor and hematological malignancies.
Collapse
Affiliation(s)
- Michael J Birrer
- Division of Hematology-Oncology, University of Alabama at Birmingham Comprehensive Cancer Center, Birmingham, AL
| | - Kathleen N Moore
- Stephenson Department of Obstetrics and Gynecology, Oklahoma Cancer Center at the University of Oklahoma Health Sciences Center, Oklahoma City, OK.,Sarah Cannon Research Institute, Nashville, TN
| | - Ilaria Betella
- Division of Hematology-Oncology, University of Alabama at Birmingham Comprehensive Cancer Center, Birmingham, AL
| | | |
Collapse
|
39
|
Abstract
Sialic acid-binding immunoglobulin-type lectins (Siglecs) are expressed on the majority of white blood cells of the immune system and play critical roles in immune cell signaling. Through recognition of sialic acid-containing glycans as ligands, they help the immune system distinguish between self and nonself. Because of their restricted cell type expression and roles as checkpoints in immune cell responses in human diseases such as cancer, asthma, allergy, neurodegeneration, and autoimmune diseases they have gained attention as targets for therapeutic interventions. In this review we describe the Siglec family, its roles in regulation of immune cell signaling, current efforts to define its roles in disease processes, and approaches to target Siglecs for treatment of human disease.
Collapse
Affiliation(s)
- Shiteng Duan
- Departments of Molecular Medicine, and Immunology and Microbiology, Scripps Research, La Jolla, California 92037, USA;
| | - James C Paulson
- Departments of Molecular Medicine, and Immunology and Microbiology, Scripps Research, La Jolla, California 92037, USA;
| |
Collapse
|
40
|
Wamhoff EC, Schulze J, Bellmann L, Rentzsch M, Bachem G, Fuchsberger FF, Rademacher J, Hermann M, Del Frari B, van Dalen R, Hartmann D, van Sorge NM, Seitz O, Stoitzner P, Rademacher C. A Specific, Glycomimetic Langerin Ligand for Human Langerhans Cell Targeting. ACS CENTRAL SCIENCE 2019; 5:808-820. [PMID: 31139717 PMCID: PMC6535779 DOI: 10.1021/acscentsci.9b00093] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Indexed: 05/30/2023]
Abstract
Langerhans cells are a subset of dendritic cells residing in the epidermis of the human skin. As such, they are key mediators of immune regulation and have emerged as prime targets for novel transcutaneous cancer vaccines. Importantly, the induction of protective T cell immunity by these vaccines requires the efficient and specific delivery of both tumor-associated antigens and adjuvants. Langerhans cells uniquely express Langerin (CD207), an endocytic C-type lectin receptor. Here, we report the discovery of a specific, glycomimetic Langerin ligand employing a heparin-inspired design strategy and structural characterization by NMR spectroscopy and molecular docking. The conjugation of this glycomimetic to liposomes enabled the specific and efficient targeting of Langerhans cells in the human skin. We further demonstrate the doxorubicin-mediated killing of a Langerin+ monocyte cell line, highlighting its therapeutic and diagnostic potential in Langerhans cell histiocytosis, caused by the abnormal proliferation of Langerin+ myeloid progenitor cells. Overall, our delivery platform provides superior versatility over antibody-based approaches and novel modalities to overcome current limitations of dendritic cell-targeted immuno- and chemotherapy.
Collapse
Affiliation(s)
- Eike-Christian Wamhoff
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, 14424 Potsdam, Germany
- Department
of Biology, Chemistry and Pharmacy, Freie
Universität Berlin, 14195 Berlin, Germany
| | - Jessica Schulze
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, 14424 Potsdam, Germany
- Department
of Biology, Chemistry and Pharmacy, Freie
Universität Berlin, 14195 Berlin, Germany
| | - Lydia Bellmann
- Department of Dermatology, Venereology and Allergology, Department of Anesthesiology
and Intensive Care Medicine, and Department of Plastic, Reconstructive and
Aesthetic Surgery, Medical University of
Innsbruck, 6020 Innsbruck, Austria
| | - Mareike Rentzsch
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Gunnar Bachem
- Department
of Chemistry, Humboldt-Universität
zu Berlin, 12489 Berlin, Germany
| | - Felix F. Fuchsberger
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, 14424 Potsdam, Germany
- Medical
Microbiology, University Medical Center
Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Juliane Rademacher
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Martin Hermann
- Department of Dermatology, Venereology and Allergology, Department of Anesthesiology
and Intensive Care Medicine, and Department of Plastic, Reconstructive and
Aesthetic Surgery, Medical University of
Innsbruck, 6020 Innsbruck, Austria
| | - Barbara Del Frari
- Department of Dermatology, Venereology and Allergology, Department of Anesthesiology
and Intensive Care Medicine, and Department of Plastic, Reconstructive and
Aesthetic Surgery, Medical University of
Innsbruck, 6020 Innsbruck, Austria
| | - Rob van Dalen
- Medical
Microbiology, University Medical Center
Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - David Hartmann
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Nina M. van Sorge
- Medical
Microbiology, University Medical Center
Utrecht, Utrecht University, 3584 CX Utrecht, Netherlands
| | - Oliver Seitz
- Department
of Chemistry, Humboldt-Universität
zu Berlin, 12489 Berlin, Germany
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology and Allergology, Department of Anesthesiology
and Intensive Care Medicine, and Department of Plastic, Reconstructive and
Aesthetic Surgery, Medical University of
Innsbruck, 6020 Innsbruck, Austria
| | - Christoph Rademacher
- Department
of Biomolecular Systems, Max Planck Institute
of Colloids and Interfaces, 14424 Potsdam, Germany
- Department
of Biology, Chemistry and Pharmacy, Freie
Universität Berlin, 14195 Berlin, Germany
| |
Collapse
|
41
|
Aujla A, Aujla R, Liu D. Inotuzumab ozogamicin in clinical development for acute lymphoblastic leukemia and non-Hodgkin lymphoma. Biomark Res 2019; 7:9. [PMID: 31011424 PMCID: PMC6458768 DOI: 10.1186/s40364-019-0160-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/27/2019] [Indexed: 12/26/2022] Open
Abstract
B cell acute lymphoblastic leukemia (ALL) and non-Hodgkin lymphoma (NHL) frequently express CD19, CD20 and CD22 on the cell surfaces. Immunotherapeutic agents including antibodies and chimeric antigen receptor T cells are widely studied in clinical trials. Several antibody-drug conjugates (ADC) have been approved for clinical use (gemtuzumab ozogamicin in acute myeloid leukemia and brentuximab vedotin in Hodgkin lymphoma as well as CD30+ anaplastic large cell lymphoma). Inotuzumab ozogamicin (INO), a CD22 antibody conjugated with calicheamicin is one of the newest ADCs. INO has been approved for treatment of relapsed /refractory B cell precursor ALL. Multiple ongoing trials are evaluating its role in the relapsed /refractory B cell NHL. This review summarized recent development in INO applications for ALL and NHL.
Collapse
Affiliation(s)
- Amandeep Aujla
- 1Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595 USA
| | - Ravijot Aujla
- 2Punjab Institute of Medical Sciences, Jalandhar, Punjab 144006 India
| | - Delong Liu
- 1Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595 USA.,3Department of Oncology, The First affiliated hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
42
|
Essandoh K, Deng S, Wang X, Jiang M, Mu X, Peng J, Li Y, Peng T, Wagner KU, Rubinstein J, Fan GC. Tsg101 positively regulates physiologic-like cardiac hypertrophy through FIP3-mediated endosomal recycling of IGF-1R. FASEB J 2019; 33:7451-7466. [PMID: 30884248 DOI: 10.1096/fj.201802338rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Development of physiologic cardiac hypertrophy has primarily been ascribed to the IGF-1 and its receptor, IGF-1 receptor (IGF-1R), and subsequent activation of the protein kinase B (Akt) pathway. However, regulation of endosome-mediated recycling and degradation of IGF-1R during physiologic hypertrophy has not been investigated. In a physiologic hypertrophy model of treadmill-exercised mice, we observed that levels of tumor susceptibility gene 101 (Tsg101), a key member of the endosomal sorting complex required for transport, were dramatically elevated in the heart compared with sedentary controls. To determine the role of Tsg101 on physiologic hypertrophy, we generated a transgenic (TG) mouse model with cardiac-specific overexpression of Tsg101. These TG mice exhibited a physiologic-like cardiac hypertrophy phenotype at 8 wk evidenced by: 1) the absence of cardiac fibrosis, 2) significant improvement of cardiac function, and 3) increased total and plasma membrane levels of IGF-1R and increased phosphorylation of Akt. Mechanistically, we identified that Tsg101 interacted with family-interacting protein 3 (FIP3) and IGF-1R, thereby stabilizing FIP3 and enhancing recycling of IGF-1R. In vitro, adenovirus-mediated overexpression of Tsg101 in neonatal rat cardiomyocytes resulted in cell hypertrophy, which was blocked by addition of monensin, an inhibitor of endosome-mediated recycling, and by small interfering RNA-mediated knockdown (KD) of FIP3. Furthermore, cardiac-specific KD of Tsg101 showed a significant reduction in levels of endosomal recycling compartment members (Rab11a and FIP3), IGF-1R, and Akt phosphorylation. Most interestingly, Tsg101-KD mice failed to develop cardiac hypertrophy after intense treadmill training. Taken together, our data identify Tsg101 as a novel positive regulator of physiologic cardiac hypertrophy through facilitating the FIP3-mediated endosomal recycling of IGF-1R.-Essandoh, K., Deng, S., Wang, X., Jiang, M., Mu, X., Peng, J., Li, Y., Peng, T., Wagner, K.-U., Rubinstein, J., Fan, G.-C. Tsg101 positively regulates physiologic-like cardiac hypertrophy through FIP3-mediated endosomal recycling of IGF-1R.
Collapse
Affiliation(s)
- Kobina Essandoh
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Shan Deng
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Department of Cardiology, Union Hospital-Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohong Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Min Jiang
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Xingjiang Mu
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jiangtong Peng
- Department of Cardiology, Union Hospital-Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yutian Li
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Tianqing Peng
- Critical Illness Research, Lawson Health Research Institute, London, Ontario, Canada
| | - Kay-Uwe Wagner
- Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Jack Rubinstein
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
43
|
Smith WS, Johnston DA, Holmes SE, Wensley HJ, Flavell SU, Flavell DJ. Augmentation of Saporin-Based Immunotoxins for Human Leukaemia and Lymphoma Cells by Triterpenoid Saponins: The Modifying Effects of Small Molecule Pharmacological Agents. Toxins (Basel) 2019; 11:toxins11020127. [PMID: 30791598 PMCID: PMC6410249 DOI: 10.3390/toxins11020127] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/11/2019] [Accepted: 02/14/2019] [Indexed: 12/22/2022] Open
Abstract
Triterpenoid saponins from Saponinum album (SA) significantly augment the cytotoxicity of saporin-based immunotoxins but the mechanism of augmentation is not fully understood. We investigated the effects of six small molecule pharmacological agents, which interfere with endocytic and other processes, on SA-mediated augmentation of saporin and saporin-based immunotoxins (ITs) directed against CD7, CD19, CD22 and CD38 on human lymphoma and leukaemia cell lines. Inhibition of clathrin-mediated endocytosis or endosomal acidification abolished the SA augmentation of saporin and of all four immunotoxins tested but the cytotoxicity of each IT or saporin alone was largely unaffected. The data support the hypothesis that endocytic processes are involved in the augmentative action of SA for saporin ITs targeted against a range of antigens expressed by leukaemia and lymphoma cells. In addition, the reactive oxygen species (ROS) scavenger tiron reduced the cytotoxicity of BU12-SAP and OKT10-SAP but had no effect on 4KB128-SAP or saporin cytotoxicity. Tiron also had no effect on SA-mediated augmentation of the saporin-based ITs or unconjugated saporin. These results suggest that ROS are not involved in the augmentation of saporin ITs and that ROS induction is target antigen-dependent and not directly due to the cytotoxic action of the toxin moiety.
Collapse
Affiliation(s)
- Wendy S Smith
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - David A Johnston
- Biomedical Imaging Unit, University of Southampton School of Medicine, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Suzanne E Holmes
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Harrison J Wensley
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Sopsamorn U Flavell
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - David J Flavell
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton SO16 6YD, UK.
| |
Collapse
|
44
|
Meyer SJ, Linder AT, Brandl C, Nitschke L. B Cell Siglecs-News on Signaling and Its Interplay With Ligand Binding. Front Immunol 2018; 9:2820. [PMID: 30559744 PMCID: PMC6286995 DOI: 10.3389/fimmu.2018.02820] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/15/2018] [Indexed: 12/11/2022] Open
Abstract
CD22 and Siglec-G are members of the Siglec family. Both are inhibitory co-receptors on the surface of B cells and inhibit B-cell receptor induced signaling, characterized by inhibition of the calcium mobilization and cellular activation. CD22 functions predominantly as an inhibitor on conventional B cells, while Siglec-G is an important inhibitor on the B1a-cell subset. These two B-cell Siglecs do not only inhibit initial signaling, but also have an important function in preventing autoimmunity, as double deficient mice develop a lupus-like phenotype with age. Siglecs are characterized by their conserved ability to bind terminal sialic acid of glycans on the cell surface, which is important to regulate the inhibitory role of Siglecs. While CD22 binds α2,6-linked sialic acids, Siglec-G can bind both α2,6-linked and α2,3-linked sialic acids. Interestingly, ligand binding is differentially regulating the ability of CD22 and Siglec-G to control B-cell activation. Within the last years, quite a few studies focused on the different functions of B-cell Siglecs and the interplay of ligand binding and signal inhibition. This review summarizes the role of CD22 and Siglec-G in regulating B-cell receptor signaling, membrane distribution with the importance of ligand binding, preventing autoimmunity and the role of CD22 beyond the naïve B-cell stage. Additionally, this review article features the long time discussed interaction between CD45 and CD22 with highlighting recent data, as well as the interplay between CD22 and Galectin-9 and its influence on B-cell receptor signaling. Moreover, therapeutical approaches targeting human CD22 will be elucidated.
Collapse
Affiliation(s)
- Sarah J Meyer
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Alexandra T Linder
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Carolin Brandl
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Lars Nitschke
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| |
Collapse
|
45
|
Clark EA, Giltiay NV. CD22: A Regulator of Innate and Adaptive B Cell Responses and Autoimmunity. Front Immunol 2018; 9:2235. [PMID: 30323814 PMCID: PMC6173129 DOI: 10.3389/fimmu.2018.02235] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/07/2018] [Indexed: 12/12/2022] Open
Abstract
CD22 (Siglec 2) is a receptor predominantly restricted to B cells. It was initially characterized over 30 years ago and named “CD22” in 1984 at the 2nd International workshop in Boston (1). Several excellent reviews have detailed CD22 functions, CD22-regulated signaling pathways and B cell subsets regulated by CD22 or Siglec G (2–4). This review is an attempt to highlight recent and possibly forgotten findings. We also describe the role of CD22 in autoimmunity and the great potential for CD22-based immunotherapeutics for the treatment of autoimmune diseases such as systemic lupus erythematosus (SLE).
Collapse
Affiliation(s)
- Edward A Clark
- Department of Immunology, University of Washington, Seattle, WA, United States.,Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Natalia V Giltiay
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
46
|
Domain II of Pseudomonas Exotoxin Is Critical for Efficacy of Bolus Doses in a Xenograft Model of Acute Lymphoblastic Leukemia. Toxins (Basel) 2018; 10:toxins10050210. [PMID: 29883379 PMCID: PMC5983266 DOI: 10.3390/toxins10050210] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/14/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022] Open
Abstract
Moxetumomab pasudotox is a fusion protein of a CD22-targeting antibody and Pseudomonas exotoxin. Minutes of exposure to Moxetumomab achieves similar cell killing than hours of exposure to a novel deimmunized variant against some acute lymphoblastic leukemia (ALL). Because blood levels fall quickly, Moxetumomab is more than 1000-fold more active than the deimmunized variant in vivo. We aimed to identify which part of Moxetumomab increases in vivo efficacy and generated five immunotoxins, tested time-dependent activity, and determined the efficacy in a KOPN-8 xenograft model. Full domain II shortened the time cells had to be exposed to die to only a few minutes for some ALL; deimmunized domain III consistently extended the time. Against KOPN-8, full domain II accelerated time to arrest protein synthesis by three-fold and tripled PARP-cleavage. In vivo efficacy was increased by more than 10-fold by domain II and increasing size, and therefore half-life enhanced efficacy two- to four-fold. In summary, in vivo efficacy is determined by the time cells have to be exposed to immunotoxin to die and serum half-life. Thus, domain II is most critical for activity against some ALL treated with bolus doses; however, immunotoxins lacking all but the furin-cleavage site of domain II may be advantageous when treating continuously.
Collapse
|
47
|
The Glycoscience of Immunity. Trends Immunol 2018; 39:523-535. [PMID: 29759949 DOI: 10.1016/j.it.2018.04.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 02/05/2023]
Abstract
Carbohydrates, or glycans, are as integral to biology as nucleic acids and proteins. In immunology, glycans are well known to drive diverse functions ranging from glycosaminoglycan-mediated chemokine presentation and selectin-dependent leukocyte trafficking to the discrimination of self and non-self through the recognition of sialic acids by Siglec (sialic acid-binding Ig-like lectin) receptors. In recent years, a number of key immunological discoveries are driving a renewed and burgeoning appreciation for the importance of glycans. In this review, we highlight these findings which collectively help to define and refine our knowledge of the function and impact of glycans within the immune response.
Collapse
|
48
|
Ereño-Orbea J, Sicard T, Cui H, Mazhab-Jafari MT, Benlekbir S, Guarné A, Rubinstein JL, Julien JP. Molecular basis of human CD22 function and therapeutic targeting. Nat Commun 2017; 8:764. [PMID: 28970495 PMCID: PMC5624926 DOI: 10.1038/s41467-017-00836-6] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022] Open
Abstract
CD22 maintains a baseline level of B-cell inhibition to keep humoral immunity in check. As a B-cell-restricted antigen, CD22 is targeted in therapies against dysregulated B cells that cause autoimmune diseases and blood cancers. Here we report the crystal structure of human CD22 at 2.1 Å resolution, which reveals that specificity for α2-6 sialic acid ligands is dictated by a pre-formed β-hairpin as a unique mode of recognition across sialic acid-binding immunoglobulin-type lectins. The CD22 ectodomain adopts an extended conformation that facilitates concomitant CD22 nanocluster formation on B cells and binding to trans ligands to avert autoimmunity in mammals. We structurally delineate the CD22 site targeted by the therapeutic antibody epratuzumab at 3.1 Å resolution and determine a critical role for CD22 N-linked glycosylation in antibody engagement. Our studies provide molecular insights into mechanisms governing B-cell inhibition and valuable clues for the design of immune modulators in B-cell dysfunction.The B-cell-specific co-receptor CD22 is a therapeutic target for depleting dysregulated B cells. Here the authors structurally characterize the ectodomain of CD22 and present its crystal structure with the bound therapeutic antibody epratuzumab, which gives insights into the mechanism of inhibition of B-cell activation.
Collapse
Affiliation(s)
- June Ereño-Orbea
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4
| | - Taylor Sicard
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada, M5S 1A8
| | - Hong Cui
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4
| | - Mohammad T Mazhab-Jafari
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4
| | - Samir Benlekbir
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4
| | - Alba Guarné
- Department of Biochemistry and Biomedical Science, McMaster University, Hamilton, ON, Canada, L8S 4L8
| | - John L Rubinstein
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada, M5S 1A8
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada, M5G 1L7
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4.
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada, M5S 1A8.
- Department of Immunology, University of Toronto, Toronto, ON, Canada, M5S 1A8.
| |
Collapse
|
49
|
Peng W, Paulson JC. CD22 Ligands on a Natural N-Glycan Scaffold Efficiently Deliver Toxins to B-Lymphoma Cells. J Am Chem Soc 2017; 139:12450-12458. [PMID: 28829594 DOI: 10.1021/jacs.7b03208] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
CD22 is a sialic acid-binding immunoglobulin-like lectin (Siglec) that is highly expressed on B-cells and B cell lymphomas, and is a validated target for antibody and nanoparticle based therapeutics. However, cell targeted therapeutics are limited by their complexity, heterogeneity, and difficulties in production. We describe here a chemically defined natural N-linked glycan scaffold that displays high affinity CD22 glycan ligands and outcompetes the natural ligand for the receptor, resulting in single molecule binding to CD22 and endocytosis into cells. Binding affinity is increased by up to 1500-fold compared to the monovalent ligand, while maintaining the selectivity for hCD22 over other Siglecs. Conjugates of these multivalent ligands with auristatin and saporin toxins are efficiently internalized via hCD22 resulting in killing of B-cell lymphoma cells. This single molecule ligand targeting strategy represents an alternative to antibody- and nanoparticle-mediated approaches for delivery of agents to cells expressing CD22 and other Siglecs.
Collapse
Affiliation(s)
- Wenjie Peng
- Departments of Molecular Medicine and Immunology & Microbiology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - James C Paulson
- Departments of Molecular Medicine and Immunology & Microbiology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
50
|
Li YQ, Zhang J, Li J, Sun L. First characterization of fish CD22: An inhibitory role in the activation of peripheral blood leukocytes. Vet Immunol Immunopathol 2017; 190:39-44. [DOI: 10.1016/j.vetimm.2017.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/24/2017] [Accepted: 07/11/2017] [Indexed: 01/05/2023]
|