1
|
Wang H, Zhang W, Sun Y, Xu X, Chen X, Zhao K, Yang Z, Liu H. Nanotherapeutic strategies exploiting biological traits of cancer stem cells. Bioact Mater 2025; 50:61-94. [PMID: 40242505 PMCID: PMC12002948 DOI: 10.1016/j.bioactmat.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) represent a distinct subpopulation of cancer cells that orchestrate cancer initiation, progression, metastasis, and therapeutic resistance. Despite advances in conventional therapies, the persistence of CSCs remains a major obstacle to achieving cancer eradication. Nanomedicine-based approaches have emerged for precise CSC targeting and elimination, offering unique advantages in overcoming the limitations of traditional treatments. This review systematically analyzes recent developments in nanomedicine for CSC-targeted therapy, emphasizing innovative nanomaterial designs addressing CSC-specific challenges. We first provide a detailed examination of CSC biology, focusing on their surface markers, signaling networks, microenvironmental interactions, and metabolic signatures. On this basis, we critically evaluate cutting-edge nanomaterial engineering designed to exploit these CSC traits, including stimuli-responsive nanodrugs, nanocarriers for drug delivery, and multifunctional nanoplatforms capable of generating localized hyperthermia or reactive oxygen species. These sophisticated nanotherapeutic approaches enhance selectivity and efficacy in CSC elimination, potentially circumventing drug resistance and cancer recurrence. Finally, we present an in-depth analysis of current challenges in translating nanomedicine-based CSC-targeted therapies from bench to bedside, offering critical insights into future research directions and clinical implementation. This review aims to provide a comprehensive framework for understanding the intersection of nanomedicine and CSC biology, contributing to more effective cancer treatment modalities.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Wenjing Zhang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Yun Sun
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xican Xu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xiaoyang Chen
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Kexu Zhao
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Zhao Yang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Huiyu Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| |
Collapse
|
2
|
Hemavathy OR, Marimuthu Ramaswamy M, Mohana Priya CD, Bhardwaj S. Role of Liquid Biopsy in Oral Premalignant and Malignant Lesions: Correlation with CD24 and CD44 Expression in Early Diagnosis of Oral Cancer. J Maxillofac Oral Surg 2025; 24:207-212. [PMID: 39902448 PMCID: PMC11787126 DOI: 10.1007/s12663-024-02398-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/21/2024] [Indexed: 02/05/2025] Open
Abstract
Aim This study examines the levels of CD24 and CD44 expression in premalignant oral lesions (PMOL) and locally advanced oral squamous cell carcinoma (OSCC) in order to improve the early detection of the disease. We utilized RT-PCR techniques to examine the expression of two cell membrane proteins, CD24 and CD44, in blood and tissue samples. Methods A quantitative real-time polymerase chain reaction was performed to evaluate the expression patterns of CD24 and CD44 in blood and tissue samples obtained from individuals with PMOL (n = 20), OSCC (n = 20), and control subjects (n = 10). An independent t-test was utilized, revealing that CD24 demonstrated statistical significance with a p-value < 0.05. Fold change analysis had been performed to quantify the differences in expression between the two groups. Results CD24 was found to be increased in both tissue and blood groups. CD44 expression was higher in the tissue group in contrast to the blood group. The results for CD24 were statistically significant, with a p-value < 0.05. The findings affirm the use of CD24 as a dependable biomarker for early detection and diagnosis of OSCC in both tissue and blood specimens. Conclusion In conjunction with histological characteristics and clinicopathological factors, CD24 serves as a dependable biomarker for the early diagnosis of OSCC.
Collapse
Affiliation(s)
- O. R. Hemavathy
- Department of Oral and Maxillofacial Surgery, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospital, Chennai, India
| | - Muthusekhar Marimuthu Ramaswamy
- Department of Oral and Maxillofacial Surgery, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospital, Chennai, India
| | - C. D. Mohana Priya
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Shrishty Bhardwaj
- Department of Oral and Maxillofacial Surgery, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospital, Chennai, India
| |
Collapse
|
3
|
Du R, Zhang J, Lukas RV, Tripathi S, Ahrendsen JT, Curran MA, Dmello C, Zhang P, Stupp R, Rao G, Heimberger AB. Is modulation of immune checkpoints on glioblastoma-infiltrating myeloid cells a viable therapeutic strategy? Neuro Oncol 2025; 27:33-49. [PMID: 39427326 PMCID: PMC11726257 DOI: 10.1093/neuonc/noae193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024] Open
Abstract
The field of immunology has traditionally focused on immune checkpoint modulation of adaptive immune cells. However, many malignancies such as glioblastoma are mostly devoid of T cells and rather are enriched with immunosuppressive myeloid cells of the innate immune system. While some immune checkpoint targets are shared between adaptive and innate immunity, myeloid-specific checkpoints could also serve as potential therapeutics. To better understand the impact of immune checkpoint blockade on myeloid cells, we systematically summarize the current literature focusing on the direct immunological effects of PD-L1/PD-1, CD24/Siglec-10, collagen/LAIR-1, CX3CL1/CX3CR1, and CXCL10/CXCR3. By synthesizing the molecular mechanisms and the translational implications, we aim to prioritize agents in this category of therapeutics for glioblastoma.
Collapse
Affiliation(s)
- Ruochen Du
- Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jianzhong Zhang
- Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Rimas V Lukas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Shashwat Tripathi
- Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jared T Ahrendsen
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA (J.T.A.)
- Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Michael A Curran
- Department of Immunology, MD Anderson Cancer Center, the University of Texas, Houston, Texas, USA
| | - Crismita Dmello
- Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Peng Zhang
- Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Roger Stupp
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ganesh Rao
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
| | - Amy B Heimberger
- Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
4
|
Hazra R, Chattopadhyay S, Mallick A, Gayen S, Roy S. Unravelling CD24-Siglec-10 pathway: Cancer immunotherapy from basic science to clinical studies. Immunology 2024; 173:442-469. [PMID: 39129256 DOI: 10.1111/imm.13847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/27/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer immunotherapy has revolutionized the treatment landscape by harnessing the power of the immune system to combat malignancies. Two of the most promising players in this field are cluster of differentiation 24 (CD24) and sialic acid-binding Ig-like lectin 10 (Siglec-10), and both of them play pivotal roles in modulating immune responses. CD24, a cell surface glycoprotein, emerges as a convincing fundamental signal transducer for therapeutic intervention, given its significant implication in the processes related to tumour progression and immunogenic evasion. Additionally, the immunomodulatory functions of Siglec-10, a prominent member within the Siglec family of immune receptors, have recently become a crucial point of interest, particularly in the context of the tumour microenvironment. Hence, the intricate interplay of both CD24 and Siglec-10 assumes a critical role in fostering tumour growth, facilitating metastasis and also orchestrating immune evasion. Recent studies have found multiple evidences supporting the therapeutic potential of targeting CD24 in cancer treatment. Siglec-10, on the other hand, exhibits immunosuppressive properties that contribute to immune tolerance within the tumour microenvironment. Therefore, we delve into the complex mechanisms through which Siglec-10 modulates immune responses and facilitates immune escape in cancer. Siglec-10 also acts as a viable target for cancer immunotherapy and presents novel avenues for the development of therapeutic interventions. Furthermore, we examine the synergy between CD24 and Siglec-10 in shaping the immunosuppressive tumour microenvironment and discuss the implications for combination therapies. Therefore, understanding the roles of CD24 and Siglec-10 in cancer immunotherapy opens exciting possibilities for the development of novel therapeutics.
Collapse
Affiliation(s)
- Rudradeep Hazra
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Soumyadeep Chattopadhyay
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Arijit Mallick
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Sakuntala Gayen
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, India
| |
Collapse
|
5
|
Wang J, Shi J, Xiang Y, Wang ZW, Qi FF, Li ZY, Zhao LL, Zhu GH, Duan YY, Yang ZY, Li JP, Liao XH. LINC00525 enhances ZNF460-regulated CD24 expression through the sponge miR-125a-5p to promote malignant progression of breast cancer. J Cancer Res Clin Oncol 2024; 150:317. [PMID: 38914670 PMCID: PMC11196364 DOI: 10.1007/s00432-024-05830-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 06/02/2024] [Indexed: 06/26/2024]
Abstract
INTRODUCTION CD24 is a highly glycosylated glycosylphosphatidylinositol anchored membrane protein that plays an important role in tumor progression. The aim of this study was to investigate the effect of abnormal expression of CD24 on the proliferation, migration and invasion of breast cancer (BC) cells, and the molecular mechanism of regulating CD24 expression in breast cancer. METHODOLOGY The bioinformatics method was used to predict the expression level of CD24 in BC and its relationship with the occurrence and development of BC. IHC, RT-qPCR and WB were used to detect the expression of CD24 in BC tissues and cells. The proliferation of CD24 was evaluated by CCK-8 and colony formation assay, and the migration and invasion of CD24 were evaluated by wound healing and transwell. In addition, the effect of CD24 on the malignancy of BC in vivo was further evaluated by subcutaneous tumorigenesis assay. Molecular mechanisms were measured by luciferase reporter assays, biotin-labeled miRNA pull-down assay, RIP, and western blotting. RESULTS The results show that CD24 is highly expressed in breast cancer tissues and cell lines, and knockdown of CD24 in vivo and in vitro can inhibit the proliferation, migration and invasion of BC cells. Mechanistically, the transcription factor ZNF460 promotes its expression by binding to the CD24 promoter, and the expression of ZNF460 is regulated by miR-125a-5p, which inhibits its expression by targeting the 3'UTR of ZNF460. In addition, LINC00525 acts as a ceRNA sponge to adsorb miR-125a-5p and regulate its expression. CONCLUSIONS Overexpression of CD24 is involved in the development and poor prognosis of BC, which can be used as a potential target for the treatment of BC and provide a theoretical basis for the treatment of BC.
Collapse
Affiliation(s)
- Jun Wang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, People's Republic of China
| | - Ji Shi
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, People's Republic of China
| | - Yuan Xiang
- Department of Medical Laboratory, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Wen Wang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, People's Republic of China
- Key Laboratory of Chronic Noncommunicable Diseases, Yueyang Vocational Technical College, Yueyang, China
| | - Fei-Fei Qi
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, People's Republic of China
| | - Zi-Yi Li
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, People's Republic of China
| | - Li-Li Zhao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, People's Republic of China
| | - Guan-Hua Zhu
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, People's Republic of China
| | - Yuan-Yuan Duan
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, People's Republic of China
| | - Zhong-Yi Yang
- Yueyang Engineering Technology Research Center of Breast Disease Diagnosis and Treatment, Yueyang People's Hospital, Yueyang Hospital, Affiliated to Hunan Normal University, Yueyang, China
| | - Jia-Peng Li
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, People's Republic of China
| | - Xing-Hua Liao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
6
|
Razavipour SF, Yoon H, Jang K, Kim M, Nawara HM, Bagheri A, Huang WC, Shin M, Zhao D, Zhou Z, Van Boven D, Briegel K, Morey L, Ince TA, Johnson M, Slingerland JM. C-terminally phosphorylated p27 activates self-renewal driver genes to program cancer stem cell expansion, mammary hyperplasia and cancer. Nat Commun 2024; 15:5152. [PMID: 38886396 PMCID: PMC11183067 DOI: 10.1038/s41467-024-48742-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
In many cancers, a stem-like cell subpopulation mediates tumor initiation, dissemination and drug resistance. Here, we report that cancer stem cell (CSC) abundance is transcriptionally regulated by C-terminally phosphorylated p27 (p27pT157pT198). Mechanistically, this arises through p27 co-recruitment with STAT3/CBP to gene regulators of CSC self-renewal including MYC, the Notch ligand JAG1, and ANGPTL4. p27pTpT/STAT3 also recruits a SIN3A/HDAC1 complex to co-repress the Pyk2 inhibitor, PTPN12. Pyk2, in turn, activates STAT3, creating a feed-forward loop increasing stem-like properties in vitro and tumor-initiating stem cells in vivo. The p27-activated gene profile is over-represented in STAT3 activated human breast cancers. Furthermore, mammary transgenic expression of phosphomimetic, cyclin-CDK-binding defective p27 (p27CK-DD) increases mammary duct branching morphogenesis, yielding hyperplasia and microinvasive cancers that can metastasize to liver, further supporting a role for p27pTpT in CSC expansion. Thus, p27pTpT interacts with STAT3, driving transcriptional programs governing stem cell expansion or maintenance in normal and cancer tissues.
Collapse
Affiliation(s)
- Seyedeh Fatemeh Razavipour
- Cancer Host Interactions Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC, USA
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
| | - Hyunho Yoon
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon-si, South Korea
| | - Kibeom Jang
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
| | - Minsoon Kim
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
| | - Hend M Nawara
- Cancer Host Interactions Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC, USA
| | - Amir Bagheri
- Cancer Host Interactions Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC, USA
| | - Wei-Chi Huang
- Cancer Host Interactions Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC, USA
| | - Miyoung Shin
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Dekuang Zhao
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
| | - Zhiqun Zhou
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
| | - Derek Van Boven
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Karoline Briegel
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, Fl, USA
| | - Lluis Morey
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Tan A Ince
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Michael Johnson
- Cancer Host Interactions Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC, USA
| | - Joyce M Slingerland
- Cancer Host Interactions Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC, USA.
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA.
| |
Collapse
|
7
|
Zhao K, Wu C, Li X, Niu M, Wu D, Cui X, Zhao H. From mechanism to therapy: the journey of CD24 in cancer. Front Immunol 2024; 15:1401528. [PMID: 38881902 PMCID: PMC11176514 DOI: 10.3389/fimmu.2024.1401528] [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: 03/15/2024] [Accepted: 04/25/2024] [Indexed: 06/18/2024] Open
Abstract
CD24 is a glycosylphosphatidylinositol-anchored protein that is expressed in a wide range of tissues and cell types. It is involved in a variety of physiological and pathological processes, including cell adhesion, migration, differentiation, and apoptosis. Additionally, CD24 has been studied extensively in the context of cancer, where it has been found to play a role in tumor growth, invasion, and metastasis. In recent years, there has been growing interest in CD24 as a potential therapeutic target for cancer treatment. This review summarizes the current knowledge of CD24, including its structure, function, and its role in cancer. Finally, we provide insights into potential clinical application of CD24 and discuss possible approaches for the development of targeted cancer therapies.
Collapse
Affiliation(s)
- Kai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Caifeng Wu
- Department of Hand and Foot, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangjun Li
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mengchao Niu
- Department of Operation Room, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dan Wu
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaofeng Cui
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
8
|
Huang S, Zhang X, Wei Y, Xiao Y. Checkpoint CD24 function on tumor and immunotherapy. Front Immunol 2024; 15:1367959. [PMID: 38487533 PMCID: PMC10937401 DOI: 10.3389/fimmu.2024.1367959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/12/2024] [Indexed: 03/17/2024] Open
Abstract
CD24 is a protein found on the surface of cells that plays a crucial role in the proliferation, invasion, and spread of cancer cells. It adheres to cell membranes through glycosylphosphatidylinositol (GPI) and is associated with the prognosis and survival rate of cancer patients. CD24 interacts with the inhibitory receptor Siglec-10 that is present on immune cells like natural killer cells and macrophages, leading to the inhibition of natural killer cell cytotoxicity and macrophage-mediated phagocytosis. This interaction helps tumor cells escape immune detection and attack. Although the use of CD24 as a immune checkpoint receptor target for cancer immunotherapy is still in its early stages, clinical trials have shown promising results. Monoclonal antibodies targeting CD24 have been found to be well-tolerated and safe. Other preclinical studies are exploring the use of chimeric antigen receptor (CAR) T cells, antibody-drug conjugates, and gene therapy to target CD24 and enhance the immune response against tumors. In summary, this review focuses on the role of CD24 in the immune system and provides evidence for CD24 as a promising immune checkpoint for cancer immunotherapy.
Collapse
Affiliation(s)
- Shiming Huang
- Department of Radiology, First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- Graduate School, Chinese PLA Medical School, Beijing, China
- Department of Nuclear Medicine, Characteristic Medical Center of the Chinese People’s Armed Police Force, Tianjin, China
| | - Xiaobo Zhang
- Department of Radiology, First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Yingtian Wei
- Department of Radiology, First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Yueyong Xiao
- Department of Radiology, First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| |
Collapse
|
9
|
Chen W, Hu Z, Guo Z. Targeting CD24 in Cancer Immunotherapy. Biomedicines 2023; 11:3159. [PMID: 38137380 PMCID: PMC10740697 DOI: 10.3390/biomedicines11123159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Immunotherapy is a hot area in cancer treatment, and one of the keys to this therapy is the identification of the right tumour-associated or tumour-specific antigen. Cluster of differentiation 24 (CD24) is an emerging tumour-associated antigen that is commonly and highly expressed in various tumours. In addition, CD24 is associated with several cancer-related signalling pathways and closely interacts with other molecules and immune cells to influence tumour progression. Monoclonal antibodies, antibody-drug conjugates (ADCs), chimeric antigen receptor (CAR) T-cell therapy, and CAR-NK cell therapy are currently available for the treatment of CD24. In this review, we summarise the existing therapeutic approaches and possible future directions targeting CD24.
Collapse
Affiliation(s)
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210023, China;
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210023, China;
| |
Collapse
|
10
|
Moon SY, Han M, Ryu G, Shin SA, Lee JH, Lee CS. Emerging Immune Checkpoint Molecules on Cancer Cells: CD24 and CD200. Int J Mol Sci 2023; 24:15072. [PMID: 37894750 PMCID: PMC10606340 DOI: 10.3390/ijms242015072] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/04/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Cancer immunotherapy strategies are based on the utilization of immune checkpoint inhibitors to instigate an antitumor immune response. The efficacy of immune checkpoint blockade, directed at adaptive immune checkpoints, has been demonstrated in select cancer types. However, only a limited subset of patients has exhibited definitive outcomes characterized by a sustained response after discontinuation of therapy. Recent investigations have highlighted the significance of immune checkpoint molecules that are overexpressed in cancer cells and inhibit myeloid lineage immune cells within a tumor microenvironment. These checkpoints are identified as potential targets for anticancer immune responses. Notably, the immune checkpoint molecules CD24 and CD200 have garnered attention owing to their involvement in tumor immune evasion. CD24 and CD200 are overexpressed across diverse cancer types and serve as signaling checkpoints by engaging their respective receptors, Siglec-10 and CD200 receptor, which are expressed on tumor-associated myeloid cells. In this review, we summarized and discussed the latest advancements and insights into CD24 and CD200 as emergent immune checkpoint moieties, further delving into their therapeutic potentials for cancer treatment.
Collapse
Affiliation(s)
- Sun Young Moon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| | - Minjoo Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| | - Gyoungah Ryu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| | - Seong-Ah Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| | - Jun Hyuck Lee
- Research Unit of Cryogenic Novel Material, Korea Polar Research Institute, Incheon 21990, Republic of Korea;
- Department of Polar Sciences, University of Science and Technology, Incheon 21990, Republic of Korea
| | - Chang Sup Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| |
Collapse
|
11
|
Söhngen C, Thomas DJ, Skowron MA, Bremmer F, Eckstein M, Stefanski A, Driessen MD, Wakileh GA, Stühler K, Altevogt P, Theodorescu D, Klapdor R, Schambach A, Nettersheim D. CD24 targeting with NK-CAR immunotherapy in testis, prostate, renal and (luminal-type) bladder cancer and identification of direct CD24 interaction partners. FEBS J 2023; 290:4864-4876. [PMID: 37254618 PMCID: PMC11129509 DOI: 10.1111/febs.16880] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 04/26/2023] [Accepted: 05/26/2023] [Indexed: 06/01/2023]
Abstract
Alternative therapeutic options targeting urologic malignancies, such as germ cell tumours, as well as urothelial, renal and prostate carcinomas, are still urgently needed. The membrane protein CD24 represents a promising immunotherapeutical approach. The present study aimed to decipher the molecular function of CD24 in vitro and evaluate the cytotoxic capacity of a third-generation natural killer (NK) cell chimeric antigen receptor (CAR) against CD24 in urologic tumour cell lines. Up to 20 urologic tumour cell lines and several non-malignant control cells were included. XTT viability assays and annexin V/propidium iodide flow cytometry analyses were performed to measure cell viability and apoptosis rates, respectively. Co-immunoprecipitation followed by mass spectrometry analyses identified direct interaction partners of CD24. Luciferase reporter assays were used to functionally validate transactivation of CD24 expression by SOX2. N- and O-glycosylation of CD24 were evaluated by enzymatic digestion and mass spectrometry. The study demonstrates that SOX2 transactivates CD24 expression in embryonal carcinoma cells. In cells of different urological origins, CD24 interacted with proteins involved in cell adhesion, ATP binding, phosphoprotein binding and post-translational modifications, such as histone acetylation and ubiquitination. Treatment of urological tumour cells with NK-CD24-CAR cells resulted in a decreased cell viability and apoptosis induction specifically in CD24+ tumour cells. Limitations of the study include the in vitro setting, which still has to be confirmed in vivo. In conclusion, we show that CD24 is a promising novel target for immune therapeutic approaches targeting urologic malignancies.
Collapse
Affiliation(s)
- Christian Söhngen
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany
| | - David J. Thomas
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany
| | - Margaretha A. Skowron
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany
| | - Felix Bremmer
- Institute of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Markus Eckstein
- Institute of Pathology, Friedrich Alexander University Erlangen-Nürnberg, University Hospital, Erlangen, Germany
| | - Anja Stefanski
- Molecular Proteomics Laboratory, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Marc D. Driessen
- Molecular Proteomics Laboratory, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Gamal A. Wakileh
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany
- Department of Urology, University Hospital Ulm, Ulm, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University Heidelberg, Germany
| | - Dan Theodorescu
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rüdiger Klapdor
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
- Institute for Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Daniel Nettersheim
- Department of Urology, Urological Research Laboratory, Translational UroOncology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany
| |
Collapse
|
12
|
Huntoon K, Lee D, Dong S, Antony A, Kim BYS, Jiang W. Targeting phagocytosis to enhance antitumor immunity. Trends Cancer 2023; 9:650-665. [PMID: 37150626 DOI: 10.1016/j.trecan.2023.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/10/2023] [Accepted: 04/14/2023] [Indexed: 05/09/2023]
Abstract
Many patients with metastatic or treatment-resistant cancer have experienced improved outcomes after immunotherapy that targets adaptive immune checkpoints. However, innate immune checkpoints, which can hinder the detection and clearance of malignant cells, are also crucial in tumor-mediated immune escape and may also serve as targets in cancer immunotherapy. In this review, we discuss the current understanding of immune evasion by cancer cells via disruption of phagocytic clearance, and the potential effects of blocking phagocytosis checkpoints on the activation of antitumor immune responses. We propose that a more effective combination immunotherapy strategy could be to exploit tumor-intrinsic processes that inhibit key innate immune surveillance processes, such as phagocytosis, and incorporate both innate and adaptive immune responses for treating patients with cancer.
Collapse
Affiliation(s)
- Kristin Huntoon
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - DaeYong Lee
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shiyan Dong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Abin Antony
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
13
|
Wang Y, Yu H, Yu M, Liu H, Zhang B, Wang Y, Zhao S, Xia Q. CD24 blockade as a novel strategy for cancer treatment. Int Immunopharmacol 2023; 121:110557. [PMID: 37379708 DOI: 10.1016/j.intimp.2023.110557] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 05/22/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
The CD24 protein is a heat-stable protein with a small core that undergoes extensive glycosylation. It is expressed on the surface of various normal cells, including lymphocytes, epithelial cells, and inflammatory cells. CD24 exerts its function by binding to different ligands. Numerous studies have demonstrated the close association of CD24 with tumor occurrence and progression. CD24 not only facilitates tumor cell proliferation, metastasis, and immune evasion but also plays a role in tumor initiation, thus, serving as a marker on the surface of cancer stem cells (CSCs). Additionally, CD24 induces drug resistance in various tumor cells following chemotherapy. To counteract the tumor-promoting effects of CD24, several treatment strategies targeting CD24 have been explored, such as the use of CD24 monoclonal antibodies (mAb) alone, the combination of CD24 and chemotoxic drugs, or the combination of these drugs with other targeted immunotherapeutic techniques. Regardless of the approach, targeting CD24 has demonstrated significant anti-tumor effects. Therefore, the present study focuses on anti-tumor therapy and provides a comprehensive review of the structure and fundamental physiological function of CD24 and its impact on tumor development, and suggests that targeting CD24 may represent an effective strategy for treating malignant tumors.
Collapse
Affiliation(s)
- Yawen Wang
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China; Henan Engineering Research Center of Pathological Diagnostic Antibody, Zhengzhou 450008, China
| | - Haoran Yu
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China; Henan Engineering Research Center of Pathological Diagnostic Antibody, Zhengzhou 450008, China
| | - Mengyuan Yu
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China; Henan Engineering Research Center of Pathological Diagnostic Antibody, Zhengzhou 450008, China
| | - Hui Liu
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Bing Zhang
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China
| | - Yuanyuan Wang
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China
| | - Simin Zhao
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China.
| | - Qingxin Xia
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou 450008, China; Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou 450008, China; Henan Engineering Research Center of Pathological Diagnostic Antibody, Zhengzhou 450008, China.
| |
Collapse
|
14
|
Gu Y, Lu Y, Xiong Y, Zhan X, Liu T, Tang M, Xie A, Liu X, Fu B. Advances in the bladder cancer research using 3D culture models. Bladder (San Franc) 2023; 10:e21200005. [PMID: 37936584 PMCID: PMC10627085 DOI: 10.14440/bladder.2023.856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/31/2023] [Accepted: 02/28/2023] [Indexed: 11/09/2023] Open
Abstract
Bladder cancer represents the most common malignancy of the urinary system, posing a significant threat to patients' life. Animal models and two-dimensional (2D) cell cultures, among other traditional models, have been used for years to study various aspects of bladder cancer. However, these methods are subject to various limitations when mimicking the tumor microenvironment in vivo, thus hindering the further improvement of bladder cancer treatments. Recently, three-dimensional (3D) culture models have attracted extensive attention since they overcome the shortcomings of their traditional counterparts. Most importantly, 3D culture models more accurately reproduce the tumor microenvironment in the human body because they can recapitulate the cell-cell and cell-extracellular matrix interactions. 3D culture models can thereby help us gain deeper insight into the bladder cancer. The 3D culture models of tumor cells can extend the culture duration and allow for co-culturing with different cell types. Study of patient-specific bladder cancer mutations and subtypes is made possible by the ability to preserve cells isolated from particular patients in 3D culture models. It will be feasible to develop customized treatments that target relevant signaling pathways or biomarkers. This article reviews the development, application, advantages, and limitations of traditional modeling systems and 3D culture models used in the study of bladder cancer and discusses the potential application of 3D culture models.
Collapse
Affiliation(s)
- Yexin Gu
- Cyberiad Intelligent Technology, Shanghai 201112, China
| | - Ye Lu
- Cyberiad Intelligent Technology, Shanghai 201112, China
| | - Yunqiang Xiong
- Department of Urology, the First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang City, Jiangxi Province, China
| | - Xiangpeng Zhan
- Department of Urology, the First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang City, Jiangxi Province, China
| | - Taobin Liu
- Department of Urology, the First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang City, Jiangxi Province, China
| | - Min Tang
- Cyberiad Intelligent Technology, Shanghai 201112, China
| | - An Xie
- Department of Urology, the First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang City, Jiangxi Province, China
| | - Xiaoqiang Liu
- Department of Urology, the First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang City, Jiangxi Province, China
| | - Bin Fu
- Department of Urology, the First Affiliated Hospital of Nanchang University, 17 Yongwai Street, Nanchang City, Jiangxi Province, China
| |
Collapse
|
15
|
Christian SL. CD24 as a Potential Therapeutic Target in Patients with B-Cell Leukemia and Lymphoma: Current Insights. Onco Targets Ther 2022; 15:1391-1402. [PMID: 36425299 PMCID: PMC9680537 DOI: 10.2147/ott.s366625] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/10/2022] [Indexed: 01/12/2024] Open
Abstract
CD24 is a highly glycosylated glycophosphatidylinositol (GPI)-anchored protein that is expressed in many types of differentiating cells and some mature cells of the immune system as well as the central nervous system. CD24 has been extensively used as a biomarker for developing B cells as its expression levels change over the course of B cell development. Functionally, engagement of CD24 induces apoptosis in developing B cells and restricts cell growth in more mature cell types. Interestingly, CD24 is also expressed on many hematological and solid tumors. As such, it has been investigated as a therapeutic target in many solid tumors including ovarian, colorectal, pancreatic, lung and others. Most of the B-cell leukemias and lymphomas studied to date express CD24 but its role as a therapeutic target in these malignancies has, thus far, been understudied. Here, I review what is known about CD24 biology with a focus on B cell development and activation followed by a brief overview of how CD24 is being targeted in solid tumors. This is followed by an assessment of the value of CD24 as a therapeutic target in B cell leukemia and lymphoma in humans, including an evaluation of the challenges in using CD24 as a target considering its pattern of expression on normal cells.
Collapse
Affiliation(s)
- Sherri L Christian
- Department of Biochemistry, Memorial University of Newfoundland, St. John’s, NL, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| |
Collapse
|
16
|
Wang Y, Johnson KCC, Gatti-Mays ME, Li Z. Emerging strategies in targeting tumor-resident myeloid cells for cancer immunotherapy. J Hematol Oncol 2022; 15:118. [PMID: 36031601 PMCID: PMC9420297 DOI: 10.1186/s13045-022-01335-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/09/2022] [Indexed: 12/11/2022] Open
Abstract
Immune checkpoint inhibitors targeting programmed cell death protein 1, programmed death-ligand 1, and cytotoxic T-lymphocyte-associated protein 4 provide deep and durable treatment responses which have revolutionized oncology. However, despite over 40% of cancer patients being eligible to receive immunotherapy, only 12% of patients gain benefit. A key to understanding what differentiates treatment response from non-response is better defining the role of the innate immune system in anti-tumor immunity and immune tolerance. Teleologically, myeloid cells, including macrophages, dendritic cells, monocytes, and neutrophils, initiate a response to invading pathogens and tissue repair after pathogen clearance is successfully accomplished. However, in the tumor microenvironment (TME), these innate cells are hijacked by the tumor cells and are imprinted to furthering tumor propagation and dissemination. Major advancements have been made in the field, especially related to the heterogeneity of myeloid cells and their function in the TME at the single cell level, a topic that has been highlighted by several recent international meetings including the 2021 China Cancer Immunotherapy workshop in Beijing. Here, we provide an up-to-date summary of the mechanisms by which major myeloid cells in the TME facilitate immunosuppression, enable tumor growth, foster tumor plasticity, and confer therapeutic resistance. We discuss ongoing strategies targeting the myeloid compartment in the preclinical and clinical settings which include: (1) altering myeloid cell composition within the TME; (2) functional blockade of immune-suppressive myeloid cells; (3) reprogramming myeloid cells to acquire pro-inflammatory properties; (4) modulating myeloid cells via cytokines; (5) myeloid cell therapies; and (6) emerging targets such as Siglec-15, TREM2, MARCO, LILRB2, and CLEVER-1. There is a significant promise that myeloid cell-based immunotherapy will help advance immuno-oncology in years to come.
Collapse
Affiliation(s)
- Yi Wang
- Division of Medical Oncology, Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | | | - Margaret E Gatti-Mays
- Division of Medical Oncology, Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
- Stefanie Spielman Comprehensive Breast Center, Columbus, OH, USA.
| | - Zihai Li
- Division of Medical Oncology, Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
17
|
Panagiotou E, Syrigos NK, Charpidou A, Kotteas E, Vathiotis IA. CD24: A Novel Target for Cancer Immunotherapy. J Pers Med 2022; 12:jpm12081235. [PMID: 36013184 PMCID: PMC9409925 DOI: 10.3390/jpm12081235] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/31/2022] Open
Abstract
Cluster of differentiation 24 (CD24) is a small, highly glycosylated cell adhesion protein that is normally expressed by immune as well as epithelial, neural, and muscle cells. Tumor CD24 expression has been linked with alterations in several oncogenic signaling pathways. In addition, the CD24/Siglec-10 interaction has been implicated in tumor immune evasion, inhibiting macrophage-mediated phagocytosis as well as natural killer (NK) cell cytotoxicity. CD24 blockade has shown promising results in preclinical studies. Although there are limited data on efficacy, monoclonal antibodies against CD24 have demonstrated clinical safety and tolerability in two clinical trials. Other treatment modalities evaluated in the preclinical setting include antibody–drug conjugates and chimeric antigen receptor (CAR) T cell therapy. In this review, we summarize current evidence and future perspectives on CD24 as a potential target for cancer immunotherapy.
Collapse
|
18
|
CD24 Is a Prognostic Marker for Multiple Myeloma Progression and Survival. J Clin Med 2022; 11:jcm11102913. [PMID: 35629039 PMCID: PMC9144978 DOI: 10.3390/jcm11102913] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022] Open
Abstract
Surface antigens are commonly used in flow cytometry assays for the diagnosis of multiple myeloma (MM). Some of these are directly involved in MM pathogenesis or interactions with the microenvironment, but most are used for either diagnostic or prognostic purposes. In a previous study, we showed that in-vitro, CD24-positive plasma cells exhibit a less tumorigenic phenotype. Here, we assessed the prognostic importance of CD24 expression in patients newly diagnosed with MM as it correlates to their clinical course. Immunophenotyping by flow cytometry of 124 patients uniformly treated by a bortezomib-based protocol was performed. The expression of CD24, CD117, CD19, CD45, and CD56 in bone marrow PCs was tested for correlations to clinical parameters. None of the CD markers correlated with the response rates to first-line therapy. However, patients with elevated CD24+ expression on their PCs at diagnosis had a significantly longer PFS (p = 0.002) and OS (p = 0.044). In contrast, the expression of CD117, CD56, or CD45 was found to have no prognostic value; CD19 expression was inversely correlated with PFS alone (p < 0.001) and not with OS. Thus, elevated CD24 expression on PCs appears to be strongly correlated with survival and can be used as a single-surface antigenic prognostic factor in MM.
Collapse
|
19
|
Geng R, Harland N, Montes-Mojarro IA, Fend F, Aicher WK, Stenzl A, Amend B. CD24: A Marker for an Extended Expansion Potential of Urothelial Cancer Cell Organoids In Vitro? Int J Mol Sci 2022; 23:5453. [PMID: 35628262 PMCID: PMC9141653 DOI: 10.3390/ijms23105453] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Bladder cancer is the most cost-intensive cancer due to high recurrence rates and long follow-up times. Bladder cancer organoids were considered interesting tools for investigating better methods for the detection and treatment of this cancer. METHODS Organoids were generated from urothelial carcinoma tissue samples, then expanded and characterized; the expression of immune modulatory antigens and tumor stem cells markers CD24 and CD44 was explored in early (P ≤ 3) and later (P ≥ 5) passages (P) by immunofluorescence and by quantitative PCR of cDNA. The expression of these factors was investigated in the corresponding cancer tissue samples by immunohistochemistry. RESULTS The expression of the PD-L1 was detected on some but not all organoids. CD276 and CD47 were observed on organoids in all passages investigated. Organoids growing beyond passage 8 expressed both CD24 and CD44 at elevated levels in early and late cultures. Organoids proliferating to the eighth passage initially expressed both CD24 and CD44, but lost CD24 expression over time, while CD44 remained. Organoids growing only up to the 6th passage failed to express CD24 but expressed CD44. CONCLUSIONS The data indicate that the expression of CD24 in urothelial cancer cell organoids may serve as an indicator for the prolonged proliferation potential of the cells.
Collapse
Affiliation(s)
- Ruizhi Geng
- Center for Medical Research, University Hospital, Eberhard Karls University, 72074 72072 Tuebingen, Germany; (R.G.); (W.K.A.)
| | - Niklas Harland
- Department of Urology, University of Tuebingen Hospital, 72076 Tuebingen, Germany; (N.H.); (A.S.)
| | - Ivonne A. Montes-Mojarro
- Institute for Pathology, Eberhard Karls University, 72074 Tuebingen, Germany; (I.A.M.-M.); (F.F.)
| | - Falko Fend
- Institute for Pathology, Eberhard Karls University, 72074 Tuebingen, Germany; (I.A.M.-M.); (F.F.)
| | - Wilhelm K. Aicher
- Center for Medical Research, University Hospital, Eberhard Karls University, 72074 72072 Tuebingen, Germany; (R.G.); (W.K.A.)
| | - Arnulf Stenzl
- Department of Urology, University of Tuebingen Hospital, 72076 Tuebingen, Germany; (N.H.); (A.S.)
| | - Bastian Amend
- Department of Urology, University of Tuebingen Hospital, 72076 Tuebingen, Germany; (N.H.); (A.S.)
| |
Collapse
|
20
|
Wang Y, Zhao C, Liu Y, Wang C, Jiang H, Hu Y, Wu J. Recent Advances of Tumor Therapy Based on the CD47-SIRPα Axis. Mol Pharm 2022; 19:1273-1293. [PMID: 35436123 DOI: 10.1021/acs.molpharmaceut.2c00073] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer is still a major disease that is currently difficult for humans to overcome. When the expression of the cluster of differentiation 47 (CD47) is upregulated, tumor cells interact with the macrophage inhibitory receptor signal regulatory protein α (SIRPα) to transmit the "Don't eat me" signal, thereby avoiding phagocytosis by the macrophages. Therefore, when the CD47-SIRPα axis is inhibited, the macrophages' phagocytic function can be restored and can also exert antitumor effects. This Review mainly introduces recent advances in tumor therapy targeted on the CD47-SIRPα axis, including the antibody and fusion protein, small molecule, gene therapy, cell therapy, and drug delivery system, to inhibit the function of CD47 expressed on tumor cells and promote tumor phagocytosis by macrophages. In addition, this Review also summarizes the current approaches to avoid anemia, a common side effect of CD47-SIRPα inhibitions, and provides ideas for clinical transformation.
Collapse
Affiliation(s)
- Yuchen Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Chenxuan Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Yang Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Chao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Haojie Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China.,Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China.,Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing 210093, China
| |
Collapse
|
21
|
Quader S, Tanabe S, Cabral H. Abnormal Glycosylation in Cancer Cells and Cancer Stem Cells as a Therapeutic Target. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1393:141-156. [PMID: 36587306 DOI: 10.1007/978-3-031-12974-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Tumor resistance and recurrence have been associated with the presence of cancer stem cells (CSCs) in tumors. The functions and survival of the CSCs have been associated with several intracellular and extracellular features. Particularly, the abnormal glycosylation of these signaling pathways and markers of CSCs have been correlated with maintaining survival, self-renewal and extravasation properties. Here, we highlight the importance of glycosylation in promoting the stemness character of CSCs and the current strategies for targeting abnormal glycosylation toward generating effective therapies against the CSC population.
Collapse
Affiliation(s)
- Sabina Quader
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Shihori Tanabe
- Division of Risk Assessment, Center for Biological Safety and Research, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, 210-9501, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
| |
Collapse
|
22
|
Animal Models in Bladder Cancer. Biomedicines 2021; 9:biomedicines9121762. [PMID: 34944577 PMCID: PMC8698361 DOI: 10.3390/biomedicines9121762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/17/2021] [Accepted: 11/21/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Bladder cancer (urothelial cancer of the bladder) is the most common malignancy affecting the urinary system with an increasing incidence and mortality. Mouse models of bladder cancer should possess a high value of reproducibility, predictability, and translatability to allow mechanistic, chemo-preventive, and therapeutic studies that can be furthered into human clinical trials. OBJECTIVES To provide an overview and resources on the origin, molecular and pathological characteristics of commonly used animal models in bladder cancer. METHODS A PubMed and Web of Science search was performed for relevant articles published between 1980 and 2021 using words such as: "bladder" and/or "urothelial carcinoma" and animal models. Animal models of bladder cancer can be categorized as autochthonous (spontaneous) and non-autochthonous (transplantable). The first are either chemically induced models or genetically engineered models. The transplantable models can be further subclassified as syngeneic (murine bladder cancer cells implanted into immunocompetent or transgenic mice) and xenografts (human bladder cancer cells implanted into immune-deficient mice). These models can be further divided-based on the site of the tumor-as orthotopic (tumor growth occurs within the bladder) and heterotopic (tumor growth occurs outside of the bladder).
Collapse
|
23
|
Ni YH, Zhao X, Wang W. CD24, A Review of its Role in Tumor Diagnosis, Progression and Therapy. Curr Gene Ther 2021; 20:109-126. [PMID: 32576128 DOI: 10.2174/1566523220666200623170738] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/28/2020] [Accepted: 06/02/2020] [Indexed: 02/08/2023]
Abstract
CD24, is a mucin-like GPI-anchored molecules. By immunohistochemistry, it is widely detected in many solid tumors, such as breast cancers, genital system cancers, digestive system cancers, neural system cancers and so on. The functional roles of CD24 are either fulfilled by combination with ligands or participate in signal transduction, which mediate the initiation and progression of neoplasms. However, the character of CD24 remains to be intriguing because there are still opposite voices about the impact of CD24 on tumors. In preclinical studies, CD24 target therapies, including monoclonal antibodies, target silencing by RNA interference and immunotherapy, have shown us brighten futures on the anti-tumor application. Nevertheless, evidences based on clinical studies are urgently needed. Here, with expectancy to spark new ideas, we summarize the relevant studies about CD24 from a tumor perspective.
Collapse
Affiliation(s)
- Yang-Hong Ni
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041, Sichuan, China
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041, Sichuan, China
| |
Collapse
|
24
|
Wu H, Liu J, Wang Z, Yuan W, Chen L. Prospects of antibodies targeting CD47 or CD24 in the treatment of glioblastoma. CNS Neurosci Ther 2021; 27:1105-1117. [PMID: 34363319 PMCID: PMC8446212 DOI: 10.1111/cns.13714] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Glioma is a malignant tumor with the highest incidence among all brain tumors (about 46% of intracranial tumors) and is the most common primary intracranial tumor. Among them, glioblastoma (GBM) is highly malignant and is one of the three refractory tumors with the highest mortality rate in the world. The survival time from glioblastoma diagnosis to death is only 14–16 months for patients with standard treatment such as surgery plus radiotherapy and chemotherapy. Due to its high malignancy and poor prognosis, in‐depth studies have been conducted to explore effective therapeutic strategies for glioblastoma. In addition to the conventional surgery, radiotherapy, and chemotherapy, the glioblastoma treatments also include targeted therapy, immunotherapy, and electric field treatment. However, current treatment methods provide limited benefits because of the heterogeneity of glioblastoma and the complexity of the immune microenvironment within a tumor. Therefore, seeking an effective treatment plan is imperative. In particular, developing an active immunotherapy for glioblastoma has become an essential objective in the field. This article reviews the feasibility of CD47/CD24 antibody treatment, either individually or in combination, to target the tumor stem cells and the antitumor immunity in glioblastoma. The potential mechanisms underlying the antitumor effects of CD47/CD24 antibodies are also discussed.
Collapse
Affiliation(s)
- Hao Wu
- The Third Xiangya Hospital of Central South University, Changsha, China.,Chinese PLA General Hospital and PLA Medical College, Chinese PLA Institute of Neurosurgery, Beijing, China
| | - Jialin Liu
- Chinese PLA General Hospital and PLA Medical College, Chinese PLA Institute of Neurosurgery, Beijing, China
| | - Zhifei Wang
- The Third Xiangya Hospital of Central South University, Changsha, China
| | - Wen Yuan
- Zhuzhou Central Hospital, Zhuzhou, China
| | - Ling Chen
- Chinese PLA General Hospital and PLA Medical College, Chinese PLA Institute of Neurosurgery, Beijing, China
| |
Collapse
|
25
|
Shapira S, Kazanov D, Mdah F, Yaakobi H, Herishanu Y, Perry C, Avivi I, Itchaki G, Shacham-Abulafia A, Raanani P, Hay-Levy M, Aiger G, Mashiah J, Lev-Ari S, Arber N. Feasibly of CD24/CD11b as a Screening Test for Hematological Malignancies. J Pers Med 2021; 11:724. [PMID: 34442367 PMCID: PMC8399145 DOI: 10.3390/jpm11080724] [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: 07/01/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 11/17/2022] Open
Abstract
An estimated 1.24 million blood cancer cases occur annually worldwide, accounting for approximately 6% of all cancer cases. Currently, there are no standardized hematology cancer screening tests that are recommended for the general population. CD24 is a mucin-like cell surface molecule and P-selectin ligand, which plays a significant role in the maturation of B-lymphocytes and was found to be overexpressed in a number of hematological malignancies. Our primary aim was to assess the sensitivity and specificity of the CD24/CD11b-based blood test for the detection of hematological malignancies. Our cohort included 488 subjects with positive hematological cancer diagnosis (n = 122) and healthy subjects (n = 366). CD24/CD11b expression in peripheral blood leukocytes (PBLs) obtained from blood samples of participants was analyzed by flow cytometry. Our results demonstrated that the average levels of CD24/CD11b in healthy patients (21.7 ± 9.0) were statistically significantly lower compared to levels of CD24/CD11b in cancer patients (29.5 ± 18.7, p < 0.001). The highest levels of CD24/CD11b were found in multiple myeloma (39.1 ± 23.6), followed by chronic myeloid leukemia (33.0 ± 13.7) and non-Hodgkin lymphoma (32.3 ± 13.3). The test had an overall sensitivity for hematologic cancers of 78.5% (95% CI, 70.7-86.3%) and specificity of 80.2% (95% CI, 76.1-84.3%). In conclusion, our findings indicate the feasibility of a CD24/CD11b-based blood test as a screening test of hematological malignancies.
Collapse
Affiliation(s)
- Shiran Shapira
- Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv 6423906, Israel; (S.S.); (D.K.); (F.M.); (H.Y.); (M.H.-L.); (G.A.); (S.L.-A.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6423906, Israel; (Y.H.); (C.P.); (I.A.); (G.I.); (A.S.-A.); (P.R.)
| | - Dina Kazanov
- Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv 6423906, Israel; (S.S.); (D.K.); (F.M.); (H.Y.); (M.H.-L.); (G.A.); (S.L.-A.)
| | - Fatin Mdah
- Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv 6423906, Israel; (S.S.); (D.K.); (F.M.); (H.Y.); (M.H.-L.); (G.A.); (S.L.-A.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6423906, Israel; (Y.H.); (C.P.); (I.A.); (G.I.); (A.S.-A.); (P.R.)
| | - Hadas Yaakobi
- Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv 6423906, Israel; (S.S.); (D.K.); (F.M.); (H.Y.); (M.H.-L.); (G.A.); (S.L.-A.)
| | - Yair Herishanu
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6423906, Israel; (Y.H.); (C.P.); (I.A.); (G.I.); (A.S.-A.); (P.R.)
- Tel Aviv Medical Center, Department of Hematology, Tel Aviv 6423906, Israel
| | - Chava Perry
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6423906, Israel; (Y.H.); (C.P.); (I.A.); (G.I.); (A.S.-A.); (P.R.)
- Tel Aviv Medical Center, Department of Hematology, Tel Aviv 6423906, Israel
| | - Irit Avivi
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6423906, Israel; (Y.H.); (C.P.); (I.A.); (G.I.); (A.S.-A.); (P.R.)
- Tel Aviv Medical Center, Department of Hematology, Tel Aviv 6423906, Israel
| | - Gilad Itchaki
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6423906, Israel; (Y.H.); (C.P.); (I.A.); (G.I.); (A.S.-A.); (P.R.)
- Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Petah Tikva 49100, Israel
| | - Adi Shacham-Abulafia
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6423906, Israel; (Y.H.); (C.P.); (I.A.); (G.I.); (A.S.-A.); (P.R.)
- Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Petah Tikva 49100, Israel
| | - Pia Raanani
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6423906, Israel; (Y.H.); (C.P.); (I.A.); (G.I.); (A.S.-A.); (P.R.)
- Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Petah Tikva 49100, Israel
| | - Mori Hay-Levy
- Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv 6423906, Israel; (S.S.); (D.K.); (F.M.); (H.Y.); (M.H.-L.); (G.A.); (S.L.-A.)
| | - Gal Aiger
- Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv 6423906, Israel; (S.S.); (D.K.); (F.M.); (H.Y.); (M.H.-L.); (G.A.); (S.L.-A.)
| | - Jacob Mashiah
- Tel Aviv Medical Center, The Pediatric Dermatology Unit, Tel Aviv 6423906, Israel;
| | - Shahar Lev-Ari
- Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv 6423906, Israel; (S.S.); (D.K.); (F.M.); (H.Y.); (M.H.-L.); (G.A.); (S.L.-A.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6423906, Israel; (Y.H.); (C.P.); (I.A.); (G.I.); (A.S.-A.); (P.R.)
| | - Nadir Arber
- Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv 6423906, Israel; (S.S.); (D.K.); (F.M.); (H.Y.); (M.H.-L.); (G.A.); (S.L.-A.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6423906, Israel; (Y.H.); (C.P.); (I.A.); (G.I.); (A.S.-A.); (P.R.)
| |
Collapse
|
26
|
Khan T, Cabral H. Abnormal Glycosylation of Cancer Stem Cells and Targeting Strategies. Front Oncol 2021; 11:649338. [PMID: 33889547 PMCID: PMC8056457 DOI: 10.3389/fonc.2021.649338] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cell (CSCs) are deemed as one of the main reasons of tumor relapse due to their resistance to standard therapies. Numerous intracellular signaling pathways along with extracellular features are crucial in regulating CSCs properties, such as heterogeneity, plasticity and differentiation. Aberrant glycosylation of these cellular signaling pathways and markers of CSCs have been directly correlated with maintaining survival, self-renewal and extravasation properties. In this review, we highlight the importance of glycosylation in promoting stemness character of CSCs, and present strategies for targeting abnormal glycosylation to eliminate the resistant CSC population.
Collapse
Affiliation(s)
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
27
|
Fu B, Wang D, Shen X, Guo C, Liu Y, Ye Y, Sun R, Li J, Tian Z, Wei H. Immunomodulation Induced During Interferon-α Therapy Impairs the Anti-HBV Immune Response Through CD24 +CD38 hi B Cells. Front Immunol 2020; 11:591269. [PMID: 33424840 PMCID: PMC7786281 DOI: 10.3389/fimmu.2020.591269] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Type I interferon is widely used for antiviral therapy, yet has yielded disappointing results toward chronic HBV infection. Here we identify that PEG-IFNα-2b therapy toward persistent infection in humans is a double-edged sword of both immunostimulation and immunomodulation. Our studies of this randomised trial showed persistent PEG-IFNα-2b therapy induced large number of CD24+CD38hi B cells and launched a CD24+CD38hi B cells centered immunosuppressive response, including downregulating functions of T cells and NK cells. Patients with low induced CD24+CD38hi B cells have achieved an improved therapeutic effect. Specifically, using the anti-CD24 antibody to deplete CD24+CD38hi B cells without harming other B cell subsets suggest a promising strategy to improve the therapeutic effects. Our findings show that PEG-IFNα-2b therapy toward persistent infection constitutes an immunomodulation effect, and strategies to identifying the molecular basis for the antiviral versus immunomodulatory effects of PEG-IFNα-2b to selectively manipulate these opposing activities provide an opportunity to ameliorate anti-virus immunity and control viral infection.
Collapse
Affiliation(s)
- Binqing Fu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Dongyao Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xiaokun Shen
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Chuang Guo
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Yanyan Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ying Ye
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Rui Sun
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Jiabin Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| |
Collapse
|
28
|
The Emerging Role of CD24 in Cancer Theranostics-A Novel Target for Fluorescence Image-Guided Surgery in Ovarian Cancer and Beyond. J Pers Med 2020; 10:jpm10040255. [PMID: 33260974 PMCID: PMC7712410 DOI: 10.3390/jpm10040255] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022] Open
Abstract
Complete cytoreductive surgery is the cornerstone of the treatment of epithelial ovarian cancer (EOC). The application of fluorescence image-guided surgery (FIGS) allows for the increased intraoperative visualization and delineation of malignant lesions by using fluorescently labeled targeting biomarkers, thereby improving intraoperative guidance. CD24, a small glycophosphatidylinositol-anchored cell surface receptor, is overexpressed in approximately 70% of solid cancers, and has been proposed as a prognostic and therapeutic tumor-specific biomarker for EOC. Recently, preclinical studies have demonstrated the benefit of CD24-targeted contrast agents for non-invasive fluorescence imaging, as well as improved tumor resection by employing CD24-targeted FIGS in orthotopic patient-derived xenograft models of EOC. The successful detection of miniscule metastases denotes CD24 as a promising biomarker for the application of fluorescence-guided surgery in EOC patients. The aim of this review is to present the clinical and preclinically evaluated biomarkers for ovarian cancer FIGS, highlight the strengths of CD24, and propose a future bimodal approach combining CD24-targeted fluorescence imaging with radionuclide detection and targeted therapy.
Collapse
|
29
|
Giulietti M, Bastianoni M, Cecati M, Ruzzo A, Bracci M, Malavolta M, Piacenza F, Giacconi R, Piva F. MetaTropismDB: a database of organ-specific metastasis induced by human cancer cell lines in mouse models. Database (Oxford) 2020; 2020:baaa100. [PMID: 33238004 PMCID: PMC7687678 DOI: 10.1093/database/baaa100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 09/25/2020] [Accepted: 11/02/2020] [Indexed: 11/12/2022]
Abstract
The organotropism is the propensity of metastatic cancer cells to colonize preferably certain distant organs, resulting in a non-random distribution of metastases. In order to shed light on this behaviour, several studies were performed by the injection of human cancer cell lines into immunocompromised mouse models. However, the information about these experiments is spread in the literature. For each xenograft experiment reported in the literature, we annotated both the experimental conditions and outcomes, including details on inoculated human cell lines, mouse models, injection methods, sites of metastasis, organs not colonized, rate of metastasis, latency time, overall survival and the involved genes. We created MetaTropismDB, a freely available database collecting hand-curated data useful to highlight the mechanisms of organ-specific metastasis. Currently, it stores the results of 513 experiments in which injections of 219 human cell lines have been carried out in mouse models. Notably, 296 genes involved in organotropic metastases have been collected. This specialized database allows the researchers to compare the current results about organotropism and plan future experiments in order to identify which tumour molecular signatures establish if and where the metastasis will develop. Database URL: http://www.introni.it/Metastasis/metastasis.html.
Collapse
Affiliation(s)
- Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy
| | - Marco Bastianoni
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy
| | - Monia Cecati
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy
| | - Annamaria Ruzzo
- Department of Biomolecular Sciences, University of Urbino “Carlo Bo”, Via Sant’Andrea 34, 61029, Urbino, Italy
| | - Massimo Bracci
- Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Via Tronto 10/a, 60126, Ancona, Italy
| | - Marco Malavolta
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, Via Birarelli 8, 60121, Ancona, Italy
| | - Francesco Piacenza
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, Via Birarelli 8, 60121, Ancona, Italy
| | - Robertina Giacconi
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, Via Birarelli 8, 60121, Ancona, Italy
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy
| |
Collapse
|
30
|
Altevogt P, Sammar M, Hüser L, Kristiansen G. Novel insights into the function of CD24: A driving force in cancer. Int J Cancer 2020; 148:546-559. [PMID: 32790899 DOI: 10.1002/ijc.33249] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/23/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
CD24 is a highly glycosylated protein with a small protein core that is linked to the plasma membrane via a glycosyl-phosphatidylinositol anchor. CD24 is primarily expressed by immune cells but is often overexpressed in human tumors. In cancer, CD24 is a regulator of cell migration, invasion and proliferation. Its expression is associated with poor prognosis and it is used as cancer stemness marker. Recently, CD24 on tumor cells was identified as a phagocytic inhibitor ("do not eat me" signal) having a suppressive role in tumor immunity via binding to Siglec-10 on macrophages. This finding is reminiscent of the demonstration that soluble CD24-Fc can dampen the immune system in autoimmune disease. In the present review, we summarize recent progress on the role of the CD24-Siglec-10 binding axis at the interface between tumor cells and the immune system, and the role of CD24 genetic polymorphisms in cancer. We describe the specific function of cytoplasmic CD24 and discuss the presence of CD24 on tumor-released extracellular vesicles. Finally, we evaluate the potential of CD24-based immunotherapy.
Collapse
Affiliation(s)
- Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Marei Sammar
- ORT Braude College for Engineering, Karmiel, Israel
| | - Laura Hüser
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | | |
Collapse
|
31
|
Characteristic molecular signature of pericardial effusion identifies malignant cancer in pericardial disorder patients. Mol Cell Toxicol 2020. [DOI: 10.1007/s13273-020-00076-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
32
|
CD24-targeted fluorescence imaging in patient-derived xenograft models of high-grade serous ovarian carcinoma. EBioMedicine 2020; 56:102782. [PMID: 32454401 PMCID: PMC7248428 DOI: 10.1016/j.ebiom.2020.102782] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 04/02/2020] [Accepted: 04/21/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The survival rate of patients with advanced high-grade serous ovarian carcinoma (HGSOC) remains disappointing. Clinically translatable orthotopic cell line xenograft models and patient-derived xenografts (PDXs) may aid the implementation of more personalised treatment approaches. Although orthotopic PDX reflecting heterogeneous molecular subtypes are considered the most relevant preclinical models, their use in therapeutic development is limited by lack of appropriate imaging modalities. METHODS We developed novel orthotopic xenograft and PDX models for HGSOC, and applied a near-infrared fluorescently labelled monoclonal antibody targeting the cell surface antigen CD24 for non-invasive molecular imaging of epithelial ovarian cancer. CD24-Alexa Fluor 680 fluorescence imaging was compared to bioluminescence imaging in three orthotopic cell line xenograft models of ovarian cancer (OV-90luc+, Skov-3luc+ and Caov-3luc+, n = 3 per model). The application of fluorescence imaging to assess treatment efficacy was performed in carboplatin-paclitaxel treated orthotopic OV-90 xenografts (n = 10), before the probe was evaluated to detect disease progression in heterogenous PDX models (n = 7). FINDINGS Application of the near-infrared probe, CD24-AF680, enabled both spatio-temporal visualisation of tumour development, and longitudinal therapy monitoring of orthotopic xenografts. Notably, CD24-AF680 facilitated imaging of multiple PDX models representing different histological subtypes of the disease. INTERPRETATION The combined implementation of CD24-AF680 and orthotopic PDX models creates a state-of-the-art preclinical platform which will impact the identification and validation of new targeted therapies, fluorescence image-guided surgery, and ultimately the outcome for HGSOC patients. FUNDING This study was supported by the H2020 program MSCA-ITN [675743], Helse Vest RHF, and Helse Bergen HF [911809, 911852, 912171, 240222, HV1269], as well as by The Norwegian Cancer Society [182735], and The Research Council of Norway through its Centers of excellence funding scheme [223250, 262652].
Collapse
|
33
|
Gao M, Bai H, Jethava Y, Wu Y, Zhu Y, Yang Y, Xia J, Cao H, Franqui-Machin R, Nadiminti K, Thomas GS, Salama ME, Altevogt P, Bishop G, Tomasson M, Janz S, Shi J, Chen L, Frech I, Tricot G, Zhan F. Identification and Characterization of Tumor-Initiating Cells in Multiple Myeloma. J Natl Cancer Inst 2020; 112:507-515. [PMID: 31406992 PMCID: PMC7225664 DOI: 10.1093/jnci/djz159] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 07/11/2019] [Accepted: 08/07/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Treatment failures in cancers, including multiple myeloma (MM), are most likely due to the persistence of a minor population of tumor-initiating cells (TICs), which are noncycling or slowly cycling and very drug resistant. METHODS Gene expression profiling and real-time quantitative reverse transcription polymerase chain reaction were employed to define genes differentially expressed between the side-population cells, which contain the TICs, and the main population of MM cells derived from 11 MM patient samples. Self-renewal potential was analyzed by clonogenicity and drug resistance of CD24+ MM cells. Flow cytometry (n = 60) and immunofluorescence (n = 66) were applied on MM patient samples to determine CD24 expression. Therapeutic effects of CD24 antibodies were tested in xenograft MM mouse models containing three to six mice per group. RESULTS CD24 was highly expressed in the side-population cells, and CD24+ MM cells exhibited high expression of induced pluripotent or embryonic stem cell genes. CD24+ MM cells showed increased clonogenicity, drug resistance, and tumorigenicity. Only 10 CD24+ MM cells were required to develop plasmacytomas in mice (n = three of five mice after 27 days). The frequency of CD24+ MM cells was highly variable in primary MM samples, but the average of CD24+ MM cells was 8.3% after chemotherapy and in complete-remission MM samples with persistent minimal residual disease compared with 1.0% CD24+ MM cells in newly diagnosed MM samples (n = 26). MM patients with a high initial percentage of CD24+ MM cells had inferior progression-free survival (hazard ratio [HR] = 3.81, 95% confidence interval [CI] = 5.66 to 18.34, P < .001) and overall survival (HR = 3.87, 95% CI = 16.61 to 34.39, P = .002). A CD24 antibody inhibited MM cell growth and prevented tumor progression in vivo. CONCLUSION Our studies demonstrate that CD24+ MM cells maintain the TIC features of self-renewal and drug resistance and provide a target for myeloma therapy.
Collapse
Affiliation(s)
- Minjie Gao
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA.,Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hua Bai
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Yogesh Jethava
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Yujie Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Yuqi Zhu
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Ye Yang
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Jiliang Xia
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Huojun Cao
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, IA
| | - Reinaldo Franqui-Machin
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Kalyan Nadiminti
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Gregory S Thomas
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Mohamed E Salama
- Department of Pathology, University of Utah, and Associated Regional University Pathologists Laboratories, Salt Lake City, UT
| | - Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center, and University Medical Center Mannheim, Germany
| | - Gail Bishop
- Department of Microbiology and Immunology, University of Iowa and VA Medical Center, Iowa City, IA
| | - Michael Tomasson
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Siegfried Janz
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Jumei Shi
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lijuan Chen
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Ivana Frech
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Guido Tricot
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Fenghuang Zhan
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|
34
|
Cole AJ, Fayomi AP, Anyaeche VI, Bai S, Buckanovich RJ. An evolving paradigm of cancer stem cell hierarchies: therapeutic implications. Theranostics 2020; 10:3083-3098. [PMID: 32194856 PMCID: PMC7053211 DOI: 10.7150/thno.41647] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/22/2020] [Indexed: 02/07/2023] Open
Abstract
Over a decade of research has confirmed the critical role of cancer stem-like cells (CSCs) in tumor initiation, chemoresistance, and metastasis. Increasingly, CSC hierarchies have begun to be defined with some recurring themes. This includes evidence that these hierarchies are 'flexible,' with both cell state transitions and dedifferentiation events possible. These findings pose therapeutic hurdles and opportunities. Here, we review cancer stem cell hierarchies and their interactions with the tumor microenvironment. We also discuss the current therapeutic approaches designed to target CSC hierarchies and initial clinical trial results for CSC targeting agents. While cancer stem cell targeted therapies are still in their infancy, we are beginning to see encouraging results that suggest a positive outlook for CSC-targeting approaches.
Collapse
Affiliation(s)
- Alexander J Cole
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adetunji P Fayomi
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Shoumei Bai
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ronald J Buckanovich
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
35
|
Takashima K, Fujii S, Komatsuzaki R, Komatsu M, Takahashi M, Kojima T, Daiko H, Minashi K, Chiwaki F, Muto M, Sasaki H, Yano T. CD24 and CK4 are upregulated by SIM2, and are predictive biomarkers for chemoradiotherapy and surgery in esophageal cancer. Int J Oncol 2020; 56:835-847. [PMID: 32124945 DOI: 10.3892/ijo.2020.4963] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 11/19/2019] [Indexed: 11/05/2022] Open
Abstract
Definitive chemoradiotherapy (CRT) is a less invasive therapy compared with surgery for some types of cancer; however, the 5‑year survival rate of patients with stages II‑III esophageal squamous cell carcinoma (ESCC) is only 37%. Therefore, prediction of CRT responders is necessary. Unfortunately, no definitive biomarker exists that is useful to predict survival outcome following CRT. From our previous microarray study, CD24 and keratin 4 (KRT4), which encodes cytokeratin 4 (CK4), were overexpressed in the favorable prognostic epithelial subtype with SIM bHLH transcription factor 2 (SIM2) expression. This study investigated the association between their mRNA and protein expression levels, and clinicopathological characteristics, and also investigated the functions of CD24 in SIM2‑mediated tumor differentiation and CRT sensitivity. High CD24 and KRT4 mRNA expression was associated with a favorable prognosis following CRT. Multivariate analyses revealed that high CD24 and CK4 protein expression, as determined by immunohistochemistry, and differentiated type were independent factors for predicting a favorable prognosis in response to CRT. Notably, in cases with low CD24 or CK4, surgery was suggested to be a good therapeutic modality compared with CRT. CD24 and KRT4 were expressed preferentially in differentiated layers of the normal esophageal mucosa, and their mRNA expression in 3D cultured ESCC cells was induced by SIM2 transfection, thus suggesting that CD24 and KRT4 were downstream differentiation markers of SIM2. Furthermore, CD24 small interfering RNA increased the mRNA expression levels of superoxide dismutase 2 and enhanced H2O2 resistance, thus indicating the involvement of CD24 in the radiosensitivity of patients with ESCC; however, it had no effect on cisplatin sensitivity. In conclusion, the two markers CD24 and CK4 may be considered predictive biomarkers for definitive CRT.
Collapse
Affiliation(s)
- Kenji Takashima
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Chiba 277‑8577, Japan
| | - Satoshi Fujii
- Department of Pathology, Exploratory Oncology Research and Clinical Trial Center, Kashiwa, Chiba 277‑8577, Japan
| | - Rie Komatsuzaki
- Department of Translational Oncology, National Cancer Center Research Institute, Tokyo 104‑0045, Japan
| | - Masayuki Komatsu
- Department of Translational Oncology, National Cancer Center Research Institute, Tokyo 104‑0045, Japan
| | - Mari Takahashi
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Chiba 277‑8577, Japan
| | - Takashi Kojima
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Chiba 277‑8577, Japan
| | - Hiroyuki Daiko
- Department of Esophageal Surgery, National Cancer Center Hospital East, Kashiwa, Chiba 277‑8577, Japan
| | - Keiko Minashi
- Department of Clinical Trial Promotion, Chiba Cancer Center, Chiba 260‑8717, Japan
| | - Fumiko Chiwaki
- Department of Translational Oncology, National Cancer Center Research Institute, Tokyo 104‑0045, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, Kyoto 606‑8507, Japan
| | - Hiroki Sasaki
- Department of Translational Oncology, National Cancer Center Research Institute, Tokyo 104‑0045, Japan
| | - Tomonori Yano
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Chiba 277‑8577, Japan
| |
Collapse
|
36
|
Alvarado-Ortiz E, Sarabia-Sánchez MÁ, García-Carrancá A. Molecular Mechanisms Underlying the Functions of Cellular Markers Associated with the Phenotype of Cancer Stem Cells. Curr Stem Cell Res Ther 2019; 14:405-420. [PMID: 30147013 DOI: 10.2174/1574888x13666180821154752] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/18/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022]
Abstract
Cancer Stem Cells (CSC) generally constitute a minor cellular population within tumors that exhibits some capacities of normal Stem Cells (SC). The existence of CSC, able to self-renew and differentiate, influences central aspects of tumor biology, in part because they can continue tumor growth, give rise to metastasis, and acquire drug and radioresistance, which open new avenues for therapeutics. It is well known that SC constantly interacts with their niche, which includes mesenchymal cells, extracellular ligands, and the Extra Cellular Matrix (ECM). These interactions regularly lead to homeostasis and maintenance of SC characteristics. However, the exact participation of each of these components for CSC maintenance is not clear, as they appear to be context- or cell-specific. In the recent past, surface cellular markers have been fundamental molecular tools for identifying CSC and distinguishing them from other tumor cells. Importantly, some of these cellular markers have been shown to possess functional roles that affect central aspects of CSC. Likewise, some of these markers can participate in regulating the interaction of CSC with their niche, particularly the ECM. We focused this review on the molecular mechanisms of surface cellular markers commonly employed to identify CSC, highlighting the signaling pathways and mechanisms involved in CSC-ECM interactions, through each of the cellular markers commonly used in the study of CSC, such as CD44, CD133, CD49f, CD24, CXCR4, and LGR5. Their presence does not necessarily implicate them in CSC biology.
Collapse
Affiliation(s)
- Eduardo Alvarado-Ortiz
- Programa de Maestría y Doctorado en Ciencias Biológicas, Facultad de Ciencias, Universidad Nacional Autónoma de México, México City, México.,Laboratory of Virus and Cancer, Unidad de Investigacion Biomedica en Cáncer, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico & Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Secretaria de Salud, Ciudad de Mexico, Mexico
| | - Miguel Á Sarabia-Sánchez
- Laboratory of Virus and Cancer, Unidad de Investigacion Biomedica en Cáncer, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico & Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Secretaria de Salud, Ciudad de Mexico, Mexico.,Programa de Maestría y Doctorado en Ciencias Bioquímicas, Facultad de Química, Universidad Nacional Autónoma de México, , México City, México
| | - Alejandro García-Carrancá
- Laboratory of Virus and Cancer, Unidad de Investigacion Biomedica en Cáncer, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico & Subdireccion de Investigacion Basica, Instituto Nacional de Cancerologia, Secretaria de Salud, Ciudad de Mexico, Mexico
| |
Collapse
|
37
|
Barash U, Spyrou A, Liu P, Vlodavsky E, Zhu C, Luo J, Su D, Ilan N, Forsberg-Nilsson K, Vlodavsky I, Yang X. Heparanase promotes glioma progression via enhancing CD24 expression. Int J Cancer 2019; 145:1596-1608. [PMID: 31032901 DOI: 10.1002/ijc.32375] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 04/01/2019] [Accepted: 04/10/2019] [Indexed: 02/05/2023]
Abstract
Heparanase is an endo-β-d-glucuronidase that cleaves heparan sulfate (HS) side chains of heparan sulfate proteoglycans. Compelling evidence tie heparanase levels with all steps of tumor formation including tumor initiation, growth, metastasis and chemo-resistance, likely involving augmentation of signaling pathways and gene transcription. In order to reveal the molecular mechanism(s) underlying the protumorigenic properties of heparanase, we established an inducible (Tet-on) system in U87 human glioma cells and applied gene array methodology in order to identify genes associated with heparanase induction. We found that CD24, a mucin-like cell adhesion protein, is consistently upregulated by heparanase and by heparanase splice variant devoid of enzymatic activity, whereas heparanase gene silencing was associated with decreased CD24 expression. This finding was further substantiated by a similar pattern of heparanase and CD24 immunostaining in glioma patients (Pearson's correlation; R = 0.66, p = 0.00001). Noteworthy, overexpression of CD24 stimulated glioma cell migration, invasion, colony formation in soft agar and tumor growth in mice suggesting that CD24 functions promote tumor growth. Likewise, anti-CD24 neutralizing monoclonal antibody attenuated glioma tumor growth, and a similar inhibition was observed in mice treated with a neutralizing mAb directed against L1 cell adhesion molecule (L1CAM), a ligand for CD24. Importantly, significant shorter patient survival was found in heparanase-high/CD24-high tumors vs. heparanase-high/CD24-low tumors for both high-grade and low-grade glioma (p = 0.02). Our results thus uncover a novel heparanase-CD24-L1CAM axis that plays a significant role in glioma tumorigenesis.
Collapse
Affiliation(s)
- Uri Barash
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Haifa, Israel
| | - Argyris Spyrou
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Pei Liu
- Shantou University Medical College, Shantou, China
| | | | - Chenchen Zhu
- Shantou University Medical College, Shantou, China
| | - Juanjuan Luo
- Shantou University Medical College, Shantou, China
| | - Dongsheng Su
- Shantou University Medical College, Shantou, China
| | - Neta Ilan
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Haifa, Israel
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Israel Vlodavsky
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Haifa, Israel
| | - Xiaojun Yang
- Shantou University Medical College, Shantou, China
| |
Collapse
|
38
|
Farid RM, Sammour SAE, Shehab ElDin ZA, Salman MI, Omran TI. Expression of CD133 and CD24 and their different phenotypes in urinary bladder carcinoma. Cancer Manag Res 2019; 11:4677-4690. [PMID: 31213893 PMCID: PMC6536712 DOI: 10.2147/cmar.s198348] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
Introduction: Several lines of evidence suggest the contribution of cancer stem cells (CSCs) to the tumorigenicity of bladder cancer. Although CD133 and CD24 CSC biomarkers are associated with survival disadvantages in some cancers, the biological attributes of a specific tumor alters the expression of these markers and any associated phenotypic characteristics. Aim: To analyze CD133 and CD24 expression and their different phenotypes in urinary bladder carcinoma. Material and methods: Expression of CD133 and CD24 and their divergent phenotypes were analyzed in patients with urinary bladder carcinoma (n=60) and correlated with clinicopathological parameters. Results: CD133+ and CD24+ tumor cells were more frequent in high grade, less differentiated carcinomas (18/22, and 15/17, p=0.022 and 0.01, respectively), muscle invasive tumors (20/22, p=0.017 and 17/17, p=0.001, respectively), and tumors with advanced stage (p=0.001 and 0.007, respectively). The expression of CD24 slightly correlated with lymphovascular invasion (p=0.04), whereas CD133 was associated with distant metastasis. The CD133+ CD24+ phenotype exhibited more aggressive tumorigenic behavior than other phenotypes. Conclusion: CD133+ and CD24+ cells correlated with determinants of aggressive behavior and may be involved in tumor progression and distant metastasis. The CD133+ subpopulation is likely to have a more potent tumorigenic capacity. Although divergent, the strong correlation between the two populations may support phenotypic plasticity among them. Compared to the CD133+ CD24− and CD133− CD24+ phenotypes, the CD133+ CD24+ phenotype is the most aggressive. These putative biomarkers can potentially aid in the selection of high-risk patients for more aggressive targeted therapy.
Collapse
Affiliation(s)
- Rola M Farid
- Department of Pathology, Ain Shams University, Cairo, Egypt
| | | | | | | | | |
Collapse
|
39
|
Enhanced metastatic potential in the MB49 urothelial carcinoma model. Sci Rep 2019; 9:7425. [PMID: 31092844 PMCID: PMC6520404 DOI: 10.1038/s41598-019-43641-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/12/2019] [Indexed: 01/24/2023] Open
Abstract
Recent data suggest that patients with a basal/stem-like bladder cancer (BC) subtype tend to have metastatic disease, but this is unconfirmed. Here we report the identification of murine MB49 cell line sub-clones with stem-like characteristics in culture. Subcutaneous implantation of S2 and S4 MB49 sub-clones into immunocompetent mice resulted in lung metastases in 50% and 80% of mice respectively, whereas none of the mice implanted with the parental cells developed metastasis. Gene profiling of cells cultured from S2 and S4 primary and metastatic tumors revealed that a panel of genes with basal/stem-like/EMT properties is amplified during metastatic progression. Among them, ITGB1, TWIST1 and KRT6B are consistently up-regulated in metastatic tumors of both MB49 sub-clones. To evaluate clinical relevance, we examined these genes in a human public dataset and found that ITGB1 and KRT6B expression in BC patient tumor samples are positively correlated with tumor grade. Likewise, the expression levels of these three genes are correlated with worse clinical outcomes. This MB49 BC metastatic pre-clinical model provides a unique opportunity to validate and recapitulate results discovered in patient studies and to pursue future mechanistic therapeutic interventions for BC metastasis.
Collapse
|
40
|
Lee YC, Kurtova AV, Xiao J, Nikolos F, Hayashi K, Tramel Z, Jain A, Chen F, Chokshi M, Lee C, Bao G, Zhang X, Shen J, Mo Q, Jung SY, Rowley D, Chan KS. Collagen-rich airway smooth muscle cells are a metastatic niche for tumor colonization in the lung. Nat Commun 2019; 10:2131. [PMID: 31086186 PMCID: PMC6513865 DOI: 10.1038/s41467-019-09878-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 03/27/2019] [Indexed: 01/04/2023] Open
Abstract
Metastases account for the majority of cancer deaths. While certain steps of the metastatic cascade are well characterized, identification of targets to block this process remains a challenge. Host factors determining metastatic colonization to secondary organs are particularly important for exploration, as those might be shared among different cancer types. Here, we showed that bladder tumor cells expressing the collagen receptor, CD167a, responded to collagen I stimulation at the primary tumor to promote local invasion and utilized the same receptor to preferentially colonize at airway smooth muscle cells (ASMCs)—a rich source of collagen III in lung. Morphologically, COL3-CD167a-driven metastatic foci are uniquely distinct from typical lung alveolar metastatic lesions and exhibited activation of the CD167a-HSP90-Stat3 axis. Importantly, metastatic lung colonization could be abrogated using an investigational drug that attenuates Stat3 activity, implicating this seed-and-soil interaction as a therapeutic target for eliminating lung metastasis. Collagen is a dynamic component of both the tumor and metastatic niche. Here, the authors show that airway smooth muscle cells are a collagen III rich niche bladder cancer cells expressing CD167a, and Stat3 is a downstream target for abrogating these collagen III/CD167a-driven metastatic foci.
Collapse
Affiliation(s)
- Yu-Cheng Lee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Antonina V Kurtova
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jing Xiao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Fotis Nikolos
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kazukuni Hayashi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zoe Tramel
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Antrix Jain
- Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Fengju Chen
- Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Mithil Chokshi
- Department of Bioengineering, Rice University Houston, Houston, TX, 77030, USA
| | - Ciaran Lee
- Department of Bioengineering, Rice University Houston, Houston, TX, 77030, USA
| | - Gang Bao
- Department of Bioengineering, Rice University Houston, Houston, TX, 77030, USA
| | - Xiang Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jianjun Shen
- University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Qianxing Mo
- Department of Biostatistics & Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Sung Yun Jung
- Department of Biochemistry and Molecular Biology, Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - David Rowley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Keith Syson Chan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
41
|
Abstract
PI3K is activated in over 60% of human cancers, mediating C-terminal p27 phosphorylation. This work reveals cooperation between PI3K and cJun pathways: p27 phosphorylation by PI3K-activated kinases stimulates p27/cJun corecruitment to chromatin and activation of transcription programs of cell adhesion, motility, TGFB2, and epithelial–mesenchymal transformation to drive tumor progression. Prior analysis showed that high p27pT157 strongly associates with activated AKTpS273 and p90RSKpT359 in human breast cancers. These cancers also differentially express p27/cJun target genes and identify a poor prognostic group. In cancers, the cell cycle-restraining effects of p27 are lost through increased proteolysis and decreased translation. We reveal a previously unknown oncogenic action of p27, in which p27 acts as a cJun coactivator to drive oncogenic gene expression programs. p27 shifts from CDK inhibitor to oncogene when phosphorylated by PI3K effector kinases. Here, we show that p27 is a cJun coregulator, whose assembly and chromatin association is governed by p27 phosphorylation. In breast and bladder cancer cells with high p27pT157pT198 or expressing a CDK-binding defective p27pT157pT198 phosphomimetic (p27CK−DD), cJun is activated and interacts with p27, and p27/cJun complexes localize to the nucleus. p27/cJun up-regulates TGFB2 to drive metastasis in vivo. Global analysis of p27 and cJun chromatin binding and gene expression shows that cJun recruitment to many target genes is p27 dependent, increased by p27 phosphorylation, and activates programs of epithelial–mesenchymal transformation and metastasis. Finally, human breast cancers with high p27pT157 differentially express p27/cJun-regulated genes of prognostic relevance, supporting the biological significance of the work.
Collapse
|
42
|
Anderson RL, Balasas T, Callaghan J, Coombes RC, Evans J, Hall JA, Kinrade S, Jones D, Jones PS, Jones R, Marshall JF, Panico MB, Shaw JA, Steeg PS, Sullivan M, Tong W, Westwell AD, Ritchie JWA. A framework for the development of effective anti-metastatic agents. Nat Rev Clin Oncol 2019; 16:185-204. [PMID: 30514977 PMCID: PMC7136167 DOI: 10.1038/s41571-018-0134-8] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Most cancer-related deaths are a result of metastasis, and thus the importance of this process as a target of therapy cannot be understated. By asking 'how can we effectively treat cancer?', we do not capture the complexity of a disease encompassing >200 different cancer types - many consisting of multiple subtypes - with considerable intratumoural heterogeneity, which can result in variable responses to a specific therapy. Moreover, we have much less information on the pathophysiological characteristics of metastases than is available for the primary tumour. Most disseminated tumour cells that arrive in distant tissues, surrounded by unfamiliar cells and a foreign microenvironment, are likely to die; however, those that survive can generate metastatic tumours with a markedly different biology from that of the primary tumour. To treat metastasis effectively, we must inhibit fundamental metastatic processes and develop specific preclinical and clinical strategies that do not rely on primary tumour responses. To address this crucial issue, Cancer Research UK and Cancer Therapeutics CRC Australia formed a Metastasis Working Group with representatives from not-for-profit, academic, government, industry and regulatory bodies in order to develop recommendations on how to tackle the challenges associated with treating (micro)metastatic disease. Herein, we describe the challenges identified as well as the proposed approaches for discovering and developing anticancer agents designed specifically to prevent or delay the metastatic outgrowth of cancer.
Collapse
Affiliation(s)
- Robin L Anderson
- Translational Breast Cancer Program, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
- Cancer Therapeutics Cooperative Research Centre (CTx), Melbourne, Victoria, Australia
| | - Theo Balasas
- Commercial Partnerships, Cancer Research UK (CRUK), London, UK
| | - Juliana Callaghan
- Research and Innovation Services, University of Portsmouth, Portsmouth, Hampshire, UK
| | - R Charles Coombes
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Jeff Evans
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Jacqueline A Hall
- Research and Development, Vivacitv Ltd, Chesham, Buckinghamshire, UK
| | - Sally Kinrade
- Cancer Therapeutics Cooperative Research Centre (CTx), Melbourne, Victoria, Australia
- Medicines Development for Global Health, Southbank, Victoria, Australia
| | - David Jones
- Medicines and Healthcare Products Regulatory Agency, London, UK
| | | | - Rob Jones
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - John F Marshall
- Queen Mary University of London, Barts Cancer Institute, London, UK
| | | | - Jacqui A Shaw
- Leicester Cancer Research Centre, University of Leicester, Leicester, Leicestershire, UK
| | - Patricia S Steeg
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Mark Sullivan
- Cancer Therapeutics Cooperative Research Centre (CTx), Melbourne, Victoria, Australia
- Medicines Development for Global Health, Southbank, Victoria, Australia
| | - Warwick Tong
- Cancer Therapeutics Cooperative Research Centre (CTx), Melbourne, Victoria, Australia
| | - Andrew D Westwell
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, Wales, UK
| | - James W A Ritchie
- Commercial Partnerships, Cancer Research UK (CRUK), London, UK.
- Centre for Drug Development, CRUK, London, UK.
| |
Collapse
|
43
|
Fafińska J, Czech A, Sitz T, Ignatova Z, Hahn U. DNA Aptamers for the Malignant Transformation Marker CD24. Nucleic Acid Ther 2018; 28:326-334. [PMID: 30407110 DOI: 10.1089/nat.2018.0748] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Cluster of differentiation 24 (CD24) is a cell surface glycoprotein, which is largely present on hematopoietic cells and many types of solid tumor cells. CD24 is known to be involved in a wide range of downstream signaling pathways and neural development, yet the underlying mechanisms are poorly understood. Moreover, its production correlates with poor cancer prognosis, and targeting of CD24 with different antibodies has been shown to inhibit disease progression. Nucleic acid aptamers are oligonucleotides that are selected from random DNA or RNA libraries for high affinity and specific binding to a certain target. Thus, they can be used as an alternative to antibodies. To gain an insight on CD24 role and its interaction partners, we performed several SELEX (systematic evolution of ligands by exponential enrichment) experiments to select CD24-specfiic DNA aptamers. We found that the cell-SELEX approach was the most useful and that using HT-29 cell line presenting CD24 along with CD24 knockdown HT-29 cells has selected six aptamers. For the selected aptamers, we determined dissociation constants in the nanomolar range (18-709 nM) using flow cytometry. These aptamers can be applied as diagnostic tools to track cancer progression and bear a potential for therapeutic use for inhibiting signaling pathways that promote the metastatic process.
Collapse
Affiliation(s)
- Joanna Fafińska
- Hamburg University, MIN Faculty, Chemistry Department, Institute for Biochemistry & Molecular Biology, Hamburg, Germany
| | - Andreas Czech
- Hamburg University, MIN Faculty, Chemistry Department, Institute for Biochemistry & Molecular Biology, Hamburg, Germany
| | - Tobias Sitz
- Hamburg University, MIN Faculty, Chemistry Department, Institute for Biochemistry & Molecular Biology, Hamburg, Germany
| | - Zoya Ignatova
- Hamburg University, MIN Faculty, Chemistry Department, Institute for Biochemistry & Molecular Biology, Hamburg, Germany
| | - Ulrich Hahn
- Hamburg University, MIN Faculty, Chemistry Department, Institute for Biochemistry & Molecular Biology, Hamburg, Germany
| |
Collapse
|
44
|
Chan SH, Tsai KW, Chiu SY, Kuo WH, Chen HY, Jiang SS, Chang KJ, Hung WC, Wang LH. Identification of the Novel Role of CD24 as an Oncogenesis Regulator and Therapeutic Target for Triple-Negative Breast Cancer. Mol Cancer Ther 2018; 18:147-161. [DOI: 10.1158/1535-7163.mct-18-0292] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/12/2018] [Accepted: 10/23/2018] [Indexed: 11/16/2022]
|
45
|
Ooki A, VandenBussche CJ, Kates M, Hahn NM, Matoso A, McConkey DJ, Bivalacqua TJ, Hoque MO. CD24 regulates cancer stem cell (CSC)-like traits and a panel of CSC-related molecules serves as a non-invasive urinary biomarker for the detection of bladder cancer. Br J Cancer 2018; 119:961-970. [PMID: 30327565 PMCID: PMC6203855 DOI: 10.1038/s41416-018-0291-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 09/07/2018] [Accepted: 09/17/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND CD24 is a cornerstone of tumour progression in urothelial carcinoma of the bladder (UCB). However, its contribution to cancer stem cell (CSC)-like traits and the clinical utility of CD24 as a urinary biomarker for cancer detection have not been determined. METHODS The functional relevance of CD24 was evaluated using in vitro and in vivo approaches. The clinical utility of CSC-related molecules was assessed in urine samples by quantitative RT-PCR. RESULTS The knockdown of CD24 attenuated cancer stemness properties. The high-CD24-expressing cells, isolated from patient-derived UCB xenograft tumours, exhibited their enhanced stemness properties. CD24 was overexpressed not only in primary tumours but also in urine from UCB subjects. By assessment of 15 candidate CSC-related molecules in urine samples of a training cohort, a panel of three molecules (CD24, CD49f, and NANOG) was selected. The combination of these three molecules yielded a sensitivity and specificity of 81.7% and 74.3%, respectively, in an independent cohort. A combined set of 84 cases and 207 controls provided a sensitivity and specificity of 82% and 76%, respectively. CONCLUSION CD24 has a crucial role in maintaining the urothelial cancer stem-like traits and a panel of CSC-related molecules has potential as a urinary biomarker for non-invasive UCB detection.
Collapse
Affiliation(s)
- Akira Ooki
- Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | | | - Max Kates
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Noah M Hahn
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Andres Matoso
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231-2410, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - David J McConkey
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Trinity J Bivalacqua
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Mohammad Obaidul Hoque
- Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA.
| |
Collapse
|
46
|
Barkeer S, Chugh S, Batra SK, Ponnusamy MP. Glycosylation of Cancer Stem Cells: Function in Stemness, Tumorigenesis, and Metastasis. Neoplasia 2018; 20:813-825. [PMID: 30015157 PMCID: PMC6037882 DOI: 10.1016/j.neo.2018.06.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/01/2018] [Accepted: 06/11/2018] [Indexed: 02/06/2023] Open
Abstract
Aberrant glycosylation plays a critical role in tumor aggressiveness, progression, and metastasis. Emerging evidence associates cancer initiation and metastasis to the enrichment of cancer stem cells (CSCs). Several universal markers have been identified for CSCs characterization; however, a specific marker has not yet been identified for different cancer types. Specific glycosylation variation plays a major role in the progression and metastasis of different cancers. Interestingly, many of the CSC markers are glycoproteins and undergo differential glycosylation. Given the importance of CSCs and altered glycosylation in tumorigenesis, the present review will discuss current knowledge of altered glycosylation of CSCs and its application in cancer research.
Collapse
Affiliation(s)
- Srikanth Barkeer
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE.
| | - Seema Chugh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
47
|
Zuiverloon TC, de Jong FC, Costello JC, Theodorescu D. Systematic Review: Characteristics and Preclinical Uses of Bladder Cancer Cell Lines. Bladder Cancer 2018; 4:169-183. [PMID: 29732388 PMCID: PMC5929350 DOI: 10.3233/blc-180167] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Bladder cancer (BC) cell lines are indispensable in basic and preclinical research. Currently, an up-to-date and comprehensive overview of available BC cell lines is not available. OBJECTIVE To provide an overview and resources on the origin, pathological and molecular characteristics of commonly used human, murine and canine BC cell lines. METHODS A PubMed search was performed for relevant articles published between 1980 and 2017 according to the following MeSH terms: cell line; cell line, tumor; urinary bladder neoplasms; carcinoma, transitional cell. The Cellosaurus database was searched, using the term "bladder" and/or "urothelial carcinoma". We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. RESULTS We provide information on 157 human, murine and canine BC cell lines. 103 human BC cell lines have molecular data available, of which 69 have been profiled by at least one "omic" technology. We outline how these cell lines are currently being used for in vitro and in vivo experimental models. These results allow direct comparison of BC cell lines to patient samples, providing information needed to make informed decisions on the most genomically appropriate cell line to answer research questions. Furthermore, we show that cross-contamination remains an issue and describe guidelines for prevention. CONCLUSIONS In the BC field, multiple human, murine and canine BC cell lines have been developed and many have become indispensable for in vitro and in vivo research. High-throughput -omic technologies have dramatically increased the amount of molecular data on these cell lines. We synthesized a comprehensive overview of these data as a resource for the BC scientific community.
Collapse
Affiliation(s)
- Tahlita C.M. Zuiverloon
- Department of Urology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Surgery (Urology), University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Comprehensive Cancer Center, Aurora, CO, USA
| | - Florus C. de Jong
- Department of Urology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pathology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - James C. Costello
- University of Colorado Comprehensive Cancer Center, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dan Theodorescu
- Department of Surgery (Urology), University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Comprehensive Cancer Center, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
48
|
Xi J, Chen Y, Huang S, Cui F, Wang X. Suppression of GRP78 sensitizes human colorectal cancer cells to oxaliplatin by downregulation of CD24. Oncol Lett 2018; 15:9861-9867. [PMID: 29805687 PMCID: PMC5958709 DOI: 10.3892/ol.2018.8549] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 01/29/2018] [Indexed: 12/11/2022] Open
Abstract
Glucose-regulated protein 78 (GRP78) is an endoplasmic reticulum stress signaling regulator with anti-apoptotic properties. It has been demonstrated to promote tumor proliferation, survival and metastasis, and to confer resistance against a large variety of therapies. CD24 is a glycosyl-phosphatidylinositol-anchored protein, which is known to have a role in tumor progression, particularly in colorectal cancer (CRC). In the present study, oxaliplatin (L-OHP) was demonstrated to decrease the expression of CD24 in HT29 cells. Knockdown of CD24 using small interfering RNA resulted in sensitization of HT29 cells to L-OHP. By contrast, overexpression of CD24 rendered SW480 cells resistant to L-OHP, which indicated that CD24 antagonized L-OHP-induced cytotoxicity. A co-immunoprecipitation assay revealed that GRP78 physically associates with CD24. L-OHP suppresses the expression of GRP78 and CD24, in part come from the inhibition of interaction between the two. Suppression of GRP78 caused downregulation of CD24 expression and enhanced L-OHP-induced CD24 inhibition. Furthermore, down-regulation of GPR78 with a pharmacological inhibitor sensitized the CRC cells to L-OHP. Collectively, the present results indicate that CD24 antagonizes L-OHP-induced cytotoxicity and that GRP78 is involved in this process. A novel mechanism via which CRC cells acquire resistance to L-OHP was thereby revealed. Use of a combination of compounds which suppress GRP78 may help to improve the effectiveness of L-OHP in the treatment of CRC.
Collapse
Affiliation(s)
- Jingle Xi
- Department of Oncology, Nanfang Hospital, Guangzhou, Guangdong 510515, P.R. China
| | - Yufan Chen
- Department of Orthopaedic Surgery, Nanfang Hospital, Guangzhou, Guangdong 510515, P.R. China
| | - Shangbin Huang
- Department of General Surgery, Taixin Hospital, Dongguan, Guangdong 523000, P.R. China
| | - Fei Cui
- Department of Oncology, Nanfang Hospital, Guangzhou, Guangdong 510515, P.R. China
| | - Xinying Wang
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| |
Collapse
|
49
|
Okabe H, Aoki K, Yogosawa S, Saito M, Marumo K, Yoshida K. Downregulation of CD24 suppresses bone metastasis of lung cancer. Cancer Sci 2017; 109:112-120. [PMID: 29095550 PMCID: PMC5765300 DOI: 10.1111/cas.13435] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/20/2017] [Accepted: 10/28/2017] [Indexed: 01/06/2023] Open
Abstract
Suppression of bone metastasis can improve patient quality of life. Current drugs for bone metastasis have been shown to prolong progression‐free survival but not overall survival; therefore, other potential therapeutic targets for bone metastasis should be investigated. Cell‐surface antigens, such as CD24, have been recently shown to be involved in the metastasis of various cancers. However, whether CD24 plays a role in bone metastasis of lung cancer remains unknown. To observe metastasis of lung cancer cells by imaging technology, we introduced a near‐infrared fluorescent protein, iRFP720, into a bone‐seeking subclone established from lung cancer cells, HARA‐B4 cells. The anchorage‐independent growth of these cells was then evaluated by colony formation assays. We also compared cancer cell tropism to bone tissue with HARA‐B4 cells in the presence or absence of CD24 by cell adhesion assays. To clarify the role of CD24 in bone metastasis, we intracardially injected CD24‐knockdown HARA‐B4 cells into mice and monitored metastasis through detection of iRFP720 using an in vivo imaging system. CD24‐knockdown HARA‐B4 cells in vitro showed reduced anchorage‐independent growth and cancer cell tropism to bone. Bone metastasis was diminished in mice inoculated with CD24‐knockdown HARA‐B4 cells, which was rescued by add‐back of CD24 in cells. Our findings indicate that iRFP720 is effective for in vivo imaging analysis of bone metastasis and that downregulation of CD24 suppresses bone metastasis of lung cancer cells. These findings collectively indicate that CD24 may be considered a promising new therapeutic candidate for the prevention of bone metastasis of lung cancer.
Collapse
Affiliation(s)
- Hinako Okabe
- Department of Biochemistry, Jikei University School of Medicine, Tokyo, Japan.,Department of Orthopedic Surgery, Jikei University School of Medicine, Tokyo, Japan
| | - Katsuhiko Aoki
- Department of Biochemistry, Jikei University School of Medicine, Tokyo, Japan
| | - Satomi Yogosawa
- Department of Biochemistry, Jikei University School of Medicine, Tokyo, Japan
| | - Mitsuru Saito
- Department of Orthopedic Surgery, Jikei University School of Medicine, Tokyo, Japan
| | - Keishi Marumo
- Department of Orthopedic Surgery, Jikei University School of Medicine, Tokyo, Japan
| | - Kiyotsugu Yoshida
- Department of Biochemistry, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
50
|
Arbajian E, Puls F, Antonescu CR, Amary F, Sciot R, Debiec-Rychter M, Sumathi VP, Järås M, Magnusson L, Nilsson J, Hofvander J, Mertens F. In-depth Genetic Analysis of Sclerosing Epithelioid Fibrosarcoma Reveals Recurrent Genomic Alterations and Potential Treatment Targets. Clin Cancer Res 2017; 23:7426-7434. [DOI: 10.1158/1078-0432.ccr-17-1856] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/01/2017] [Accepted: 09/15/2017] [Indexed: 11/16/2022]
|