1
|
Ustjanzew A, Nedwed AS, Sandhoff R, Faber J, Marini F, Paret C. Unraveling the glycosphingolipid metabolism by leveraging transcriptome-weighted network analysis on neuroblastic tumors. Cancer Metab 2024; 12:29. [PMID: 39449099 PMCID: PMC11515559 DOI: 10.1186/s40170-024-00358-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Glycosphingolipids (GSLs) are membrane lipids composed of a ceramide backbone linked to a glycan moiety. Ganglioside biosynthesis is a part of the GSL metabolism, which involves sequential reactions catalyzed by specific enzymes that in part have a poor substrate specificity. GSLs are deregulated in cancer, thus playing a role as potential biomarkers for personalized therapy or subtype classification. However, the analysis of GSL profiles is complex and requires dedicated technologies, that are currently not included in the commonly utilized high-throughput assays adopted in contexts such as molecular tumor boards. METHODS In this study, we developed a method to discriminate the enzyme activity among the four series of the ganglioside metabolism pathway by incorporating transcriptome data and topological information of the metabolic network. We introduced three adjustment options for reaction activity scores (RAS) and demonstrated their application in both exploratory and comparative analyses by applying the method on neuroblastic tumors (NTs), encompassing neuroblastoma (NB), ganglioneuroblastoma (GNB), and ganglioneuroma (GN). Furthermore, we interpreted the results in the context of earlier published GSL measurements in the same tumors. RESULTS By adjusting RAS values using a weighting scheme based on network topology and transition probabilities (TPs), the individual series of ganglioside metabolism can be differentiated, enabling a refined analysis of the GSL profile in NT entities. Notably, the adjustment method we propose reveals the differential engagement of the ganglioside series between NB and GNB. Moreover, MYCN gene expression, a well-known prognostic marker in NTs, appears to correlate with the expression of therapeutically relevant gangliosides, such as GD2. Using unsupervised learning, we identified subclusters within NB based on altered GSL metabolism. CONCLUSION Our study demonstrates the utility of adjusting RAS values in discriminating ganglioside metabolism subtypes, highlighting the potential for identifying novel cancer subgroups based on sphingolipid profiles. These findings contribute to a better understanding of ganglioside dysregulation in NT and may have implications for stratification and targeted therapeutic strategies in these tumors and other tumor entities with a deregulated GSL metabolism.
Collapse
Affiliation(s)
- Arsenij Ustjanzew
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany.
| | - Annekathrin Silvia Nedwed
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany
| | - Roger Sandhoff
- Lipid Pathobiochemistry, German Cancer Research Center, Heidelberg, 69120, Germany
| | - Jörg Faber
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany
- University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany
| | - Federico Marini
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany
- Research Center for Immunotherapy (FZI), Mainz, 55131, Germany
| | - Claudia Paret
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany
- University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, 55131, Germany
- Research Center for Immunotherapy (FZI), Mainz, 55131, Germany
| |
Collapse
|
2
|
Khamrui E, Banerjee S, Mukherjee DD, Biswas K. Emerging role of MAPK signaling in glycosphingolipid-associated tumorigenesis. Glycoconj J 2024; 41:343-360. [PMID: 39368037 DOI: 10.1007/s10719-024-10168-5] [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: 08/16/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024]
Abstract
Glycosphingolipids (GSLs) are a type of amphipathic lipid molecules consisting of hydrophobic ceramide backbone bound to carbohydrate moiety clustered in the cell surface microdomains named 'lipid rafts' and are known to participate in cell-cell communication as well as intra-cellular signaling, thereby facilitating critical normal cellular processes and functions. Over the past several decades, various GSLs have been reported to be aberrantly expressed in different cancers, many of which have been associated with their prognosis. The wide implication of MAPK signaling in controlling tumor growth, progression, and metastasis through activation of an upstream signaling cascade, often originating in the cell membrane, justifies the rationale for its plausible influence on MAPK signaling. This review highlights the role of GSLs and their metabolites in regulating different signaling pathways towards modulation of tumor cell growth, migration, and adhesion by interacting with various receptors [epidermal growth factor receptor (EGFR), and platelet derived growth factor receptor (PDGFR), and other receptor tyrosine kinases (RTKs)] leading to activation of the MAPK pathway. Furthermore, GSLs can influence the activity and localization of downstream signaling components in the MAPK pathway by regulating the activation state of kinases, which in turn, regulate the activity of MAPKs. Additionally, this review further consolidates the GSL-mediated modulation of MAPK pathway components through the regulation of gene expression. Finally, recent findings on GSL-MAPK crosstalk will be explored in this article for the identification of potential anti-cancer therapeutic targets.
Collapse
Affiliation(s)
- Elora Khamrui
- Department of Biological Sciences, Bose Institute, EN-80, Bidhan Nagar, Salt Lake, Sector-V, Kolkata, West Bengal, 700091, India
| | - Sounak Banerjee
- Department of Biological Sciences, Bose Institute, EN-80, Bidhan Nagar, Salt Lake, Sector-V, Kolkata, West Bengal, 700091, India
| | - Dipanwita Das Mukherjee
- Department of Biological Sciences, Bose Institute, EN-80, Bidhan Nagar, Salt Lake, Sector-V, Kolkata, West Bengal, 700091, India
| | - Kaushik Biswas
- Department of Biological Sciences, Bose Institute, EN-80, Bidhan Nagar, Salt Lake, Sector-V, Kolkata, West Bengal, 700091, India.
| |
Collapse
|
3
|
Markotić A, Omerović J, Marijan S, Režić-Mužinić N, Čikeš Čulić V. Biochemical Pathways Delivering Distinct Glycosphingolipid Patterns in MDA-MB-231 and MCF-7 Breast Cancer Cells. Curr Issues Mol Biol 2024; 46:10200-10217. [PMID: 39329960 PMCID: PMC11430773 DOI: 10.3390/cimb46090608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024] Open
Abstract
The complex structure of glycosphingolipids (GSLs) supports their important role in cell function as modulators of growth factor receptors and glutamine transporters in plasma membranes. The aberrant composition of clustered GSLs within signaling platforms, so-called lipid rafts, inevitably leads to tumorigenesis due to disturbed growth factor signal transduction and excessive uptake of glutamine and other molecules needed for increased energy and structural molecule cell supply. GSLs are also involved in plasma membrane processes such as cell adhesion, and their transition converts cells from epithelial to mesenchymal with features required for cell migration and metastasis. Glutamine activates the mechanistic target of rapamycin complex 1 (mTORC1), resulting in nucleotide synthesis and proliferation. In addition, glutamine contributes to the cancer stem cell GD2 ganglioside-positive phenotype in the triple-negative breast cancer cell line MDA-MB-231. Thieno[2,3-b]pyridine derivative possesses higher cytotoxicity against MDA-MB-231 than against MCF-7 cells and induces a shift to aerobic metabolism and a decrease in S(6)nLc4Cer GSL-positive cancer stem cells in the MDA-MB-231 cell line. In this review, we discuss findings in MDA-MB-231, MCF-7, and other breast cancer cell lines concerning their differences in growth factor receptors and recent knowledge of the main biochemical pathways delivering distinct glycosphingolipid patterns during tumorigenesis and therapy.
Collapse
Affiliation(s)
- Anita Markotić
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, 21000 Split, Croatia
| | - Jasminka Omerović
- Department of Immunology, University of Split School of Medicine, 21000 Split, Croatia
| | - Sandra Marijan
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, 21000 Split, Croatia
| | - Nikolina Režić-Mužinić
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, 21000 Split, Croatia
| | - Vedrana Čikeš Čulić
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, 21000 Split, Croatia
| |
Collapse
|
4
|
Nguyen Van Long F, Le T, Caron P, Valcourt-Gendron D, Sergerie R, Laverdière I, Vanura K, Guillemette C. Targeting sphingolipid metabolism in chronic lymphocytic leukemia. Clin Exp Med 2024; 24:174. [PMID: 39078421 PMCID: PMC11289351 DOI: 10.1007/s10238-024-01440-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024]
Abstract
Elevated levels of circulating C16:0 glucosylceramides (GluCer) and increased mRNA expression of UDP-glucose ceramide glycosyltransferase (UGCG), the enzyme responsible for converting ceramides (Cer) to GluCer, represent unfavorable prognostic markers in chronic lymphocytic leukemia (CLL) patients. To evaluate the therapeutic potential of inhibiting GluCer synthesis, we genetically repressed the UGCG pathway using in vitro models of leukemic B cells, in addition to UGCG pharmacological inhibition with approved drugs such as eliglustat and ibiglustat, both individually and in combination with ibrutinib, assessed in cell models and primary CLL patient cells. Cell viability, apoptosis, and proliferation were evaluated in vitro, and survival and apoptosis were examined ex vivo. UGCG inhibition efficacy was confirmed by quantifying intracellular sphingolipid levels through targeted lipidomics using mass spectrometry. Other inhibitors of sphingolipid biosynthesis pathways were similarly assessed. Blocking UGCG significantly decreased cell viability and proliferation, highlighting the oncogenic role of UGCG in CLL. The efficient inhibition of UGCG was confirmed by a significant reduction in GluCer intracellular levels. The combination of UGCG inhibitors with ibrutinib demonstrated synergistic effect. Inhibitors that target alternative pathways within sphingolipid metabolism, like sphingosine kinases inhibitor SKI-II, also demonstrated promising therapeutic effects both alone and when used in combination with ibrutinib, reinforcing the oncogenic impact of sphingolipids in CLL cells. Targeting sphingolipid metabolism, especially the UGCG pathway, represents a promising therapeutic strategy and as a combination therapy for potential treatment of CLL patients, warranting further investigation.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Humans
- Sphingolipids/metabolism
- Cell Survival/drug effects
- Glucosyltransferases/antagonists & inhibitors
- Glucosyltransferases/metabolism
- Glucosyltransferases/genetics
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Piperidines/pharmacology
- Adenine/analogs & derivatives
- Adenine/pharmacology
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Glucosylceramides/metabolism
- Pyrazoles/pharmacology
- Pyrimidines/pharmacology
Collapse
Affiliation(s)
- Flora Nguyen Van Long
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Pharmacy and Université Laval Cancer Research Center, Université Laval, R4701.5, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada
| | - Trang Le
- Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Patrick Caron
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Pharmacy and Université Laval Cancer Research Center, Université Laval, R4701.5, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada
| | - Délya Valcourt-Gendron
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Pharmacy and Université Laval Cancer Research Center, Université Laval, R4701.5, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada
| | - Roxanne Sergerie
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Pharmacy and Université Laval Cancer Research Center, Université Laval, R4701.5, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada
| | - Isabelle Laverdière
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Pharmacy and Université Laval Cancer Research Center, Université Laval, R4701.5, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada
| | - Katrina Vanura
- Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| | - Chantal Guillemette
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Faculty of Pharmacy and Université Laval Cancer Research Center, Université Laval, R4701.5, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada.
- Canada Research Chair in Pharmacogenomics, Quebec, Canada.
| |
Collapse
|
5
|
Wenyao C, Shuai M, Yifeng F, Xinzhi L, Xiangyong Q. Combined use of niraparib enhanced the inhibitory effect of Anti-GD2 antibody on osteosarcoma cells. Discov Oncol 2024; 15:304. [PMID: 39048747 PMCID: PMC11269552 DOI: 10.1007/s12672-024-01166-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 07/16/2024] [Indexed: 07/27/2024] Open
Abstract
PURPOSE This study aims to investigate the effect of Niraparib in combination with an Anti-GD2 Antibody on osteosarcoma cells. METHODS Scratch test was utilized to assess cell migration capacity, while the Transwell experiment was utilized to evaluate cell invasion potential. Cell proliferation was measured using the CCK8 experiment. The affinity between the anti-GD2 antibody and its antigen was determined via ELISA. Tumor growth was evaluated through animal experiments. Western blotting, QRT-PCR, and histological analysis were conducted to examine the expression of relevant proteins and mRNAs. RESULTS MG63 cell line was used for an example. The scratch test showed that the migration rate of osteosarcoma cells in Niraparib + Anti-GD2 group was 1.07 ± 0.04 after 48 h, and 0.34 ± 0.04 in the Control group. Transwell experiment showed that the invasion ability of osteosarcoma cells in Niraparib + Anti-GD2 group was 21.0 ± 1.5, and that in Control group was 87.7 ± 2.9. CCK8 experiment showed that the absorbance value of Niraparib + Anti-GD2 group was 0.16 ± 0.10 on day 5, and that of the Control group was 0.76 ± 0.09. Western blotting showed that the expression levels of BALP and CICP in Niraparib + Anti-GD2 group were 0.751 ± 0.135 and 1.086 ± 0.115, respectively, and those in Control group were 1.025 ± 0.143 and 1.216 ± 0.168, respectively. QRT-PCR results showed that the absorbance values of Niraparib + Anti-GD2 group were 0.173 ± 0.065 and 0.170 ± 0.078 on day 14. The results of animal experiments showed that on day 5, the tumor volume of the Control group was 2433 ± 391, and that of the Niraparib + Anti-GD2 group was 1137 ± 148. Histological analysis showed that the mean density values of Niraparib + Anti-GD2 group were 0.19 ± 0.08 and 0.22 ± 0.07, and those of Control group were 0.26 ± 0.09 and 0.29 ± 0.10. CONCLUSION The combination of Niraparib and Anti-GD2 antibody significantly inhibits Osteosarcoma cells.
Collapse
Affiliation(s)
- Chen Wenyao
- Affiliated Renhe Hospital of China Three Gorges University, No. 410, Yiling Avenue, Yichang, 443001, China
| | - Ma Shuai
- Affiliated Renhe Hospital of China Three Gorges University, No. 410, Yiling Avenue, Yichang, 443001, China
| | - Fan Yifeng
- Affiliated Renhe Hospital of China Three Gorges University, No. 410, Yiling Avenue, Yichang, 443001, China
| | - Li Xinzhi
- Affiliated Renhe Hospital of China Three Gorges University, No. 410, Yiling Avenue, Yichang, 443001, China
| | - Que Xiangyong
- Affiliated Renhe Hospital of China Three Gorges University, No. 410, Yiling Avenue, Yichang, 443001, China.
| |
Collapse
|
6
|
Philippova J, Shevchenko J, Sennikov S. GD2-targeting therapy: a comparative analysis of approaches and promising directions. Front Immunol 2024; 15:1371345. [PMID: 38558810 PMCID: PMC10979305 DOI: 10.3389/fimmu.2024.1371345] [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: 01/16/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Disialoganglioside GD2 is a promising target for immunotherapy with expression primarily restricted to neuroectodermal and epithelial tumor cells. Although its role in the maintenance and repair of neural tissue is well-established, its functions during normal organism development remain understudied. Meanwhile, studies have shown that GD2 plays an important role in tumorigenesis. Its functions include proliferation, invasion, motility, and metastasis, and its high expression and ability to transform the tumor microenvironment may be associated with a malignant phenotype. Structurally, GD2 is a glycosphingolipid that is stably expressed on the surface of tumor cells, making it a suitable candidate for targeting by antibodies or chimeric antigen receptors. Based on mouse monoclonal antibodies, chimeric and humanized antibodies and their combinations with cytokines, toxins, drugs, radionuclides, nanoparticles as well as chimeric antigen receptor have been developed. Furthermore, vaccines and photoimmunotherapy are being used to treat GD2-positive tumors, and GD2 aptamers can be used for targeting. In the field of cell therapy, allogeneic immunocompetent cells are also being utilized to enhance GD2 therapy. Efforts are currently being made to optimize the chimeric antigen receptor by modifying its design or by transducing not only αβ T cells, but also γδ T cells, NK cells, NKT cells, and macrophages. In addition, immunotherapy can combine both diagnostic and therapeutic methods, allowing for early detection of disease and minimal residual disease. This review discusses each immunotherapy method and strategy, its advantages and disadvantages, and highlights future directions for GD2 therapy.
Collapse
Affiliation(s)
| | | | - Sergey Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| |
Collapse
|
7
|
Hengst JA, Nduwumwami AJ, Sharma A, Yun JK. Fanning the Flames of Endoplasmic Reticulum (ER) Stress: Can Sphingolipid Metabolism Be Targeted to Enhance ER Stress-Associated Immunogenic Cell Death in Cancer? Mol Pharmacol 2024; 105:155-165. [PMID: 38164594 PMCID: PMC10877730 DOI: 10.1124/molpharm.123.000786] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 01/03/2024] Open
Abstract
The three arms of the unfolded protein response (UPR) surveil the luminal environment of the endoplasmic reticulum (ER) and transmit information through the lipid bilayer to the cytoplasm to alert the cell of stress conditions within the ER lumen. That same lipid bilayer is the site of de novo synthesis of phospholipids and sphingolipids. Thus, it is no surprise that lipids are modulated by and are modulators of ER stress. Given that sphingolipids have both prosurvival and proapoptotic effects, they also exert opposing effects on life/death decisions in the face of prolonged ER stress detected by the UPR. In this review, we will focus on several recent studies that demonstrate how sphingolipids affect each arm of the UPR. We will also discuss the role of sphingolipids in the process of immunogenic cell death downstream of the protein kinase RNA-like endoplasmic reticulum kinase (PERK)/eukaryotic initiating factor 2α (eIF2α) arm of the UPR. Furthermore, we will discuss strategies to target the sphingolipid metabolic pathway that could potentially act synergistically with agents that induce ER stress as novel anticancer treatments. SIGNIFICANCE STATEMENT: This review provides the readers with a brief discussion of the sphingolipid metabolic pathway and the unfolded protein response. The primary focus of the review is the mechanism(s) by which sphingolipids modulate the endoplasmic reticulum (ER) stress response pathways and the critical role of sphingolipids in the process of immunogenic cell death associated with the ER stress response.
Collapse
Affiliation(s)
- Jeremy A Hengst
- Departments of Pediatrics (J.A.H.) and Pharmacology (A.S., J.K.Y.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Drug Metabolism and Pharmacokinetics, National Center for Advancing Translational Science, Rockville, Maryland (A.J.N.)
| | - Asvelt J Nduwumwami
- Departments of Pediatrics (J.A.H.) and Pharmacology (A.S., J.K.Y.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Drug Metabolism and Pharmacokinetics, National Center for Advancing Translational Science, Rockville, Maryland (A.J.N.)
| | - Arati Sharma
- Departments of Pediatrics (J.A.H.) and Pharmacology (A.S., J.K.Y.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Drug Metabolism and Pharmacokinetics, National Center for Advancing Translational Science, Rockville, Maryland (A.J.N.)
| | - Jong K Yun
- Departments of Pediatrics (J.A.H.) and Pharmacology (A.S., J.K.Y.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Drug Metabolism and Pharmacokinetics, National Center for Advancing Translational Science, Rockville, Maryland (A.J.N.)
| |
Collapse
|
8
|
Machy P, Mortier E, Birklé S. Biology of GD2 ganglioside: implications for cancer immunotherapy. Front Pharmacol 2023; 14:1249929. [PMID: 37670947 PMCID: PMC10475612 DOI: 10.3389/fphar.2023.1249929] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023] Open
Abstract
Part of the broader glycosphingolipid family, gangliosides are composed of a ceramide bound to a sialic acid-containing glycan chain, and locate at the plasma membrane. Gangliosides are produced through sequential steps of glycosylation and sialylation. This diversity of composition is reflected in differences in expression patterns and functions of the various gangliosides. Ganglioside GD2 designates different subspecies following a basic structure containing three carbohydrate residues and two sialic acids. GD2 expression, usually restrained to limited tissues, is frequently altered in various neuroectoderm-derived cancers. While GD2 is of evident interest, its glycolipid nature has rendered research challenging. Physiological GD2 expression has been linked to developmental processes. Passing this stage, varying levels of GD2, physiologically expressed mainly in the central nervous system, affect composition and formation of membrane microdomains involved in surface receptor signaling. Overexpressed in cancer, GD2 has been shown to enhance cell survival and invasion. Furthermore, binding of antibodies leads to immune-independent cell death mechanisms. In addition, GD2 contributes to T-cell dysfunction, and functions as an immune checkpoint. Given the cancer-associated functions, GD2 has been a source of interest for immunotherapy. As a potential biomarker, methods are being developed to quantify GD2 from patients' samples. In addition, various therapeutic strategies are tested. Based on initial success with antibodies, derivates such as bispecific antibodies and immunocytokines have been developed, engaging patient immune system. Cytotoxic effectors or payloads may be redirected based on anti-GD2 antibodies. Finally, vaccines can be used to mount an immune response in patients. We review here the pertinent biological information on GD2 which may be of use for optimizing current immunotherapeutic strategies.
Collapse
Affiliation(s)
| | | | - Stéphane Birklé
- Nantes Université, Univ Angers, INSERM, CNRS, CRCI2NA, Nantes, France
| |
Collapse
|
9
|
Jeremiasse B, Rijs Z, Angoelal KR, Hiemcke-Jiwa LS, de Boed EA, Kuppen PJK, Sier CFM, van Driel PBAA, van de Sande MAJ, Wijnen MHWA, Rios AC, van der Steeg AFW. Evaluation of Potential Targets for Fluorescence-Guided Surgery in Pediatric Ewing Sarcoma: A Preclinical Proof-of-Concept Study. Cancers (Basel) 2023; 15:3896. [PMID: 37568714 PMCID: PMC10417270 DOI: 10.3390/cancers15153896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Fluorescence-guided surgery (FGS), based on fluorescent tracers binding to tumor-specific biomarkers, could assist surgeons to achieve complete tumor resections. This study evaluated potential biomarkers for FGS in pediatric Ewing sarcoma (ES). Immunohistochemistry (IHC) was performed to assess CD99, CXCR4, CD117, NPY-R-Y1, and IGF-1R expression in ES biopsies and resection specimens. LINGO-1 and GD2 evaluation did not work on the acquired tissue. Based on the immunoreactive scores, anti-CD99 and anti-CD117 were evaluated for binding specificity using flow cytometry and immunofluorescence microscopy. Anti-GD2, a tracer in the developmental phase, was also tested. These three tracers were topically applied to a freshly resected ES tumor and adjacent healthy tissue. IHC demonstrated moderate/strong CD99 and CD117 expression in ES tumor samples, while adjacent healthy tissue had limited expression. Flow cytometry and immunofluorescence microscopy confirmed high CD99 expression, along with low/moderate CD117 and low GD2 expression, in ES cell lines. Topical anti-CD99 and anti-GD2 application on ES tumor showed fluorescence, while anti-CD117 did not show fluorescence for this patient. In conclusion, CD99-targeting tracers hold promise for FGS of ES. CD117 and GD2 tracers could be potential alternatives. The next step towards development of ES-specific FGS tracers could be ex vivo topical application experiments on a large cohort of ES patients.
Collapse
Affiliation(s)
- Bernadette Jeremiasse
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (B.J.); (K.R.A.); (M.A.J.v.d.S.); (M.H.W.A.W.); (A.F.W.v.d.S.)
| | - Zeger Rijs
- Department of Orthopedic Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Karieshma R. Angoelal
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (B.J.); (K.R.A.); (M.A.J.v.d.S.); (M.H.W.A.W.); (A.F.W.v.d.S.)
| | - Laura S. Hiemcke-Jiwa
- Department of Pathology, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (L.S.H.-J.); (E.A.d.B.)
- Department of Pathology, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Ella A. de Boed
- Department of Pathology, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (L.S.H.-J.); (E.A.d.B.)
| | - Peter J. K. Kuppen
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (P.J.K.K.); (C.F.M.S.)
| | - Cornelis F. M. Sier
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (P.J.K.K.); (C.F.M.S.)
| | | | - Michiel A. J. van de Sande
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (B.J.); (K.R.A.); (M.A.J.v.d.S.); (M.H.W.A.W.); (A.F.W.v.d.S.)
- Department of Orthopedic Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marc H. W. A. Wijnen
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (B.J.); (K.R.A.); (M.A.J.v.d.S.); (M.H.W.A.W.); (A.F.W.v.d.S.)
| | - Anne C. Rios
- Research Department, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands;
- Oncode Institute, Jaarbeursplein 6, 3521 AL Utrecht, The Netherlands
| | - Alida F. W. van der Steeg
- Department of Surgery, Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (B.J.); (K.R.A.); (M.A.J.v.d.S.); (M.H.W.A.W.); (A.F.W.v.d.S.)
| |
Collapse
|
10
|
El Malki K, Wehling P, Alt F, Sandhoff R, Zahnreich S, Ustjanzew A, Wilzius C, Brockmann MA, Wingerter A, Russo A, Beck O, Sommer C, Ottenhausen M, Frauenknecht KBM, Paret C, Faber J. Glucosylceramide Synthase Inhibitors Induce Ceramide Accumulation and Sensitize H3K27 Mutant Diffuse Midline Glioma to Irradiation. Int J Mol Sci 2023; 24:9905. [PMID: 37373053 DOI: 10.3390/ijms24129905] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
H3K27M mutant (mut) diffuse midline glioma (DMG) is a lethal cancer with no effective cure. The glycosphingolipids (GSL) metabolism is altered in these tumors and could be exploited to develop new therapies. We tested the effect of the glucosylceramide synthase inhibitors (GSI) miglustat and eliglustat on cell proliferation, alone or in combination with temozolomide or ionizing radiation. Miglustat was included in the therapy protocol of two pediatric patients. The effect of H3.3K27 trimethylation on GSL composition was analyzed in ependymoma. GSI reduced the expression of the ganglioside GD2 in a concentration and time-dependent manner and increased the expression of ceramide, ceramide 1-phosphate, sphingosine, and sphingomyelin but not of sphingosine 1-phosphate. Miglustat significantly increased the efficacy of irradiation. Treatment with miglustat according to dose recommendations for patients with Niemann-Pick disease was well tolerated with manageable toxicities. One patient showed a mixed response. In ependymoma, a high concentration of GD2 was found only in the presence of the loss of H3.3K27 trimethylation. In conclusion, treatment with miglustat and, in general, targeting GSL metabolism may offer a new therapeutic opportunity and can be administered in close proximity to radiation therapy. Alterations in H3K27 could be useful to identify patients with a deregulated GSL metabolism.
Collapse
Affiliation(s)
- Khalifa El Malki
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Pia Wehling
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Francesca Alt
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Roger Sandhoff
- Lipid Pathobiochemistry, German Cancer Research Center, 69120 Heidelberg, Germany
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
| | - Sebastian Zahnreich
- Department of Radiation Oncology and Radiation Therapy, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Arsenij Ustjanzew
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Carolin Wilzius
- Lipid Pathobiochemistry, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Marc A Brockmann
- Department of Neuroradiology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Arthur Wingerter
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Alexandra Russo
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- German Cancer Consortium (DKTK), Site Frankfurt/Mainz, Germany, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Olaf Beck
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Clemens Sommer
- Institute of Neuropathology, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Malte Ottenhausen
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Katrin B M Frauenknecht
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
- Institute of Neuropathology, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- National Center of Pathology (NCP), Laboratoire National de Santé, 3555 Dudelange, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Laboratoire National de Santé, 3555 Dudelange, Luxembourg
| | - Claudia Paret
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
- German Cancer Consortium (DKTK), Site Frankfurt/Mainz, Germany, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Research Center of Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Jörg Faber
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
- German Cancer Consortium (DKTK), Site Frankfurt/Mainz, Germany, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
11
|
Cao S, Hu X, Ren S, Wang Y, Shao Y, Wu K, Yang Z, Yang W, He G, Li X. The biological role and immunotherapy of gangliosides and GD3 synthase in cancers. Front Cell Dev Biol 2023; 11:1076862. [PMID: 36824365 PMCID: PMC9941352 DOI: 10.3389/fcell.2023.1076862] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Gangliosides are a large subfamily of glycosphingolipids that broadly exist in the nervous system and interact with signaling molecules in the lipid rafts. GD3 and GD2 are two types of disialogangliosides (GDs) that include two sialic acid residues. The expression of GD3 and GD2 in various cancers is mostly upregulated and is involved in tumor proliferation, invasion, metastasis, and immune responses. GD3 synthase (GD3S, ST8SiaI), a subclass of sialyltransferases, regulates the biosynthesis of GD3 and GD2. GD3S is also upregulated in most tumors and plays an important role in the development and progression of tumors. Many clinical trials targeting GD2 are ongoing and various immunotherapy studies targeting gangliosides and GD3S are gradually attracting much interest and attention. This review summarizes the function, molecular mechanisms, and ongoing clinical applications of GD3, GD2, and GD3S in abundant types of tumors, which aims to provide novel targets for future cancer therapy.
Collapse
Affiliation(s)
- Shangqi Cao
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xu Hu
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Shangqing Ren
- 2Robotic Minimally Invasive Surgery Center, Sichuan Academy of Medical Sciences and Sichuan Provincial Peoples Hospital, Chengdu, China
| | - Yaohui Wang
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yanxiang Shao
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Kan Wu
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Zhen Yang
- 3Department of Urology, Chengdu Second People’s Hospital, Chengdu, China
| | - Weixiao Yang
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Gu He
- 4State Key Laboratory of Biotherapy and Department of Pharmacy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China,*Correspondence: Gu He, ; Xiang Li,
| | - Xiang Li
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China,*Correspondence: Gu He, ; Xiang Li,
| |
Collapse
|
12
|
Abdul Rashid K, Ibrahim K, Wong JHD, Mohd Ramli N. Lipid Alterations in Glioma: A Systematic Review. Metabolites 2022; 12:metabo12121280. [PMID: 36557318 PMCID: PMC9783089 DOI: 10.3390/metabo12121280] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/08/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Gliomas are highly lethal tumours characterised by heterogeneous molecular features, producing various metabolic phenotypes leading to therapeutic resistance. Lipid metabolism reprogramming is predominant and has contributed to the metabolic plasticity in glioma. This systematic review aims to discover lipids alteration and their biological roles in glioma and the identification of potential lipids biomarker. This systematic review was conducted using the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. Extensive research articles search for the last 10 years, from 2011 to 2021, were conducted using four electronic databases, including PubMed, Web of Science, CINAHL and ScienceDirect. A total of 158 research articles were included in this study. All studies reported significant lipid alteration between glioma and control groups, impacting glioma cell growth, proliferation, drug resistance, patients' survival and metastasis. Different lipids demonstrated different biological roles, either beneficial or detrimental effects on glioma. Notably, prostaglandin (PGE2), triacylglycerol (TG), phosphatidylcholine (PC), and sphingosine-1-phosphate play significant roles in glioma development. Conversely, the most prominent anti-carcinogenic lipids include docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and vitamin D3 have been reported to have detrimental effects on glioma cells. Furthermore, high lipid signals were detected at 0.9 and 1.3 ppm in high-grade glioma relative to low-grade glioma. This evidence shows that lipid metabolisms were significantly dysregulated in glioma. Concurrent with this knowledge, the discovery of specific lipid classes altered in glioma will accelerate the development of potential lipid biomarkers and enhance future glioma therapeutics.
Collapse
Affiliation(s)
- Khairunnisa Abdul Rashid
- Department of Biomedical Imaging, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Kamariah Ibrahim
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Jeannie Hsiu Ding Wong
- Department of Biomedical Imaging, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Norlisah Mohd Ramli
- Department of Biomedical Imaging, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Correspondence: ; Tel.: +60-379673238
| |
Collapse
|
13
|
Paret C, Ustjanzew A, Ersali S, Seidmann L, Jennemann R, Ziegler N, Malki KE, Russo A, Wingerter A, Ortmüller F, Bornas A, Wehling PC, Lepădatu A, Ottenhausen M, Roth W, Sommer C, Fliss B, Frauenknecht KBM, Sandhoff R, Faber J. GD2 Expression in Medulloblastoma and Neuroblastoma for Personalized Immunotherapy: A Matter of Subtype. Cancers (Basel) 2022; 14:cancers14246051. [PMID: 36551537 PMCID: PMC9775636 DOI: 10.3390/cancers14246051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Neuroblastoma (NBL) and medulloblastoma (MB) are aggressive pediatric cancers which can benefit from therapies targeting gangliosides. Therefore, we compared the ganglioside profile of 9 MB and 14 NBL samples by thin layer chromatography and mass spectrometry. NBL had the highest expression of GD2 (median 0.54 nmol GD2/mg protein), and also expressed complex gangliosides. GD2-low samples expressed GD1a and were more differentiated. MB mainly expressed GD2 (median 0.032 nmol GD2/mg protein) or GM3. Four sonic hedgehog-activated (SHH) as well as one group 4 and one group 3 MBs were GD2-positive. Two group 3 MB samples were GD2-negative but GM3-positive. N-glycolyl neuraminic acid-containing GM3 was neither detected in NBL nor MB by mass spectrometry. Furthermore, a GD2-phenotype predicting two-gene signature (ST8SIA1 and B4GALNT1) was applied to RNA-Seq datasets, including 86 MBs and validated by qRT-PCR. The signature values were decreased in group 3 and wingless-activated (WNT) compared to SHH and group 4 MBs. These results suggest that while NBL is GD2-positive, only some MB patients can benefit from a GD2-directed therapy. The expression of genes involved in the ganglioside synthesis may allow the identification of GD2-positive MBs. Finally, the ganglioside profile may reflect the differentiation status in NBL and could help to define MB subtypes.
Collapse
Affiliation(s)
- Claudia Paret
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Correspondence:
| | - Arsenij Ustjanzew
- University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Sara Ersali
- Lipid Pathobiochemistry, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Larissa Seidmann
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
- Institute of Pathology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Richard Jennemann
- Lipid Pathobiochemistry, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Nicole Ziegler
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Khalifa El Malki
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Alexandra Russo
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Arthur Wingerter
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Franziska Ortmüller
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
| | - Angelina Bornas
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Pia Charlotte Wehling
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Adina Lepădatu
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Malte Ottenhausen
- Department of Neurosurgery, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Wilfried Roth
- University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Institute of Pathology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Clemens Sommer
- University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Barbara Fliss
- Institute of Forensic Medicine, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Katrin B. M. Frauenknecht
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- National Center of Pathology (NCP), Laboratoire National de Santé, 3555 Dudelange, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Laboratoire National de Santé, 3555 Dudelange, Luxembourg
| | - Roger Sandhoff
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
- Lipid Pathobiochemistry, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Jörg Faber
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| |
Collapse
|
14
|
Spasov NJ, Dombrowski F, Lode HN, Spasova M, Ivanova L, Mumdjiev I, Burnusuzov H, Siebert N. First-line Anti-GD2 Therapy Combined With Consolidation Chemotherapy in 3 Patients With Newly Diagnosed Metastatic Ewing Sarcoma or Ewing-like Sarcoma. J Pediatr Hematol Oncol 2022; 44:e948-e953. [PMID: 35622995 PMCID: PMC9323561 DOI: 10.1097/mph.0000000000002488] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 03/31/2022] [Indexed: 12/04/2022]
Abstract
Despite multimodal therapy, the prognosis of patients with metastatic Ewing sarcoma (ES) remains poor, with new treatments urgently needed. The disialoganglioside GD2, a well-established tumor-associated antigen, is expressed in 40% to 90% of ES cells, making it a suitable therapeutic target. Here we report 3 cases with newly diagnosed, metastatic, GD2-positive ES or Ewing-like sarcoma treated with the anti-GD2 antibody dinutuximab beta in addition to standard chemotherapeutic regimens. Treatment was well-tolerated, and all patients achieved complete remission, without evidence of relapse. First-line anti-GD2 immunotherapy in patients with metastatic, GD2-positive ES or Ewing-like sarcoma represents a promising therapeutic option that warrants further clinical evaluation.
Collapse
Affiliation(s)
- Neofit J. Spasov
- Division of Pediatric Oncohematology, Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, Plovdiv, Bulgaria
| | | | - Holger N. Lode
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| | - Mariya Spasova
- Division of Pediatric Oncohematology, Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Liliya Ivanova
- Division of Pediatric Oncohematology, Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Ivan Mumdjiev
- Division of Pediatric Oncohematology, Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Hassan Burnusuzov
- Division of Pediatric Oncohematology, Department of Pediatrics and Medical Genetics, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Nikolai Siebert
- Department of Pediatric Hematology and Oncology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
15
|
Ingley KM, Maleddu A, Grange FL, Gerrand C, Bleyer A, Yasmin E, Whelan J, Strauss SJ. Current approaches to management of bone sarcoma in adolescent and young adult patients. Pediatr Blood Cancer 2022; 69:e29442. [PMID: 34767314 DOI: 10.1002/pbc.29442] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/02/2021] [Accepted: 10/16/2021] [Indexed: 01/05/2023]
Abstract
Bone tumors are a group of histologically diverse diseases that occur across all ages. Two of the commonest, osteosarcoma (OS) and Ewing sarcoma (ES), are regarded as characteristic adolescent and young adult (AYA) cancers with an incidence peak in AYAs. They are curable for some but associated with unacceptably high rates of treatment failure and morbidity. The introduction of effective new therapeutics for bone sarcomas is slow, and to date, complex biology has been insufficiently characterized to allow more rapid therapeutic exploitation. This review focuses on current standards of care, recent advances that have or may soon change that standard of care and challenges to the expert clinical research community that we suggest must be met.
Collapse
Affiliation(s)
- Katrina M Ingley
- London Sarcoma Service, University College London Hospitals NHS Trust, London, UK
| | - Alessandra Maleddu
- London Sarcoma Service, University College London Hospitals NHS Trust, London, UK
| | - Franel Le Grange
- London Sarcoma Service, University College London Hospitals NHS Trust, London, UK
| | - Craig Gerrand
- London Sarcoma Service, Department of Orthopaedic Oncology, Royal National Orthopaedic Hospital NHS Trust, Stanmore, UK
| | - Archie Bleyer
- Oregon Health and Science University, Portland, Oregon
| | - Ephia Yasmin
- Reproductive Medicine Unit, University College London Hospitals NHS Trust, London, UK
| | - Jeremy Whelan
- London Sarcoma Service, University College London Hospitals NHS Trust, London, UK
| | - Sandra J Strauss
- London Sarcoma Service, University College London Hospitals NHS Trust, London, UK.,UCL Cancer Institute, London, UK
| |
Collapse
|
16
|
Single-Cell Proteomic Analysis Dissects the Complexity of Tumor Microenvironment in Muscle Invasive Bladder Cancer. Cancers (Basel) 2021; 13:cancers13215440. [PMID: 34771607 PMCID: PMC8582554 DOI: 10.3390/cancers13215440] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary The tumor microenvironment (TME) is considered to play a key role in the development of many types of tumors. Muscle invasive bladder cancer (MIBC), which is well known for its heterogeneity, has a highly complex TME. Herein, we integrated mass cytometry and imaging mass cytometry to systematically investigate the complexity of the MIBC TME. Our investigation revealed tumor and immune cells with diverse phenotypes. We identified a specific cancer stem-like cell cluster (ALDH+PD-L1+ER-β−), which is associated with poor prognosis and highlighted the importance of the spatial distribution patterns of MIBC TME components. The present study comprehensively elucidated the complexity of the MIBC TME and provides potentially valuable information for future research. Abstract Muscle invasive bladder cancer (MIBC) is a malignancy with considerable heterogeneity. The MIBC tumor microenvironment (TME) is highly complex, comprising diverse phenotypes and spatial architectures. The complexity of the MIBC TME must be characterized to provide potential targets for precision therapy. Herein, an integrated combination of mass cytometry and imaging mass cytometry was used to analyze tumor cells, immune cells, and TME spatial characteristics of 44 MIBC patients. We detected tumor and immune cell clusters with abnormal phenotypes. In particular, we identified a previously overlooked cancer stem-like cell cluster (ALDH+PD-L1+ER-β−) that was strongly associated with poor prognosis. We elucidated the different spatial architectures of immune cells (excluded, infiltrated, and deserted) and tumor-associated collagens (curved, stretched, directionally distributed, and chaotic) in the MIBC TME. The present study is the first to provide in-depth insight into the complexity of the MIBC TME at the single-cell level. Our results will improve the general understanding of the heterogeneous characteristics of MIBC, potentially facilitating patient stratification and personalized therapy.
Collapse
|
17
|
Jennemann R, Volz M, Bestvater F, Schmidt C, Richter K, Kaden S, Müthing J, Gröne HJ, Sandhoff R. Blockade of Glycosphingolipid Synthesis Inhibits Cell Cycle and Spheroid Growth of Colon Cancer Cells In Vitro and Experimental Colon Cancer Incidence In Vivo. Int J Mol Sci 2021; 22:ijms221910539. [PMID: 34638879 PMCID: PMC8508865 DOI: 10.3390/ijms221910539] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most frequently diagnosed cancers in humans. At early stages CRC is treated by surgery and at advanced stages combined with chemotherapy. We examined here the potential effect of glucosylceramide synthase (GCS)-inhibition on CRC biology. GCS is the rate-limiting enzyme in the glycosphingolipid (GSL)-biosynthesis pathway and overexpressed in many human tumors. We suppressed GSL-biosynthesis using the GCS inhibitor Genz-123346 (Genz), NB-DNJ (Miglustat) or by genetic targeting of the GCS-encoding gene UDP-glucose-ceramide-glucosyltransferase- (UGCG). GCS-inhibition or GSL-depletion led to a marked arrest of the cell cycle in Lovo cells. UGCG silencing strongly also inhibited tumor spheroid growth in Lovo cells and moderately in HCT116 cells. MS/MS analysis demonstrated markedly elevated levels of sphingomyelin (SM) and phosphatidylcholine (PC) that occurred in a Genz-concentration dependent manner. Ultrastructural analysis of Genz-treated cells indicated multi-lamellar lipid storage in vesicular compartments. In mice, Genz lowered the incidence of experimentally induced colorectal tumors and in particular the growth of colorectal adenomas. These results highlight the potential for GCS-based inhibition in the treatment of CRC.
Collapse
Affiliation(s)
- Richard Jennemann
- Lipid Pathobiochemistry Group, German Cancer Research Center, 69120 Heidelberg, Germany; (M.V.); (R.S.)
- Correspondence:
| | - Martina Volz
- Lipid Pathobiochemistry Group, German Cancer Research Center, 69120 Heidelberg, Germany; (M.V.); (R.S.)
| | - Felix Bestvater
- Light Microscopy Facility, German Cancer Research Center, 69120 Heidelberg, Germany; (F.B.); (C.S.)
| | - Claudia Schmidt
- Light Microscopy Facility, German Cancer Research Center, 69120 Heidelberg, Germany; (F.B.); (C.S.)
| | - Karsten Richter
- Core Facility Electron Microscopy, German Cancer Research Center, 69120 Heidelberg, Germany; (K.R.); (S.K.)
| | - Sylvia Kaden
- Core Facility Electron Microscopy, German Cancer Research Center, 69120 Heidelberg, Germany; (K.R.); (S.K.)
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, 48149 Münster, Germany;
| | - Hermann-Josef Gröne
- Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany;
- Institute of Pharmacology, University of Marburg, 35043 Marburg, Germany
| | - Roger Sandhoff
- Lipid Pathobiochemistry Group, German Cancer Research Center, 69120 Heidelberg, Germany; (M.V.); (R.S.)
| |
Collapse
|
18
|
Tang F, Tie Y, Wei YQ, Tu CQ, Wei XW. Targeted and immuno-based therapies in sarcoma: mechanisms and advances in clinical trials. Biochim Biophys Acta Rev Cancer 2021; 1876:188606. [PMID: 34371128 DOI: 10.1016/j.bbcan.2021.188606] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/04/2021] [Accepted: 08/02/2021] [Indexed: 02/08/2023]
Abstract
Sarcomas represent a distinct group of rare malignant tumors with high heterogeneity. Limited options with clinical efficacy for the metastatic or local advanced sarcoma existed despite standard therapy. Recently, targeted therapy according to the molecular and genetic phenotype of individual sarcoma is a promising option. Among these drugs, anti-angiogenesis therapy achieved favorable efficacy in sarcomas. Inhibitors targeting cyclin-dependent kinase 4/6, poly-ADP-ribose polymerase, insulin-like growth factor-1 receptor, mTOR, NTRK, metabolisms, and epigenetic drugs are under clinical evaluation for sarcomas bearing the corresponding signals. Immunotherapy represents a promising and favorable method in advanced solid tumors. However, most sarcomas are immune "cold" tumors, with only alveolar soft part sarcoma and undifferentiated pleomorphic sarcoma respond to immune checkpoint inhibitors. Cellular therapies with TCR-engineered T cells, chimeric antigen receptor T cells, tumor infiltrating lymphocytes, and nature killer cells transfer show therapeutic potential. Identifying tumor-specific antigens and exploring immune modulation factors arguing the efficacy of these immunotherapies are the current challenges. This review focuses on the mechanisms, advances, and potential strategies of targeted and immune-based therapies in sarcomas.
Collapse
Affiliation(s)
- Fan Tang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China; Department of Orthopeadics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Tie
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yu-Quan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Chong-Qi Tu
- Department of Orthopeadics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Xia-Wei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|