1
|
Jia W, Yuan J, Zhang J, Li S, Lin W, Cheng B. Bioactive sphingolipids as emerging targets for signal transduction in cancer development. Biochim Biophys Acta Rev Cancer 2024; 1879:189176. [PMID: 39233263 DOI: 10.1016/j.bbcan.2024.189176] [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: 06/06/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Sphingolipids, crucial components of cellular membranes, play a vital role in maintaining cellular structure and signaling integrity. Disruptions in sphingolipid metabolism are increasingly implicated in cancer development. Key bioactive sphingolipids, such as ceramides, sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), and glycosphingolipids, profoundly impact tumor biology. They influence the behavior of tumor cells, stromal cells, and immune cells, affecting tumor aggressiveness, angiogenesis, immune modulation, and extracellular matrix remodeling. Furthermore, abnormal expression of sphingolipids and their metabolizing enzymes modulates the secretion of tumor-derived extracellular vesicles (TDEs), which are key players in creating an immunosuppressive tumor microenvironment, remodeling the extracellular matrix, and facilitating oncogenic signaling within in situ tumors and distant pre-metastatic niches (PMNs). Understanding the role of sphingolipids in the biogenesis of tumor-derived extracellular vesicles (TDEs) and their bioactive contents can pave the way for new biomarkers in cancer diagnosis and prognosis, ultimately enhancing comprehensive tumor treatment strategies.
Collapse
Affiliation(s)
- Wentao Jia
- Department of General Practice, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China
| | - Jiaying Yuan
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jinbo Zhang
- Department of Pharmacy, Tianjin Rehabilitation and Recuperation Center, Joint Logistics Support Force, Tianjin 300000, China
| | - Shu Li
- Department of Gastroenterology, Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Wanfu Lin
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| | - Binbin Cheng
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| |
Collapse
|
2
|
Romanò C, Jiang H, Tahvili S, Wei P, Keiding UB, Clergeaud G, Skovbakke SL, Blomberg AL, Hafkenscheid L, Henriksen JR, Andresen TL, Goletz S, Hansen AE, Christensen D, Clausen MH. Chemical synthesis and immunological evaluation of cancer vaccines based on ganglioside antigens and α-galactosylceramide. RSC Med Chem 2024; 15:2718-2728. [PMID: 39149099 PMCID: PMC11324045 DOI: 10.1039/d4md00387j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 06/20/2024] [Indexed: 08/17/2024] Open
Abstract
iNKT cells - often referred as the "Swiss Army knife" of the immune system - have emerged as central players in cancer vaccine therapies. Glycolipids activating iNKT cells, such as α-galactosylceramide (αGalCer), can enhance the immune response against co-delivered cancer antigens and have been applied in the design of self-adjuvanting anti-tumor vaccines. In this context, this work focuses on the chemical synthesis of ganglioside tumor-associated carbohydrate antigens (TACAs), namely GM3 and (Neu5Gc)GM3 antigens, their conjugation to αGalCer, and their formulation into liposomes as an efficient platform for their in vivo delivery. Liposomes containing GM3-αGalCer, (Neu5Gc)GM3-αGalCer, and equimolar amounts of the two conjugates have been fully characterized and their ability to activate iNKT cell has been confirmed ex vivo in mouse and human cell assays. The candidates were tested in in vivo immunization studies, demonstrating an ability to induce both TH1 and TH2 cytokines leading to the production of all subclasses of IgG antibodies. Notably, the study also demonstrated that serum antibodies raised against the two TACAs, alone and in combination, were cross-reactive. This finding has consequences for future vaccine designs - even if a highly tumor-selective antigen is chosen, the resulting antibody response may be broader than anticipated.
Collapse
Affiliation(s)
- Cecilia Romanò
- Center for Nanomedicine & Theranostics, Department of Chemistry, Technical University of Denmark Kemitorvet 207 2800 Kgs. Lyngby Denmark
| | - Hao Jiang
- Center for Nanomedicine & Theranostics, Department of Chemistry, Technical University of Denmark Kemitorvet 207 2800 Kgs. Lyngby Denmark
| | - Sahar Tahvili
- Center for Nanomedicine & Theranostics, Department of Chemistry, Technical University of Denmark Kemitorvet 207 2800 Kgs. Lyngby Denmark
| | - Peng Wei
- Center for Nanomedicine & Theranostics, Department of Chemistry, Technical University of Denmark Kemitorvet 207 2800 Kgs. Lyngby Denmark
| | - Ulrik B Keiding
- Center for Nanomedicine & Theranostics, Department of Chemistry, Technical University of Denmark Kemitorvet 207 2800 Kgs. Lyngby Denmark
| | - Gael Clergeaud
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark Ørsteds Plads 2800 Kgs Lyngby Denmark
| | - Sarah Line Skovbakke
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark Søltofts Plads 2800 Kgs Lyngby Denmark
| | - Anne Louise Blomberg
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark Søltofts Plads 2800 Kgs Lyngby Denmark
| | - Lise Hafkenscheid
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark Søltofts Plads 2800 Kgs Lyngby Denmark
| | - Jonas R Henriksen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark Ørsteds Plads 2800 Kgs Lyngby Denmark
| | - Thomas L Andresen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark Ørsteds Plads 2800 Kgs Lyngby Denmark
| | - Steffen Goletz
- Department of Biotechnology and Biomedicine, Section for Medical Biotechnology, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark Søltofts Plads 2800 Kgs Lyngby Denmark
| | - Anders E Hansen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark Ørsteds Plads 2800 Kgs Lyngby Denmark
| | - Dennis Christensen
- Adjuvant Systems Research & Development, Croda Pharma 2800 Lyngby Denmark
| | - Mads H Clausen
- Center for Nanomedicine & Theranostics, Department of Chemistry, Technical University of Denmark Kemitorvet 207 2800 Kgs. Lyngby Denmark
| |
Collapse
|
3
|
Heinzelbecker J, Fauskanger M, Jonson I, Krengel U, Løset GÅ, Munthe L, Tveita A. Chimeric antigen receptor T cells targeting the GM3(Neu5Gc) ganglioside. Front Immunol 2024; 15:1331345. [PMID: 38370401 PMCID: PMC10869436 DOI: 10.3389/fimmu.2024.1331345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/19/2024] [Indexed: 02/20/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell technology has ushered in a new era of immunotherapy, enabling the targeting of a broad range of surface antigens, surpassing the limitations of traditional T cell epitopes. Despite the wide range of non-protein tumor-associated antigens, the advancement in crafting CAR T cells for these targets has been limited. Owing to an evolutionary defect in the CMP-Neu5Ac hydroxylase (CMAH) that abolishes the synthesis of CMP-Neu5Gc from CMP-Neu5Ac, Neu5Gc is generally absent in human tissues. Despite this, Neu5Gc-containing antigens, including the ganglioside GM3(Neu5Gc) have consistently been observed on tumor cells across a variety of human malignancies. This restricted expression makes GM3(Neu5Gc) an appealing and highly specific target for immunotherapy. In this study, we designed and evaluated 14F7-28z CAR T cells, with a targeting unit derived from the GM3(Neu5Gc)-specific murine antibody 14F7. These cells exhibited exceptional specificity, proficiently targeting GM3(Neu5Gc)-expressing murine tumor cells in syngeneic mouse models, ranging from B cell malignancies to epithelial tumors, without compromising safety. Notably, human tumor cells enhanced with murine Cmah were effectively targeted and eliminated by the 14F7 CAR T cells. Nonetheless, despite the detectable presence of GM3(Neu5Gc) in unmodified human tumor xenografts, the levels were insufficient to trigger a tumoricidal T-cell response with the current CAR T cell configuration. Overall, our findings highlight the potential of targeting the GM3(Neu5Gc) ganglioside using CAR T cells across a variety of cancers and set the stage for the optimization of 14F7-based therapies for future human clinical application.
Collapse
Affiliation(s)
- Julia Heinzelbecker
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Centre for B cell malignancies, University of Oslo, Oslo, Norway
| | - Marte Fauskanger
- K.G. Jebsen Centre for B cell malignancies, University of Oslo, Oslo, Norway
| | - Ida Jonson
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| | - Ute Krengel
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Geir Åge Løset
- Department of Biosciences, Faculty of Mathematics and Natural Sciences University of Oslo, Oslo, Norway
- Nextera AS, Oslo, Norway
| | - Ludvig Munthe
- K.G. Jebsen Centre for B cell malignancies, University of Oslo, Oslo, Norway
| | - Anders Tveita
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Centre for B cell malignancies, University of Oslo, Oslo, Norway
| |
Collapse
|
4
|
Manni M, Mantuano NR, Zingg A, Kappos EA, Behrens AJ, Back J, Follador R, Faridmoayer A, Läubli H. Detection of N-glycolyl-neuraminic acid-containing glycolipids in human skin. Front Immunol 2023; 14:1291292. [PMID: 38094289 PMCID: PMC10716299 DOI: 10.3389/fimmu.2023.1291292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023] Open
Abstract
Humans lack the enzyme that produces the sialic acid N-glycolyl neuraminic acid (Neu5Gc), but several lines of evidence have shown that Neu5Gc can be taken up by mammalian food sources and replace the common human sialic acid N-acetyl neuraminic acid (Neu5Ac) in glycans. Cancer tissue has been shown to have increased the presence of Neu5Gc and Neu5Gc-containing glycolipids such as the ganglioside GM3, which have been proposed as tumor-specific antigens for antibody treatment. Here, we show that a previously described antibody against Neu5Gc-GM3 is binding to Neu5GC-containing gangliosides and is strongly staining different cancer tissues. However, we also found a strong intracellular staining of keratinocytes of healthy skin. We confirmed this staining on freshly isolated keratinocytes by flow cytometry and detected Neu5Gc by mass spectrometry. This finding implicates that non-human Neu5Gc can be incorporated into gangliosides in human skin, and this should be taken into consideration when targeting Neu5Gc-containing gangliosides for cancer immunotherapy.
Collapse
Affiliation(s)
- Michela Manni
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
- Glycoera AG, Wädenswil, Switzerland
| | | | - Andreas Zingg
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Elisabeth A. Kappos
- Department of Plastic, Reconstructive, Aesthetic and Handsurgery, University Hospital and University of Basel, Basel, Switzerland
| | | | | | | | | | - Heinz Läubli
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
- Division of Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
5
|
Sarkar A, Banerjee S, Biswas K. Multi-dimensional role of gangliosides in modulating cancer hallmarks and their prospects in targeted cancer therapy. Front Pharmacol 2023; 14:1282572. [PMID: 38089042 PMCID: PMC10711107 DOI: 10.3389/fphar.2023.1282572] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/14/2023] [Indexed: 12/10/2024] Open
Abstract
Gangliosides are glycosphingolipids with prevalence in nervous tissue and their involvement in certain neuronal diseases have been widely known. Interestingly, many recent studies highlighted their importance in the development and progression of various cancers through orchestration of multiple attributes of tumorigenesis, i.e., promoting migration, invasion, escaping the host immune system, and influencing other cancer hallmarks. Therefore, the multidimensional role of gangliosides in different cancers has established them as potential cancer targets. However, the tremendous structural complexity and functional heterogeneity are the major challenges in ganglioside research. Moreover, despite numerous immunotherapeutic attempts to target different gangliosides, it has failed to yield consistent results in clinical trials owing to their poor immunogenicity, a broad range of cross-reactivity, severe side effects, lack of uniform expression as well as heterogeneity. The recent identification of selective O-acetylated ganglioside expression in cancer tissues, but not in normal tissues, has strengthened their potential as a better and specific target for treating cancer patients. It was further supported by reduced cross-reactivity and side effects in clinical trials, although poor immunogenicity remains a major concern. Therefore, in addition to characterization and identification of the biological importance of O-acetylated gangliosides, their specific and efficient targeting in cancer through engineered antibodies is an emerging area of glycobiology research. This review highlights the modulatory effect of select gangliosides on different hallmarks of cancer and presents the overall development of ganglioside targeted immunotherapies along with recent progress. Here, we have also discussed its potential for future modifications aimed towards improvement in ganglioside-based cancer therapies.
Collapse
Affiliation(s)
| | | | - Kaushik Biswas
- Department of Biological Sciences, Bose Institute, Kolkata, India
| |
Collapse
|
6
|
Martynowycz MW, Andreev K, Mor A, Gidalevitz D. Cancer-Associated Gangliosides as a Therapeutic Target for Host Defense Peptide Mimics. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:12541-12549. [PMID: 37647566 DOI: 10.1021/acs.langmuir.3c01008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Aberrant levels of glycolipids expressed on cellular surfaces are characteristic of different types of cancers. The oligomer of acylated lysine (OAK) mimicking antimicrobial peptides displays in vitro activity against human and murine melanoma cell lines with upregulated GD3 and GM3 gangliosides. Herein, we demonstrate the capability of OAK to intercalate into the sialo-oligosaccharides of DPPC/GD3 and DPPC/GM3 lipid monolayers using X-ray scattering. The lack of insertion into monolayers containing phosphatidylserine suggests that the mechanism of action by OAKs against glycosylated lipid membranes is not merely driven by charge effects. The fluorescence microscopy data demonstrates the membrane-lytic activity of OAK. Understanding the molecular basis for selectivity toward GD3 and GM3 gangliosides by antimicrobial lipopeptides will contribute to the development of novel therapies to cure melanoma and other malignancies.
Collapse
Affiliation(s)
- Michael W Martynowycz
- Department of Physics, Center for Molecular Study of Condensed Soft Matter (μCoSM), Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, 10 W 35th Street, Chicago, Illinois 60616, United States
| | - Konstantin Andreev
- Department of Physics, Center for Molecular Study of Condensed Soft Matter (μCoSM), Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, 10 W 35th Street, Chicago, Illinois 60616, United States
| | - Amram Mor
- Department of Biotechnology and Food Engineering, Technion─Israel Institute of Technology, Technion City, Haifa 32000, Israel
| | - David Gidalevitz
- Department of Physics, Center for Molecular Study of Condensed Soft Matter (μCoSM), Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, 10 W 35th Street, Chicago, Illinois 60616, United States
| |
Collapse
|
7
|
Jin X, Yang GY. Pathophysiological roles and applications of glycosphingolipids in the diagnosis and treatment of cancer diseases. Prog Lipid Res 2023; 91:101241. [PMID: 37524133 DOI: 10.1016/j.plipres.2023.101241] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Glycosphingolipids (GSLs) are major amphiphilic glycolipids present on the surface of living cell membranes. They have important biological functions, including maintaining plasma membrane stability, regulating signal transduction, and mediating cell recognition and adhesion. Specific GSLs and related enzymes are abnormally expressed in many cancer diseases and affect the malignant characteristics of tumors. The regulatory roles of GSLs in signaling pathways suggest that they are involved in tumor pathogenesis. GSLs have therefore been widely studied as diagnostic markers of cancer diseases and important targets of immunotherapy. This review describes the tumor-related biological functions of GSLs and systematically introduces recent progress in using diverse GSLs and related enzymes to diagnose and treat tumor diseases. Development of drugs and biomarkers for personalized cancer therapy based on GSL structure is also discussed. These advances, combined with recent progress in the preparation of GSLs derivatives through synthetic biology technologies, suggest a strong future for the use of customized GSL libraries in treating human diseases.
Collapse
Affiliation(s)
- Xuefeng Jin
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; Department of Clinical Pharmaceutics, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Guang-Yu Yang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
8
|
Wang J, Shewell LK, Day CJ, Jennings MP. N-glycolylneuraminic acid as a carbohydrate cancer biomarker. Transl Oncol 2023; 31:101643. [PMID: 36805917 PMCID: PMC9971276 DOI: 10.1016/j.tranon.2023.101643] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/31/2023] [Accepted: 02/11/2023] [Indexed: 02/20/2023] Open
Abstract
One of the forms of aberrant glycosylation in human tumors is the expression of N-glycolylneuraminic acid (Neu5Gc). The only known enzyme to biosynthesize Neu5Gc in mammals, cytidine-5'-monophosphate-N-acetylneuraminic acid (CMAH), appears to be genetically inactivated in humans. Regardless, low levels of Neu5Gc have been detected in healthy humans. Therefore, it is proposed that the presence of Neu5Gc in humans is from dietary acquisition, such as red meat. Notably, detection of elevated Neu5Gc levels has been repeatedly found in cancer tissues, cells and serum samples, thereby Neu5Gc-containing antigens may be exploited as a class of cancer biomarkers. Here we review the findings to date on using Neu5Gc-containing tumor glycoconjugates as a class of cancer biomarkers for cancer detection, surveillance, prognosis and therapeutic targets. We review the evidence that supports an emerging hypothesis of de novo Neu5Gc biosynthesis in human cancer cells as a source of Neu5Gc in human tumors, generated under certain metabolic conditions.
Collapse
Affiliation(s)
- Jing Wang
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Lucy K Shewell
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | | | | |
Collapse
|
9
|
Doud EH, Yeh ES. Mass Spectrometry-Based Glycoproteomic Workflows for Cancer Biomarker Discovery. Technol Cancer Res Treat 2023; 22:15330338221148811. [PMID: 36740994 PMCID: PMC9903044 DOI: 10.1177/15330338221148811] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Glycosylation has a clear role in cancer initiation and progression, with numerous studies identifying distinct glycan features or specific glycoproteoforms associated with cancer. Common findings include that aggressive cancers tend to have higher expression levels of enzymes that regulate glycosylation as well as glycoproteins with greater levels of complexity, increased branching, and enhanced chain length1. Research in cancer glycoproteomics over the last 50-plus years has mainly focused on technology development used to observe global changes in glycosylation. Efforts have also been made to connect glycans to their protein carriers as well as to delineate the role of these modifications in intracellular signaling and subsequent cell function. This review discusses currently available techniques utilizing mass spectrometry-based technologies used to study glycosylation and highlights areas for future advancement.
Collapse
Affiliation(s)
- Emma H. Doud
- Center for Proteome Analysis, Indiana University School of Medicine, Indianapolis, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, USA
- IU Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, USA
| | - Elizabeth S. Yeh
- IU Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, USA
| |
Collapse
|
10
|
Bousquet PA, Manna D, Sandvik JA, Arntzen MØ, Moreno E, Sandvig K, Krengel U. SILAC-based quantitative proteomics and microscopy analysis of cancer cells treated with the N-glycolyl GM3-specific anti-tumor antibody 14F7. Front Immunol 2022; 13:994790. [PMID: 36439103 PMCID: PMC9682173 DOI: 10.3389/fimmu.2022.994790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/20/2022] [Indexed: 08/11/2024] Open
Abstract
Cancer immunotherapy represents a promising approach to specifically target and treat cancer. The most common mechanisms by which monoclonal antibodies kill cells include antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity and apoptosis, but also other mechanisms have been described. 14F7 is an antibody raised against the tumor-associated antigen NeuGc GM3, which was previously reported to kill cancer cells without inducing apoptotic pathways. The antibody was reported to induce giant membrane lesions in tumor cells, with apparent changes in the cytoskeleton. Here, we investigated the effect of humanized 14F7 on HeLa cells using stable isotope labeling with amino acids in cell culture (SILAC) in combination with LC-MS and live cell imaging. 14F7 did not kill the HeLa cells, however, it caused altered protein expression (MS data are available via ProteomeXchange with identifier PXD024320). Several cytoskeletal and nucleic-acid binding proteins were found to be strongly down-regulated in response to antibody treatment, suggesting how 14F7 may induce membrane lesions in cells that contain higher amounts of NeuGc GM3. The altered expression profile identified in this study thus contributes to an improved understanding of the unusual killing mechanism of 14F7.
Collapse
Affiliation(s)
| | - Dipankar Manna
- Department of Chemistry, University of Oslo, Oslo, Norway
| | | | | | - Ernesto Moreno
- Facultad de Ciencias Básicas, Universidad de Medellín, Medellín, Colombia
| | - Kirsten Sandvig
- Department of Biosciences, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ute Krengel
- Department of Chemistry, University of Oslo, Oslo, Norway
| |
Collapse
|
11
|
Wen R, Zhao H, Zhang D, Chiu CL, Brooks JD. Sialylated glycoproteins as biomarkers and drivers of progression in prostate cancer. Carbohydr Res 2022; 519:108598. [PMID: 35691122 DOI: 10.1016/j.carres.2022.108598] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/20/2022] [Accepted: 05/20/2022] [Indexed: 01/27/2023]
Abstract
Sialic acids have been implicated in cancer initiation, progression, and immune evasion in diverse human malignancies. Sialylation of terminal glycans on cell surface and secreted glycoproteins is a long-recognized feature of cancer cells. Recently, immune checkpoint inhibitor immunotherapy has tremendously improved the outcomes of patients with various cancers. However, available immunotherapy approaches have had limited efficacy in metastatic castration-resistant prostate cancer. Sialic acid modified glycoproteins in prostate cancers and their interaction with Siglec receptors on tumor infiltrating immune cells might underlie immunosuppressive signaling in prostate cancer. Here, we summarize the function of sialic acids and relevant glycosynthetic enzymes in cancer initiation and progression. We also discuss the possible uses of sialic acids as biomarkers in prostate cancer and the potential methods for targeting Siglec-sialic acid interactions for prostate cancer treatment.
Collapse
Affiliation(s)
- Ru Wen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Hongjuan Zhao
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Dalin Zhang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Chun-Lung Chiu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - James D Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
12
|
Guo Y, Jia W, Yang J, Zhan X. Cancer glycomics offers potential biomarkers and therapeutic targets in the framework of 3P medicine. Front Endocrinol (Lausanne) 2022; 13:970489. [PMID: 36072925 PMCID: PMC9441633 DOI: 10.3389/fendo.2022.970489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/02/2022] [Indexed: 11/30/2022] Open
Abstract
Glycosylation is one of the most important post-translational modifications (PTMs) in a protein, and is the most abundant and diverse biopolymer in nature. Glycans are involved in multiple biological processes of cancer initiation and progression, including cell-cell interactions, cell-extracellular matrix interactions, tumor invasion and metastasis, tumor angiogenesis, and immune regulation. As an important biomarker, tumor-associated glycosylation changes have been extensively studied. This article reviews recent advances in glycosylation-based biomarker research, which is useful for cancer diagnosis and prognostic assessment. Truncated O-glycans, sialylation, fucosylation, and complex branched structures have been found to be the most common structural patterns in malignant tumors. In recent years, immunochemical methods, lectin recognition-based methods, mass spectrometry (MS)-related methods, and fluorescence imaging-based in situ methods have greatly promoted the discovery and application potentials of glycomic and glycoprotein biomarkers in various cancers. In particular, MS-based proteomics has significantly facilitated the comprehensive research of extracellular glycoproteins, increasing our understanding of their critical roles in regulating cellular activities. Predictive, preventive and personalized medicine (PPPM; 3P medicine) is an effective approach of early prediction, prevention and personalized treatment for different patients, and it is known as the new direction of medical development in the 21st century and represents the ultimate goal and highest stage of medical development. Glycosylation has been revealed to have new diagnostic, prognostic, and even therapeutic potentials. The purpose of glycosylation analysis and utilization of biology is to make a fundamental change in health care and medical practice, so as to lead medical research and practice into a new era of 3P medicine.
Collapse
Affiliation(s)
- Yuna Guo
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Wenshuang Jia
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Jingru Yang
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Xianquan Zhan
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| |
Collapse
|
13
|
Cribioli E, Giordano Attianese GMP, Coukos G, Irving M. CAR T cells targeting the ganglioside NGcGM3 control ovarian tumors in the absence of toxicity against healthy tissues. Front Immunol 2022; 13:951143. [PMID: 35990626 PMCID: PMC9389107 DOI: 10.3389/fimmu.2022.951143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/30/2022] [Indexed: 11/30/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells have emerged as a powerful immunotherapeutic tool against certain hematological malignancies but a significant proportion of patients either do not respond or they relapse, sometimes as a result of target antigen loss. Moreover, limited clinical benefit has been reported for CAR therapy against epithelial derived solid tumors. A major reason for this is the paucity of solid tumor antigens identified to date that are broadly, homogeneously and stably expressed but not found on healthy tissues. To address this, here we describe the development and evaluation of CAR T cells directed against N-glycoslylated ganglioside monosialic 3 (NGcGM3). NGcGM3 derives from the enzymatic hydroxylation of N-acetylneuraminic acid (NAc) GM3 (NAcGM3) and it is present on the surface of a range of cancers including ovarian, breast, melanoma and lymphoma. However, while NAcGM3 is found on healthy human cells, NGcGM3 is not due to the 7deletion of an exon in the gene encoding for the enzyme cytidine monophospho-N-acetylneuraminic acid hydroxylase (CMAH). Indeed, unlike for most mammals, in humans NGcGM3 is considered a neoantigen as its presence on tumors is the result of metabolic incorporation from dietary sources. Here, we have generated 3 CARs comprising different single chain variable fragments (scFvs) originating from the well-characterized monoclonal antibody (mAb) 14F7. We show reactivity of the CAR T cells against a range of patient tumor fragments and we demonstrate control of NGcGM3+ SKOV3 ovarian tumors in the absence of toxicity despite the expression of CMAH and presence of NGcGM3+ on healthy tissues in NSG mice. Taken together, our data indicate clinical potential for 14F7-based CAR T cells against a range of cancers, both in terms of efficacy and of patient safety.
Collapse
|
14
|
Li Z, Liu L, Unione L, Lang Y, de Groot RJ, Boons GJ. Synthetic O-Acetyl- N-glycolylneuraminic Acid Oligosaccharides Reveal Host-Associated Binding Patterns of Coronaviral Glycoproteins. ACS Infect Dis 2022; 8:1041-1050. [PMID: 35416033 DOI: 10.1021/acsinfecdis.2c00046] [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: 11/28/2022]
Abstract
A panel of O-acetylated N-glycolylneuraminic acid oligosaccharides has been prepared by diversification of common synthetic precursors by regioselective de-O-acetylation by coronaviral hemagglutinin-esterase (HE) combined with C7-to-C9 acetyl ester migration. The resulting compound library was printed on streptavidin-coated glass slides to give a microarray to investigate receptor binding specificities of viral envelope glycoproteins, including spike proteins and HEs from animal and human coronaviruses. It was found that the binding patterns of the viral proteins for N-glycolylated sialosides differ considerable from those of the previously synthesized N-acetylated counterparts. Generally, the spike proteins tolerate N-glycolyl modification, but selectivities differ among viruses targeting different hosts. On the other hand, the lectin domain of the corresponding HEs showed a substantial decrease or loss of binding of N-glycolylated sialosides. MD simulations indicate that glycolyl recognition by HE is mediated by polar residues in a loop region (109-119) that interacts with the 5-N-glycolyl moiety. Collectively, the results indicate that coronaviruses have adjusted their receptor fine specificities to adapt to the sialoglycome of their host species.
Collapse
Affiliation(s)
- Zeshi Li
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Lin Liu
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
| | - Luca Unione
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Yifei Lang
- Virology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - Raoul J. de Groot
- Virology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - Geert-Jan Boons
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
- Bijvoet Center for Biomolecular Research, Utrecht University, 3584 CHUtrecht, The Netherlands
- Chemistry Department, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
15
|
Abou Khouzam R, Zaarour RF, Brodaczewska K, Azakir B, Venkatesh GH, Thiery J, Terry S, Chouaib S. The Effect of Hypoxia and Hypoxia-Associated Pathways in the Regulation of Antitumor Response: Friends or Foes? Front Immunol 2022; 13:828875. [PMID: 35211123 PMCID: PMC8861358 DOI: 10.3389/fimmu.2022.828875] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 01/19/2022] [Indexed: 12/15/2022] Open
Abstract
Hypoxia is an environmental stressor that is instigated by low oxygen availability. It fuels the progression of solid tumors by driving tumor plasticity, heterogeneity, stemness and genomic instability. Hypoxia metabolically reprograms the tumor microenvironment (TME), adding insult to injury to the acidic, nutrient deprived and poorly vascularized conditions that act to dampen immune cell function. Through its impact on key cancer hallmarks and by creating a physical barrier conducive to tumor survival, hypoxia modulates tumor cell escape from the mounted immune response. The tumor cell-immune cell crosstalk in the context of a hypoxic TME tips the balance towards a cold and immunosuppressed microenvironment that is resistant to immune checkpoint inhibitors (ICI). Nonetheless, evidence is emerging that could make hypoxia an asset for improving response to ICI. Tackling the tumor immune contexture has taken on an in silico, digitalized approach with an increasing number of studies applying bioinformatics to deconvolute the cellular and non-cellular elements of the TME. Such approaches have additionally been combined with signature-based proxies of hypoxia to further dissect the turbulent hypoxia-immune relationship. In this review we will be highlighting the mechanisms by which hypoxia impacts immune cell functions and how that could translate to predicting response to immunotherapy in an era of machine learning and computational biology.
Collapse
Affiliation(s)
- Raefa Abou Khouzam
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Rania Faouzi Zaarour
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Klaudia Brodaczewska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Bilal Azakir
- Faculty of Medicine, Beirut Arab University, Beirut, Lebanon
| | - Goutham Hassan Venkatesh
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Jerome Thiery
- INSERM U1186, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.,Faculty of Medicine, University Paris Sud, Le Kremlin Bicêtre, France
| | - Stéphane Terry
- INSERM U1186, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.,Faculty of Medicine, University Paris Sud, Le Kremlin Bicêtre, France.,Research Department, Inovarion, Paris, France
| | - Salem Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates.,INSERM U1186, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
16
|
Berois N, Pittini A, Osinaga E. Targeting Tumor Glycans for Cancer Therapy: Successes, Limitations, and Perspectives. Cancers (Basel) 2022; 14:cancers14030645. [PMID: 35158915 PMCID: PMC8833780 DOI: 10.3390/cancers14030645] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Aberrant glycosylation is a common feature of many cancers, and it plays crucial roles in tumor development and biology. Cancer progression can be regulated by several physiopathological processes controlled by glycosylation, such as cell–cell adhesion, cell–matrix interaction, epithelial-to-mesenchymal transition, tumor proliferation, invasion, and metastasis. Different mechanisms of aberrant glycosylation lead to the formation of tumor-associated carbohydrate antigens (TACAs), which are suitable for selective cancer targeting, as well as novel antitumor immunotherapy approaches. This review summarizes the strategies developed in cancer immunotherapy targeting TACAs, analyzing molecular and cellular mechanisms and state-of-the-art methods in clinical oncology. Abstract Aberrant glycosylation is a hallmark of cancer and can lead to changes that influence tumor behavior. Glycans can serve as a source of novel clinical biomarker developments, providing a set of specific targets for therapeutic intervention. Different mechanisms of aberrant glycosylation lead to the formation of tumor-associated carbohydrate antigens (TACAs) suitable for selective cancer-targeting therapy. The best characterized TACAs are truncated O-glycans (Tn, TF, and sialyl-Tn antigens), gangliosides (GD2, GD3, GM2, GM3, fucosyl-GM1), globo-serie glycans (Globo-H, SSEA-3, SSEA-4), Lewis antigens, and polysialic acid. In this review, we analyze strategies for cancer immunotherapy targeting TACAs, including different antibody developments, the production of vaccines, and the generation of CAR-T cells. Some approaches have been approved for clinical use, such as anti-GD2 antibodies. Moreover, in terms of the antitumor mechanisms against different TACAs, we show results of selected clinical trials, considering the horizons that have opened up as a result of recent developments in technologies used for cancer control.
Collapse
Affiliation(s)
- Nora Berois
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay;
- Correspondence: (N.B.); (E.O.)
| | - Alvaro Pittini
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay;
- Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Eduardo Osinaga
- Laboratorio de Glicobiología e Inmunología Tumoral, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay;
- Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
- Correspondence: (N.B.); (E.O.)
| |
Collapse
|
17
|
Bjerregaard-Andersen K, Abraha F, Johannesen H, Oscarson S, Moreno E, Krengel U. Key role of a structural water molecule for the specificity of 14F7-An antitumor antibody targeting the NeuGc GM3 ganglioside. Glycobiology 2021; 31:1500-1509. [PMID: 34735569 PMCID: PMC8684480 DOI: 10.1093/glycob/cwab076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/19/2021] [Accepted: 06/28/2021] [Indexed: 12/22/2022] Open
Abstract
Tumor-associated glycolipids such as NeuGc GM3 are auspicious molecular targets in antineoplastic therapies and vaccine strategies. 14F7 is a monoclonal IgG1 with high clinical potential in cancer immunotherapy as it displays extraordinary specificity for NeuGc GM3, while it does not recognize the very similar, ubiquitous NeuAc GM3. Here we present the 2.3 Å crystal structure of the 14F7 antigen-binding domain (14F7 scFv) in complex with the NeuGc GM3 trisaccharide. Modeling analysis and previous mutagenesis data suggest that 14F7 may also bind to an alternative NeuGc GM3 conformation, not observed in the crystal structure. The most intriguing finding, however, was that a water molecule centrally placed in the complementarity-determining region directly mediates the specificity of 14F7 to NeuGc GM3. This has profound impact on the complexity of engineering in the binding site and provides an excellent example of the importance in understanding the water structure in antibody-antigen interactions.
Collapse
Affiliation(s)
| | - Fana Abraha
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland.,Recipharm OT Chemistry, Uppsala, Sweden
| | - Hedda Johannesen
- Department of Chemistry, University of Oslo, NO-0315 Oslo, Norway.,Department of Biosciences, University of Oslo, NO-0316 Oslo, Norway
| | - Stefan Oscarson
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Ernesto Moreno
- Facultad de Ciencias Básicas, Universidad de Medellín, Medellín 050026, Colombia
| | - Ute Krengel
- Department of Chemistry, University of Oslo, NO-0315 Oslo, Norway
| |
Collapse
|
18
|
Hugonnet M, Singh P, Haas Q, von Gunten S. The Distinct Roles of Sialyltransferases in Cancer Biology and Onco-Immunology. Front Immunol 2021; 12:799861. [PMID: 34975914 PMCID: PMC8718907 DOI: 10.3389/fimmu.2021.799861] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/02/2021] [Indexed: 12/24/2022] Open
Abstract
Aberrant glycosylation is a key feature of malignant transformation. Hypersialylation, the enhanced expression of sialic acid-terminated glycoconjugates on the cell surface, has been linked to immune evasion and metastatic spread, eventually by interaction with sialoglycan-binding lectins, including Siglecs and selectins. The biosynthesis of tumor-associated sialoglycans involves sialyltransferases, which are differentially expressed in cancer cells. In this review article, we provide an overview of the twenty human sialyltransferases and their roles in cancer biology and immunity. A better understanding of the individual contribution of select sialyltransferases to the tumor sialome may lead to more personalized strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Marjolaine Hugonnet
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland
| | - Pushpita Singh
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Quentin Haas
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Stephan von Gunten
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland
| |
Collapse
|
19
|
Nishikawa Y, Toda S, Matsui T, Takada H, Takemoto K, Hara O. Site-Selective Acylations of α- and β-Hydroxyamides in Complex Molecules: Application of Template-Driven Acylation to Disaccharides and a Glycopeptide. Org Lett 2021; 23:2715-2719. [PMID: 33734719 DOI: 10.1021/acs.orglett.1c00612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Site-selective acylations of α-and β-hydroxyamides in complex polyols are described. The combination of a pyridine aldoxime ester and Zn(OTf)2 facilitates the acylation of two types of N-glycolyl disaccharides, namely, Gal-GlcNGc and Neu5Gc-Gal, both of which are partial structures of polysaccharides responsible for biological actions, with highly site-selective modifications achieved. Furthermore, biotinylation, one of the most important techniques in chemical biology, is used to site-selectively acylate the β-hydroxyl group in a glycopeptide.
Collapse
Affiliation(s)
- Yasuhiro Nishikawa
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi 468-8503, Japan
| | - Shione Toda
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi 468-8503, Japan
| | - Takami Matsui
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi 468-8503, Japan
| | - Hanae Takada
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi 468-8503, Japan
| | - Kohei Takemoto
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi 468-8503, Japan
| | - Osamu Hara
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi 468-8503, Japan
| |
Collapse
|
20
|
Phenylboronic acid conjugated to doxorubicin nanocomplexes as an anti-cancer drug delivery system in hepatocellular carcinoma. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 34:102389. [PMID: 33753281 DOI: 10.1016/j.nano.2021.102389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 01/11/2021] [Accepted: 03/03/2021] [Indexed: 12/22/2022]
Abstract
Anti-cancer strategies using nanocarrier systems have been explored in a variety of cancers; these systems can easily be incorporated into tumors via the enhanced permeability and retention (EPR) effect leading to enhanced anti-tumor activity while reducing systemic toxicity by specific tumor-targeting. The prognosis of hepatocellular carcinoma (HCC) is extremely poor when the condition is diagnosed at the unresectable stage as treatment options are limited. In order to improve the treatment of cancer and the overall anti-cancer effect, polymerized phenylboronic acid conjugated doxorubicin (pPBA-Dox) nanocomplexes were generated, and conjugated doxorubicin, which is conventionally used in HCC. The nanocomplexes exhibited enhanced anti-tumor activity via tumor-specific targeting in the subcutaneous and orthotopic HCC syngeneic mice tumor model, implying that the nanocomplexes facilitate the targeted Dox delivery to liver cancer in which the sialic acid is over-expressed. Therefore, this study provides insight into the novel targeted therapy using the nanocomplexes for the treatment of HCC.
Collapse
|
21
|
Abstract
Introduction: Targeting immune checkpoints with antibodies has significantly improved the outcome of cancer patients, but only few patients have long-term benefits from currently used PD-1/PD-L1 and CTLA-4 inhibitors. New approaches are needed to increase the number of patients going into long-term remission after cancer immunotherapy. Glyco-immune checkpoints are new targets for cancer immunotherapy. They are defined as immune-modulatory pathways including interactions of glycans with glycan-binding proteins or lectins. The most prominent pathway is the sialoglycan-Siglec axis and inhibitors of this axis are already successfully tested in early clinical trials.Area covered: Here, we summarize the current knowledge on glyco-immune checkpoints with a focus on the sialoglycan-Siglec axis. We also provide an overview on current approaches to clinically target glyco-immune checkpoints and give an outlook for the further clinical development of glyco-immune checkpoint targeting agents.Expert opinion: Glyco-immune checkpoints are interesting new targets to improve cancer immunotherapy. Antibodies targeting the sialoglycan-Siglec axis are already in clinical development. Other approaches with higher risk of toxicity including tumor-targeted sialidases are in late stage pre-clinical development. Despite the challenges, targeting of glyco-immune checkpoints could lead to the development of a new class of drugs providing improved anti-cancer immunity and eventually benefit cancer patients.
Collapse
Affiliation(s)
- Michela Manni
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University of Basel, and Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University of Basel, and Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
22
|
Sarbu M, Clemmer DE, Zamfir AD. Ion mobility mass spectrometry of human melanoma gangliosides. Biochimie 2020; 177:226-237. [DOI: 10.1016/j.biochi.2020.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/05/2020] [Accepted: 08/18/2020] [Indexed: 02/09/2023]
|
23
|
Prognostic Significance of Tumor Tissue NeuGcGM3 Ganglioside Expression in Patients Receiving Racotumomab Immunotherapy. JOURNAL OF ONCOLOGY 2020; 2020:1360431. [PMID: 32670370 PMCID: PMC7341415 DOI: 10.1155/2020/1360431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/01/2020] [Accepted: 05/19/2020] [Indexed: 01/05/2023]
Abstract
Expression of N-glycolyl GM3 (NeuGcGM3) ganglioside was detected in the tumor specimens of patients who were on Racotumomab anti-idiotype vaccine maintenance treatment, and prognostic significance as a biomarker was investigated. No statistically significant association was observed in the multivariate analysis between overall survival and tissue NeuGcGM3 IHC levels. Although numerically there was a difference favoring less intense IHC for better prognosis, this did not reach statistical power. However, there was a strong correlation between Racotumomab doses and overall survival (OS). Mean OS of the patient with more than 10 Racotumomab application was significantly longer than the patient who had less than 10 injections (70.7 months vs. 31.1 months, p < 0.001). We propose that, regardless of staining intensity, the presence of NeuGcGM3 in patient tissues might be an indicator of benefit in Racotumomab treatment.
Collapse
|
24
|
Boligan KF, Oechtering J, Keller CW, Peschke B, Rieben R, Bovin N, Kappos L, Cummings RD, Kuhle J, von Gunten S, Lünemann JD. Xenogeneic Neu5Gc and self-glycan Neu5Ac epitopes are potential immune targets in MS. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 7:7/2/e676. [PMID: 32014849 PMCID: PMC7051216 DOI: 10.1212/nxi.0000000000000676] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To explore the repertoire of glycan-specific immunoglobulin G (IgG) antibodies in treatment-naive patients with relapsing-remitting multiple sclerosis (RRMS). METHODS A systems-level approach combined with glycan array technologies was used to determine specificities and binding reactivities of glycan-specific IgGs in treatment-naive patients with RRMS compared with patients with noninflammatory and other inflammatory neurologic diseases. RESULTS We identified a unique signature of glycan-binding IgG in MS with high reactivities to the dietary xenoglycan N-glycolylneuraminic acid (Neu5Gc) and the self-glycan N-acetylneuraminic acid (Neu5Ac). Increased reactivities of serum IgG toward Neu5Gc and Neu5Ac were additionally observed in an independent, treatment-naive cohort of patients with RRMS. CONCLUSION Patients with MS show increased IgG reactivities to structurally related xenogeneic and human neuraminic acids. The discovery of these glycan-specific epitopes as immune targets and potential biomarkers in MS merits further investigation.
Collapse
Affiliation(s)
- Kayluz F Boligan
- From the Institute of Pharmacology (K.F.B., S.v.G.), University of Bern, Switzerland; Neurologic Clinic and Policlinic (J.O., L.K., J.K.), Departments of Medicine, Clinical Research, Biomedicine and Biomedical Engineering, University Hospital Basel, University of Basel, Switzerland; Department of Neurology with Institute of Translational Neurology (C.W.K., J.D.L.), University Hospital Münster, University of Münster, Germany; Laboratory of Neuroinflammation (C.W.K., B.P., J.D.L.), Institute of Experimental Immunology, University of Zurich, Switzerland; Department for BioMedical Research (DBMR) (R.R.), University of Bern, Switzerland; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Science (N.B.), Moscow, Russia; Auckland University of Technology (N.B.), New Zealand; and Department of Surgery (R.D.C.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Johanna Oechtering
- From the Institute of Pharmacology (K.F.B., S.v.G.), University of Bern, Switzerland; Neurologic Clinic and Policlinic (J.O., L.K., J.K.), Departments of Medicine, Clinical Research, Biomedicine and Biomedical Engineering, University Hospital Basel, University of Basel, Switzerland; Department of Neurology with Institute of Translational Neurology (C.W.K., J.D.L.), University Hospital Münster, University of Münster, Germany; Laboratory of Neuroinflammation (C.W.K., B.P., J.D.L.), Institute of Experimental Immunology, University of Zurich, Switzerland; Department for BioMedical Research (DBMR) (R.R.), University of Bern, Switzerland; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Science (N.B.), Moscow, Russia; Auckland University of Technology (N.B.), New Zealand; and Department of Surgery (R.D.C.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Christian W Keller
- From the Institute of Pharmacology (K.F.B., S.v.G.), University of Bern, Switzerland; Neurologic Clinic and Policlinic (J.O., L.K., J.K.), Departments of Medicine, Clinical Research, Biomedicine and Biomedical Engineering, University Hospital Basel, University of Basel, Switzerland; Department of Neurology with Institute of Translational Neurology (C.W.K., J.D.L.), University Hospital Münster, University of Münster, Germany; Laboratory of Neuroinflammation (C.W.K., B.P., J.D.L.), Institute of Experimental Immunology, University of Zurich, Switzerland; Department for BioMedical Research (DBMR) (R.R.), University of Bern, Switzerland; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Science (N.B.), Moscow, Russia; Auckland University of Technology (N.B.), New Zealand; and Department of Surgery (R.D.C.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Benjamin Peschke
- From the Institute of Pharmacology (K.F.B., S.v.G.), University of Bern, Switzerland; Neurologic Clinic and Policlinic (J.O., L.K., J.K.), Departments of Medicine, Clinical Research, Biomedicine and Biomedical Engineering, University Hospital Basel, University of Basel, Switzerland; Department of Neurology with Institute of Translational Neurology (C.W.K., J.D.L.), University Hospital Münster, University of Münster, Germany; Laboratory of Neuroinflammation (C.W.K., B.P., J.D.L.), Institute of Experimental Immunology, University of Zurich, Switzerland; Department for BioMedical Research (DBMR) (R.R.), University of Bern, Switzerland; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Science (N.B.), Moscow, Russia; Auckland University of Technology (N.B.), New Zealand; and Department of Surgery (R.D.C.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Robert Rieben
- From the Institute of Pharmacology (K.F.B., S.v.G.), University of Bern, Switzerland; Neurologic Clinic and Policlinic (J.O., L.K., J.K.), Departments of Medicine, Clinical Research, Biomedicine and Biomedical Engineering, University Hospital Basel, University of Basel, Switzerland; Department of Neurology with Institute of Translational Neurology (C.W.K., J.D.L.), University Hospital Münster, University of Münster, Germany; Laboratory of Neuroinflammation (C.W.K., B.P., J.D.L.), Institute of Experimental Immunology, University of Zurich, Switzerland; Department for BioMedical Research (DBMR) (R.R.), University of Bern, Switzerland; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Science (N.B.), Moscow, Russia; Auckland University of Technology (N.B.), New Zealand; and Department of Surgery (R.D.C.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Nicolai Bovin
- From the Institute of Pharmacology (K.F.B., S.v.G.), University of Bern, Switzerland; Neurologic Clinic and Policlinic (J.O., L.K., J.K.), Departments of Medicine, Clinical Research, Biomedicine and Biomedical Engineering, University Hospital Basel, University of Basel, Switzerland; Department of Neurology with Institute of Translational Neurology (C.W.K., J.D.L.), University Hospital Münster, University of Münster, Germany; Laboratory of Neuroinflammation (C.W.K., B.P., J.D.L.), Institute of Experimental Immunology, University of Zurich, Switzerland; Department for BioMedical Research (DBMR) (R.R.), University of Bern, Switzerland; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Science (N.B.), Moscow, Russia; Auckland University of Technology (N.B.), New Zealand; and Department of Surgery (R.D.C.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Ludwig Kappos
- From the Institute of Pharmacology (K.F.B., S.v.G.), University of Bern, Switzerland; Neurologic Clinic and Policlinic (J.O., L.K., J.K.), Departments of Medicine, Clinical Research, Biomedicine and Biomedical Engineering, University Hospital Basel, University of Basel, Switzerland; Department of Neurology with Institute of Translational Neurology (C.W.K., J.D.L.), University Hospital Münster, University of Münster, Germany; Laboratory of Neuroinflammation (C.W.K., B.P., J.D.L.), Institute of Experimental Immunology, University of Zurich, Switzerland; Department for BioMedical Research (DBMR) (R.R.), University of Bern, Switzerland; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Science (N.B.), Moscow, Russia; Auckland University of Technology (N.B.), New Zealand; and Department of Surgery (R.D.C.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Richard D Cummings
- From the Institute of Pharmacology (K.F.B., S.v.G.), University of Bern, Switzerland; Neurologic Clinic and Policlinic (J.O., L.K., J.K.), Departments of Medicine, Clinical Research, Biomedicine and Biomedical Engineering, University Hospital Basel, University of Basel, Switzerland; Department of Neurology with Institute of Translational Neurology (C.W.K., J.D.L.), University Hospital Münster, University of Münster, Germany; Laboratory of Neuroinflammation (C.W.K., B.P., J.D.L.), Institute of Experimental Immunology, University of Zurich, Switzerland; Department for BioMedical Research (DBMR) (R.R.), University of Bern, Switzerland; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Science (N.B.), Moscow, Russia; Auckland University of Technology (N.B.), New Zealand; and Department of Surgery (R.D.C.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Jens Kuhle
- From the Institute of Pharmacology (K.F.B., S.v.G.), University of Bern, Switzerland; Neurologic Clinic and Policlinic (J.O., L.K., J.K.), Departments of Medicine, Clinical Research, Biomedicine and Biomedical Engineering, University Hospital Basel, University of Basel, Switzerland; Department of Neurology with Institute of Translational Neurology (C.W.K., J.D.L.), University Hospital Münster, University of Münster, Germany; Laboratory of Neuroinflammation (C.W.K., B.P., J.D.L.), Institute of Experimental Immunology, University of Zurich, Switzerland; Department for BioMedical Research (DBMR) (R.R.), University of Bern, Switzerland; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Science (N.B.), Moscow, Russia; Auckland University of Technology (N.B.), New Zealand; and Department of Surgery (R.D.C.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Stephan von Gunten
- From the Institute of Pharmacology (K.F.B., S.v.G.), University of Bern, Switzerland; Neurologic Clinic and Policlinic (J.O., L.K., J.K.), Departments of Medicine, Clinical Research, Biomedicine and Biomedical Engineering, University Hospital Basel, University of Basel, Switzerland; Department of Neurology with Institute of Translational Neurology (C.W.K., J.D.L.), University Hospital Münster, University of Münster, Germany; Laboratory of Neuroinflammation (C.W.K., B.P., J.D.L.), Institute of Experimental Immunology, University of Zurich, Switzerland; Department for BioMedical Research (DBMR) (R.R.), University of Bern, Switzerland; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Science (N.B.), Moscow, Russia; Auckland University of Technology (N.B.), New Zealand; and Department of Surgery (R.D.C.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Jan D Lünemann
- From the Institute of Pharmacology (K.F.B., S.v.G.), University of Bern, Switzerland; Neurologic Clinic and Policlinic (J.O., L.K., J.K.), Departments of Medicine, Clinical Research, Biomedicine and Biomedical Engineering, University Hospital Basel, University of Basel, Switzerland; Department of Neurology with Institute of Translational Neurology (C.W.K., J.D.L.), University Hospital Münster, University of Münster, Germany; Laboratory of Neuroinflammation (C.W.K., B.P., J.D.L.), Institute of Experimental Immunology, University of Zurich, Switzerland; Department for BioMedical Research (DBMR) (R.R.), University of Bern, Switzerland; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Science (N.B.), Moscow, Russia; Auckland University of Technology (N.B.), New Zealand; and Department of Surgery (R.D.C.), Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA.
| |
Collapse
|