1
|
Li J, Ye C, Li H, Li J. Targeting the IKZF1/BCL-2 axis as a novel therapeutic strategy for treating acute T-cell lymphoblastic leukemia. Cancer Biol Ther 2025; 26:2457777. [PMID: 39862423 PMCID: PMC11776473 DOI: 10.1080/15384047.2025.2457777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/06/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025] Open
Abstract
OBJECTIVES Acute T-cell lymphoblastic leukemia (T-ALL) is a severe hematologic malignancy with limited treatment options and poor long-term survival. This study explores the role of IKZF1 in regulating BCL-2 expression in T-ALL. METHODS CUT&Tag and CUT&Run assays were employed to assess IKZF1 binding to the BCL-2 promoter. IKZF1 overexpression and knockdown experiments were performed in T-ALL cell lines. The effects of CX-4945 and venetoclax, alone and in combination, were evaluated in vitro and in vivo T-ALL models. RESULTS CUT&Tag sequencing identified IKZF1 binding to the BCL-2 promoter, establishing it as a transcriptional repressor. Functional assays demonstrated that IKZF1 overexpression reduced BCL-2 mRNA levels and increased repressive histone marks at the BCL-2 promoter, while IKZF1 knockdown led to elevated BCL-2 expression. CX-4945, a CK2 inhibitor, could reduced BCL-2 levels in T-ALL cells. Notably, knockdown of IKZF1 partially rescued the CX-4945-induced repression of BCL-2. These results underscore the CK2-IKZF1 signaling axis as a key regulator of BCL-2 expression. In vitro, CX-4945 enhanced the cytotoxicity of venetoclax, with the combination showing significant synergistic effects and increased apoptosis in T-ALL cell lines. In vivo studies with cell line-derived xenograft (CDX) and patient-derived xenograft (PDX) models demonstrated that CX-4945 and venetoclax combined therapy provided superior therapeutic efficacy, reducing tumor burden and prolonging survival compared to single-agent treatments. CONCLUSIONS IKZF1 represses BCL-2 in T-ALL, and targeting the CK2-IKZF1 axis with CX-4945 and venetoclax offers a promising therapeutic strategy, showing enhanced efficacy and potential as a novel treatment approach for T-ALL.
Collapse
Affiliation(s)
- Juan Li
- Department of Hematology, Taixing People’s Hospital Affiliated to Yangzhou University, Taixing, China
- Institute of Hematology, Affiliated hospital of Yangzhou University, Taixing, China
| | - Chunmei Ye
- Department of Hematology, Taixing People’s Hospital Affiliated to Yangzhou University, Taixing, China
- Institute of Hematology, Affiliated hospital of Yangzhou University, Taixing, China
| | - Hui Li
- Department of Hematology, Taixing People’s Hospital Affiliated to Yangzhou University, Taixing, China
| | - Jun Li
- Department of Hematology, Taixing People’s Hospital Affiliated to Yangzhou University, Taixing, China
- Institute of Hematology, Affiliated hospital of Yangzhou University, Taixing, China
| |
Collapse
|
2
|
Ma W, Baran N. Checkpoint kinase 1 as a promising target in colorectal cancer management. World J Clin Oncol 2025; 16:104213. [DOI: 10.5306/wjco.v16.i4.104213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 03/26/2025] Open
Abstract
This editorial provides insights into the pivotal role of checkpoint kinase 1 (CHEK1) as both a biomarker and therapeutic target in colorectal cancer (CRC), based on findings from a recent study by Pang et al. Using single-cell RNA sequencing and immunohistochemistry, the study demonstrates significant CHEK1 overexpression in CRC tissues and identifies nitidine chloride as a potent CHEK1 inhibitor that disrupts DNA damage repair pathways. These findings underscore the therapeutic potential of CHEK1 inhibition and highlight the need for further research to address gaps in CRC treatment.
Collapse
Affiliation(s)
- Wenxue Ma
- Department of Medicine, Sanford Stem Cell Institute, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, United States
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw 00-791, Mazowieckie, Poland
| |
Collapse
|
3
|
Wang Q, Boccalatte F, Xu J, Gambi G, Nadorp B, Akter F, Mullin C, Melnick AF, Choe E, McCarter AC, Jerome NA, Chen S, Lin K, Khan S, Kodgule R, Sussman JH, Pölönen P, Rodriguez-Hernaez J, Narang S, Avrampou K, King B, Tsirigos A, Ryan RJ, Mullighan CG, Teachey DT, Tan K, Aifantis I, Chiang MY. Native stem cell transcriptional circuits define cardinal features of high-risk leukemia. J Exp Med 2025; 222:e20231349. [PMID: 39969525 PMCID: PMC11837855 DOI: 10.1084/jem.20231349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/11/2024] [Accepted: 01/02/2025] [Indexed: 02/20/2025] Open
Abstract
While the mutational landscape across early T-cell precursor acute lymphoblastic leukemia (ETP-ALL) and ETP-like leukemia is known, establishing a unified framework that activates stem cell genes characteristic of these tumors remains elusive. Using complementary mouse and human models, chromatin mapping, and enhancer profiling, we show that the coactivator ZMIZ1 promotes normal and malignant ETP population growth by inducing the transcription factor MYB in feedforward circuits to convergently activate oncogenes (MEF2C, MYCN, and BCL2) through essential enhancers. A key superenhancer, the N-Myc regulating enhancer (NMRE), drives malignant ETP population growth but is dispensable for normal lymphopoiesis. This network of stem cell superenhancers identifies treatment-resistant tumors and poor survival outcomes; unifies diverse ETP-ALLs; and contributes to cardinal features of the recently genomically identified high-risk bone marrow progenitor-like (BMP-like) ETP-ALL tumor-stem cell/myeloid gene expression, inhibited NOTCH1-induced T-cell development, aggressive clinical behavior, and venetoclax sensitivity. Since ZMIZ1 is dispensable for essential homeostasis, it might be possible to safely target this network to treat high-risk diseases.
Collapse
Affiliation(s)
- Qing Wang
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Francesco Boccalatte
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Jason Xu
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Giovanni Gambi
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Bettina Nadorp
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Fatema Akter
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Carea Mullin
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Ashley F. Melnick
- Cellular and Molecular Biology Program, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Elizabeth Choe
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Anna C. McCarter
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Nicole A. Jerome
- Cancer Biology Program, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Siyi Chen
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Karena Lin
- Cellular and Molecular Biology Program, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Sarah Khan
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Rohan Kodgule
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan H. Sussman
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Petri Pölönen
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Javier Rodriguez-Hernaez
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Sonali Narang
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Kleopatra Avrampou
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Bryan King
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Aristotelis Tsirigos
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | | | | | - David T. Teachey
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kai Tan
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Iannis Aifantis
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Mark Y. Chiang
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| |
Collapse
|
4
|
Li T, Wu X, Li X, Chen M. Cancer-associated fungi: An emerging powerful player in cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2025; 1880:189287. [PMID: 39971202 DOI: 10.1016/j.bbcan.2025.189287] [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: 04/30/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
The role of the human microbiome in cancer has been extensively studied, focusing mainly on bacteria-host interactions and their impact on tumor development and treatment response. However, fungi, an immune-active component of the human microbiome, have received less attention regarding their roles in cancer. Recent studies have identified the widespread and specific colonization and distribution of fungi in multiple sites in patients across various cancer types. Importantly, host-fungal immune interactions significantly influence immune regulation within the tumor microenvironment. The rapid advancement of immune-checkpoint blockade (ICB)-based cancer immunotherapy creates an urgent need for effective biomarkers and synergistic therapeutic targets. Cancer-associated fungi and their associated antifungal immunity demonstrate significant potential and efficacy in enhancing cancer immunotherapy. This review summarizes and discusses the growing evidence of the functions and mechanisms of commensal and pathogenic cancer-associated fungi in cancer immunotherapy. Additionally, we emphasize the potential of fungi as predictive biomarkers and therapeutic targets in cancer immunotherapy.
Collapse
Affiliation(s)
- Tianhang Li
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China; Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.
| | - Xiangyu Wu
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiangyang Li
- Department of Gastrointestinal Tumor Surgery, Nanjing Tianyinshan Hospital, Affiliated Hospital of China Pharmaceutical University, Nanjing, China.
| | - Ming Chen
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China; Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.
| |
Collapse
|
5
|
Karimi S, Bakhshali R, Bolandi S, Zahed Z, Mojtaba Zadeh SS, Kaveh Zenjanab M, Jahanban Esfahlan R. For and against tumor microenvironment: Nanoparticle-based strategies for active cancer therapy. Mater Today Bio 2025; 31:101626. [PMID: 40124335 PMCID: PMC11926801 DOI: 10.1016/j.mtbio.2025.101626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Cancer treatment is challenged by the tumor microenvironment (TME), which promotes drug resistance and cancer cell growth. This review offers a comprehensive and innovative perspective on how nanomedicine can modify the TME to enhance therapy. Strategies include using nanoparticles to improve oxygenation, adjust acidity, and alter the extracellular matrix, making treatments more effective. Additionally, nanoparticles can enhance immune responses by activating immune cells and reducing suppression within tumors. By integrating these approaches with existing therapies, such as chemotherapy and radiotherapy, nanoparticles show promise in overcoming traditional treatment barriers. The review discusses how changes in the TME can enhance the effectiveness of nanomedicine itself, creating a reciprocal relationship that boosts overall efficacy. We also highlight novel strategies aimed at exploiting and overcoming the TME, leveraging nanoparticle-based approaches for targeted cancer therapy through precise TME modulation.
Collapse
Affiliation(s)
- Soroush Karimi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | | | - Zahra Zahed
- Department of Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Masoumeh Kaveh Zenjanab
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Jiang X, Ge X, Huang Y, Xie F, Chen C, Wang Z, Tao W, Zeng S, Lv L, Zhan Y, Bao L. Drug resistance in TKI therapy for hepatocellular carcinoma: Mechanisms and strategies. Cancer Lett 2025; 613:217472. [PMID: 39832650 DOI: 10.1016/j.canlet.2025.217472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Tyrosine kinase inhibitors (TKIs) are such as sorafenib the first-line therapeutic drugs for patients with advanced hepatocellular carcinoma. However, patients with TKI-resistant advanced liver cancer are insensitive to TKI treatment, resulting in limited survival benefits. This paper comprehensively reviewed the mechanisms underlying TKI resistance in hepatocytes, investigating activation of tumor signaling pathways, epigenetic regulation, tumor microenvironment, and metabolic reprogramming. Based on resistance mechanisms, it also reviews preclinical and clinical studies of drug resistance strategies and summarizes targeted therapy combined with immunotherapy currently in investigational clinical trials. Understanding the interactions and clinical studies of these resistance mechanisms offers new hope for improving and prolonging patient survival.
Collapse
Affiliation(s)
- Xue Jiang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Xiaoying Ge
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Yueying Huang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Fangyuan Xie
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Chun Chen
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Zijun Wang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Wanru Tao
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Sailiang Zeng
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Lei Lv
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Yangyang Zhan
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| | - Leilei Bao
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200438, China.
| |
Collapse
|
7
|
Perner F, Pahl HL, Zeiser R, Heidel FH. Malignant JAK-signaling: at the interface of inflammation and malignant transformation. Leukemia 2025:10.1038/s41375-025-02569-8. [PMID: 40140631 DOI: 10.1038/s41375-025-02569-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/21/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025]
Abstract
The JAK pathway is central to mammalian cell communication, characterized by rapid responses, receptor versatility, and fine-tuned regulation. It involves Janus kinases (JAK1, JAK2, JAK3, TYK2), which are activated when natural ligands bind to receptors, leading to autophosphorylation and activation of STAT transcription factors [1, 2]. JAK-dependent signaling plays a pivotal role in coordinating cell communication networks across a broad spectrum of biological systems including development, immune responses, cell growth, and differentiation. JAKs are frequently mutated in the aging hematopoietic system [3, 4] and in hematopoietic cancers [5]. Thus, dysregulation of the pathway results in various diseases, including cancers and immune disorders. The binding of extracellular ligands to class I and II cytokine receptors initiates a critical signaling cascade through the activation of Janus kinases (JAKs). Upon ligand engagement, JAKs become activated and phosphorylate specific tyrosine residues on the receptor, creating docking sites for signal transducer and activator of transcription (STAT) proteins. Subsequent JAK-mediated phosphorylation of STATs enables their dimerization and nuclear translocation, where they function as transcription factors to modulate gene expression. Under physiological conditions, JAK-signaling is a tightly regulated mechanism that governs cellular responses to external cues, such as cytokines and growth factors, ensuring homeostasis and maintaining the functional integrity of tissues and organs. Highly defined regulation of JAK-signaling is essential for balancing cellular responses to inflammatory stimuli and growth signals, thus safeguarding tissue health. In contrast, dysregulated JAK-signaling results in chronic inflammation and unrestrained cellular proliferation associated with various diseases. Understanding the qualitative and quantitative differences at the interface of physiologic JAK-signaling and its aberrant activation in disease is crucial for the development of targeted therapies that precisely tune this pathway to target pathologic activation patterns while leaving homeostatic processes largely unaffected. Consequently, pharmaceutical research has targeted this pathway for drug development leading to the approval of several substances with different selectivity profiles towards individual JAKs. Yet, the precise impact of inhibitor selectivity and the complex interplay of different functional modules within normal and malignant cells remains incompletely understood. In this review, we summarize the current knowledge on JAK-signaling in health and disease and highlight recent advances and future directions in the field.
Collapse
Affiliation(s)
- Florian Perner
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Hannover, Germany
| | - Heike L Pahl
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Florian H Heidel
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School (MHH), Hannover, Germany.
- Leibniz-Institute on Aging, Fritz-Lipmann-Institute (FLI), Jena, Germany.
- Cellular Therapy Center (CTC), Hannover Medical School (MHH), Hannover, Germany.
| |
Collapse
|
8
|
Nguyen QTT, Kim J, Yoo HC, Lee EJ. Strategies to overcome chemoresistance in epithelial ovarian cancer: Navigating beyond challenges. Crit Rev Oncol Hematol 2025; 210:104706. [PMID: 40127787 DOI: 10.1016/j.critrevonc.2025.104706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 02/04/2025] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
Epithelial ovarian cancer (EOC) is the most fetal gynecological malignancy. The main causes of treatment failure are primary and acquired chemoresistance that remains a major therapeutic challenge. The mechanisms underlying chemoresistance in EOC are complex and not fully understood. This review explores novel therapeutic strategies targeting chemoresistant EOC, including advanced drug delivery systems, targeting non-coding RNAs, peptide-based therapies, immunotherapy, and the use of poly-ADP ribose polymerase inhibitors. By summarizing the latest research and potential treatments, this review aims to contribute to the development of more effective therapies for patients with chemoresistant EOC.
Collapse
Affiliation(s)
- Que Thanh Thanh Nguyen
- Department of Obstetrics and Gynecology, School of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea; Organoid Medical Research Center, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jihye Kim
- Department of Obstetrics and Gynecology, School of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hee Chan Yoo
- Organoid Medical Research Center, Chung-Ang University, Seoul 06974, Republic of Korea; College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.
| | - Eun-Ju Lee
- Department of Obstetrics and Gynecology, School of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea; Organoid Medical Research Center, Chung-Ang University, Seoul 06974, Republic of Korea.
| |
Collapse
|
9
|
Henriques-Pons A, Castro MCS, Silva VS, Costa MOC, Silva HSIL, Walter MEMT, Carvalho ACC, Melo ACMA, Ocaña K, dos Santos MT, Nicolas MF, Silva FAB. Pulmonary Myeloid Cells in Mild Cases of COVID-19 Upregulate the Intracellular Fc Receptor TRIM21 and Transcribe Proteasome-Associated Molecules. Int J Mol Sci 2025; 26:2769. [PMID: 40141410 PMCID: PMC11943277 DOI: 10.3390/ijms26062769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/17/2025] [Accepted: 02/17/2025] [Indexed: 03/28/2025] Open
Abstract
Much remains to be understood about COVID-19, but the protective role of antibodies (Igs) is widely accepted in SARS-CoV-2 infection. Igs' functions are mainly carried out by receptors that bind to their Fc portion (FcR), and less attention has been dedicated to the cytoplasmic members of this family. In this work, we used single-cell RNA sequencing (scRNA-seq) data to discern cell populations in bronchoalveolar lavage fluid obtained from healthy individuals and patients with mild or severe COVID-19. Then, we evaluated the transcription of neonatal FcR (FcRn, FCGRT gene) and tripartite motif-containing protein 21 (TRIM21) and its downstream signaling components. The TRIM21 pathway is vital for virus infections as it has a dual function, leading opsonized viruses to degradation by proteasomes and the activation of innate inflammatory anti-virus response. The transcriptional level of FCGRT showed no statistical differences in any cell population comparing the three groups of patients. On the other hand, TRIM21 transcription was significantly higher in myeloid cells collected from patients with mild COVID-19. When comparing mild with severe cases, there was no statistical difference in TRIM21 transcription in lung adaptive lymphoid cells and innate lymphoid cells (ILC). Yet, we analyzed the transcription of all downstream signaling molecules in myeloid and, as most cells expressed the receptor, in adaptive lymphoid cells. Moreover, ILCs from mild cases and all cell populations from severe cases were missing most downstream components of the pathway. We observed that members of the ubiquitin-proteasome system (UPS) and other components associated with TRIM21 proteasomal degradation were transcribed in mild cases. Despite the transcription of the danger sensors DDX58 and IFIH1, the transcriptional level of inflammatory IL1B and IL18 was generally very low, along with the NLRP3 danger sensor, members of the NF-κB pathway, and TNF. Therefore, our data suggest that TRIM21 may contribute to SARS-CoV-2 protection by reducing the viral load, while the inflammatory branch of the pathway would be silenced, leading to no pathogenic cytokine production.
Collapse
Affiliation(s)
- Andrea Henriques-Pons
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil;
| | - Maria Clicia S. Castro
- Department of Informatics and Computer Science, State University of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil;
| | - Vanessa S. Silva
- Scientific Computing Program, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil;
| | - Maiana O. C. Costa
- Computational Modeling Department, National Laboratory for Scientific Computing, Petropolis 15651-075, Brazil; (M.O.C.C.); (K.O.); (M.T.d.S.); (M.F.N.)
| | - Helena S. I. L. Silva
- Department of Computer Science, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasilia 70910-900, Brazil; (H.S.I.L.S.); (M.E.M.T.W.); (A.C.M.A.M.)
| | - Maria Emilia M. T. Walter
- Department of Computer Science, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasilia 70910-900, Brazil; (H.S.I.L.S.); (M.E.M.T.W.); (A.C.M.A.M.)
| | - Anna Cristina C. Carvalho
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil;
| | - Alba C. M. A. Melo
- Department of Computer Science, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasilia 70910-900, Brazil; (H.S.I.L.S.); (M.E.M.T.W.); (A.C.M.A.M.)
| | - Kary Ocaña
- Computational Modeling Department, National Laboratory for Scientific Computing, Petropolis 15651-075, Brazil; (M.O.C.C.); (K.O.); (M.T.d.S.); (M.F.N.)
| | - Marcelo T. dos Santos
- Computational Modeling Department, National Laboratory for Scientific Computing, Petropolis 15651-075, Brazil; (M.O.C.C.); (K.O.); (M.T.d.S.); (M.F.N.)
| | - Marisa F. Nicolas
- Computational Modeling Department, National Laboratory for Scientific Computing, Petropolis 15651-075, Brazil; (M.O.C.C.); (K.O.); (M.T.d.S.); (M.F.N.)
| | - Fabrício A. B. Silva
- Scientific Computing Program, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil;
| |
Collapse
|
10
|
Cao C, Yang L, Song J, Liu Z, Li H, Li L, Fu J, Liu J. Cardiomyocyte regeneration after infarction: changes, opportunities and challenges. Mol Cell Biochem 2025:10.1007/s11010-025-05251-w. [PMID: 40097887 DOI: 10.1007/s11010-025-05251-w] [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: 11/14/2024] [Accepted: 03/08/2025] [Indexed: 03/19/2025]
Abstract
Myocardial infarction is a cardiovascular disease that poses a serious threat to human health. The traditional view is that adult mammalian cardiomyocytes have almost no regenerative ability, but recent studies have shown that they have regenerative potential under specific conditions. This article comprehensively describes the research progress of post-infarction cardiomyocyte regeneration, including the characteristics of cardiomyocytes and post-infarction changes, regeneration mechanisms, influencing factors, potential therapeutic strategies, challenges and future development directions, and deeply discusses the specific pathways and targets included in the regeneration mechanism, aiming to provide new ideas and methods for the treatment of myocardial infarction.
Collapse
Affiliation(s)
- Ce Cao
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Lili Yang
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Jianshu Song
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Zixin Liu
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Haoran Li
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Lei Li
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Jianhua Fu
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Jianxun Liu
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China.
| |
Collapse
|
11
|
Peng H, Wang D, Huang S, Yu A. Dual-targeting Aggregation-induced emission polymer micelles mediate immunogenic sonodynamic therapy for Tumor cell growth inhibition and macrophage reprogramming. Acta Biomater 2025; 195:321-337. [PMID: 39900272 DOI: 10.1016/j.actbio.2025.01.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
Sonodynamic therapy (SDT) is a promising cancer treatment known for its deep tumor penetration and high efficacy. However, developing highly efficient sonosensitizers remains a significant challenge. Reports on SDT using aggregation-induced emission luminogens (AIEgens) are rare, highlighting the urgent need for novel AIE-active sonosensitizers. For the first time, we have developed tumor- and macrophage-targeting nano micelles, AIE/Biotin/Mannose-M (ABM-M), utilizing aggregation-induced emission polymers. The ABM-M mediate immunogenic cell death through SDT. By reprogramming tumor-associated macrophages (TAMs), they promote the conversion of M2 macrophages into M1 macrophages, reversing the tumor's immunosuppressive environment. We optimized the ratio of functional molecules to achieve maximum fluorescence intensity and reactive oxygen species (ROS) generation. The multi-targeting nature of ABM-M enables them to bind to relevant antibodies or other molecules, enhancing the capture and presentation of tumor antigens. This, in turn, activates the immune responses of dendritic cells and T cells while inhibiting angiogenesis, creating a more favorable microenvironment for antitumor therapy. Furthermore, ABM-M can be combined with immune checkpoint inhibitors, such as anti-PD-L1 antibodies, to achieve promising outcomes in cancer immunotherapy. The ABM-M nanomaterials offer multi-layered and multi-targeting immune regulation. This study provides a blueprint for developing next-generation cancer diagnostic and therapeutic strategies. STATEMENT OF SIGNIFICANCE: Our research pioneers the use of nanomicelles to simultaneously target both tumor cells and tumor-associated macrophages (TAMs), integrated with sonodynamic therapy. Through precise ratio adjustments, we engineered nanomicelles capable of multi-target regulation. These micelles uniquely induce immunogenic cell death (ICD) and repolarize macrophages from an immunosuppressive M2 phenotype to an immunostimulatory M1 phenotype, reversing the tumor's immunosuppressive microenvironment. This dual mechanism can be enhanced by combining with immune checkpoint inhibitors, such as anti-PD-L1 antibodies, offering a promising strategy to treat refractory cancers. Extensive in vitro and in vivo validation confirms their therapeutic potential, providing a solid foundation for clinical application. This innovative approach shows significant promise for revolutionizing cancer treatment and improving patient outcomes.
Collapse
Affiliation(s)
- Haiheng Peng
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430070, China
| | - Dandan Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Shiwen Huang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430070, China; Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, China.
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430070, China.
| |
Collapse
|
12
|
Koh MY, Chung TH, Tang NXN, Toh SHM, Zhou J, Tan TK, Chen L, Chng WJ, Teoh PJ. The ADAR1-regulated cytoplasmic dsRNA-sensing pathway is a novel mechanism of lenalidomide resistance in multiple myeloma. Blood 2025; 145:1164-1181. [PMID: 39652772 PMCID: PMC11923435 DOI: 10.1182/blood.2024024429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/31/2024] [Indexed: 03/14/2025] Open
Abstract
ABSTRACT Immunomodulatory drugs (IMiDs) are a major class of drugs for treating multiple myeloma (MM); however, acquired resistance to IMiDs remains a significant clinical challenge. Although alterations in cereblon and its pathway are known to contribute to IMiD resistance, they account for only 20% to 30% of cases, and the underlying mechanisms in the majority of the resistance cases remain unclear. Here, we identified adenosine deaminase acting on RNA1 (ADAR1) as a novel driver of lenalidomide resistance in MM. We showed that lenalidomide activates the MDA5-mediated double-stranded RNA (dsRNA)-sensing pathway in MM cells, leading to interferon (IFN)-mediated apoptosis, with ADAR1 as the key regulator. Mechanistically, ADAR1 loss increased lenalidomide sensitivity through endogenous dsRNA accumulation, which in turn triggered dsRNA-sensing pathways and enhanced IFN responses. Conversely, ADAR1 overexpression reduced lenalidomide sensitivity, attributed to increased RNA editing frequency, reduced dsRNA accumulation, and suppression of the dsRNA-sensing pathways. In summary, we report the involvement of ADAR1-regulated dsRNA sensing in modulating lenalidomide sensitivity in MM. These findings highlight a novel RNA-related mechanism underlying lenalidomide resistance and underscore the potential of targeting ADAR1 as a novel therapeutic strategy.
Collapse
Affiliation(s)
- Mun Yee Koh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Tae-Hoon Chung
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Nicole Xin Ning Tang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Sabrina Hui Min Toh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Jianbiao Zhou
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Tze King Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Leilei Chen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wee Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Hematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Phaik Ju Teoh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
13
|
Luo H, Sun Y, Xu T. Application status and research progress of targeted therapy drugs for hormone receptor-positive breast cancer. Front Med (Lausanne) 2025; 12:1513836. [PMID: 40134916 PMCID: PMC11933059 DOI: 10.3389/fmed.2025.1513836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/10/2025] [Indexed: 03/27/2025] Open
Abstract
Breast cancer (BC) is the most common malignant tumor in women and the leading cause of cancer-related deaths in women. As one of the most common subtypes of breast cancer, patients with hormone receptor-positive (HR+) breast cancer usually experience disease progression over an extended period of time, triggering the search for therapeutic strategies other than endocrine therapy. In recent years, continuous research on various targets has led to dramatic changes in the treatment of hormone receptor-positive breast cancer patients, resulting in prolonged clinical survival. With the redefinition of human epidermal growth factor-2 (HER2) expression, more precise and individualized treatment is possible. This review comprehensively reviews targeted therapies and critical clinical trials for HR+ breast cancer and tracks the latest advances. It also provides valuable insights into the future direction of targeted therapies.
Collapse
Affiliation(s)
- Han Luo
- Department of Breast Surgery, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, China
| | - Yue Sun
- Department of Breast Surgery, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, China
| | - Tiefeng Xu
- Department of Breast Surgery, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, China
- Department of Breast Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
14
|
Herbert A, Cherednichenko O, Lybrand TP, Egli M, Poptsova M. Zα and Zβ Localize ADAR1 to Flipons That Modulate Innate Immunity, Alternative Splicing, and Nonsynonymous RNA Editing. Int J Mol Sci 2025; 26:2422. [PMID: 40141064 PMCID: PMC11942513 DOI: 10.3390/ijms26062422] [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: 01/10/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
The double-stranded RNA editing enzyme ADAR1 connects two forms of genetic programming, one based on codons and the other on flipons. ADAR1 recodes codons in pre-mRNA by deaminating adenosine to form inosine, which is translated as guanosine. ADAR1 also plays essential roles in the immune defense against viruses and cancers by recognizing left-handed Z-DNA and Z-RNA (collectively called ZNA). Here, we review various aspects of ADAR1 biology, starting with codons and progressing to flipons. ADAR1 has two major isoforms, with the p110 protein lacking the p150 Zα domain that binds ZNAs with high affinity. The p150 isoform is induced by interferon and targets ALU inverted repeats, a class of endogenous retroelement that promotes their transcription and retrotransposition by incorporating Z-flipons that encode ZNAs and G-flipons that form G-quadruplexes (GQ). Both p150 and p110 include the Zβ domain that is related to Zα but does not bind ZNAs. Here we report strong evidence that Zβ binds the GQ that are formed co-transcriptionally by ALU repeats and within R-loops. By binding GQ, ADAR1 suppresses ALU-mediated alternative splicing, generates most of the reported nonsynonymous edits and promotes R-loop resolution. The recognition of the various alternative nucleic acid conformations by ADAR1 connects genetic programming by flipons with the encoding of information by codons. The findings suggest that incorporating G-flipons into editmers might improve the therapeutic editing efficacy of ADAR1.
Collapse
Affiliation(s)
- Alan Herbert
- Discovery, InsideOutBio, Charlestown, MA 02129, USA
| | - Oleksandr Cherednichenko
- International Laboratory of Bioinformatics, HSE University, 101000 Moscow, Russia; (O.C.); (M.P.)
| | - Terry P. Lybrand
- Department of Chemistry, School of Medicine, Vanderbilt University, Nashville, TN 37232-0146, USA;
- Center for Structural Biology, School of Medicine, Vanderbilt University, Nashville, TN 37232-0146, USA
| | - Martin Egli
- Department of Biochemistry, School of Medicine, Vanderbilt University, Nashville, TN 37232-0146, USA;
| | - Maria Poptsova
- International Laboratory of Bioinformatics, HSE University, 101000 Moscow, Russia; (O.C.); (M.P.)
| |
Collapse
|
15
|
Wu S, Bu X, Chen D, Wu X, Wu H, Caiyin Q, Qiao J. Molecules-mediated bidirectional interactions between microbes and human cells. NPJ Biofilms Microbiomes 2025; 11:38. [PMID: 40038292 PMCID: PMC11880406 DOI: 10.1038/s41522-025-00657-2] [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: 10/06/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
Complex molecules-mediated interactions, which are based on the bidirectional information exchange between microbes and human cells, enable the defense against diseases and health maintenance. Recently, diverse single-direction interactions based on active metabolites, immunity factors, and quorum sensing signals have largely been summarized separately. In this review, according to a simplified timeline, we proposed the framework of Molecules-mediated Bidirectional Interactions (MBI) between microbe and humans to decipher and understand their intricate interactions systematically. About the microbe-derived interactions, we summarized various molecules, such as short-chain fatty acids, bile acids, tryptophan catabolites, and quorum sensing molecules, and their corresponding human receptors. Concerning the human-derived interactions, we reviewed the effect of human molecules, including hormones, cytokines, and other circulatory metabolites on microbial characteristics and phenotypes. Finally, we discussed the challenges and trends for developing and deciphering molecule-mediated bidirectional interactions and their potential applications in the guard of human health.
Collapse
Affiliation(s)
- Shengbo Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China
| | - Xueying Bu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
| | - Danlei Chen
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China
| | - Xueyan Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
| | - Hao Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China.
| | - Qinggele Caiyin
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China.
- Key Laboratory of Systems Bioengineering, Ministry of Education (Tianjin University), Tianjin, 300072, China.
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300072, China.
| | - Jianjun Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China.
- Key Laboratory of Systems Bioengineering, Ministry of Education (Tianjin University), Tianjin, 300072, China.
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300072, China.
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
16
|
Yang J, Zhou F, Luo X, Fang Y, Wang X, Liu X, Xiao R, Jiang D, Tang Y, Yang G, You L, Zhao Y. Enhancer reprogramming: critical roles in cancer and promising therapeutic strategies. Cell Death Discov 2025; 11:84. [PMID: 40032852 DOI: 10.1038/s41420-025-02366-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 01/24/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025] Open
Abstract
Transcriptional dysregulation is a hallmark of cancer initiation and progression, driven by genetic and epigenetic alterations. Enhancer reprogramming has emerged as a pivotal driver of carcinogenesis, with cancer cells often relying on aberrant transcriptional programs. The advent of high-throughput sequencing technologies has provided critical insights into enhancer reprogramming events and their role in malignancy. While targeting enhancers presents a promising therapeutic strategy, significant challenges remain. These include the off-target effects of enhancer-targeting technologies, the complexity and redundancy of enhancer networks, and the dynamic nature of enhancer reprogramming, which may contribute to therapeutic resistance. This review comprehensively encapsulates the structural attributes of enhancers, delineates the mechanisms underlying their dysregulation in malignant transformation, and evaluates the therapeutic opportunities and limitations associated with targeting enhancers in cancer.
Collapse
Affiliation(s)
- Jinshou Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Feihan Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Xiyuan Luo
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Yuan Fang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Xing Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Xiaohong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Ruiling Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Decheng Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Yuemeng Tang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China.
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, PR China.
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing, PR China.
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing, PR China.
| |
Collapse
|
17
|
Mi Y, Jiang P, Luan J, Feng L, Zhang D, Gao X. Peptide‑based therapeutic strategies for glioma: Current state and prospects. Peptides 2025; 185:171354. [PMID: 39922284 DOI: 10.1016/j.peptides.2025.171354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Glioma is a prevalent form of primary malignant central nervous system tumor, characterized by its cellular invasiveness, rapid growth, and the presence of the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB). Current therapeutic approaches, such as chemotherapy and radiotherapy, have shown limited efficacy in achieving significant antitumor effects. Therefore, there is an urgent demand for new treatments. Therapeutic peptides represent an innovative class of pharmaceutical agents with lower immunogenicity and toxicity. They are easily modifiable via chemical means and possess deep tissue penetration capabilities which reduce side effects and drug resistance. These unique pharmacokinetic characteristics make peptides a rapidly growing class of new therapeutics that have demonstrated significant progress in glioma treatment. This review outlines the efforts and accomplishments in peptide-based therapeutic strategies for glioma. These therapeutic peptides can be classified into four types based on their anti-tumor function: tumor-homing peptides, inhibitor/antagonist peptides targeting cell surface receptors, interference peptides, and peptide vaccines. Furthermore, we briefly summarize the results from clinical trials of therapeutic peptides in glioma, which shows that peptide-based therapeutic strategies exhibit great potential as multifunctional players in glioma therapy.
Collapse
Affiliation(s)
- Yajing Mi
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Pengtao Jiang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Jing Luan
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Lin Feng
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Dian Zhang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Xingchun Gao
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China.
| |
Collapse
|
18
|
Zhang F, Li Z, Fang F, Hu Y, He Z, Tao Y, Li Y, Zhang Z, Zhou B, Yang Y, Wu Y, Wu Y, Wei Z, Guo A, Xu L, Zhang Y, Li X, Li Y, Yang C, Zhou M, Pan J, Hu S, Yang X. IRF1 is a core transcriptional regulatory circuitry member promoting AML progression by regulating lipid metabolism. Exp Hematol Oncol 2025; 14:25. [PMID: 40025540 PMCID: PMC11871635 DOI: 10.1186/s40164-025-00612-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/11/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a prevalent malignancy of the hematologic system. Despite advancements in therapeutic approaches, significant heterogeneity and therapeutic resistance pose substantial challenges to treatment. Tumors driven by core transcription factors through super-enhancers can establish core transcriptional regulatory circuits (CRCs) that modulate oncogene expression programs. Identifying CRC is crucial for understanding disease-related transcriptional regulation. This study sought to predict and establish a CRC model for AML, identify genes critical for AML survival and explore their regulatory mechanisms in AML progression. METHODS The dbCoRC tool was used for predictive analysis of H3K27ac ChIP-seq data from 11 AML samples to construct and validate the CRC model in AML patients. To elucidate the functional role of the CRC member IRF1, we utilized short hairpin RNA (shRNA) to knock down IRF1 in AML cells. RNA-seq, CUT&Tag and lipidomics technologies were subsequently used to investigate the regulatory roles and downstream mechanisms of IRF1 in AML. RESULTS This study established a core transcriptional regulatory circuit consisting of IRF1, ELF1, ETV6, RUNX2, and MEF2D, which formed an interconnected autoregulatory loop. Further investigations revealed up-regulated expression of IRF1 in AML patients, which was associated with poor prognosis. Inhibition of IRF1 expression resulted in decreased AML cell proliferation and induced apoptosis, indicating its essential role in the survival of AML cells. Additionally, this study revealed that IRF1 directly regulates the transcription of key genes such as FASN, SCD, and SREBF1 for lipid synthesis, thereby affecting lipid metabolism in AML cells. CONCLUSION In summary, this study identified IRF1 as a novel core transcription factor involved in AML pathogenesis. IRF1 collaborates with ELF1, ETV6, RUNX2, and MEF2D to form a core transcriptional regulatory circuit that promotes AML progression. Furthermore, we demonstrated that IRF1 directly regulates the expression of key genes involved in lipid metabolism, influencing the synthesis of diverse lipid molecules crucial for AML survival.
Collapse
Affiliation(s)
- Fenli Zhang
- Department of Pediatrics, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, 550000, Guizhou, China
| | - Zhiheng Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Fang Fang
- Institute of Pediatric Research, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Yixin Hu
- Department of Hematology, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, Jiangsu, China
| | - Zhixu He
- Department of Pediatrics, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, 550000, Guizhou, China
| | - Yanfang Tao
- Department of Traditional Chinese Medicine, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Yizhen Li
- Department of Hematology, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, Jiangsu, China
- Pediatric Hematology & Oncology Key Laboratory of Higher Education Institutions in Jiangsu Province, Suzhou, 215003, China
| | - Zimu Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Bi Zhou
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Department of Pediatrics, Suzhou Hospital of Anhui Medical University, Suzhou, 234000, China
| | - Ying Yang
- Department of Pediatrics, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, 550000, Guizhou, China
| | - Yumeng Wu
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Department of Pediatrics, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233004, China
| | - Yijun Wu
- Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Zhongling Wei
- Department of Hematology, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, Jiangsu, China
| | - Ailian Guo
- Department of Hematology, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, Jiangsu, China
| | - Ling Xu
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Department of Pediatrics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Yongping Zhang
- Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Xiaolu Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, China
| | - Yan Li
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Department of Pediatrics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Chunxia Yang
- Department of Pediatrics, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, 550000, Guizhou, China
| | - Man Zhou
- Department of Pediatrics, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, 550000, Guizhou, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, China.
| | - Shaoyan Hu
- Department of Hematology, Children's Hospital of Soochow University, No.92 Zhongnan Street, SIP, Suzhou, 215003, Jiangsu, China.
| | - Xiaoyan Yang
- Department of Pediatrics, Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, 550000, Guizhou, China.
| |
Collapse
|
19
|
Cheng YC, Chen MY, Yadav VK, Pikatan NW, Fong IH, Kuo KT, Yeh CT, Tsai JT. Targeting FABP4/UCP2 axis to overcome cetuximab resistance in obesity-driven CRC with drug-tolerant persister cells. Transl Oncol 2025; 53:102274. [PMID: 39823981 PMCID: PMC11787020 DOI: 10.1016/j.tranon.2025.102274] [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: 08/18/2024] [Revised: 11/24/2024] [Accepted: 01/05/2025] [Indexed: 01/20/2025] Open
Abstract
Colorectal cancer (CRC) is closely linked to obesity, a condition that significantly impacts tumor progression and therapeutic resistance. Although cetuximab, an EGFR-targeting monoclonal antibody, is a cornerstone in metastatic CRC treatment, resistance often emerges, leading to poor outcomes. This study investigated the role of drug-tolerant persister (DTP) cells and their metabolic interactions within the tumor microenvironment (TME) in cetuximab resistance. Using patient-derived organoids and in vivo models, we identified the FABP4/UCP2 axis as a critical mediator of resistance. Organoids derived from cetuximab non-responders revealed upregulated FABP4 and UCP2 expression post-treatment. Coculture experiments with adipocytes showed that FABP4 and UCP2 promote lipid metabolic reprogramming, facilitating cancer cell survival in a dormant state. CRISPR/Cas9 mediated inhibition of FABP4 disrupted this metabolic interaction, sensitising resistant cells to cetuximab. In vivo, the FABP4 inhibitor BMS309403, either alone or in combination with cetuximab, significantly reduced tumor growth in resistant CRC models, highlighting its therapeutic potential. These findings establish the FABP4/UCP2 axis as a pivotal driver of cetuximab resistance in obesity-associated CRC and suggest that targeting this metabolic pathway could improve outcomes in DTP-resistant CRC patients.
Collapse
Affiliation(s)
- Yi-Chiao Cheng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Division of Colon and Rectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Ming-Yao Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang Ho Hospital, New Taipei City 23561, Taiwan
| | - Vijesh Kumar Yadav
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang Ho Hospital, New Taipei City 23561, Taiwan
| | - Narpati Wesa Pikatan
- Division of Urology, Department of Surgery, Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Iat-Hang Fong
- Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan; Continuing Education Program of Food Biotechnology Applications, College of Science and Engineering, National Taitung University, Taitung 95092, Taiwan
| | - Kuang-Tai Kuo
- Department of Surgery, Division of Thoracic Surgery, Taipei Medical University Shuang-Ho Hospital, New Taipei City 23561, Taiwan
| | - Chi-Tai Yeh
- Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan; Continuing Education Program of Food Biotechnology Applications, College of Science and Engineering, National Taitung University, Taitung 95092, Taiwan.
| | - Jo-Ting Tsai
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Department of Radiology, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan.
| |
Collapse
|
20
|
Antonsen KW, Jensen AG, Sorensen BS, Etzerodt A, Moestrup SK, Møller HJ. In vitro ovarian tumor-conditioned CD163+ human macrophages retain phagocytic response to CD47 blockade. Cell Immunol 2025; 409-410:104932. [PMID: 39985839 DOI: 10.1016/j.cellimm.2025.104932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/02/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
INTRODUCTION CD163-expressing macrophages are abundant in ovarian cancer where they accelerate tumor growth and metastasis. CD47 blockade is a novel immunotherapy aiming to activate macrophage phagocytosis of tumor cells, but it is currently unknown if the tumor-associated macrophages expressing CD163 respond poorly to CD47 blockade. METHODS Human monocyte-derived macrophages were exposed to tumor-conditioned medium from A2780 ovarian cancer cells during differentiation. Effects on gene expression, membrane protein levels, release of soluble proteins and macrophage phagocytosis of A2780 cells in response to CD47 blockade were measured and compared to control macrophages. RESULTS Tumor cell conditioning induced macrophage expression of CD163 on both the mRNA and protein level. Furthermore, tumor conditioning simultaneously increased protein expression of the phenotype markers CD206 and CD80, and the phagocytosis checkpoint LILRB1. However, tumor conditioning did not reduce phagocytic capacity, as CD47 blockade induced macrophage phagocytosis of A2780 cells to similar degrees in both control and tumor cell-conditioned macrophages. DISCUSSION In vitro tumor conditioning did not reduce the phagocytic response to CD47 blockade, suggesting that induction of a macrophage phenotype with increased expression of CD163 does not directly limit the capacity for phagocytosis of tumor cells. In conclusion, these findings suggest that CD163+ macrophages remain responsive to CD47 blockade, highlighting their potential as targets for immunotherapy in ovarian cancer.
Collapse
Affiliation(s)
- Kristian W Antonsen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Anne G Jensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Boe S Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Holger J Møller
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
21
|
Shi Z, Li J, Ding J, Zhang Y, Min W, Zhu Y, Hou Y, Yuan K, Sun C, Wang X, Shen H, Wang L, Liang SQ, Kuang W, Wang X, Yang P. ADAR1 is required for acute myeloid leukemia cell survival by modulating post-transcriptional Wnt signaling through impairing miRNA biogenesis. Leukemia 2025; 39:599-613. [PMID: 39702795 DOI: 10.1038/s41375-024-02500-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024]
Abstract
Recent extensive studies on the genomic and molecular profiles of acute myeloid leukemia (AML) have expanded the treatment options, including, a range of compounds represented by fms-like tyrosine kinase 3 and isocitrate dehydrogenase 1/2 inhibitors. However, despite this progress, further treatments for AML are still required. Adenosine deaminase acting on RNA 1 (ADAR1) has been shown to play an important oncogenic role in many cancers, but its involvement in AML progression remains underexplored. In this study, we demonstrated that ADAR1 was overexpressed in AML and served as a crucial oncogenic target. Loss of ADAR1 inhibited the Wnt signaling pathway, blocked AML cell proliferation, and induced apoptosis. Importantly, we demonstrate that ADAR1, as an RNA-binding protein, interacts with pri-miR-766 independently of its editing function, regulating the maturation of miR-766-3p and enhancing the expression of WNT5B. Genetic inhibition or use of the ADAR1 inhibitor ZYS-1 significantly suppressed AML cell growth both in vitro and in vivo. Overall, these results elucidated the tumorigenic mechanism of ADAR1 and validated it as a potential drug target in AML.
Collapse
Affiliation(s)
- Zhongrui Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiaxing Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiayu Ding
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yiwen Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wenjian Min
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yasheng Zhu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yi Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kai Yuan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chengliang Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xuejiao Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hao Shen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Liping Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shun-Qing Liang
- Department of Medicine, University of Minnesota Twin Cities, Minneapolis, MN, USA.
| | - Wenbin Kuang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Xiao Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, China.
| | - Peng Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
22
|
Leck LYW, Abd El-Aziz YS, McKelvey KJ, Park KC, Sahni S, Lane DJR, Skoda J, Jansson PJ. Cancer stem cells: Masters of all traits. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167549. [PMID: 39454969 DOI: 10.1016/j.bbadis.2024.167549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Cancer is a heterogeneous disease, which contributes to its rapid progression and therapeutic failure. Besides interpatient tumor heterogeneity, tumors within a single patient can present with a heterogeneous mix of genetically and phenotypically distinct subclones. These unique subclones can significantly impact the traits of cancer. With the plasticity that intratumoral heterogeneity provides, cancers can easily adapt to changes in their microenvironment and therapeutic exposure. Indeed, tumor cells dynamically shift between a more differentiated, rapidly proliferating state with limited tumorigenic potential and a cancer stem cell (CSC)-like state that resembles undifferentiated cellular precursors and is associated with high tumorigenicity. In this context, CSCs are functionally located at the apex of the tumor hierarchy, contributing to the initiation, maintenance, and progression of tumors, as they also represent the subpopulation of tumor cells most resistant to conventional anti-cancer therapies. Although the CSC model is well established, it is constantly evolving and being reshaped by advancing knowledge on the roles of CSCs in different cancer types. Here, we review the current evidence of how CSCs play a pivotal role in providing the many traits of aggressive tumors while simultaneously evading immunosurveillance and anti-cancer therapy in several cancer types. We discuss the key traits and characteristics of CSCs to provide updated insights into CSC biology and highlight its implications for therapeutic development and improved treatment of aggressive cancers.
Collapse
Affiliation(s)
- Lionel Y W Leck
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Yomna S Abd El-Aziz
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Kelly J McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Kyung Chan Park
- Proteina Co., Ltd./Seoul National University, Seoul, South Korea
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
| | - Patric J Jansson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
23
|
Chen J, Ma N, Chen B, Huang Y, Li J, Li J, Chen Z, Wang P, Ran B, Yang J, Bai J, Ning S, Ai J, Wei Q, Liu L, Cao D. Synergistic effects of immunotherapy and adjunctive therapies in prostate cancer management. Crit Rev Oncol Hematol 2025; 207:104604. [PMID: 39732304 DOI: 10.1016/j.critrevonc.2024.104604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024] Open
Abstract
In recent years, cancer immunotherapy has received widespread attention due to significant tumor clearance in some malignancies. Various immunotherapy approaches, including vaccines, immune checkpoint inhibitors, oncolytic virotherapy, bispecific T cell engagers, and adoptive T cell transfer, have completed or are undergoing clinical trials for prostate cancer. Despite immune checkpoint blockade's extraordinary effectiveness in treating a variety of cancers, targeted prostate cancer treatment using the immune system is still in its infancy. Multiple factors including the heterogeneity of prostate cancer, the cold tumor microenvironment, and a low level of neoantigens, contribute to the poor immunotherapy response. Significant effort is being devoted to improving immune-based prostate cancer therapy. Recently, several key discoveries demonstrate that prostate cancer immunotherapy agents may be used to promise better prognosis for patients as part of combination strategies with other agents targeting tumor-associated immune mechanism of resistance. Here, this review comprehensively examines the recent advancements in immunotherapy for prostate cancer, exploring its potential synergistic effects when combined with other treatment modalities to enhance clinical efficacy.
Collapse
Affiliation(s)
- Jie Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Ma
- Department of Pediatrics, West China Second University Hospital, Sichuan University, No. 20, 3rd section, South Renmin Road, Chengdu 610041, China
| | - Bo Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yin Huang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinze Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zeyu Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Puze Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Biao Ran
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiahao Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingxing Bai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shu Ning
- Department of Urologic Surgery, University of California Davis, Davis, CA, USA
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liangren Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Dehong Cao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
24
|
Certa F, Horn PA, Keyl J, Mende B, Lueong S, Hilser T, Theurer S, Virchow I, Zaun Y, Pogorzelski M, Metzenmacher M, Kalkavan H, Kasper S, Schuler M, Wiesweg M, Zaun G. ABO-Blood Group Associates With Survival Outcomes in Patients With Metastatic Non-Small Cell Lung Cancer Treated With Pembrolizumab Monotherapy. Thorac Cancer 2025; 16:e70037. [PMID: 40114329 PMCID: PMC11925720 DOI: 10.1111/1759-7714.70037] [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: 01/21/2025] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025] Open
Abstract
PURPOSE In patients with metastatic non-small cell lung cancer (NSCLC) with high programmed death-ligand 1 (PD-L1) expression, there is still a lack of biomarkers to identify patients with maximum benefit from first-line treatment with checkpoint inhibitor therapy (CIT) alone. This work examines the impact of different ABO blood groups (BG) on the response to CIT monotherapy. METHODS Retrospective analysis of patients with stage IV NSCLC and high PD-L1 expression (tumor proportional score/TPS ≥ 50%), receiving first-line therapy with pembrolizumab alone or in combination with chemotherapy at the West German Cancer Center from 2017 to 2022. Study endpoints were overall survival (OS) and progression-free survival (PFS). RESULTS Eighty-two patients were included in the analysis. Twenty-two patients (27%) received first-line therapy with pembrolizumab alone (monoimmunotherapy cohort/MIC), of which seven patients (32%) had BGO. Sixty patients (73%) were treated with pembrolizumab combined with platinum-based chemotherapy (chemoimmunotherapy cohort/CIC), of which 38 (63%) had BGO. In MIC, younger age and BGO were independent predictors of favorable OS (BGO vs. other ABO-BG: HR 0.22, 95% CI: 0.1-0.9; p = 0.037; median OS 62 versus 19 months) and PFS (BGO vs. other ABO-BG: HR 0.21, 95% CI: 0.1-0.8; p = 0.024; median PFS 39 vs. 4 months). There was no significant impact of ABO-BG in patients treated with CIC. In support, a historical control group treated with chemotherapy alone also showed no prognostic impact of the ABO-BG. CONCLUSION BGO associates with favorable survival in patients with NSCLC receiving pembrolizumab monotherapy, but not in patients with chemo-immunotherapy or chemotherapy. Further validation of this promising strategy for personalized decision-making is warranted.
Collapse
Affiliation(s)
- Franziska Certa
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - Peter A Horn
- Medical Faculty, University Duisburg-Essen, Essen, Germany
- Institute for Transfusion Medicine, University Hospital of Essen, Essen, Germany
| | - Julius Keyl
- Medical Faculty, University Duisburg-Essen, Essen, Germany
- West German Cancer Center, Institute of Pathology Essen, University Hospital Essen, Essen, Germany
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| | - Bastian Mende
- Medical Faculty, University Duisburg-Essen, Essen, Germany
- Central Pharmacy, University Hospital Essen, Essen, Germany
| | - Smiths Lueong
- Medical Faculty, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
- West German Cancer Center, Institute for Developmental Cancer Therapeutics, University Hospital Essen, Essen, Germany
| | - Thomas Hilser
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - Sarah Theurer
- Medical Faculty, University Duisburg-Essen, Essen, Germany
- West German Cancer Center, Institute of Pathology Essen, University Hospital Essen, Essen, Germany
| | - Isabel Virchow
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - Yasmin Zaun
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - Michael Pogorzelski
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - Martin Metzenmacher
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - Halime Kalkavan
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
- National Center for Tumor Diseases (NCT) West, Essen, Germany
| | - Stefan Kasper
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
- National Center for Tumor Diseases (NCT) West, Essen, Germany
| | - Martin Schuler
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
- National Center for Tumor Diseases (NCT) West, Essen, Germany
| | - Marcel Wiesweg
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
- National Center for Tumor Diseases (NCT) West, Essen, Germany
| | - Gregor Zaun
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
25
|
Gui Y, Wu H, Fan H. Pseudostellaria heterophylla (Miq.) Pax, a traditional folk medicine, ameliorates colorectal cancer by remodeling the tumor immune microenvironment. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119362. [PMID: 39828144 DOI: 10.1016/j.jep.2025.119362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pseudostellaria heterophylla (Miq.) Pax (PH) is a traditional folk medicine, which is widely used clinically for digestive system tumors such as esophageal, gastric, colorectal, and liver cancers. The anti-tumor effect and mechanism of PH in colorectal cancer (CRC) deserves further study. AIM OF THE STUDY The objective of this study is to examine the effects and the underlying mechanisms of aqueous extract of Pseudostellaria heterophylla (Miq.) Pax (AEPH) in the CRC. MATERIALS AND METHODS The components of AEPH were fully resolved using ultra-high-performance liquid chromatography coupled with quadrupole-orbitrap high-resolution mass spectrometry (UHPLC-Q/Orbitrap HRMS). The effect of AEPH was evaluated in vivo using the MC38 mouse colon cancer model, and its impact on the tumor microenvironment was analyzed by flow cytometry. Bioinformatics analysis, combined with transcriptome sequencing, was utilized to further investigate the signaling pathways of AEPH in CRC cells. RESULTS A mass spectrometry analysis identified 371 compounds in AEPH, each with a comprehensive score exceeding 60. In vivo experiments demonstrated that AEPH suppressed the growth of MC38 tumors without exhibiting obvious toxicity. Mechanistic studies revealed that AEPH inhibited the JNK signaling pathway, reduced Chemokine C-C Motif Chemokine Ligand 5 (CCL5) secreted by CRC cells, hindered the recruitment of M2-like tumor-associated macrophages (TAMs), promoted the infiltration of IFN-γ+ CD8+ T cells, and improved the immunosuppressive microenvironment of CRC. CONCLUSION AEPH contributes to the remodeling of the tumor immune microenvironment primarily through the inhibition of CCL5-mediated recruitment of M2-like TAMs. The findings of this study offer a novel perspective on the potential development of AEPH as a therapeutic agent for CRC.
Collapse
Affiliation(s)
- Yang Gui
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Wu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
26
|
Cruz-Rodriguez N, Deininger MW. Novel treatment strategies for chronic myeloid leukemia. Blood 2025; 145:931-943. [PMID: 39729529 PMCID: PMC11952011 DOI: 10.1182/blood.2024026312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024] Open
Abstract
ABSTRACT Starting with imatinib, tyrosine kinase inhibitors (TKIs) have turned chronic myeloid leukemia (CML) from a lethal blood cancer into a chronic condition. As patients with access to advanced CML care have an almost normal life expectancy, there is a perception that CML is a problem of the past, and one should direct research resources elsewhere. However, a closer look at the current CML landscape reveals a more nuanced picture. Most patients still require life-long TKI therapy to avoid recurrence of active CML. Chronic TKI toxicity and the high costs of the well-tolerated agents remain challenging. Progression to blast phase still occurs, particularly in socioeconomically disadvantaged parts of the world, where high-risk CML at diagnosis is common. Here, we review the prospects of further improving TKIs to achieve optimal suppression of BCR::ABL1 kinase activity, the potential of combining different classes of TKIs, and the current state of BCR::ABL1 degraders. We cover combination therapy approaches to address TKI resistance in the setting of residual leukemia and in advanced CML. Despite the unprecedented success of TKIs in CML, more work is needed to truly finish the job, and we hope to stimulate innovative research aiming to achieve this goal.
Collapse
Affiliation(s)
| | - Michael W. Deininger
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Medicine, Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
27
|
Li X, Zeng Q, Liu C, Yi X, Luo H, Tong Q, Chen H, You X. The Immune Modulatory Role of Surfactants in Mycoplasma pneumoniae Infection. J Inflamm Res 2025; 18:2909-2922. [PMID: 40034686 PMCID: PMC11873027 DOI: 10.2147/jir.s507526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/08/2025] [Indexed: 03/05/2025] Open
Abstract
Mycoplasma pneumoniae is a prevalent respiratory microbe that causes acute inflammation in the respiratory system. Surfactant proteins (SP), particularly SP-A and SP-D, are essential for the immunological protection against M. pneumoniae infection. Variant SP-A2 may lead to immune reactions, which could account for the variability in clinical manifestations among individuals. Mechanistically, these surfactant proteins may act as candidate receptors, facilitating both the adhesion of M. pneumoniae and internalization of community-acquired respiratory distress syndrome toxin. They also exhibit a high affinity for lipid ligands on the surface of M. pneumoniae membranes via their carbohydrate recognition domains, which aid in the direct clearing of the bacteria. In addition, SP-A and SP-D demonstrated synergistic effects in augmenting the intake and elimination of M. pneumoniae by alveolar macrophages. Furthermore, these surfactant proteins negatively regulate pulmonary inflammation by influencing lymphocyte and dendritic cell activities, reducing airway eosinophilic infiltration, and managing asthma-related inflammatory responses. A thorough understanding of the immunomodulatory roles of surfactant proteins in M. pneumoniae infection will shed light on how homeostasis is preserved during mycoplasma pneumonia and may guide the development of novel therapeutic strategies against this organism.
Collapse
Affiliation(s)
- Xinru Li
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, People’s Republic of China
| | - Qianrui Zeng
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, People’s Republic of China
| | - Chang Liu
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, People’s Republic of China
| | - Xinchao Yi
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, People’s Republic of China
| | - Haodang Luo
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, People’s Republic of China
| | - Qin Tong
- Department of Oncology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, 421001, People’s Republic of China
| | - Hongliang Chen
- Chenzhou No. 1 People’s Hospital (The Affiliated Chenzhou Hospital), Hengyang Medical College, University of South China, Chenzhou, People’s Republic of China
| | - Xiaoxing You
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, People’s Republic of China
| |
Collapse
|
28
|
Khizar H, Ali K, Wang J. From silent partners to potential therapeutic targets: macrophages in colorectal cancer. Cancer Immunol Immunother 2025; 74:121. [PMID: 39998578 PMCID: PMC11861851 DOI: 10.1007/s00262-025-03965-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/30/2025] [Indexed: 02/27/2025]
Abstract
Cancer cells grow and survive in the tumor microenvironment, which is a complicated process. As a key part of how colorectal cancer (CRC) progresses, tumor-associated macrophages (TAMs) exhibit a double role. Through angiogenesis, this TAM can promote the growth of cancers. Although being able to modify and adjust immune cells is a great advantage, these cells can also exhibit anti-cancer properties including direct killing of cancer cells, presenting antigens, and aiding T cell-mediated responses. The delicate regulatory mechanisms between the immune system and tumors are composed of a complex network of pathways regulated by several factors including hypoxia, metabolic reprogramming, cytokine/chemokine signaling, and cell interactions. Decoding and figuring out these complex systems become significant in building targeted treatment programs. Targeting TAMs in CRC involves disrupting chemokine signaling or adhesion molecules, reprogramming them to an anti-tumor phenotype using TLR agonists, CD40 agonists, or metabolic modulation, and selectively removing TAM subsets that promote tumor growth. Multi-drug resistance, the absence of an accurate biomarker, and drug non-specificity are also major problems. Combining macrophage-targeted therapies with chemotherapy and immunotherapy may revolutionize treatment. Macrophage studies will advance with new technology and multi-omics methodologies to help us understand CRC and build specific and efficient treatments.
Collapse
Affiliation(s)
- Hayat Khizar
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Kamran Ali
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jianwei Wang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, 2nd Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310009, China.
| |
Collapse
|
29
|
Zhou B, Fang F, Zhang Y, Li Z, Hu Y, Li Y, Jiao W, Wu Y, Wan X, Yang Y, Zhang F, Xu L, Ji T, Pan J, Hu S. Core transcriptional regulatory circuitry molecule ZNF217 promotes AML cell proliferation by up-regulating MYB. Int J Biol Sci 2025; 21:1966-1983. [PMID: 40083704 PMCID: PMC11900805 DOI: 10.7150/ijbs.103211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/02/2025] [Indexed: 03/16/2025] Open
Abstract
Leukemia is characterized by multiple rearrangements of signal transduction genes and overexpression of nonmutated genes, such as transcription factors (TFs) genes. Super-enhancers (SEs) are prevalent in human cancers and are associated with the accumulation of numerous core TFs. SEs drive the expression of core TF genes by delivering robust transcriptional activation signals. Additionally, core TFs sustain the stability and activity of SEs through mutual auto-regulation loops, creating a positive feedback loop known as the Core Transcriptional Regulation Circuit (CRC). Using ChIP-seq data, we identified the involvement of the SE-related gene ZNF217 in acute myeloid leukemia (AML), in which its functional role and underlying mechanism remain unclear. We demonstrated that ZNF217, ELF1, MEF2D, RUNX2, and FOXP1 are likely integral components of the AML CRC through various experimental techniques, including CUT&Tag, short hairpin RNA (shRNA) transduction, and Luciferase reporter assays. Notably, ZNF217 was determined to be indispensable for the proliferation and viability of AML cells both in vitro and in vivo. Subsequent analysis of RNA-seq and CUT&Tag results identified MYB as a key direct target of ZNF217. Overall, our research highlights ZNF217 as a critical oncogene in AML and offers new insights into the transcriptional regulatory mechanisms at play in AML.
Collapse
Affiliation(s)
- Bi Zhou
- Children's Hospital of Soochow University, Suzhou, 215003, China
- Dept. of Pediatric, Suzhou Hospital of AnHui Medical University, Suzhou, 234000, China
| | - Fang Fang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215003, China
- Pediatric Hematology & Oncology Key Laboratory of Higher Education Institutions in Jiangsu Province, Jiangsu, China
| | - YongPing Zhang
- Dept. of Hematology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - ZhiHeng Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215003, China
- Pediatric Hematology & Oncology Key Laboratory of Higher Education Institutions in Jiangsu Province, Jiangsu, China
| | - YiXin Hu
- Dept. of Hematology, Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Yan Li
- Dept. of Pediatric, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - WanYan Jiao
- Dept. of Pediatric, Yancheng Third People' Hospital, YanCheng, 224000, China
| | - YuMeng Wu
- Dept. of Pediatric, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - XiaoMei Wan
- Dept. of Pediatric, The First Affiliated Hospital of Wannan Medical College, Wuhu, 234100, China
| | - Ying Yang
- Clinical Medicine, Guizhou Medical University, Guiyang 550000, China
| | - FenLi Zhang
- Clinical Medicine, Guizhou Medical University, Guiyang 550000, China
| | - Ling Xu
- Dept. of Pediatric, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - TongTing Ji
- Children's Hospital of Soochow University, Suzhou, 215003, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215003, China
- Pediatric Hematology & Oncology Key Laboratory of Higher Education Institutions in Jiangsu Province, Jiangsu, China
| | - ShaoYan Hu
- Dept. of Hematology, Children's Hospital of Soochow University, Suzhou, 215003, China
| |
Collapse
|
30
|
Chen S, Wu S, Li N, Qiang X, Tang Y, Feng Y, Zhang C, Peng X, Liu S, Zhang X. SIL-TAL1-Positive Adult T-ALL with t(11;14)(p15;q11.2): A Rare Case Report Highlighting Prognostic Challenges and Treatment Implications. J Inflamm Res 2025; 18:2339-2347. [PMID: 39991668 PMCID: PMC11844270 DOI: 10.2147/jir.s497615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/25/2025] [Indexed: 02/25/2025] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) with coexisting SIL-TAL1 fusion and t(11;14)(p15;q11.2) is exceedingly rare. There are limited data on the clinical course and outcomes of such patients. We report a case of a 19-year-old male presenting with aggressive T-ALL harboring these abnormalities, along with NOTCH1 and PTEN mutations. SIL-TAL1 positivity is associated with poor prognosis in T-ALL. Despite achieving remission with intensive chemotherapy, the patient experienced rapid relapse and poor overall survival, reflecting the ineffectiveness of conventional treatments. The findings highlight the synergistic role of SIL-TAL1 and t(11;14) in disease progression and underscore the urgent need for targeted therapies and immunotherapies to improve outcomes in such high-risk cases.
Collapse
Affiliation(s)
- Siyu Chen
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, People’s Republic of China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, People’s Republic of China
| | - Shengwang Wu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, People’s Republic of China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, People’s Republic of China
| | - Nan Li
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, People’s Republic of China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, People’s Republic of China
| | - Xing Qiang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, People’s Republic of China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, People’s Republic of China
| | - Yongjie Tang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, People’s Republic of China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, People’s Republic of China
| | - Yimei Feng
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, People’s Republic of China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, People’s Republic of China
| | - Cheng Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, People’s Republic of China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, People’s Republic of China
| | - Xiangui Peng
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, People’s Republic of China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, People’s Republic of China
| | - Shuiqing Liu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, People’s Republic of China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, People’s Republic of China
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, People’s Republic of China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, People’s Republic of China
- Jinfeng Laboratory, Chongqing, 401329, People’s Republic of China
| |
Collapse
|
31
|
Lim JU, Jung J, Kim YW, Kim CY, Lee SH, Park DW, Choi SI, Ji W, Yeo CD, Lee SH. Targeting the Tumor Microenvironment in EGFR-Mutant Lung Cancer: Opportunities and Challenges. Biomedicines 2025; 13:470. [PMID: 40002883 PMCID: PMC11852785 DOI: 10.3390/biomedicines13020470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have transformed the treatment of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer. However, treatment resistance remains a major challenge in clinical practice. The tumor microenvironment (TME) is a complex system composed of tumor cells, immune and non-immune cells, and non-cellular components. Evidence indicates that dynamic changes in TME during TKI treatment are associated with the development of resistance. Research has focused on identifying how each component of the TME interacts with tumors and TKIs to understand therapeutic targets that could address TKI resistance. In this review, we describe how TME components, such as immune cells, fibroblasts, blood vessels, immune checkpoint proteins, and cytokines, interact with EGFR-mutant tumors and how they can promote resistance to TKIs. Furthermore, we discuss potential strategies targeting TME as a novel therapeutic approach.
Collapse
Affiliation(s)
- Jeong Uk Lim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yeon Wook Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Chi Young Kim
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sang Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Institute of Chest Diseases, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dong Won Park
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul 04763, Republic of Korea;
| | - Sue In Choi
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Wonjun Ji
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 44610, Republic of Korea
| | - Chang Dong Yeo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03083, Republic of Korea
| | - Seung Hyeun Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
32
|
Guoyun J, Yuefeng Q, Zhenglan H, Zuowei Y, Hongyan Z, Ying Y, Wenli F. CAR-macrophages targets CD26 to eliminate chronic myeloid leukemia stem cells. Exp Hematol Oncol 2025; 14:14. [PMID: 39948620 PMCID: PMC11823019 DOI: 10.1186/s40164-025-00608-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 02/05/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Chronic myeloid leukemia stem cells (CML-LSCs), which exhibit resistance to tyrosine kinase inhibitors (TKIs), are the leading cause of treatment failure and recurrence in chronic myeloid leukemia (CML). This highlights the urgent need for novel therapies aimed at eliminating these CML-LSCs. Chimeric antigen receptor macrophages (CAR-M) not only perform phagocytosis on target cells but also function as antigen-presenting cells, thereby activating the anti-tumor immune response.CD26 (dipeptidyl peptidase 4, DPP IV) is abundantly expressed in CML-LSCs and functions as a tumor-specific antigen (TSA) in CAR-M treatment. The purpose of this study is to evaluate CAR-M's efficacy in targeting CD26-positive CML cells and to develop a novel strategy for CML treatment. METHODS CD26 CAR-M was constructed using mouse-derived macrophage Raw264.7 cells. CD26 was overexpressed in CML cell lines BP210 and BP210-T315I. The targeting phagocytosis of CAR-M was verified using confocal microscopy and flow cytometry. X-ray was used to eliminate the tumorigenicity of CAR-M, and the safety of CAR-M was verified through CCK-8, clone formation assays, and animal experiments. To assess the anti-leukemia ability of CAR-M in the CML mouse model, the survival, peripheral blood white blood cell counts, and CML cell infiltration in the liver, spleen, and bone marrow (BM) were measured. Additionally, CD26 CAR-THP1 was constructed, and its phagocytic ability against CD26-positive cells NCI-H2452 was confirmed by confocal microscopy. RESULTS We successfully constructed CD26 CAR-M and validated its targeted phagocytosis of CD26-positive CML cells both in vitro and in vivo. The data indicate that CAR-M has higher phagocytic efficiency in CD26-positive CML cells than in CD26-negative cells. CAR-M-treated CML mice demonstrated extended survival and reduced CML invasion. In addition, CAR-THP1 demonstrated targeted phagocytosis of NCI-H2452 cells that normally express CD26. CONCLUSION This study demonstrates that CD26 CAR-M effectively targets and phagocytizes CD26-positive CML cells, implying that targeting CD26 with CAR-M could be a viable method for eradicating CML-LSCs. Furthermore, our discoveries illuminate the potential application of CAR-M in treating hematological malignancies.
Collapse
Affiliation(s)
- Jiang Guoyun
- Department of Clinical Hematology, School of Laboratory Medicine, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Qin Yuefeng
- Department of Clinical Hematology, School of Laboratory Medicine, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Huang Zhenglan
- Department of Clinical Hematology, School of Laboratory Medicine, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yuan Zuowei
- Department of Clinical Hematology, School of Laboratory Medicine, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Zhou Hongyan
- Department of Clinical Hematology, School of Laboratory Medicine, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yuan Ying
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1, Youyi Road, Yuzhong District, Chongqing, 400016, China.
| | - Feng Wenli
- Department of Clinical Hematology, School of Laboratory Medicine, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
33
|
Caragine CM, Le VT, Mustafa M, Diaz BJ, Morris JA, Müller S, Mendez-Mancilla A, Geller E, Liscovitch-Brauer N, Sanjana NE. Comprehensive dissection of cis-regulatory elements in a 2.8 Mb topologically associated domain in six human cancers. Nat Commun 2025; 16:1611. [PMID: 39948336 PMCID: PMC11825950 DOI: 10.1038/s41467-025-56568-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Cis-regulatory elements (CREs), such as enhancers and promoters, are fundamental regulators of gene expression and, across different cell types, the MYC locus utilizes a diverse regulatory architecture driven by multiple CREs. To better understand differences in CRE function, we perform pooled CRISPR inhibition (CRISPRi) screens to comprehensively probe the 2.8 Mb topologically-associated domain containing MYC in 6 human cancer cell lines with nucleotide resolution. We map 32 CREs where inhibition leads to changes in cell growth, including 8 that overlap previously identified enhancers. Targeting specific CREs decreases MYC expression by as much as 60%, and cell growth by as much as 50%. Using 3-D enhancer contact mapping, we find that these CREs almost always contact MYC but less than 10% of total MYC contacts impact growth when silenced, highlighting the utility of our approach to identify phenotypically-relevant CREs. We also detect an enrichment of lineage-specific transcription factors (TFs) at MYC CREs and, for some of these TFs, find a strong, tumor-specific correlation between TF and MYC expression not found in normal tissue. Taken together, these CREs represent systematically identified, functional regulatory regions and demonstrate how the same region of the human genome can give rise to complex, tissue-specific gene regulation.
Collapse
Affiliation(s)
- Christina M Caragine
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Victoria T Le
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Meer Mustafa
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Bianca Jay Diaz
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - John A Morris
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Simon Müller
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Alejandro Mendez-Mancilla
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Evan Geller
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Noa Liscovitch-Brauer
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Neville E Sanjana
- New York Genome Center, New York, NY, USA.
- Department of Biology, New York University, New York, NY, USA.
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA.
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
34
|
Twardowska M, Łyskowski A, Misiorek M, Szymaszek Ż, Wołowiec S, Dąbrowska M, Uram Ł. Human Embryonic Kidney HEK293 Cells as a Model to Study SMVT-Independent Transport of Biotin and Biotin-Furnished Nanoparticles in Targeted Therapy. Int J Mol Sci 2025; 26:1594. [PMID: 40004058 PMCID: PMC11855691 DOI: 10.3390/ijms26041594] [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: 12/31/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
The aim of this study was to investigate the usefulness of human embryonic kidney HEK293 cells as a model of normal cells in biotin-mediated therapy. The expression and role of sodium multivitamin transporter (SMVT) in the uptake and accumulation of free biotin, as well as cationic and neutral biotinylated PAMAM dendrimers of the fourth generation synthesized in our laboratory, were assessed in HEK293 cells in comparison to other immortalized (HaCaT) and cancer cells (HepG2, U-118 MG). The obtained data showed that a higher level of SMVT in HEK293 cells was not associated with a stronger uptake of biotin and biotinylated PAMAM dendrimers. Biotinylation increased the selective uptake of neutral dendrimers in an inversely proportional manner to the concentration used; however, the accumulation in HEK293 cells was lower than that in cells of other cell lines. The time-dependent biotin and biotinylated dendrimers uptake profiles differed significantly. Therefore, it should be assumed that the efficiency of biotinylated nanoparticles' uptake depends on multiple cellular transport mechanisms. Toxicity tests showed significantly higher sensitivity to PAMAM conjugates for HEK293 cells than for HepG2 and HaCaT cells. Molecular modeling studies and the profile of biotin uptake suggest that not only SMVT but also monocarboxylate transporter 1 (MCT-1) may play an important role in the selective transport of biotin and biotinylated nanoparticles into cells. Due to the complexity of the problem, further studies are necessary. In summary, HEK293 cells can be considered a valuable model of normal cells in the study of biotin- targeted therapy using nanoparticles based on PAMAM dendrimers.
Collapse
Affiliation(s)
- Magdalena Twardowska
- The Faculty of Chemistry, Rzeszow University of Technology, Powstancow Warszawy 6 Ave., 35-959 Rzeszow, Poland; (M.M.); (Ż.S.); (Ł.U.)
| | - Andrzej Łyskowski
- The Faculty of Chemistry, Rzeszow University of Technology, Powstancow Warszawy 6 Ave., 35-959 Rzeszow, Poland; (M.M.); (Ż.S.); (Ł.U.)
| | - Maria Misiorek
- The Faculty of Chemistry, Rzeszow University of Technology, Powstancow Warszawy 6 Ave., 35-959 Rzeszow, Poland; (M.M.); (Ż.S.); (Ł.U.)
| | - Żaneta Szymaszek
- The Faculty of Chemistry, Rzeszow University of Technology, Powstancow Warszawy 6 Ave., 35-959 Rzeszow, Poland; (M.M.); (Ż.S.); (Ł.U.)
| | - Stanisław Wołowiec
- Medical College, University of Rzeszow, 1a Warzywna Street, 35-310 Rzeszow, Poland;
| | - Magdalena Dąbrowska
- Laboratory of Molecular Bases of Ageing, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warszawa, Poland;
| | - Łukasz Uram
- The Faculty of Chemistry, Rzeszow University of Technology, Powstancow Warszawy 6 Ave., 35-959 Rzeszow, Poland; (M.M.); (Ż.S.); (Ł.U.)
| |
Collapse
|
35
|
Ebaid NF, Abdelkawy KS, Said ASA, Al-Ahmad MM, Shehata MA, Salem HF, Hussein RRS. Is the Neutrophil-to-Lymphocyte Ratio a Predictive Factor of Pathological Complete Response in Egyptian Breast Cancer Patients Treated with Neoadjuvant Chemotherapy? MEDICINA (KAUNAS, LITHUANIA) 2025; 61:327. [PMID: 40005444 PMCID: PMC11857557 DOI: 10.3390/medicina61020327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/01/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025]
Abstract
Background and Objectives: The role of the neutrophil-to-lymphocyte ratio (NLR) as a predictor of response in breast cancers after neoadjuvant chemotherapy is controversial. This study aims to explore the relationship of NLR with pathological complete response (pCR) in a cohort of Egyptian breast cancer patients who received neoadjuvant chemotherapy. Materials and Methods: Forty-six breast cancer females received preoperative neoadjuvant chemotherapy and then underwent surgery. All resected tumors were evaluated to determine the pathologic effect of the neoadjuvant chemotherapy. A complete blood count was carried out at baseline before beginning the neoadjuvant chemotherapy. The absolute count of neutrophils was divided by the absolute count of lymphocytes to calculate the NLR. Results: Of the study patients, 18 (39.1%) were considered to have a low NLR (NLR < 1.76), and 28 (60.9%) were considered to have a high NLR (NLR ≥ 1.76). Patients with a low NLR had 18-fold higher rates of pCR when compared to patients with a high NLR (OR 18.1; 95% CI (1.058-310.757); p = 0.046). Conclusions: Our findings indicate that the pretreatment NLR is a pivotal predictor factor of the pathological complete response in Egyptian breast cancer patients treated with neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Naglaa F. Ebaid
- Clinical Pharmacy Department, Faculty of Pharmacy, Menoufia University, Menoufia 32511, Egypt;
| | - Khaled S. Abdelkawy
- Clinical Pharmacy Department, Faculty of Pharmacy, Kafrelsheikh University, Kafr El Sheikh 33516, Egypt;
| | - Amira S. A. Said
- Department of Clinical Pharmacy, College of Pharmacy, Al Ain University, Al Ain P.O. Box 64141, United Arab Emirates; (A.S.A.S.); (M.M.A.-A.)
| | - Mohamad M. Al-Ahmad
- Department of Clinical Pharmacy, College of Pharmacy, Al Ain University, Al Ain P.O. Box 64141, United Arab Emirates; (A.S.A.S.); (M.M.A.-A.)
| | - Mohamed A. Shehata
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Menoufia University, Menofia 32511, Egypt;
| | - Heba F. Salem
- Pharmaceutics and Industrial Pharmacy Department, Beni-Suef University, Beni-Suef 62574, Egypt;
| | - Raghda R. S. Hussein
- Clinical Pharmacy Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62574, Egypt
| |
Collapse
|
36
|
Wu L, Lin L, Yu M, Li H, Dang Y, Liang H, Chen G, Muhetaer H, Zheng G, Li J, Jia X, Wu B, Li C. Antitumor Activity of USP7 Inhibitor GNE-6776 in Non-Small Cell Lung Cancer Involves Regulation of Epithelial-Mesenchymal Transition, Cell Cycle, Wnt/β-Catenin, and PI3K/AKT/mTOR Pathways. Pharmaceuticals (Basel) 2025; 18:245. [PMID: 40006058 PMCID: PMC11858873 DOI: 10.3390/ph18020245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Objective: Non-small cell lung cancer (NSCLC) is a major cause of cancer-related deaths worldwide. This study investigated the effects and mechanisms of the USP7 inhibitor GNE-6776 on human NSCLC A549 and H1299 cells, providing insights for anti-NSCLC drug development. Methods: USP7 expression was analyzed in lung cancer tissue using data from public databases. RNA sequencing and functional enrichment analyses were conducted to explore differentially expressed genes (DEGs) and potentially related pathways. A549 and H1299 cells were treated with GNE-6776 at different concentrations, and its effects on cell proliferation, migration, invasion, apoptosis, mitochondrial membrane potential, and cell cycle were evaluated. Changes in protein expression following GNE-6776 treatment were assessed by Western blot. A xenograft tumor model in nude mice was used to evaluate the in vivo effects of GNE-6776. Results: GNE-6776 inhibited the proliferation, migration, and invasion of A549 and H1299 cells, induced apoptosis, and caused cells to arrest in the G1 phase in a concentration-dependent manner. GNE-6776 decreased the mitochondrial membrane potential, suppressed epithelial-mesenchymal transition (EMT) markers, and downregulated the PI3K/AKT/mTOR and Wnt/β-catenin signaling pathways. GNE-6776 significantly inhibited tumor growth without affecting body weight, reduced expression of CDK6, C-myc, and N-cadherin, and increased GSK3β expression in tumor tissue. Conclusions: In summary, GNE-6776 demonstrated potent anti-tumor activity in NSCLC both in vitro and in vivo. GNE-6776 suppresses NSCLC cell proliferation, invasion, and migration while promoting apoptosis by inhibiting the EMT and modulating the PI3K/AKT/mTOR and Wnt/β-catenin pathways. These findings support its potential as a therapeutic agent for treating NSCLC.
Collapse
Affiliation(s)
- Lipeng Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Long Lin
- Phase I Clinical Trial Center, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou 510440, China
| | - Meng Yu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Huajian Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yuanye Dang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Huosheng Liang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Guangyang Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Halimulati Muhetaer
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Guodong Zheng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong SAR 999077, China
| | - Xuejing Jia
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Bo Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Chuwen Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
37
|
Deng Z, Jin X, Liu B, Zhen H, Wang X. Unveiling the prognostic significance of RNA editing-related genes in colon cancer: evidence from bioinformatics and experiment. Eur J Med Res 2025; 30:94. [PMID: 39940052 PMCID: PMC11823094 DOI: 10.1186/s40001-025-02335-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 01/26/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND RNA editing is recognized as a crucial factor in cancer biology. Its potential application in predicting the prognosis of colon adenocarcinoma (COAD) remains unexplored. METHODS RNA editing data of COAD patients were downloaded from the Synapse database. LASSO regression was used to construct the risk model and verified by the receiver operating characteristic (ROC) curve. GO and KEGG enrichment analyses were performed to delineate the biological significance of the differentially expressed genes. Finally, differential analysis and immunohistochemistry were used to verify the expression of adenosine deaminase 1 (ADAR1). RESULTS We evaluated a total of 4079 RNA editing sites in 514 COAD patients from Synapse database. A prognostic signature was constructed based on five genes were significantly associated with the prognosis of COAD patients including GNL3L, NUP43, MAGT1, EMP2, and ARSD. Univariate and multivariate Cox regression analysis revealed that RNA editing-related genes (RERGs)-related signature was an independent risk factor for COAD. Moreover, Experimental evidence shows that ADAR1 is highly expressed in colon adenocarcinoma and silencing ADAR1 can inhibit cancer cell proliferation. CONCLUSION We established a prognostic model based on five RERGs with strong predictive value. This model not only serves as a foundation for a novel prognostic tool but also facilitates the identification of potential drug candidates for treating COAD.
Collapse
Affiliation(s)
- Zhengcong Deng
- Hubei Third People's Hospital, Wuhan, 430033, Hubei, China
- Wuhan Donghu New Technology Development Zone Disease Prevention and Control Center, Wuhan, 430200, Hubei, China
| | - Xueqin Jin
- Hubei Third People's Hospital, Wuhan, 430033, Hubei, China
| | - Bingxue Liu
- Medical School, Jianghan University, Wuhan, 430056, Hubei, China
| | - Hongyan Zhen
- Medical School, Jianghan University, Wuhan, 430056, Hubei, China
| | - Xiang Wang
- Medical School, Jianghan University, Wuhan, 430056, Hubei, China.
- Wuhan University of Arts and Science, Wuhan, 430345, Hubei, China.
| |
Collapse
|
38
|
Xu P, Yu H, Bian H, Jia D, Li W, Dong H, Peng X. Bibliometric insight into neoadjuvant immunotherapy in non-small cell lung cancer: trends, collaborations, and future avenues. Front Immunol 2025; 16:1533651. [PMID: 39995679 PMCID: PMC11847801 DOI: 10.3389/fimmu.2025.1533651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
Background Neoadjuvant immunotherapy (NIT) is a rapidly emerging paradigm for advanced resectable non-small cell lung cancer (NSCLC). However, there is no bibliometric analysis in this research field. Objective To analyze the hotspots and trends in the research of NIT for NSCLC and provide a reference for the study of NIT for lung cancer in China. Methods Retrieve literature related to NIT for NSCLC from Web of Science, PubMed, and Scopus databases up to September 10, 2024. Use CiteSpace and VOSviewer software visualization software to analyze the keywords of country, author, institution, and literature. Results There were 1575 references, and the overall annual publication volume showed an upward trend; Forde and Patrick M have published the most articles in the literature. The research hotspots mainly focus on chemotherapy, NIT for NSCLC, immunotherapy, neoadjuvant chemotherapy, pathological reactions, etc. Conclusions This is the first bibliometric study comprehensively summarizing NIT's research trends and development in NSCLC. Our bibliometric assessment provides a panoramic view of the research milieu surrounding NIT for NSCLC, encapsulating the present state, evolving trends, and potential future directions, particularly emphasizing the promise of immunochemotherapy.
Collapse
Affiliation(s)
- Pengliang Xu
- Department of Thoracic Surgery, The First People’s Hospital of Huzhou, Huzhou, China
| | - Huanming Yu
- Department of Thoracic Surgery, The First People’s Hospital of Huzhou, Huzhou, China
| | - Hupo Bian
- Department of Radiology, The First People’s Hospital of Huzhou, Huzhou, China
| | - Dan Jia
- Department of Respiratory Medicine, The First People’s Hospital of Huzhou, Huzhou, China
| | - Wenhui Li
- Department of Thoracic Surgery, The First People’s Hospital of Huzhou, Huzhou, China
| | - Hongfeng Dong
- Department of Radiology, The First People’s Hospital of Huzhou, Huzhou, China
| | - Xiuhua Peng
- Department of Radiology, The First People’s Hospital of Huzhou, Huzhou, China
| |
Collapse
|
39
|
Liu M, Li Y, Deng Z, Zhang K, Huang S, Xia J, Feng Y, Liang Y, Sun C, Liu X, Li S, Su B, Dong Y, Huang S. Mcm5 mutation leads to silencing of Stat1-bcl2 which accelerating apoptosis of immature T lymphocytes with DNA damage. Cell Death Dis 2025; 16:84. [PMID: 39929806 PMCID: PMC11811017 DOI: 10.1038/s41419-025-07392-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 01/08/2025] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
Mutation in genes involved in DNA replication continuously disrupt DNA replication and give rise to genomic instability, a critical driver of oncogenesis. To prevent leukemia, immature T lymphocytes with genomic instability often undergo rapid cell death during development. However, the mechanism by which immature T lymphocytes undergo rapid cell death upon genomic instability has been enigmatic. Here we show that zebrafish mcm5 mutation leads to DNA damage in immature T lymphocytes and the immature T cells sensitively undergo rapid cell death. Detailed analyses demonstrated that the immature T lymphocytes undergo rapid apoptosis via upregulation of tp53 and downregulation of bcl2 transcription in mcm5 mutants. Mechanistically, Mcm5 directly binds to Stat1a and facilitates its phosphorylation to enhance bcl2a expression under the conditions of DNA replication stress. However, in mcm5 mutants, the absence of the Mcm5-Stat1 complex decreases Stat1 phosphorylation and subsequent bcl2a transcription, accelerating apoptosis of immature T lymphocytes with genomic instability. Furthermore, our study shows that the role of Mcm5 in T-cell development is conserved in mice. In conclusion, our work identifies a role of Mcm5 in regulating T cell development via Stat1-Bcl2 cascade besides its role in DNA replication, providing a kind of mechanism by which immature T cells with gene mutation-induced DNA damage are rapidly cleared during T lymphocyte development.
Collapse
Affiliation(s)
- Min Liu
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, China
- Department of Cardiology, The First Affiliated Hospital, Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Yuanyuan Li
- Department of Neurology, the Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610000, China
| | - Zhilin Deng
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, China
| | - Ke Zhang
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, China
| | - Shuying Huang
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, China
| | - Jiamin Xia
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, China
| | - Yi Feng
- Centre for Inflammation Research, Queen's Medical Research Institute, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Yundan Liang
- Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, 610500, China
| | - Chengfu Sun
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, China
| | - Xindong Liu
- Department of Neurology, the Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610000, China
| | - Shurong Li
- Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, 610500, China
| | - Bingyin Su
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, China
| | - Yong Dong
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, China.
| | - Sizhou Huang
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610500, China.
- Department of Neurology, the Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610000, China.
- Centre for Inflammation Research, Queen's Medical Research Institute, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
40
|
Qin L, Li Y, Liu J, An X. Advancements in cellular immunotherapy: overcoming resistance in lung and colorectal cancer. Front Immunol 2025; 16:1554256. [PMID: 39975543 PMCID: PMC11835964 DOI: 10.3389/fimmu.2025.1554256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 01/17/2025] [Indexed: 02/21/2025] Open
Abstract
Immunotherapy has revolutionized cancer treatment, offering hope for patients with otherwise treatment-resistant tumors. Among the most promising approaches are cellular therapies, particularly chimeric antigen receptor T-cell (CAR-T) therapy, which has shown remarkable success in hematologic malignancies. However, the application of these therapies to solid tumors, such as lung and colorectal cancers, has faced significant challenges. Tumor resistance mechanisms-ranging from immune evasion, antigen loss, and immune checkpoint upregulation, to tumor microenvironment immunosuppression-remain major obstacles. This mini-review highlights the latest advancements in tumor immunotherapy, with a focus on cellular therapies, and addresses the resistance mechanisms that hinder their effectiveness in lung and colorectal cancers. We examine the evolution of CAR-T cell therapy, as well as the potential of engineered natural killer (NK) cells and macrophages in solid tumor treatment. The review also explores cutting-edge strategies aimed at overcoming resistance, including combination therapies, gene editing technologies, and nanotechnology for targeted drug delivery. By discussing the molecular, cellular, and microenvironmental factors contributing to resistance, we aim to provide a comprehensive overview of how these challenges can be overcome, paving the way for more effective, personalized immunotherapies in lung and colorectal cancer treatment.
Collapse
Affiliation(s)
- Lijuan Qin
- Department of Radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuan Li
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Juan Liu
- Department of Special needs Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoqin An
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
41
|
Cao X, Luo N, Liu X, Guo K, Deng M, Lv C. Crosstalk of SPINK4 Expression With Patient Mortality, Immunotherapy and Metastasis in Pan-Cancer Based on Integrated Multi-Omics Analyses. Onco Targets Ther 2025; 18:161-177. [PMID: 39926372 PMCID: PMC11806753 DOI: 10.2147/ott.s487126] [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: 08/27/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Background Cancer remains a major global health challenge, with early detection and prompt treatment being crucial for reducing mortality rates. The SPINK4 has been linked to the development of several tumors, and there is growing evidence of its involvement. However, its specific functions and effects in different cancer types remain unclear. Methods The association between SPINK4 expression levels and tumor progression was investigated and confirmed using the TCGA dataset. Kaplan-Meier curves were utilized to examine the correlation between SPINK4 expression with survival outcomes in pan-cancer patients. The Pearson method was employed to investigate the association of SPINK4 expression with the tumor microenvironment, stemness score, immunoinfiltrating subtype, and chemotherapy sensitivity in human different cancer types. Wound healing and Transwell assays were performed to confirm the roles of the model gene in colon adenocarcinoma cells. Results The expression of SPINK4 shows heterogeneity across pan-cancer tissues, and is closely associated with poor prognosis, immune cell invasion, tumor cell resistance, and tumor metastasis in a various human cancer. Mutation of SPINK4 hold significant predictive value for poor prognosis of pan-cancer patients. In addition, SPINK4 expression was significantly correlated with the tumor microenvironment (stromal cells and immune cells) and stemness score (DNAss and RNAss) in human pan-cancer tissues, particularly in BLCA and COAD. Single-cell sequencing analysis showed that SPINK4 is mainly expressed in endothelial cells in BLCA and in malignant cells in COAD. Drug resistance analysis showed a significant association between SPINK4 expression and sensitivity to several cancer chemotherapy drugs. Importantly, overexpression of SPINK4 promoted the metastasis of colon cancer cell lines (HCT116 and RKO), whereas SPINK4 knockout markedly inhibited their metastasis. Conclusion These findings reveal the crucial role of SPINK4 in the pan-cancer process and may have significant implications for the diagnosis and treatment of cancer in the future.
Collapse
Affiliation(s)
- Xiuhua Cao
- Center for Basic Medical Research, Southwest Medical University, Luzhou, People’s Republic of China
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Na Luo
- Center for Basic Medical Research, Southwest Medical University, Luzhou, People’s Republic of China
| | - Xiaoyan Liu
- Center for Basic Medical Research, Southwest Medical University, Luzhou, People’s Republic of China
| | - Kan Guo
- Center for Basic Medical Research, Southwest Medical University, Luzhou, People’s Republic of China
| | - Mingming Deng
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
| | - Chaoxiang Lv
- Center for Basic Medical Research, Southwest Medical University, Luzhou, People’s Republic of China
| |
Collapse
|
42
|
Fisher MS, Sennikov SV. T-regulatory cells for the treatment of autoimmune diseases. Front Immunol 2025; 16:1511671. [PMID: 39967659 PMCID: PMC11832489 DOI: 10.3389/fimmu.2025.1511671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
Autoimmune diseases result from imbalances in the immune system and disturbances in the mechanisms of immune tolerance. T-regulatory cells (Treg) are key factors in the formation of immune tolerance. Tregs modulate immune responses and repair processes, controlling the innate and adaptive immune system. The use of Tregs in the treatment of autoimmune diseases began with the manipulation of endogenous Tregs using immunomodulatory drugs. Then, a method of adoptive transfer of Tregs grown in vitro was developed. Adoptive transfer of Tregs includes polyclonal Tregs with non-specific effects and antigen-specific Tregs in the form of CAR-Treg and TCR-Treg. This review discusses non-specific and antigen-specific approaches to the use of Tregs, their advantages, disadvantages, gaps in development, and future prospects.
Collapse
Affiliation(s)
- Marina S. Fisher
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University under the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Sergey V. Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University under the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| |
Collapse
|
43
|
Nguyen HX, Kipping T, Banga AK. Enhancement of Transdermal Drug Delivery: Integrating Microneedles with Biodegradable Microparticles. Mol Pharm 2025; 22:984-1009. [PMID: 39823349 DOI: 10.1021/acs.molpharmaceut.4c01202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
This investigation aimed to enhance transdermal methotrexate delivery through human skin by employing Dr. Pen microneedles and poly(d,l-lactide-co-glycolide) acid microparticles formulated from eight polymer grades (Expansorb DLG 95-4A, DLG 75-5A, DLG 50-2A, DLG 50-5A, DLG 50-8A, DLG 50-6P, DLG 50-7P, and DLL 10-15A). A comprehensive characterization of the microparticles was performed, encompassing various parameters such as size, charge, morphology, microencapsulation efficiency, yield, release kinetics, and chemical composition. The efficacy of microneedles in disrupting skin integrity was demonstrated by scanning electron microscopy, dye binding, histological examination, confocal laser microscopy, and pore size analysis. Microneedle-mediated skin microporation led to a substantial reduction in skin electrical resistance and a concomitant increase in transepidermal water loss. In vitro permeation experiments using human skin delivered microparticles into microporated skin and demonstrated a considerable difference in methotrexate delivery among the polymer groups. Microneedle treatment significantly amplified cumulative drug delivery, steady-state flux, diffusion coefficient, permeability coefficient, and drug concentration within skin layers while concurrently diminishing lag time (p < 0.05). Furthermore, a robust correlation was established between microparticle properties (cumulative release, release rate, encapsulation efficiency) and drug deposition in the skin. In conclusion, the synergistic combination of Dr. Pen microneedles and PLGA microparticles facilitated enhanced and regulated transdermal methotrexate delivery.
Collapse
Affiliation(s)
- Hiep X Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, Georgia 30341, United States
- Faculty of Pharmacy, Phenikaa University, Yen Nghia, Ha Dong, Hanoi 12116, Vietnam
| | - Thomas Kipping
- MilliporeSigma, a Business of Merck KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Ajay K Banga
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, Georgia 30341, United States
| |
Collapse
|
44
|
Yu B, Shao S, Ma W. Frontiers in pancreatic cancer on biomarkers, microenvironment, and immunotherapy. Cancer Lett 2025; 610:217350. [PMID: 39581219 DOI: 10.1016/j.canlet.2024.217350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Pancreatic cancer remains one of the most challenging malignancies to treat due to its late-stage diagnosis, aggressive progression, and high resistance to existing therapies. This review examines the latest advancements in early detection, and therapeutic strategies, with a focus on emerging biomarkers, tumor microenvironment (TME) modulation, and the integration of artificial intelligence (AI) in data analysis. We highlight promising biomarkers, including microRNAs (miRNAs) and circulating tumor DNA (ctDNA), that offer enhanced sensitivity and specificity for early-stage diagnosis when combined with multi-omics panels. A detailed analysis of the TME reveals how components such as cancer-associated fibroblasts (CAFs), immune cells, and the extracellular matrix (ECM) contribute to therapy resistance by creating immunosuppressive barriers. We also discuss therapeutic interventions that target these TME components, aiming to improve drug delivery and overcome immune evasion. Furthermore, AI-driven analyses are explored for their potential to interpret complex multi-omics data, enabling personalized treatment strategies and real-time monitoring of treatment response. We conclude by identifying key areas for future research, including the clinical validation of biomarkers, regulatory frameworks for AI applications, and equitable access to innovative therapies. This comprehensive approach underscores the need for integrated, personalized strategies to improve outcomes in pancreatic cancer.
Collapse
Affiliation(s)
- Baofa Yu
- Taimei Baofa Cancer Hospital, Dongping, Shandong, 271500, China; Jinan Baofa Cancer Hospital, Jinan, Shandong, 250000, China; Beijing Baofa Cancer Hospital, Beijing, 100010, China; Immune Oncology Systems, Inc, San Diego, CA, 92102, USA.
| | - Shengwen Shao
- Institute of Microbiology and Immunology, Huzhou University School of Medicine, Huzhou, Zhejiang, 313000, China.
| | - Wenxue Ma
- Department of Medicine, Sanford Stem Cell Institute, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
45
|
Zhang G, Yan S, Liu Y, Du Z, Min Q, Qin S. PROTACs coupled with oligonucleotides to tackle the undruggable. Bioanalysis 2025; 17:261-276. [PMID: 39895280 PMCID: PMC11864318 DOI: 10.1080/17576180.2025.2459528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025] Open
Abstract
Undruggable targets account for roughly 85% of human disease-related targets and represent a category of therapeutic targets that are difficult to tackle with traditional methods, but their considerable clinical importance. These targets are generally defined by planar functional interfaces and the absence of efficient ligand-binding pockets, making them unattainable for conventional pharmaceutical strategies. The advent of oligonucleotide-based proteolysis-targeting chimeras (PROTACs) has instilled renewed optimism in addressing these challenges. These PROTACs facilitate the targeted degradation of undruggable entities, including transcription factors (TFs) and RNA-binding proteins (RBPs), via proteasome-dependent mechanisms, thereby presenting novel therapeutic approaches for diseases linked to these targets. This review offers an in-depth examination of recent progress in the integration of PROTAC technology with oligonucleotides to target traditionally undruggable proteins, emphasizing the design principles and mechanisms of action of these innovative PROTACs.
Collapse
Affiliation(s)
- Guangshuai Zhang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Si Yan
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Yan Liu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Ziwei Du
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Qin Min
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Shuanglin Qin
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, P.R. China
| |
Collapse
|
46
|
Jaing TH, Hsiao YW, Wang YL. Chimeric Antigen Receptor Cell Therapy: Empowering Treatment Strategies for Solid Tumors. Curr Issues Mol Biol 2025; 47:90. [PMID: 39996811 PMCID: PMC11854309 DOI: 10.3390/cimb47020090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
Chimeric antigen receptor-T (CAR-T) cell therapy has demonstrated impressive efficacy in the treatment of blood cancers; however, its effectiveness against solid tumors has been significantly limited. The differences arise from a range of difficulties linked to solid tumors, including an unfriendly tumor microenvironment, variability within the tumors, and barriers to CAR-T cell infiltration and longevity at the tumor location. Research shows that the reasons for the decreased effectiveness of CAR-T cells in treating solid tumors are not well understood, highlighting the ongoing need for strategies to address these challenges. Current strategies frequently incorporate combinatorial therapies designed to boost CAR-T cell functionality and enhance their capacity to effectively target solid tumors. However, these strategies remain in the testing phase and necessitate additional validation to assess their potential benefits. CAR-NK (natural killer), CAR-iNKT (invariant natural killer T), and CAR-M (macrophage) cell therapies are emerging as promising strategies for the treatment of solid tumors. Recent studies highlight the construction and optimization of CAR-NK cells, emphasizing their potential to overcome the unique challenges posed by the solid tumor microenvironment, such as hypoxia and metabolic barriers. This review focuses on CAR cell therapy in the treatment of solid tumors.
Collapse
Affiliation(s)
- Tang-Her Jaing
- Division of Hematology and Oncology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 33315, Taiwan;
| | - Yi-Wen Hsiao
- Division of Nursing, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 33315, Taiwan;
| | - Yi-Lun Wang
- Division of Hematology and Oncology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 33315, Taiwan;
| |
Collapse
|
47
|
Liao YY, Tsai CL, Huang HP. Optimizing Osimertinib for NSCLC: Targeting Resistance and Exploring Combination Therapeutics. Cancers (Basel) 2025; 17:459. [PMID: 39941826 PMCID: PMC11815769 DOI: 10.3390/cancers17030459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Non-small-cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide, with epidermal growth factor receptor (EGFR) mutations present in a substantial proportion of patients. Third-generation EGFR tyrosine kinase inhibitors (EGFR TKI), exemplified by osimertinib, have dramatically improved outcomes by effectively targeting the T790M mutation-a primary driver of acquired resistance to earlier-generation EGFR TKI. Despite these successes, resistance to third-generation EGFR TKIs inevitably emerges. Mechanisms include on-target mutations such as C797S, activation of alternative pathways like MET amplification, histologic transformations, and intricate tumor microenvironment (TME) alterations. These resistance pathways are compounded by challenges in tolerability, adverse events, and tumor heterogeneity. In light of these hurdles, this review examines the evolving landscape of combination therapies designed to enhance or prolong the effectiveness of third-generation EGFR TKIs. We explore key strategies that pair osimertinib with radiotherapy, anti-angiogenic agents, immune checkpoint inhibitors, and other molecularly targeted drugs, and we discuss the biological rationale, preclinical evidence, and clinical trial data supporting these approaches. Emphasis is placed on how these combinations may circumvent diverse resistance mechanisms, improve survival, and maintain a favorable safety profile. By integrating the latest findings, this review aims to guide clinicians and researchers toward more individualized and durable treatment options, ultimately enhancing both survival and quality of life for patients with EGFR-mutated NSCLC.
Collapse
Affiliation(s)
- Yan-You Liao
- Department of Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan;
| | - Chia-Luen Tsai
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100233, Taiwan;
| | - Hsiang-Po Huang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100233, Taiwan;
| |
Collapse
|
48
|
Huang Y, Zhang P, Wang H, Chen Y, Liu T, Luo X. Genetic Code Expansion: Recent Developments and Emerging Applications. Chem Rev 2025; 125:523-598. [PMID: 39737807 PMCID: PMC11758808 DOI: 10.1021/acs.chemrev.4c00216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/01/2025]
Abstract
The concept of genetic code expansion (GCE) has revolutionized the field of chemical and synthetic biology, enabling the site-specific incorporation of noncanonical amino acids (ncAAs) into proteins, thus opening new avenues in research and applications across biology and medicine. In this review, we cover the principles of GCE, including the optimization of the aminoacyl-tRNA synthetase (aaRS)/tRNA system and the advancements in translation system engineering. Notable developments include the refinement of aaRS/tRNA pairs, enhancements in screening methods, and the biosynthesis of noncanonical amino acids. The applications of GCE technology span from synthetic biology, where it facilitates gene expression regulation and protein engineering, to medicine, with promising approaches in drug development, vaccine production, and gene editing. The review concludes with a perspective on the future of GCE, underscoring its potential to further expand the toolkit of biology and medicine. Through this comprehensive review, we aim to provide a detailed overview of the current state of GCE technology, its challenges, opportunities, and the frontier it represents in the expansion of the genetic code for novel biological research and therapeutic applications.
Collapse
Affiliation(s)
- Yujia Huang
- State
Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular
and Cellular Pharmacology, School of Pharmaceutical Sciences, Chemical
Biology Center, Peking University, Beijing 100191, China
| | - Pan Zhang
- Shenzhen
Key Laboratory for the Intelligent Microbial Manufacturing of Medicines,
Key Laboratory of Quantitative Synthetic Biology, Center for Synthetic
Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese
Academy of Sciences, Shenzhen 518055, P.R. China
| | - Haoyu Wang
- State
Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular
and Cellular Pharmacology, School of Pharmaceutical Sciences, Chemical
Biology Center, Peking University, Beijing 100191, China
| | - Yan Chen
- Shenzhen
Key Laboratory for the Intelligent Microbial Manufacturing of Medicines,
Key Laboratory of Quantitative Synthetic Biology, Center for Synthetic
Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese
Academy of Sciences, Shenzhen 518055, P.R. China
- University
of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Tao Liu
- State
Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular
and Cellular Pharmacology, School of Pharmaceutical Sciences, Chemical
Biology Center, Peking University, Beijing 100191, China
| | - Xiaozhou Luo
- Shenzhen
Key Laboratory for the Intelligent Microbial Manufacturing of Medicines,
Key Laboratory of Quantitative Synthetic Biology, Center for Synthetic
Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese
Academy of Sciences, Shenzhen 518055, P.R. China
- University
of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
49
|
Rafanan J, Ghani N, Kazemeini S, Nadeem-Tariq A, Shih R, Vida TA. Modernizing Neuro-Oncology: The Impact of Imaging, Liquid Biopsies, and AI on Diagnosis and Treatment. Int J Mol Sci 2025; 26:917. [PMID: 39940686 PMCID: PMC11817476 DOI: 10.3390/ijms26030917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Advances in neuro-oncology have transformed the diagnosis and management of brain tumors, which are among the most challenging malignancies due to their high mortality rates and complex neurological effects. Despite advancements in surgery and chemoradiotherapy, the prognosis for glioblastoma multiforme (GBM) and brain metastases remains poor, underscoring the need for innovative diagnostic strategies. This review highlights recent advancements in imaging techniques, liquid biopsies, and artificial intelligence (AI) applications addressing current diagnostic challenges. Advanced imaging techniques, including diffusion tensor imaging (DTI) and magnetic resonance spectroscopy (MRS), improve the differentiation of tumor progression from treatment-related changes. Additionally, novel positron emission tomography (PET) radiotracers, such as 18F-fluoropivalate, 18F-fluoroethyltyrosine, and 18F-fluluciclovine, facilitate metabolic profiling of high-grade gliomas. Liquid biopsy, a minimally invasive technique, enables real-time monitoring of biomarkers such as circulating tumor DNA (ctDNA), extracellular vesicles (EVs), circulating tumor cells (CTCs), and tumor-educated platelets (TEPs), enhancing diagnostic precision. AI-driven algorithms, such as convolutional neural networks, integrate diagnostic tools to improve accuracy, reduce interobserver variability, and accelerate clinical decision-making. These innovations advance personalized neuro-oncological care, offering new opportunities to improve outcomes for patients with central nervous system tumors. We advocate for future research integrating these tools into clinical workflows, addressing accessibility challenges, and standardizing methodologies to ensure broad applicability in neuro-oncology.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Department of Medical Education, Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (J.R.); (N.G.); (S.K.); (A.N.-T.); (R.S.)
| |
Collapse
|
50
|
Zohora FT, Pathmanathan R, Chowdhury EH. Application of Strontium Chloride Hexahydrate to Synthesize Strontium-Substituted Carbonate Apatite as a pH-Sensitive, Biologically Safe, and Highly Efficient siRNA Nanocarrier. ACS APPLIED BIO MATERIALS 2025; 8:348-367. [PMID: 39723844 DOI: 10.1021/acsabm.4c01319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Naked siRNAs are sensitive to enzymatic degradation, phagocytic entrapment, quick renal excretion, membrane impermeability, endosomal escape, and off-target effects. Designing a safe and efficient nanocarrier for siRNA delivery to the target site without toxicity remains a significant hurdle in gene therapy. CA is a unique derivative of hydroxyapatite and a highly pH-sensitive nanocarrier with strong particle aggregation and a high polydispersity index. Strontium (Sr2+), a group two divalent metal in the periodic table, has been reported for substituting calcium (Ca2+) ions from the apatite lattice and limiting particle growth/aggregation. This study used strontium chloride hexahydrate (SrCl2·6H2O) salt to develop a Sr-substituted CA (Sr-CA) nanocarrier with ∼30 nm size, spherical shape, less aggregation, homodispersity, and a fair anionic charge. Sr-CA demonstrated a large surface area-to-volume ratio, an improved cargo loading efficiency, and enhanced cellular uptake in HEK-293 cells. Moreover, Sr-CA is a pH-responsive nanocarrier responsible for its long physiological stability, efficient endosomal escape, and optimal cargo delivery within cells. These NPs have differential effects on MAPK1, MAP2K4, PIK3Ca, CAMK4, and p53 gene expression in HEK-293 cells without showing any significant cytotoxicity in cell growth properties. Gene silencing by Sr-CA-mediated siRNA delivery against MAPK1, MAP2K4, PIK3Ca, and CAMK4 genes significantly decreased the level of target gene expression and cell survival, demonstrating successful intracellular siRNA delivery in HEK-293 cells. Additionally, biocompatibility testing confirmed the biological safety of the Sr-CA nanocarrier in mice. These findings suggest that Sr-CA nanocarriers are a promising siRNA delivery system, combining high efficiency with pH-sensitive release and excellent biocompatibility, making them a viable option for future therapeutic applications.
Collapse
Affiliation(s)
- Fatema Tuz Zohora
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Rajadurai Pathmanathan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
- Daffodil International University, Daffodil Smart City, Birulia 1216, Bangladesh
- Nanoflex LLC, 31756 Broadwater Avenue, Leesburg, Florida 34748, United States
| |
Collapse
|