1
|
Huang P, Wang B, Xing Y, Wu Z, Li B, Liu L. Detection with Active Capture of Breast Cancer Marker MUC1 Using a MXene-Based Electrochemical Actuator. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40432219 DOI: 10.1021/acsami.5c05585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
The electrochemical aptamer-based detection of mucin 1 (MUC1) has been widely used for the early diagnosis and detection of breast cancer, which is of vital significance for reducing the incidence and mortality of this disease. However, the immutability of electrodes restricts the binding efficiency between electrodes and target biomolecules during the incubation process, limiting the ability of electrochemical aptasensors to efficiently and rapidly detect target molecules. Herein, the fabrication of a three-layer MXene-Au electrochemical actuator is reported for the active MUC1 detection. Using cyclic actuation characteristics achieved via the reversible insertion and detachment of ions in electrodes, the MXene-Au electrochemical actuator actively captures MUC1 in the region distant from the electrode during the incubation process. This improves the binding efficiency between the electrode and MUC1 and significantly reduces the aptamer-MUC1 binding time. The ultrasensitive and active detection of MUC1 was achieved with a detection limit of 0.65 fg mL-1, which was 2 orders of magnitude higher than that achieved during passive detection using the same structure.
Collapse
Affiliation(s)
- Peng Huang
- School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China
- Suzhou Research Institute of Southeast University, Suzhou 215123, People's Republic of China
| | - Bozheng Wang
- School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China
- Suzhou Research Institute of Southeast University, Suzhou 215123, People's Republic of China
| | - Youqiang Xing
- School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China
- Suzhou Research Institute of Southeast University, Suzhou 215123, People's Republic of China
| | - Ze Wu
- School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China
- Suzhou Research Institute of Southeast University, Suzhou 215123, People's Republic of China
| | - Bingjue Li
- School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China
- Suzhou Research Institute of Southeast University, Suzhou 215123, People's Republic of China
| | - Lei Liu
- School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China
- Suzhou Research Institute of Southeast University, Suzhou 215123, People's Republic of China
| |
Collapse
|
2
|
Gibadullin R, Suárez Ó, Lazaris FS, Gutiez N, Atondo E, Araujo-Aris S, Eguskiza A, Niu J, Kuhn AJ, Grosso AS, Rodriguez H, García-Martín F, Marcelo F, Santos T, Avenoza A, Busto JH, Peregrina JM, Gellman SH, Anguita J, Fiammengo R, Corzana F. Enhancing Cancer Vaccine Efficacy: Backbone Modification with β‑Amino Acids Alters the Stability and Immunogenicity of MUC1-Derived Glycopeptide Formulations. JACS AU 2025; 5:2270-2284. [PMID: 40443897 PMCID: PMC12117419 DOI: 10.1021/jacsau.5c00224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 05/07/2025] [Accepted: 05/07/2025] [Indexed: 06/02/2025]
Abstract
Glycopeptides derived from the mucin-1 (MUC1) glycoprotein hold significant promise as cancer vaccine candidates, but their clinical utility is limited by proteolytic degradation and the poor bioavailability of L-α-amino acid-based peptides. In this study, we demonstrate that substitution of multiple α-amino acids with homologous β-amino acids (same side chain, but extended backbone) in O-glycosylated MUC1 derivatives significantly enhances their proteolytic stability. We further show that α-to-β substitutions within the most immunogenic epitope of MUC1 impede binding to an anti-MUC1 antibody, while substitutions outside the same epitope preserve antibody recognition. Structural investigations using circular dichroism, NMR spectroscopy, and molecular dynamics simulations reveal that the strongest α/β-peptide binders retain native-like conformations in the epitope region, both in their unbound state and when bound to the anti-MUC1 antibody. Conjugation of these high-affinity α/β-peptide analogs to gold nanoparticles induces robust immune responses in mice comparable to that of the native glycopeptide. Additionally, these α/β-analogs elicit elevated levels of the cytokine IFNγ, one of the key proteins for tumor cell elimination, surpassing levels produced by the native MUC1 glycopeptide. In contrast, a low-affinity α/β-analogue with lower proteolytic stability produces minimal cytokine responses, underscoring the critical role of these biochemical properties in vaccine efficacy. Collectively, our findings highlight that α-to-β modifications in the peptide backbone offer an effective strategy for developing biostable, highly immunogenic glycopeptide-based cancer vaccines, exemplifying the power of structure-based rational design in advancing next-generation vaccines.
Collapse
Affiliation(s)
- Ruslan Gibadullin
- Department
of Chemistry, University of Wisconsin, 1101 University Avenue, Madison, Wisconsin53706, United States
| | - Óscar Suárez
- Departamento
de Química and Instituto de Investigación en Química
de la Universidad de La Rioja (IQUR), Universidad
de La Rioja, 26006Logroño, Spain
| | - Foivos S. Lazaris
- Departamento
de Química and Instituto de Investigación en Química
de la Universidad de La Rioja (IQUR), Universidad
de La Rioja, 26006Logroño, Spain
| | - Naiara Gutiez
- Inflammation
and Macrophage Plasticity Laboratory, CIC
BioGUNE, BRTA, 48160Derio, Spain
| | - Estibaliz Atondo
- Inflammation
and Macrophage Plasticity Laboratory, CIC
BioGUNE, BRTA, 48160Derio, Spain
| | - Sarai Araujo-Aris
- Inflammation
and Macrophage Plasticity Laboratory, CIC
BioGUNE, BRTA, 48160Derio, Spain
| | - Ander Eguskiza
- Department
of Biotechnology, University of Verona, 37134Verona, Italy
| | - Jiani Niu
- Department
of Chemistry, University of Wisconsin, 1101 University Avenue, Madison, Wisconsin53706, United States
| | - Ariel J. Kuhn
- Department
of Chemistry, University of Wisconsin, 1101 University Avenue, Madison, Wisconsin53706, United States
| | - Ana S. Grosso
- UCIBIO
- Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, 2829-516Caparica, Portugal
- Associate
Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, 2829-516Caparica, Portugal
| | - Héctor Rodriguez
- Inflammation
and Macrophage Plasticity Laboratory, CIC
BioGUNE, BRTA, 48160Derio, Spain
| | - Fayna García-Martín
- Departamento
de Química and Instituto de Investigación en Química
de la Universidad de La Rioja (IQUR), Universidad
de La Rioja, 26006Logroño, Spain
| | - Filipa Marcelo
- UCIBIO
- Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, 2829-516Caparica, Portugal
- Associate
Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, 2829-516Caparica, Portugal
| | - Tanausú Santos
- Departamento
de Química and Instituto de Investigación en Química
de la Universidad de La Rioja (IQUR), Universidad
de La Rioja, 26006Logroño, Spain
| | - Alberto Avenoza
- Departamento
de Química and Instituto de Investigación en Química
de la Universidad de La Rioja (IQUR), Universidad
de La Rioja, 26006Logroño, Spain
| | - Jesús H. Busto
- Departamento
de Química and Instituto de Investigación en Química
de la Universidad de La Rioja (IQUR), Universidad
de La Rioja, 26006Logroño, Spain
| | - Jesús M. Peregrina
- Departamento
de Química and Instituto de Investigación en Química
de la Universidad de La Rioja (IQUR), Universidad
de La Rioja, 26006Logroño, Spain
| | - Samuel H. Gellman
- Department
of Chemistry, University of Wisconsin, 1101 University Avenue, Madison, Wisconsin53706, United States
| | - Juan Anguita
- Inflammation
and Macrophage Plasticity Laboratory, CIC
BioGUNE, BRTA, 48160Derio, Spain
- Ikerbasque, Basque Foundation for Science, Maria Diaz de Haro 13, 48009Bilbao, Spain
| | - Roberto Fiammengo
- Department
of Biotechnology, University of Verona, 37134Verona, Italy
| | - Francisco Corzana
- Departamento
de Química and Instituto de Investigación en Química
de la Universidad de La Rioja (IQUR), Universidad
de La Rioja, 26006Logroño, Spain
| |
Collapse
|
3
|
Pandey SN, Babu MA, Ali H, H M, Maharana L, Goyal K, Rana M, Imran M. MUC1 as a diagnostic biomarker and siRNA-based therapeutic target in breast cancer: A clinical chemistry perspective. Clin Chim Acta 2025; 576:120387. [PMID: 40425136 DOI: 10.1016/j.cca.2025.120387] [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: 04/19/2025] [Revised: 05/24/2025] [Accepted: 05/24/2025] [Indexed: 05/29/2025]
Abstract
Breast cancer remains the leading cause of cancer mortality in women, and early detection coupled with real-time monitoring of tumor burden are clinical imperatives; yet existing imaging-based screening (e.g., mammography, ultrasound) suffers from sensitivities as low as 60-80% and even lower in dense breasts plus substantial false-positive rates, underscoring the critical need for molecular assays with higher accuracy. Current clinical assays for circulating MUC1 (CA15-3) achieve high specificity but exhibit limited sensitivity in early-stage disease, underscoring a critical unmet need for more sensitive, multiplexed biomarkers to enable timely intervention. Mass spectrometry-based glycoproteomic workflows offer multiplexed quantification of tumour-associated MUC1 glycoforms, substantially improving analytical specificity and dynamic range. Complementary liquid-biopsy platforms that detect anti-MUC1 autoantibodies further extend lead time for recurrence detection. Concurrently, small interfering RNA (siRNA) therapies targeting MUC1 delivered via ionizable lipid nanoparticles demonstrate efficient tumor accumulation, robust mRNA knockdown, and favourable safety in phaseI solid tumor trials. In this review, we critically assess the analytical performance and standardization challenges of current MUC1 assays, evaluate emerging mass spectrometry and immunoarray techniques, and examine chemical and nanocarrier strategies that surmount biological barriers to siRNA delivery. We propose a co-development framework for harmonized companion diagnostics and MUC1-directed RNAi therapeutics under unified regulatory pathways, paving the way for precision, biomarker-driven interventions in breast cancer care.
Collapse
Affiliation(s)
- Surya Nath Pandey
- Department of Pharmacology, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad 244001 Uttar Pradesh, India
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Malathi H
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Laxmidhar Maharana
- Department of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751030, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Mohd Imran
- Center For Health Research, Northern Border University, Arar 73213, Saudi Arabia.
| |
Collapse
|
4
|
Qusairy Z, Rada M. Glycosylation in cancer: mechanisms, diagnostic markers, and therapeutic applications. Mol Cell Biochem 2025:10.1007/s11010-025-05303-1. [PMID: 40389792 DOI: 10.1007/s11010-025-05303-1] [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: 03/04/2025] [Accepted: 05/04/2025] [Indexed: 05/21/2025]
Abstract
Glycosylation, a key post-translational modification, plays a pivotal role in cancer progression by influencing critical processes such as protein folding, immune modulation, and intercellular signaling. Altered glycosylation patterns are increasingly recognized as fundamental drivers of tumorigenesis, contributing to key cancer hallmarks like enhanced tumor migration, metastasis, and immune evasion. These aberrant glycosylation signatures not only offer insights into cancer biology but also serve as valuable diagnostic markers and potential therapeutic targets across a range of malignancies. This review explores the mechanisms underlying glycosylation alterations in cancer. We discuss the molecular basis of these changes, including genetic mutations, epigenetic regulation, and oncogene-driven shifts in glycosylation pathways. Additionally, we highlight recent advancements in glycomics research, with a focus on how these alterations influence tumor progression, angiogenesis, and the tumor microenvironment. Furthermore, the review considers the clinical implications of glycosylation changes, including their role in resistance to anti-cancer therapies and their potential as biomarkers for personalized treatment strategies. By bridging fundamental glycosylation research with clinical applications, this review underscores the promise of glycosylation as both a diagnostic tool and a therapeutic target in oncology, offering new avenues for improved patient stratification and precision medicine.
Collapse
Affiliation(s)
- Zahraa Qusairy
- McGill University Health Center Research Institute, Montreal, QC, H4A 3J1, Canada
| | - Miran Rada
- Medical Laboratory Science, Komar University of Science and Technology, Qularaisi, Sulaimani, Sulaymaniyah, Kurdistan Region, Iraq.
- Komar Cancer Research Program, Komar University of Science and Technology, Qularaisi, Sulaimani, Sulaymaniyah, Kurdistan Region, Iraq.
| |
Collapse
|
5
|
Yuan X, Li C, Gao J, Yang L, Wang B, Li Z. Glycosylation in T2 high and Th17 Asthma: A Narrative Review. J Asthma Allergy 2025; 18:545-558. [PMID: 40248104 PMCID: PMC12003201 DOI: 10.2147/jaa.s509940] [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: 12/03/2024] [Accepted: 03/03/2025] [Indexed: 04/19/2025] Open
Abstract
Glycosylation, a fundamental biochemical process, entails the covalent attachment of sugar molecules to proteins, DNA, or RNA. Beginning with an overview of the pathophysiological features of asthma, this review proceeds to elucidate various facets of glycosylation in asthma pathology, specifically in T2 high asthma and Th17-mediated responses. We examined glycosylation's involvement in regulating airway inflammation, encompassing the modulation of pro-inflammatory cytokine release such as IL-4, IL-5, and IL-13, key components of T2 inflammation, as well as its significance in modulating immune cell functionality, notably T cells and dendritic cells. Moreover, we explored glycosylation's impact on airway remodeling processes, including its regulation of airway smooth muscle cell proliferation and migration. Addressing molecular mechanisms, this review delved into several glycosylation modifications of proteins and genes implicated in asthma pathogenesis, including IgE, IL-4 receptor, TGF-β, and the regulation of select glycosylation enzymes. Additionally, the review highlights the role of Th17 cells in T2 high asthma and their modulation through glycosylation. We underscored future research imperatives, including biomarker discovery, therapeutic realization, and the potential utility of glycosylation modifications in asthma prevention and management. In short, this review provides an in-depth analysis of the critical role of glycosylation in the pathogenesis of T2 high asthma and Th17 responses.
Collapse
Affiliation(s)
- Xingxing Yuan
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
- Department of Medicine, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, People’s Republic of China
| | - Chaofan Li
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Jiawei Gao
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Liuxin Yang
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Bingyu Wang
- Department of Medicine, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, People’s Republic of China
| | - Zhuying Li
- Department of Respiratory, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| |
Collapse
|
6
|
Godfrey TM, Shanneik Y, Zhang W, Tran T, Verbeeck N, Patterson NH, Jackobs FE, Nagi C, Ramineni M, Eberlin LS. Integrating Ambient Ionization Mass Spectrometry Imaging and Spatial Transcriptomics on the Same Cancer Tissues to Identify RNA-Metabolite Correlations. Angew Chem Int Ed Engl 2025:e202502028. [PMID: 40216587 DOI: 10.1002/anie.202502028] [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: 01/24/2025] [Revised: 03/24/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025]
Abstract
Innovations in spatial omics technologies applied to human tissues have led to breakthrough discoveries in various diseases, including cancer. Two of these approaches-spatial transcriptomics and spatial metabolomics-have blossomed independently, fueled by technologies such as spatial transcriptomics (ST) and mass spectrometry imaging (MSI). Although powerful, these technologies only offer insights into the spatial distributions of restricted classes of molecules and have not yet been integrated to provide more holistic insights into biological questions. These techniques can be performed on adjacent serial sections from the same sample, but section-to-section variability can convolute data integration. We present a novel method combining desorption electrospray ionization mass spectrometry imaging (DESI-MSI) spatial metabolomics and Visium spatial transcriptomics on the same tissue sections. We show that RNA quality is maintained after performing DESI-MSI on a tissue under ambient conditions and that ST data is unperturbed following DESI-MSI. We demonstrate this workflow on human breast and lung cancer tissues and identify novel correlations between metabolites and mRNA transcripts in cancer-specific tissue regions.
Collapse
Affiliation(s)
- Trevor M Godfrey
- Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yasmin Shanneik
- Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | | | | | | | - Faith E Jackobs
- Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chandandeep Nagi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Maheshwari Ramineni
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Livia S Eberlin
- Department of Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
7
|
Yadav P, Rajendrasozhan S, Lajimi RH, Patel RR, Heymann D, Prasad NR. Circulating tumor cell markers for early detection and drug resistance assessment through liquid biopsy. Front Oncol 2025; 15:1494723. [PMID: 40260304 PMCID: PMC12009936 DOI: 10.3389/fonc.2025.1494723] [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: 09/11/2024] [Accepted: 03/18/2025] [Indexed: 04/23/2025] Open
Abstract
Circulating tumor cells (CTCs) are cancerous cells that extravasate from the primary tumor or metastatic foci and travel through the bloodstream to distant organs. CTCs provide crucial insights into cancer metastasis, the evolution of tumor genotypes during treatment, and the development of chemo- and/or radio-resistance during disease progression. The process of Epithelial-to-mesenchymal transition (EMT) plays a key role in CTCs formation, as this process enhances cell's migration properties and is often associated with increased invasiveness thereby leading to chemotherapy resistance. During the EMT process, tumor cells lose epithelial markers like EpCAM and acquire mesenchymal markers such as vimentin driven by transcription factors like Snail and Twist. CTCs are typically identified using specific cell surface markers, which vary depending on the cancer type. Common markers include EpCAM, used for epithelial cancers; CD44 and CD24, which are associated with cancer stem cells; and cytokeratins, such as CK8 and CK18. Other markers like HER2/neu and vimentin can also be used to target CTCs in specific cancer types and stages. Commonly, immune-based isolation techniques are being implemented for the isolation and enrichment of CTCs. This review emphasizes the clinical relevance of CTCs, particularly in understanding drug resistance mechanisms, and underscores the importance of EMT-derived CTCs in multidrug resistance (MDR). Moreover, the review also discusses CTCs-specific surface markers that are crucial for their isolation and enrichment. Ultimately, the EMT-specific markers found in CTCs could provide significant information to halt the disease progression and enable personalized therapies.
Collapse
Affiliation(s)
- Priya Yadav
- Department of Biochemistry and Biotechnology, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Saravanan Rajendrasozhan
- Department of Chemistry, College of Science, University of Ha’il, Ha’il, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Ha’il, Ha’il, Saudi Arabia
| | - Ramzi Hadj Lajimi
- Department of Chemistry, College of Science, University of Ha’il, Ha’il, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Ha’il, Ha’il, Saudi Arabia
| | - Raja Ramadevi Patel
- Medical and Diagnostic Research Centre, University of Ha’il, Ha’il, Saudi Arabia
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| | - Dominique Heymann
- Nantes Université, CNRS, US2B, UMR 6286, Nantes, France
- Institut de Cancérologie de l’Ouest, Tumor Heterogeneity and Precision Medecine Laboratory, Saint-Herblain, France
- Medical School, University of Sheffield, Sheffield, United Kingdom
| | - N. Rajendra Prasad
- Department of Biochemistry and Biotechnology, Annamalai University, Chidambaram, Tamil Nadu, India
| |
Collapse
|
8
|
Rajput SK, Minhas K, Azam I, Habib S, Shaikh U, Lalani EN. Prognostic implications of MUC1 and XBP1 concordant expression in multiple myeloma: A retrospective study. PLoS One 2025; 20:e0320934. [PMID: 40179083 PMCID: PMC11967961 DOI: 10.1371/journal.pone.0320934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Multiple myeloma (MM) is a disease of malignant plasma cells (PC) with poor survival. Disease progression and treatment relapse are attributed to MM cancer stem cells (CSCs) and signaling molecules such as MUC1 and XBP1. The study aimed to determine the prognostic value of expression of CSC-associated biomarkers, MUC1 and XBP1 in MM, which has not been explored previously. In this study, we determined the immunohistochemical expression of CSC markers (ALDH1, CD117, and CD34), MUC1, and XBP1 in 128 MM formalin-fixed paraffin-embedded bone marrow archival blocks. The expression of biomarkers was assessed for association with clinicopathological variables and patient survival. Descriptive analysis, survival plots and crude association between outcome and independent variables were assessed using Kaplan Meier and Log rank test. Univariate and multivariable analyses were performed using simple and multiple Cox regression models. The results are reported as crude and adjusted hazard ratios with 95% confidence intervals. Expression of ALDH1 and CD117 was found in 51% and 48% of the tumors, respectively. ALDH1 expression was associated with 1.83 years of reduced survival for patients with CD56-negative tumors. MUC1 expression was observed in 62%, whereas XBP1 was expressed in 48% of tumors. Combinatorial group analysis of XBP1 and MUC1 stratified patients into two prognostic groups. Cases with tumors negative for expression of MUC1 and XBP1 (XBP1-/ MUC1-) were categorized as a good prognostic group with increased survival of 3.42 years compared to cases with tumors expressing both (Worst prognosis, XBP1 + /MUC1+). Concordant expression of MUC1 and XBP1 in MM defines a subset of patients with adverse outcomes. The adjusted hazard ratio showed a four-fold increased risk of mortality associated with the concordant expression of MUC1 and XBP1 in patients > 65 years of age.
Collapse
Affiliation(s)
- Sheerien Kareem Rajput
- Centre for Regenerative Medicine and Stem Cell Research, The Aga Khan University, Karachi, Pakistan
| | - Khurram Minhas
- Department of Pathology and Laboratory Medicine, The Aga Khan University, Karachi, Pakistan
| | - Iqbal Azam
- Department of Community Health Sciences, The Aga Khan University, Karachi, Pakistan
| | - Sadia Habib
- Centre for Regenerative Medicine and Stem Cell Research, The Aga Khan University, Karachi, Pakistan
| | - Usman Shaikh
- Department of Pathology and Laboratory Medicine, The Aga Khan University, Karachi, Pakistan
| | - El-Nasir Lalani
- Centre for Regenerative Medicine and Stem Cell Research, The Aga Khan University, Karachi, Pakistan
| |
Collapse
|
9
|
Liu C, Liu N, Zhang T, Tu Y. Adoptive immune cell therapy for colorectal cancer. Front Immunol 2025; 16:1557906. [PMID: 40236691 PMCID: PMC11996668 DOI: 10.3389/fimmu.2025.1557906] [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: 01/09/2025] [Accepted: 02/28/2025] [Indexed: 04/17/2025] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related morbidity and mortality worldwide, with limited options for patients at advanced stages. Immunotherapy, particularly immune cell-based therapies, has gained significant attention as an innovative approach for targeting CRC. This review summarizes the progress in various immune cell therapies, including DC vaccine, CAR/TCR-T cells, CAR-NK cells et al, each engineered to recognize and attack cancer cells expressing specific antigens. CAR-T cell therapy, which has been successful in hematologic cancers, faces challenges in CRC due to the solid tumor microenvironment, which limits cell infiltration and persistence. CAR-NK cells, CAR-M and CAR-γδ T cells, however, offer alternative strategies due to their unique properties, such as the ability to target tumor cells without prior sensitization and a lower risk of inducing severe cytokine release syndrome. Recent advances in lentiviral transduction have enabled effective expression of CARs on NK and γδ T cells, providing promising preclinical results in CRC models. This review explores the mechanisms, tumor targets, preclinical studies, and early-phase clinical trials of these therapies, addressing key challenges such as enhancing specificity to tumor antigens and overcoming the immunosuppressive tumor microenvironment. The potential of combination therapies, including immune checkpoint inhibitors and cytokine therapy, is also discussed some as a means to improve the effectiveness of immune cell-based treatments for CRC. Continued research is essential to translate these promising approaches into clinical settings, offering new hope for CRC patients.
Collapse
Affiliation(s)
- Chenxiao Liu
- Guangdong Province Science and Technology Expert Workstation, Huizhou Central People’s Hospital, Huizhou, Guangdong, China
| | - Nan Liu
- Guangdong Province Science and Technology Expert Workstation, Huizhou Central People’s Hospital, Huizhou, Guangdong, China
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - Tongcun Zhang
- Guangdong Province Science and Technology Expert Workstation, Huizhou Central People’s Hospital, Huizhou, Guangdong, China
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - Yanyang Tu
- Science Research Center, Huizhou Central People’s Hospital, Huizhou, Guangdong, China
- Huizhou Central People’s Hospital Academy of Medical Sciences, Huizhou Central People’s Hospital, Huizhou, Guangdong, China
| |
Collapse
|
10
|
Teng C, Ma W, Liu J, Hou J, Zhang Y, Meng X, Xue Y, Wang Z, Wang J, Chen D, Sui Q, Gao Q, Li X, Li T, Zong C. Chemoenzymatic liquid-phase synthesis and immunogenic assessment of tumor-associated complex MUC1 glycopeptide variants. Int J Biol Macromol 2025; 302:140525. [PMID: 39892541 DOI: 10.1016/j.ijbiomac.2025.140525] [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: 10/02/2024] [Revised: 01/15/2025] [Accepted: 01/29/2025] [Indexed: 02/03/2025]
Abstract
Aberrantly glycosylated Mucin 1 (MUC1) is frequently over-expressed in epithelial cancers, making it an attractive target for cancer vaccines. Over the past two decades, multiple MUC1-based vaccines have been investigated clinically, yet they have generally shown limited efficacy due to challenges such as low immunogenicity, difficulty in overcoming immune tolerance, and potential issues related to glycosylation effects and antigen presentations. To advance the understanding of MUC1 vaccines, we report an efficient chemo-enzymatic approach for the preparation of four MUC1 antigen variants with different glycoforms through liquid-phase glycopeptide synthesis. These antigen were conjugated with CRM197 to generate various glycopeptide-protein conjugate vaccines, and their immunogenicity was evaluated based on total and subtype antibody titers, binding affinity, complement-dependent cytotoxicity (CDC) activity, and antibody-dependent cellular phagocytosis (ADCP). The combination of MPL and QS21 adjuvants with STn-MUC1-CRM197 conjugate induced a potent Th1-biased immune response, evidenced by elevating IgG2a titers and stronger antibody binding to MCF-7 cells. This formulation demonstrated superior CDC activity, ADCP activity and binding affinity to human breast cancer tissues in immuno-histochemical assays. The synergistic effect of specific adjuvants and glycosylated MUC1 conjugates offers a strategic avenue for MUC1 cancer vaccine development.
Collapse
Affiliation(s)
- Changcai Teng
- School of Pharmaceutical Sciences, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Wenjing Ma
- State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jinfeng Liu
- State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China; Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng 475004, China
| | - Juan Hou
- Clinical Laboratory, Binzhou Medical University Hospital, Binzhou 256600, China
| | - Yalong Zhang
- Department of Pathology, Binzhou Medical University Hospital, Binzhou 256600, China
| | - Xiongyan Meng
- School of Pharmaceutical Sciences, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Yannan Xue
- School of Pharmaceutical Sciences, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Zhen Wang
- School of Pharmaceutical Sciences, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - Jiajia Wang
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng 475004, China
| | - Dexiang Chen
- Maxvax Biotechnology Co., LTD, Chengdu 610200, China
| | - Qiang Sui
- Maxvax Biotechnology Co., LTD, Chengdu 610200, China
| | - Qi Gao
- Maxvax Biotechnology Co., LTD, Chengdu 610200, China
| | - Xia Li
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng 475004, China.
| | - Tiehai Li
- State Key Laboratory of Chemical Biology, Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Chengli Zong
- School of Pharmaceutical Sciences, Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China.
| |
Collapse
|
11
|
Li H, Yang T, Chen Y, Xie Z. Single cell RNA-seq and bulk RNA-seq analysis identifies MUC1 as a key gene for lung adenocarcinoma to neuroendocrine transformation. Transl Lung Cancer Res 2025; 14:824-841. [PMID: 40248742 PMCID: PMC12000963 DOI: 10.21037/tlcr-24-806] [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: 09/25/2024] [Accepted: 02/13/2025] [Indexed: 04/19/2025]
Abstract
Background Tyrosine kinase inhibitors (TKIs) have demonstrated significant effectiveness in treating advanced non-small cell lung cancer (NSCLC) harboring epidermal growth factor receptor (EGFR) mutations. Despite initial success, resistance to EGFR-TKIs inevitably occurs. One observed phenomenon in resistant lung cancers is the histological transformation from NSCLC to neuroendocrine carcinoma (NEC). The objective of this study is to explore and delineate the genetic and immune features linked to the transition from lung adenocarcinoma (LUAD) to NEC. Methods Bulk RNA-sequencing and Mendelian randomization (MR) analysis were utilized to identify candidate genes associated with the progression from LUAD to NEC. Expression quantitative trait locus data from publicly available databases were leveraged to pinpoint key genes in relevant tissues. Furthermore, the immune microenvironment was explored using weighted gene co-expression network analysis (WGCNA) and cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) databases. Single-cell RNA sequencing data from 16,765 cells across six tissue biopsy samples of LUAD and NEC were scrutinized to investigate cell interaction networks during histological transformation. The molecular pathways involved in dynamic cellular processes were elucidated through the analysis of cellular communication and pseudotime trajectory. Results Through the use of RNA-sequencing and MR analysis, it was determined that mucin-1 (MUC1) displayed a negative correlation with the progression of LUAD to NEC, as evidenced by its downregulation in clinical specimens. Additionally, MUC1 expression was found to be significantly correlated with the infiltration of diverse immune cell populations, notably CD8+ T cells. These results suggest a notable enrichment of neuron-related signaling pathways in the context of transformed NEC. Examination of immune cell infiltration in NEC indicated a reduction in the proportion of CD8+ central memory T cells, which has implications for the immune microenvironment and may point to potential therapeutic targets. Further investigation into cell-cell interactions among subpopulations identified the MIF-CD74 axis as the principal signaling pathway facilitating intercellular communication between immune cells and epithelial cells. Conclusions In conclusion, our study provides insights into the molecular landscape governing the LUAD-to-NEC transition, highlighting MUC1 as a potential biomarker. The immune microenvironment is believed to exert a substantial influence on histological transformation, particularly with regards to T cells.
Collapse
Affiliation(s)
- Hongxia Li
- Department of Pathology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tiantian Yang
- Department of Pathology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu Chen
- Department of Pathology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhiqin Xie
- Department of Hepatobiliary and Pancreatic Surgery, Medical Center of Digestive Disease, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| |
Collapse
|
12
|
Zhang N, Liu Q, Wang D, Wang X, Pan Z, Han B, He G. Multifaceted roles of Galectins: from carbohydrate binding to targeted cancer therapy. Biomark Res 2025; 13:49. [PMID: 40134029 PMCID: PMC11934519 DOI: 10.1186/s40364-025-00759-1] [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: 11/04/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Galectins play pivotal roles in cellular recognition and signaling processes by interacting with glycoconjugates. Extensive research has highlighted the significance of Galectins in the context of cancer, aiding in the identification of biomarkers for early detection, personalized therapy, and predicting treatment responses. This review offers a comprehensive overview of the structural characteristics, ligand-binding properties, and interacting proteins of Galectins. We delve into their biological functions and examine their roles across various cancer types. Galectins, characterized by a conserved carbohydrate recognition domain (CRD), are divided into prototype, tandem-repeat, and chimera types based on their structural configurations. Prototype Galectins contain a single CRD, tandem-repeat Galectins contain two distinct CRDs linked by a peptide, and the chimera-type Galectin-3 features a unique structural arrangement. The capacity of Galectins to engage in multivalent interactions allows them to regulate a variety of signaling pathways, thereby affecting cell fate and function. In cancer, Galectins contribute to tumor cell transformation, angiogenesis, immune evasion, and metastasis, making them critical targets for therapeutic intervention. This review discusses the multifaceted roles of Galectins in cancer progression and explores current advancements in the development of Galectin-targeted therapies. We also address the challenges and future directions for integrating Galectin research into clinical practice to enhance cancer treatment outcomes. In brief, understanding the complex functions of Galectins in cancer biology opens new avenues for therapeutic strategies. Continued research on Galectin interactions and their pathological roles is essential for developing effective carbohydrate-based treatments and improving clinical interventions for cancer patients.
Collapse
Affiliation(s)
- Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Qiao Liu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Daihan Wang
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Xiaoyun Wang
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Zhaoping Pan
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Gu He
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
13
|
Cheng L, Yang H, Tan S, Shi C, Zeng F, Yang W, Kong W. E2F4 Promotes Malignant Behaviors of Prostate Cancer Through Activating MUC1 Expression Transcriptionally. Asia Pac J Clin Oncol 2025. [PMID: 40110904 DOI: 10.1111/ajco.14164] [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: 02/14/2025] [Accepted: 02/27/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND The malignant features of prostate cancer (PC) threaten the patient's life. MUC1 was observably enhanced in PC. However, the reason for higher MUC1 expression in PC is still unclear and deserves to be further investigated. METHODS The abundance of MUC1 and E2F4 was evaluated using RT-qPCR in PC patients and PC cells. Pearson correlation coefficient analyzed the relationship between E2F4 and MUC1 in tissues from PC patients. Malignant phenotypes were examined using clone formation, scratch tests, transwell, and flow cytometry. The JASPAR website, luciferase activity assay, and ChIP were employed for validating interplays between E2F4 and the MUC1 promoter. RESULTS MUC1 and E2F4 were abnormally elevated in samples of PC patients and PC cells. MUC1 silencing resulted in suppression of growth and metastasis and promotion of cell apoptosis of PC cells. Additionally, E2F4 could provoke the transcriptional activity of MUC1 to enhance MUC1 expression. Furthermore, E2F4 knockdown inhibited malignant features of PC cells, which was abolished by MUC1 overexpression. CONCLUSION Our findings revealed that E2F4 silencing led to the suppression of growth and metastasis and the promotion of cell apoptosis of PC cells through reducing MUC1 expression, which offered targeting molecules for PC treatment.
Collapse
Affiliation(s)
- Long Cheng
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou City, China
- Department of Urology, The Affiliated Huizhou Hospital, Guangzhou Medical University, Huizhou City, China
| | - Haichao Yang
- Department of Urology, Huizhou No.2 Women's and Children's Healthcare Hospital, Huizhou City, China
| | - Shuoguo Tan
- Department of Hepatobiliary Surgery, The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou City, China
| | - Chongjun Shi
- Department of Urology, The Affiliated Huizhou Hospital, Guangzhou Medical University, Huizhou City, China
| | - Fanfei Zeng
- Department of Urology, The Affiliated Huizhou Hospital, Guangzhou Medical University, Huizhou City, China
| | - Weizhong Yang
- Department of Urology, The Affiliated Huizhou Hospital, Guangzhou Medical University, Huizhou City, China
| | - Weiqin Kong
- Department of Urology, The Affiliated Huizhou Hospital, Guangzhou Medical University, Huizhou City, China
| |
Collapse
|
14
|
Osman TE, Guo Y, Li S. Exploring the combined roles of GALNT1 and GALNT2 in hepatocellular carcinoma malignancy and EGFR modulation. Discov Oncol 2025; 16:337. [PMID: 40095226 PMCID: PMC11914428 DOI: 10.1007/s12672-025-02069-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC), the most formidable subtype of primary liver cancers, is becoming increasingly concerning due to its rising incidence worldwide. HCC ranks as the sixth most diagnosed cancer globally and is the third leading cause of cancer-related deaths. Glycosylation, a common post-translational modification of proteins, is frequently altered in tumors and is associated with the progression of malignancies. GALNT1 and GALNT2 are GalNAc-transferases that initiate protein O-glycosylation and are closely linked to cancer development. Investigating the relationship between GALNT1 and GALNT2 in HCC could provide new insights into the disease's pathogenesis. Thus, this study aimed to explore the combined effects of GALNT1 and GALNT2 transfection on HCC, compared to the effects of modifying each gene individually. MATERIALS AND METHODS GALNT1 and GALNT2 were assessed by bioinformatics, qPCR, and Western blot analyses to detect their expression in HCC tissues and cell lines. The effects of GALNT1/GALNT2 overexpression and knockdown on cell viability, proliferation, migration, invasion, and apoptosis were evaluated in HCC cells using CCK8, colony formation, transwell migration and invasion, wound healing, TUNEL, and flow cytometry assays. EGFR protein levels were also analyzed by Western blotting. RESULTS Co-transfection of GALNT1 knockdown with GALNT2 overexpression significantly suppressed proliferation, migration, and invasion, while promoting apoptosis in HCC cells. Conversely, co-transfection of GALNT1 overexpression with GALNT2 knockdown enhanced these malignant characteristics compared to the modified single gene. Notably, we observed that GALNT1 and GALNT2 modulated EGFR protein expression. Overall, our findings suggest that the combined activity of GALNT1 and GALNT2 is critical in regulating HCC malignant behaviors.
Collapse
Affiliation(s)
- Tagwa E Osman
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
- Clinical Laboratory Department, Dalian Medical University First Affiliated Hospital, Dalian, 116011, Liaoning Province, China
| | - Yanru Guo
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
- Clinical Laboratory Department, Dalian Medical University First Affiliated Hospital, Dalian, 116011, Liaoning Province, China
| | - Shijun Li
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China.
- Clinical Laboratory Department, Dalian Medical University First Affiliated Hospital, Dalian, 116011, Liaoning Province, China.
| |
Collapse
|
15
|
Renata S, Verma N, Peddinti RK. Surface-enhanced Raman spectroscopy as effective tool for detection of sialic acid as cancer biomarker. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 329:125631. [PMID: 39736186 DOI: 10.1016/j.saa.2024.125631] [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: 10/01/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 01/01/2025]
Abstract
Sialic acid, a negatively charged nine-carbon monosaccharide, is mainly located at the terminal end of glycan chains on glycoproteins and glycolipids of cell surface and most secreted proteins. Elevated levels of sialylated glycans have been known as a hallmark in numerous cancers. As a result, sialic acid acts as a useful and accessible cancer biomarker for early cancer detection and monitoring the disease development during cancer treatment which is crucial in elevating the survival rate. The detection of sialic acid has been done by many tools including surface-enhanced Raman spectroscopy (SERS) which gained incredible attention due to its high selectivity and sensitivity. However, currently, comprehensive reviews of sialic acid detection and imaging as a cancer biomarker using SERS are still lacking. Here, we present the significant breakthroughs in SERS-based detection of sialic acid levels on cells, tissues, and body fluids due to the presence of cancer, different cancer metastasis stages, and in response to the external stimuli. This review covers the SERS substrate and novel SERS strategies, using lectin, boronic acid, metabolic glycan labelling and label-free methods, for sialic acid detection as cancer biomarker. The remaining challenges to detect sialic acid and prospect of future development of SERS for other carbohydrate-based cancer biomarker, for instance fucose, are also discussed.
Collapse
Affiliation(s)
- Septila Renata
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| | - Nitish Verma
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India; Department of Chemistry, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria
| | - Rama Krishna Peddinti
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India.
| |
Collapse
|
16
|
Li F, Li Z, Zhao J, Zhang Q, Wu M, Guo Y. Nanocapsules with dual-targeting of cell and mitochondria functions for enhanced hypoxia-activated drug therapy. Chem Commun (Camb) 2025; 61:4375-4378. [PMID: 39989182 DOI: 10.1039/d5cc00337g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The cell and mitochondria dual-targeting nanocapsules reported here could exacerbate tumor hypoxia to activate a hypoxia-activated drug, tirapazamine, producing benzotriazinyl radicals and ˙OH, which are capable of killing tumor cells via DNA damage. In addition, mitochondrial dysfunction can result from the accumulation of ˙OH. This chain reaction triggers a surge in ROS that can effectively increase the therapeutic efficiency of the nanocapsules.
Collapse
Affiliation(s)
- Fen Li
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| | - Ziyi Li
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| | - Jing Zhao
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| | - Qingqing Zhang
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| | - Mengting Wu
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| | - Yingshu Guo
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
| |
Collapse
|
17
|
Arras W, Breugelmans T, Oosterlinck B, De Man JG, Malhotra-Kumar S, Abrams S, Van Laere S, Macken E, Somers M, Jauregui-Amezaga A, De Winter BY, Smet A. The Intestinal Mucin Isoform Landscape Reveals Region-Specific Biomarker Panels for Inflammatory Bowel Disease Patient Stratification. J Crohns Colitis 2025; 19:jjae155. [PMID: 39330996 PMCID: PMC11945306 DOI: 10.1093/ecco-jcc/jjae155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/26/2024] [Accepted: 09/26/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND AND AIMS Mucosal healing is considered a key therapeutic endpoint in inflammatory bowel diseases (IBD) and comprises endoscopic improvement of inflammation without taking barrier healing into account. Mucins are critical components of the mucosal barrier function that give rise to structurally diverse isoforms. Unraveling disease-associated mucin isoforms that could act as an indication for barrier function would greatly enhance IBD management. METHODS We present the intestinal mucin RNA isoform landscape in IBD and control patients using a targeted mucin isoform sequencing approach on a discovery cohort (n = 106). Random Forest modeling (n = 1683 samples) with external validation (n = 130 samples) identified unique mucin RNA isoform panels that accurately stratified IBD patients in multiple subpopulations based on inflammation, IBD subtype (Crohn's disease [CD], ulcerative colitis [UC]), and anatomical location of the intestinal tract (i.e. ileum, proximal colon, distal colon, and rectum). RESULTS Particularly, the mucin RNA isoform panels obtained from the inflamed UC and CD distal colon showed high performance in distinguishing inflamed biopsies from their control counterparts (AUC of 93.3% and 91.1% in the training, 95.0% and 96.0% in the test, and 89.5% and 78.3% in the external validation datasets, respectively). Furthermore, the differentially expressed MUC4 (PB.1238.363), MUC5AC (PB.2811.15), MUC16 (ENST00000397910.8), and MUC1 (ENST00000462317.5 and ENST00000620103.4) RNA isoforms frequently occurred throughout the different panels highlighting their role in IBD pathogenesis. CONCLUSIONS We unveiled region-specific mucin RNA isoform panels capturing the heterogeneity of the IBD patient population and showing great potential to indicate barrier function in IBD patients.
Collapse
Affiliation(s)
- Wout Arras
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Tom Breugelmans
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Baptiste Oosterlinck
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Joris G De Man
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Steven Abrams
- Global Health Institute, Department of Family Medicine and Population Health, University of Antwerp, Antwerp, Belgium
- Data Science Institute, Interuniversity Institute for Biostatistics and statistical Bioinformatics, University of Hasselt, Diepenbeek, Belgium
| | - Steven Van Laere
- Center for Oncological Research, Integrated Personalized and Precision Oncology Network, University of Antwerp, Antwerp, Belgium
| | - Elisabeth Macken
- Division of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Michaël Somers
- Division of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | | | - Benedicte Y De Winter
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Division of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Annemieke Smet
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
18
|
Ajadee A, Mahmud S, Sarkar A, Noor T, Ahmmed R, Haque Mollah MN. Screening of common genomic biomarkers to explore common drugs for the treatment of pancreatic and kidney cancers with type-2 diabetes through bioinformatics analysis. Sci Rep 2025; 15:7363. [PMID: 40025145 PMCID: PMC11873208 DOI: 10.1038/s41598-025-91875-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
Type 2 diabetes (T2D) is a crucial risk factor for both pancreatic cancer (PC) and kidney cancer (KC). However, effective common drugs for treating PC and/or KC patients who are also suffering from T2D are currently lacking, despite the probability of their co-occurrence. Taking disease-specific multiple drugs during the co-existence of multiple diseases may lead to adverse side effects or toxicity to the patients due to drug-drug interactions. This study aimed to identify T2D-, PC and KC-causing common genomic biomarkers (cGBs) highlighting their pathogenetic mechanisms to explore effective drugs as their common treatment. We analyzed transcriptomic profile datasets, applying weighted gene co-expression network analysis (WGCNA) and protein-protein interaction (PPI) network analysis approaches to identify T2D-, PC-, and KC-causing cGBs. We then disclosed common pathogenetic mechanisms through gene ontology (GO) terms, KEGG pathways, regulatory networks, and DNA methylation of these cGBs. Initially, we identified 78 common differentially expressed genes (cDEGs) that could distinguish T2D, PC, and KC samples from controls based on their transcriptomic profiles. From these, six top-ranked cDEGs (TOP2A, BIRC5, RRM2, ALB, MUC1, and E2F7) were selected as cGBs and considered targets for exploring common drug molecules for each of three diseases. Functional enrichment analyses, including GO terms, KEGG pathways, and regulatory network analyses involving transcription factors (TFs) and microRNAs, along with DNA methylation and immune infiltration studies, revealed critical common molecular mechanisms linked to PC, KC, and T2D. Finally, we identified six top-ranked drug molecules (NVP.BHG712, Irinotecan, Olaparib, Imatinib, RG-4733, and Linsitinib) as potential common treatments for PC, KC and T2D during their co-existence, supported by the literature reviews. Thus, this bioinformatics study provides valuable insights and resources for developing a genome-guided common treatment strategy for PC and/or KC patients who are also suffering from T2D.
Collapse
Affiliation(s)
- Alvira Ajadee
- Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Sabkat Mahmud
- Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Arnob Sarkar
- Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
- Department of Biochemistry & Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Tasfia Noor
- Department of Computer Science and Engineering, Rajshahi University of Engineering & Technology (RUET), Rajshahi, 6204, Bangladesh
| | - Reaz Ahmmed
- Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
- Department of Biochemistry & Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Nurul Haque Mollah
- Bioinformatics Lab, Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| |
Collapse
|
19
|
Li X, Yang F, Wang M, Huang X, Zeng X, Zhou L, Peng S, Zhang J. Unleashing the power of peptides in prostate cancer immunotherapy: mechanism, facts and perspectives. Front Pharmacol 2025; 16:1478331. [PMID: 40078274 PMCID: PMC11897510 DOI: 10.3389/fphar.2025.1478331] [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: 08/12/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Prostate cancer, the second most common cancer in men, often progresses to castration-resistant prostate cancer despite androgen deprivation therapy. Immunotherapy, revolutionary in cancer treatment, has limited efficacy in prostate cancer due to its "cold tumor" nature. Peptides, with unique advantages, offer new hope. This review explores how peptide-based tumor immunotherapy can transform prostate cancer from a "cold" to a "hot" state. It modulates the immunosuppressive tumor microenvironment by regulating non-immune cells (such as cancer-associated fibroblasts, endothelial cells, and adipose stromal cells), repolarizing tumor-associated macrophages, activating NK cells, and tuning cytokines. Additionally, peptides can induce immunogenic cell death (ICD) in prostate cancer cells through ferroptosis, pyroptosis, and autophagy modulation. The review also revisits existing prostate cancer immunotherapies, including immune checkpoint blockade, CAR T cell therapy, and dendritic cell vaccines, highlighting how peptides can enhance their effectiveness and safety. Finally, two peptide-based immunotherapy strategies in the development stage, peptide-integrated Proteolysis-Targeting Chimera therapy and peptide-involved epigenomic therapy, are introduced, showing great potential for future prostate cancer treatment.
Collapse
Affiliation(s)
- Xiaoya Li
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fang Yang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meijing Wang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaopeng Huang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Zeng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Zhou
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sixue Peng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingyi Zhang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
20
|
Wu Z, Chen Q, Lin Z, Chen Y, Gan X, He Y. Dual-functional probe for sensitive detection of MCF-7 cells and mendelian randomization analysis of MUC1 association with multiple cancers. Sci Rep 2025; 15:6167. [PMID: 39979505 PMCID: PMC11842835 DOI: 10.1038/s41598-025-90575-2] [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: 11/22/2024] [Accepted: 02/13/2025] [Indexed: 02/22/2025] Open
Abstract
The present study successfully developed a method based on a dual-functional probe for detecting breast cancer cells MCF-7 by recognizing the MUC1 protein on the cell surface. This method integrated inductively coupled plasma mass spectrometry (ICP-MS) and fluorescence imaging technology, enhancing the sensitivity, specificity, and accuracy of breast cancer cell detection. Additionally, through two-sample Mendelian randomization (MR) analysis, we verified a potential correlation between breast cancer and MUC1 (PIVW<0.05), while also proving no potential correlation between liver cancer and MUC1 (PIVW>0.05). Furthermore, this study explored the relationship between other cancers and MUC1, indicating a potential correlation between ovarian cancer and colorectal cancer with MUC1 (PIVW<0.05). In summary, this study provides new strategies for the early diagnosis and treatment of breast cancer and offers new insights into the potential of MUC1 as a biomarker for the detection of other cancers.
Collapse
Affiliation(s)
- Zhuzheng Wu
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, Fujian, China
| | - Qingquan Chen
- The School of Public Health, Fujian Medical University, Fuzhou, 350108, Fujian Province, China
| | - Zhifeng Lin
- The School of Public Health, Fujian Medical University, Fuzhou, 350108, Fujian Province, China
| | - Yating Chen
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaohao Gan
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Ye He
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
21
|
Sheikh KA, Amjad M, Irfan MT, Anjum S, Majeed T, Riaz MU, Jassim AY, Sharif EAM, Ibrahim WN. Exploring TGF-β Signaling in Cancer Progression: Prospects and Therapeutic Strategies. Onco Targets Ther 2025; 18:233-262. [PMID: 39989503 PMCID: PMC11846535 DOI: 10.2147/ott.s493643] [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: 08/29/2024] [Accepted: 11/19/2024] [Indexed: 02/25/2025] Open
Abstract
Cancer persists as a ubiquitous global challenge despite the remarkable advances. It is caused by uncontrolled cell growth and metastasis. The Transforming Growth Factor-beta (TGF-β) signaling pathway is considered a primary regulator of various normal physiological processes in the human body. Recently, factors determining the nature of TGF-β response have received attention, specifically its signaling pathway which can be an attractive therapeutic target for various cancer treatments. The TGF-β receptor is activated by its ligands and undergoes transduction of signals via canonical (SMAD dependent) or non-canonical (SMAD independent) signaling pathways regulating several cellular functions. Furthermore, the cross talk of the TGF-β signaling pathway cross with other signaling pathways has shown the controlled regulation of cellular functions. This review highlights the cross talk between various major signaling pathways and TGF-β. These signaling pathways include Wnt, NF-κB, PI3K/Akt, and Hedgehog (Hh). TGF-β signaling pathway has a dual role at different stages. It can suppress tumor formation at early stages and promote progression at advanced stages. This complex behaviour of TGF-β has made it a promising target for therapeutic interventions. Moreover, many strategies have been designed to control TGF-β signaling pathways at different levels, inhibiting tumor-promoting while enhancing tumor-suppressive effects, each with unique molecular mechanisms and clinical implications. This review also discusses various therapeutic inhibitors including ligand traps, small molecule inhibitors (SMIs), monoclonal antibodies (mAbs), and antisense oligonucleotides which target specific components of TGF-β signaling pathway to inhibit TGF-β signaling and are studied in both preclinical and clinical trials for different types of cancer. The review also highlights the prospect of TGF-β signaling in normal physiology and in the case of dysregulation, TGF-β inhibitors, and different therapeutic effects in cancer therapy along with the perspective of combinational therapies to treat cancer.
Collapse
Affiliation(s)
- Khansa Ali Sheikh
- Department of Biotechnology, Kinnaird College for Women, Lahore, Pakistan
| | - Momna Amjad
- Department of Biotechnology, Kinnaird College for Women, Lahore, Pakistan
| | | | - Sumaira Anjum
- Department of Biotechnology, Kinnaird College for Women, Lahore, Pakistan
| | - Tanveer Majeed
- Department of Biotechnology, Kinnaird College for Women, Lahore, Pakistan
| | - Muhammad Usman Riaz
- School of Computer Science, University College Dublin, Belfield, Dublin 4, Ireland
| | | | - Elham Abdullatif M Sharif
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Wisam Nabeel Ibrahim
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
22
|
Flores Banda JS, Gangane S, Raza F, Massarelli E. Current Development of Therapeutic Vaccines in Lung Cancer. Vaccines (Basel) 2025; 13:185. [PMID: 40006732 PMCID: PMC11860707 DOI: 10.3390/vaccines13020185] [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: 01/05/2025] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Cancer vaccines have a potential to change the current landscape of immunotherapy research and development. They target and neutralize specific tumor cells by utilizing the body's own immune system which offers a promising modality in treating various cancers including lung cancer. Historically, prior vaccination approaches specifically towards lung cancer have posed several challenges but also potential with early phase I/II trials showing improved overall survival. With better understanding of the body's immune system as well as advancements in vaccine development, the use of vaccines to target lung cancer cells in both non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) has shown promise but also challenges in the setting of advanced stage cancers, tumor resistance mechanisms, immune evasion, and tumor heterogeneity. The proposed solution is to enroll patients in the early stages of the disease, rather than waiting until progression occurs. Additionally, future efforts will focus on the targeted identification of specific and novel tumor neo-antigens. This review offers discussion and analysis of both completed and ongoing trials utilizing different strategies for vaccine development in relation to treating lung cancer as well as current challenges faced.
Collapse
Affiliation(s)
| | | | | | - Erminia Massarelli
- Department of Medicine, University of Texas at Tyler School of Medicine, 11937 US Hwy 271, Tyler, TX 75799, USA; (J.S.F.B.); (S.G.); (F.R.)
| |
Collapse
|
23
|
Dieli R, Lioy R, Crispo F, Cascelli N, Martinelli M, Lerose R, Telesca D, Milella MR, Colella M, Loperte S, Mazzoccoli C. The Oncoprotein Mucin 1 in Pancreatic Cancer Onset and Progression: Potential Clinical Implications. Biomolecules 2025; 15:275. [PMID: 40001578 PMCID: PMC11853026 DOI: 10.3390/biom15020275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy characterized by poor prognosis, therapeutic resistance, and frequent recurrence. Current therapeutic options for PDAC include surgery, radiotherapy, immunological and targeted approaches. However, all these therapies provide only a slight improvement in patient survival. Consequently, the discovery of novel specific targets is becoming a priority to develop more effective treatments for PDAC. Mucin 1 (MUC1), a transmembrane glycoprotein, is aberrantly glycosylated and frequently overexpressed in pancreatic cancer. Recent studies highlighted the role of this oncoprotein in pancreatic carcinogenesis and its involvement in the acquisition of typical aggressive features of PDAC, like local invasion, metastases, and drug resistance. This review explores the mechanisms by which MUC1 contributes to cancer onset and progression, with a focus on its potential role as a biomarker and novel therapeutic target for pancreatic adenocarcinoma treatment.
Collapse
Affiliation(s)
- Rosalia Dieli
- Laboratory of Pre-Clinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.D.); (R.L.); (N.C.); (M.M.); (C.M.)
| | - Rosa Lioy
- Laboratory of Pre-Clinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.D.); (R.L.); (N.C.); (M.M.); (C.M.)
| | - Fabiana Crispo
- Laboratory of Pre-Clinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.D.); (R.L.); (N.C.); (M.M.); (C.M.)
| | - Nicoletta Cascelli
- Laboratory of Pre-Clinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.D.); (R.L.); (N.C.); (M.M.); (C.M.)
| | - Mara Martinelli
- Laboratory of Pre-Clinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.D.); (R.L.); (N.C.); (M.M.); (C.M.)
| | - Rosa Lerose
- Hospital Pharmacy, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.L.); (D.T.); (M.R.M.)
| | - Donatella Telesca
- Hospital Pharmacy, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.L.); (D.T.); (M.R.M.)
| | - Maria Rita Milella
- Hospital Pharmacy, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.L.); (D.T.); (M.R.M.)
| | - Marco Colella
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Simona Loperte
- Institute of Methodologies for Environmental Analysis, National Research Council, 85050 Tito Scalo, Italy;
| | - Carmela Mazzoccoli
- Laboratory of Pre-Clinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (R.D.); (R.L.); (N.C.); (M.M.); (C.M.)
| |
Collapse
|
24
|
Wang F, Zhang R, Zhou Z, Shi R, Peng F, Xu Y, Yang S, Wang Z, Zhang P, Tu R, Zhang C, Liu X, Cai J. CAR-T therapy for endocrine neoplasms: novel targets and combination of therapies. Front Endocrinol (Lausanne) 2025; 16:1517525. [PMID: 40007813 PMCID: PMC11850254 DOI: 10.3389/fendo.2025.1517525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Endocrine malignancies constitute a heterogeneous tumour group with diverse biological characteristics. While typically indolent, they encompass aggressive types and presence of any metastatic sign indicates a high probability of recurrence and a diminished response to conventional therapies. Chimeric antigen receptor (CAR)-T cell immunotherapy has constituted a revolutionary advance in cancer treatment and exhibited significant potential for application in endocrine cancer. However, limited effectiveness was displayed in clinical application, which necessitates the exploration of novel modalities. Identification of specific and safe targets for endocrine cancer is the initial stage towards establishing a successful CAR-T treatment. Various therapies under investigation offer potential enhancements to CAR T cell efficacy through diverse mechanisms. Herein, we summarize recent advances in identifying targets of endocrine cancer for CAR therapy and provide an overview of combinatorial approaches.
Collapse
Affiliation(s)
- Fang Wang
- Department of Otolaryngology-Head and Neck Surgery, Xinyang Central Hospital, Xinyang, Henan, China
| | - Ruiqi Zhang
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Henan Joint International Pediatric Urodynamic Laboratory, Zhengzhou, China
| | - Zhaokai Zhou
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Henan Joint International Pediatric Urodynamic Laboratory, Zhengzhou, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuai Yang
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Henan Joint International Pediatric Urodynamic Laboratory, Zhengzhou, China
| | - Zhan Wang
- Department of Urology, First Affiliated Hospital of Zhengzhou University, Henan Joint International Pediatric Urodynamic Laboratory, Zhengzhou, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Rui Tu
- Department of Ultrasound, Xinyang Central Hospital, Xinyang, Henan, China
| | - Chun Zhang
- Department of Gastroenterology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xingchen Liu
- Department of Gynaecology, Xinyang Central Hospital, Xinyang, Henan, China
| | - Jun Cai
- Department of Gynaecology, Xinyang Central Hospital, Xinyang, Henan, China
| |
Collapse
|
25
|
Baker K, Eastwood TA, Garcia E, Lennon C, Mulvihill DP. Simple recombinant monoclonal antibody production from Escherichia coli. Open Biol 2025; 15:240229. [PMID: 39965660 PMCID: PMC11835484 DOI: 10.1098/rsob.240229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
Antibodies are valuable biological reagents used in a wide range of discovery research, biotechnology, diagnostic and therapeutic applications. Currently, both commercial and laboratory-scale antibody production is reliant on expression from mammalian cells, which can be time-consuming and requires the use of specialist facilities and costly growth reagents. Here, we describe a simple, rapid and cheap method for producing and isolating functional monoclonal antibodies and antibody fragments from bacterial cells that can be used in a range of laboratory applications. This simple method only requires access to basic microbial cell culture and molecular biology equipment, making scalable in-house antibody production accessible to the global diagnostics, therapeutics and molecular bioscience research communities.
Collapse
Affiliation(s)
- Karen Baker
- School of Biosciences, University of Kent, CanterburyCT2 7NJ, UK
| | - Tara A. Eastwood
- School of Biosciences, University of Kent, CanterburyCT2 7NJ, UK
| | - Esther Garcia
- Central Laser Facility, Research Complex at Harwell, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Harwell, Didcot, OxfordOX11 0FA, UK
| | - Chris Lennon
- Fujifilm-Diosynth Biotechnologies UK Ltd, Belasis Avenue, BillinghamTS23 1LH, UK
| | | |
Collapse
|
26
|
Mehrotra S, Kaur N, Kaur S, Matharoo K, Pandey RK. From antibodies to nanobodies: The next frontier in cancer theranostics for solid tumors. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:287-329. [PMID: 39978969 DOI: 10.1016/bs.apcsb.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
The field of cancer therapeutics has witnessed significant advancements over the past decades, particularly with the emergence of immunotherapy. This chapter traces the transformative journey from traditional antibody-based therapies to the innovative use of nanobodies in the treatment and diagnosis of solid tumors. Nanobodies are the smallest fragments of antibodies derived from camelid immunoglobulins and have redefined the possibilities in cancer theranostics due to their unique structural and functional properties. We provide an overview of the biochemical characteristics of nanobodies that make them particularly suitable for theranostic applications, such as their small size, high stability, enhanced infiltration into the complex tumor microenvironment (TME) and ability to bind with high affinity to epitopes that are inaccessible to conventional antibodies. Further, their ease of modification and functionalization has enabled the development of nanobody-based drug conjugates/toxins and radiolabeled compounds for precise imaging and targeted radiotherapy. We elucidate how nanobodies are being served as valuable tools for prognostic assessment, enabling clinicians to predict disease aggressiveness, monitor treatment response, and stratify patients for personalized therapeutic interventions.
Collapse
Affiliation(s)
- Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India.
| | - Navdeep Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Sukhpreet Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Kawaljit Matharoo
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | | |
Collapse
|
27
|
Ren G, Fan Y, Zhong R, Zou G, Huang X, Zhang Y. Relationship between mucin gene polymorphisms and different types of gallbladder stones. BMC Med Genomics 2025; 18:22. [PMID: 39885433 PMCID: PMC11783967 DOI: 10.1186/s12920-025-02090-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 01/21/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Gallstones, a common surgical condition globally, affect around 20% of patients. The development of gallstones is linked to abnormal cholesterol and bilirubin metabolism, reduced gallbladder function, insulin resistance, biliary infections, and genetic factors. In addition to these factors, research has shown that mucins play a role in gallstone formation. This study aims to explore the connection between different types of gallstones and mucin gene polymorphisms. METHODS For this purpose, a total of 121 patients with gallbladder stones PNS and 107 patients with healthy controls PNS were enrolled in this case-control study. One SNPs (rs4072037) of MUC1 gene、 three SNPs (rs2856111、rs41532344、rs41349846) of MUC2 gene、four SNPs (rs712005、rs2246980、rs2258447、rs2259292) of MUC4 gene、seven SNPs (rs28415193、rs56047977、rs2037089、rs2075854、rs3829224、rs2672785、rs2735709) of MUC5 gene、eight SNPs (rs10902268、rs61869016、rs573849895、rs59257210、rs7396383、rs74644072、rs7481521、rs9704308) of MUC6 gene、five SNPs (rs10229731、rs73168398、rs4729655、rs55903219、rs74974199) of MUC17 gene. We amplified SNP sites by polymerase chain reaction (PCR) using specific primer sets followed by DNA sequencing. RESULTS The frequencies of MUC2 rs2856111 C/T genotype (OR = 3.81, 95%CI: 1.06-13.68) was higher than the control group. MUC17 rs10229731 A/C genotype (OR = 0.33, 95%CI: 0.12-0.95), rs73168398 G/A genotype (OR = 0.26, 95%CI: 0.07-0.98), MUC6 rs10902268 G/A genotype (OR = 0.40, 95%CI: 0.17-0.95) at lower frequencies than controls. The frequencies of MUC2 rs41532344 T allele (OR = 2.55, 95%CI: 1.06-6.13), MUC4 rs712005 G allele (OR = 2.51, 95%CI: 1.20-5.22), MUC5B rs2037089 C allele (OR = 3.54, 95%CI: 1.14-11.01) and MUC5AC rs28415193 G allele (OR = 1.77, 95%CI: 1.02-3.07) were higher than the control group. MUC6 rs10902268 A allele (OR = 0.004, 95%CI: 0.00-0.27), rs61869016 C allele (OR = 0.07, 95%CI: 0.01-0.63) at lower frequencies than controls. CONCLUSIONS Polymorphisms in the mucin gene were linked to the formation of gallbladder stones. The MUC4 rs712005 G allele, MUC5B rs2037089 C allele, MUC2 rs41532344 T allele and MUC5AC rs28415193 G allele were found to predispose individuals to the development of the disease. MUC6 rs10902268 A allele and rs61869016 C allele were identified as protective factors. Meanwhile, MUC2 rs2856111 CT genotype was found to predispose individuals to the development of the disease. MUC17 rs10229731 AC genotype, rs73168398 GA genotype and MUC6 rs10902268 GA genotype were identified as protective factors.
Collapse
Affiliation(s)
- Gongqing Ren
- The Second Clinical Medical College of Jinan University, Department of Hepatobiliary Pancreatic Surgery, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Yongmao Fan
- Department of Thoracic Surgery, Shenzhen Luohu Hospital Group Luohu People's Hospital, Shenzhen, China
| | - Ruizi Zhong
- Breast Surgery, Shenzhen Futian District Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Gang Zou
- Department of Burns and Plastic Surgery, Shenzhen People's Hospital, Shenzhen, China
| | - Xiaojun Huang
- Department of Hepatobiliary Pancreatic Surgery, Shenzhen People's Hospital, No.1017 Dongmen North Road, Shenzhen, 518020, Guangdong Province, China
| | - Yue Zhang
- Department of Hepatobiliary Pancreatic Surgery, Shenzhen People's Hospital, No.1017 Dongmen North Road, Shenzhen, 518020, Guangdong Province, China.
| |
Collapse
|
28
|
Wu S, Wang Y, Yang Y, Yang C, Jiensi A, Geng C, Ju H, Chen Y. In Situ and In Vivo Evaluation of Multiplex Protein-Specific Glycosylation of Tumors with a Dual-SERS Encoding Strategy. Anal Chem 2025; 97:936-944. [PMID: 39705316 DOI: 10.1021/acs.analchem.4c05695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
A dual-SERS encoding strategy was designed for in situ and in vivo evaluation of multiplex protein-specific glycosylation of tumors. The dual-SERS encoding strategy consisted of two pairs of dual gold nanoprobes with different diameters of 10 and 30 nm, which were encoded with four different and distinguishable Raman signal molecules. The 10 and 30 nm gold nanoprobes (Au10 and Au30 probes, respectively) were further modified with lectins and aptamers to recognize the target glycans and proteins, respectively. After sequential binding to the target glycans and proteins, the adjacent Au10 and Au30 probes could emit strong surface-enhanced Raman scattering (SERS) signals to indicate the multiplex protein-specific glycosylation information on cells and in vivo, which can reveal in situ the distribution differences of different tumor markers in the central and marginal regions of tumors. This strategy has been successfully applied for in situ imaging and evaluation of the MUC1 and EpCAM-specific Sia and Gal/GalNAc information on cell surfaces and tumor xenografted mice, providing a convenient and powerful tool to study protein-specific glycosylation-related physiological and pathological mechanisms.
Collapse
Affiliation(s)
- Shan Wu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yuru Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yuhui Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Chaoyi Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ayidana Jiensi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Chengyao Geng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yunlong Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
29
|
Han H, Su H, Lv Z, Zhu C, Huang J. Identifying MTHFD1 and LGALS4 as Potential Therapeutic Targets in Prostate Cancer Through Multi-Omics Mendelian Randomization Analysis. Biomedicines 2025; 13:185. [PMID: 39857769 PMCID: PMC11759815 DOI: 10.3390/biomedicines13010185] [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: 12/18/2024] [Revised: 12/28/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Prostate cancer remains one of the leading causes of cancer-related mortality in men worldwide. The treatment of it is currently based on surgical removal, radiotherapy, and hormone therapy. It is crucial to improve therapeutic prospects for the diagnosis and treatment of prostate cancer via drug target screening. Methods: We integrated eQTL data from the eQTLGen Consortium and pQTL data from UK Biobank Proteome Plasma Proteins (UKB-PPP) and deCODE health datasets. MR analyses (SMR, heterogeneity in dependent instruments (HEIDI), IVW, Wald ratio, weighted median, and MR-Egger) were used to screen candidate genes associated with prostate adenocarcinoma (PRAD) risk. Candidate genes were further verified through TCGA-based gene expression profile, survival analysis, and immune microenvironment evaluations. TIDE analysis was utilized to investigate gene immunotherapy response. Single-cell RNA sequencing data from the GSE176031 dataset were used to investigate the gene expression patterns. The Drug Bank, Therapeutic Target Database and Drug Signatures Database were utilized to predict targeted drugs for candidate genes. Results: MTHFD1 and LGALS4 were identified as promising therapeutic targets for PRAD, with evidence provided at multi-omics levels. LGALS4 was predominantly expressed in malignant cells and was correlated with enhanced immune checkpoint pathways, increased TIDE scores, and immunotherapy resistance. In contrast, MTHFD1was expressed in both tumor and microenvironmental cells and was associated with poor survival. Drug target prediction suggested that there are no currently approved drugs specifically targeting MTHFD1 and LGALS4. Conclusions: Our study identified MTHFD1 and LGALS4 as potential preventive targets for PRAD. However, future experiments are warranted to assess the utility and effectiveness of these candidate proteins.
Collapse
Affiliation(s)
| | | | | | - Chengliang Zhu
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; (H.H.); (H.S.); (Z.L.)
| | - Jingtao Huang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; (H.H.); (H.S.); (Z.L.)
| |
Collapse
|
30
|
Hao Y, Hou DY, Zhou L, Fan YL, Wu XH, Liu YX, Xu YS, Song BL, Yi L, Qiao ZY, Wang H, Xu SP. Tumor-Specific Protein Induced in Situ Self-Assembly of Peptide Drugs for Synergistic Mitochondria Disruption. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413069. [PMID: 39604290 DOI: 10.1002/adma.202413069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/31/2024] [Indexed: 11/29/2024]
Abstract
Mitochondria-targeted cancer therapy is an effective method for controlling tumor growth. However, the presence of repair mechanisms in tumor cells in response to mitochondrial damage poses significant challenges for treatment. By taking advantage of intracellular self-assembly technology, a peptide nanomaterial, RC-K-FX, that enters tumor cells in a monomeric form is designed. After binding to MUC1-C inside the cell membrane, RC-K-FX assembles into a spherical structure that stably encapsulates MUC1-C, inhibiting its dimerization and blocking the repair of stress-induced mitochondrial damage in tumor cells. Moreover, the self-assembled mitochondrial toxic peptide effectively destroys the mitochondria, and the loss of mitochondrial repair significantly increases tumor cytotoxicity by disrupting the redox balance, enhancing reactive oxygen species (ROS), inhibiting the nuclear factor (NF)-κB pathway, and suppressing the epithelial-mesenchymal transition (EMT) process. After intravenous administration, RC-G-FX accumulated at the tumor site, exhibiting improved anti-tumor effects and extending the overall survival of tumor-bearing mice. Therefore, the integration of the in situ self-assembly of peptide drugs and damage to mitochondrial repair mechanisms provides effective therapeutic options for malignancy.
Collapse
Affiliation(s)
- Yi Hao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, P. R. China
| | - Da-Yong Hou
- National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Lei Zhou
- National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Yan-Lei Fan
- National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Xiu-Hai Wu
- National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Yi-Xuan Liu
- National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Yin-Sheng Xu
- National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Ben-Li Song
- National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Li Yi
- National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Zeng-Ying Qiao
- National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Hao Wang
- National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Shou-Ping Xu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, P. R. China
| |
Collapse
|
31
|
Ren XH, Guo T, Xu MF, Huang Y, Liao XR, Qi LJ, Cheng SX. A Multiple Targeting Genome Editing System for Remodulation of Circulating Malignant Cells to Eliminate Cancer Immunosuppression and Restore Immune Responses. Adv Healthc Mater 2025; 14:e2401223. [PMID: 39440615 DOI: 10.1002/adhm.202401223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/29/2024] [Indexed: 10/25/2024]
Abstract
Cancer immunotherapy, which aims to eliminate cancer immunosuppression and reactivate anticancer immunity, holds great promise in oncology treatments. However, it is challenging to accurately study the efficacy of immunotherapy based on human-derived cells through animal experiments due to xenogeneic immune rejection. Herein, a personalized and precise strategy to evaluate the effectiveness of immunotherapy using the blood samples of cancer patients is presented. Through the utilization of multiple cancer-targeting delivery system decorated with the epidermal growth factor receptor (EGFR)-specific aptamer CL4 and the AXL-specific aptamer GL21.T to achieve superior efficiency in delivering the genome editing plasmid for MUC1 knockout, effective modulation on the behavior of circulating malignant cells (CMCs) is realized. After genome editing, both mucin 1 (MUC1) and programmed death-ligand 1 (PD-L1) are significantly downregulated in CMCs. The elimination of immunosuppression results in markedly enhanced secretion of pro-inflammatory anticancer cytokines encompassing interleukins 2, 12, and 15 and interferon-γ by immune cells. The study not only provides a strategy to overcome immunosuppression but also yields critical insights for personalized immunotherapy approaches.
Collapse
Affiliation(s)
- Xiao-He Ren
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230011, China
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Tao Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, Anhui, 230011, China
| | - Ma-Fei Xu
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230011, China
| | - Yun Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, Anhui, 230011, China
| | - Xin-Ru Liao
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Li-Jin Qi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Si-Xue Cheng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| |
Collapse
|
32
|
Salido EM. Critical Roles of SEA Domains. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1468:477-481. [PMID: 39930241 DOI: 10.1007/978-3-031-76550-6_78] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
This mini-review delves into the multifaceted roles of SEA (sea urchin sperm protein, enterokinase, and agrin) domains, ubiquitous protein modules critical to the structure, and function of a wide range of membrane-associated and secreted proteins in organisms from yeast to humans. We explore the structural and functional characteristics of SEA domains based on their two types of fundamental characteristics: proteolytic and non-proteolytic SEA domains. We also examine the significance of SEA domains in different protein families, particularly in mucins and extracellular matrix proteins, emphasizing their roles in glycosylation, cell adhesion, and signal transduction. The review also highlights the crucial impact of SEA domains in health and disease contexts, with a focus on their implications in cancer progression and retinal health. Mutations within these domains are linked to a range of pathologies, including various cancers and congenital disorders, underscoring their clinical importance. Through this review, we aim to provide a deeper understanding of SEA domains, shedding light on their diverse biological functions and their potential as targets for therapeutic interventions in diseases where they play a pivotal role.
Collapse
Affiliation(s)
- Ezequiel M Salido
- Department of Biochemistry and Molecular Medicine, Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
33
|
Wu X, Xiong S, Tao L, Huang J, Shen X. Hairpin aptamer and ROS-sensitive microcapsule-mediated glycoprotein determination for the prognosis of colorectal cancer. Mikrochim Acta 2024; 192:21. [PMID: 39708094 DOI: 10.1007/s00604-024-06885-1] [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: 09/12/2024] [Accepted: 12/08/2024] [Indexed: 12/23/2024]
Abstract
A novel glycoprotein assay was developed by integrating the hairpin aptamer (H-APT)-mediated glycoprotein recognition and the reactive oxygen species-sensitive microcapsule (ROS-MC)-induced signal amplification. The analyzing process begins with the transfer of the target glycoprotein to a chlorin e6 (Ce6)-labeled DNA sequence via H-APT-mediated DNA displacement. Subsequently, the Ce6-labeled DNA was used to induce the disassembly of fluorophore-loaded ROS-MC under 650-nm light irradiation. Leveraging the rapid release of the fluorophore and the high loading capacity of the MC, this glycoprotein assay is capable of quantifying glycoprotein content in native biofluids within 2.5 h, achieving a detection limit of 0.034 ng/mL. We applied this assay to determine the glycoprotein composition in plasma samples of colorectal cancer patients, revealing a significant increase in glycoprotein content for those with a poor prognosis. In summary, we have developed an innovative method for glycoprotein determination that shows potential for clinical translation.
Collapse
Affiliation(s)
- Xingjie Wu
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang City and Guian New District, No.6 Ankang Avenue, Guizhou, 561113, China.
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang City and Guian New District, No.6 Ankang Avenue, Guizhou, 561113, China.
| | - Shasha Xiong
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang City and Guian New District, No.6 Ankang Avenue, Guizhou, 561113, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang City and Guian New District, No.6 Ankang Avenue, Guizhou, 561113, China
| | - Ling Tao
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang City and Guian New District, No.6 Ankang Avenue, Guizhou, 561113, China
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang City and Guian New District, No.6 Ankang Avenue, Guizhou, 561113, China
| | - Jian Huang
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China.
- School of Clinical Laboratory Science, Guian New District, Guizhou Medical University, University Town, Guizhou, 550025, China.
| | - Xiangchun Shen
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang City and Guian New District, No.6 Ankang Avenue, Guizhou, 561113, China.
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang City and Guian New District, No.6 Ankang Avenue, Guizhou, 561113, China.
| |
Collapse
|
34
|
Erickson A, Jackson LR, Camphausen K, Krauze AV. Mucins as Precision Biomarkers in Glioma: Emerging Evidence for Their Potential in Biospecimen Analysis and Outcome Prediction. Biomedicines 2024; 12:2806. [PMID: 39767713 PMCID: PMC11673638 DOI: 10.3390/biomedicines12122806] [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: 11/05/2024] [Revised: 12/06/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
Despite attempts at improving survival by employing novel therapies, progression in glioma is nearly universal. Precision biomarkers are critical to advancing outcomes; however, biomarkers for glioma are currently unknown. Most data on which the field can draw for biomarker identification comprise tissue-based analysis requiring the biospecimen to be removed from the tumor. Non-invasive specimen-based precision biomarkers are needed. Mucins are captured in tissue and blood and are increasingly studied in cancer, with several studies exploring their role as biomarkers to detect disease and monitor disease progression. CA125, also known as MUC16, is implemented as a biomarker in the clinic for ovarian cancer. Similarly, several mucins are membrane-bound, facilitating downstream signaling associated with tumor resistance and hallmarks of cancer. Evidence supports mucin expression in glioma cells with relationships to tumor detection, progression, resistance, and patient outcomes. The differential expression of mucins across tissues and organs could also provide a means of attributing signals measured in serum or plasma. In this review, we compiled existing research on mucins as candidate precision biomarkers in glioma, focusing on promising mucins in relationship to glioma and leading to a framework for mucin analysis in biospecimens as well as avenues for validation as data evolve.
Collapse
|
35
|
Wu Y, Ji X, Yang Y, Wu B. Discovery of a fully human antibody to the proximal membrane terminus of MUC1 based on a B-cell high-throughput screening technique. Int Immunopharmacol 2024; 142:113204. [PMID: 39317052 DOI: 10.1016/j.intimp.2024.113204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
Mucin 1 plays an important role in tumor signaling and is overexpressed in adenocarcinoma and the digestive system. Many antibodies have been developed against MUC1 targets. Previously developed antibodies were mainly directed against distal membrane-terminal MUC1-N, but distal membrane-terminal MUC1-N is shed during cell growth and therefore binds to antibodies developed against tandem repeat sequences and becomes ineffective. Here, we provide a simple and rapid method for preparing antibodies targeting the proximal membrane end of MUC1. Immunological target antigens were designed based on Biocytogen Renlite KO mice. With the help of B-cell high-throughput screening technology, we rapidly screened and prepared fully human antibodies with human-macaque cross-reactivity, high affinity, high specificity, and endocytosis. Using this method, we screened 40 antibodies with human-monkey cross-reactivity, which specifically recognized breast cancer cell lines with human and monkey affinities ranging from (1.04E-07-2.91E-09). Of these, the antibodies with germline genes IGHV4-59*01 and IGHV3-30*03 had nanomolar affinities, with high endocytosis effects in breast cancer cells. Ab.07 (IGHV3-30*03) coupled with monomethyl auristatin E (MMAE) showed good anti-tumor activity in different tumor cells. In summary, we describe a method for designing and producing excellent antibodies that can be assembled into antibody-drug conjugates and bispecific antibodies by proximal-membrane-end immunization and B-cell high-throughput screening that can rapidly generate high-quality antibodies.
Collapse
Affiliation(s)
- Yilin Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Institute of Antibody and Drug Research, Biocytogen (Beijing) Pharmaceutical Technology Co., Ltd, Beijing 102609, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226133, China
| | - Xin Ji
- Institute of Antibody and Drug Research, Biocytogen (Beijing) Pharmaceutical Technology Co., Ltd, Beijing 102609, China.
| | - Yi Yang
- Institute of Antibody and Drug Research, Biocytogen (Beijing) Pharmaceutical Technology Co., Ltd, Beijing 102609, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226133, China.
| | - Bo Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
36
|
Guerreiro A, Compañón I, Lazaris FS, Labão-Almeida C, Oroz P, Ghirardello M, Marques MC, Corzana F, Bernardes GJL. Non-Natural MUC1 Glycopeptide Homogeneous Cancer Vaccine with Enhanced Immunogenicity and Therapeutic Activity. Angew Chem Int Ed Engl 2024; 63:e202411009. [PMID: 39275921 DOI: 10.1002/anie.202411009] [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: 06/11/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 09/16/2024]
Abstract
Glycopeptides derived from the glycoprotein mucin-1 (MUC1) have shown potential as tumor-associated antigens for cancer vaccine development. However, their low immunogenicity and non-selective conjugation to carriers present significant challenges for the clinical efficacy of MUC1-based vaccines. Here, we introduce a novel vaccine candidate based on a structure-guided design of an artificial antigen derived from MUC1 glycopeptide. This engineered antigen contains two non-natural amino acids and has an α-S-glycosidic bond, where sulfur replaces the conventional oxygen atom linking the peptide backbone to the sugar N-acetylgalactosamine. The glycopeptide is then specifically conjugated to the immunogenic protein carrier CRM197 (Cross-Reactive Material 197), a protein approved for human use. Conjugation involves selective reduction and re-bridging of a disulfide in CRM197, allowing the attachment of a single copy of MUC1. This strategy results in a chemically defined vaccine while maintaining both the structural integrity and immunogenicity of the protein carrier. The vaccine elicits a robust Th1-like immune response in mice and generates antibodies capable of recognizing human cancer cells expressing tumor-associated MUC1. When tested in mouse models of colon adenocarcinoma and pancreatic cancer, the vaccine is effective both as a prophylactic and therapeutic use, significantly delaying tumor growth. In therapeutic applications, improved outcomes were observed when the vaccine was combined with an anti-programmed cell death protein 1 (anti-PD-1) checkpoint inhibitor. Our strategy reduces batch-to-batch variability and enhances both immunogenicity and therapeutic potential. This site-specific approach disputes a prevailing dogma where glycoconjugate vaccines require multivalent display of antigens.
Collapse
Affiliation(s)
- Ana Guerreiro
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
- Basinnov Lifesciences, Av. José Malhoa 2, Escritório 3.7, 1070-325, Lisboa, Portugal
| | - Ismael Compañón
- Departamento de Química and Instituto de Investigación en Química de la Universidad de La Rioja (IQUR), Madre de Dios, 53, 26006, Logroño, Spain
| | - Foivos S Lazaris
- Departamento de Química and Instituto de Investigación en Química de la Universidad de La Rioja (IQUR), Madre de Dios, 53, 26006, Logroño, Spain
| | - Carlos Labão-Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Paula Oroz
- Departamento de Química and Instituto de Investigación en Química de la Universidad de La Rioja (IQUR), Madre de Dios, 53, 26006, Logroño, Spain
| | - Mattia Ghirardello
- Departamento de Química and Instituto de Investigación en Química de la Universidad de La Rioja (IQUR), Madre de Dios, 53, 26006, Logroño, Spain
| | - Marta C Marques
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Francisco Corzana
- Departamento de Química and Instituto de Investigación en Química de la Universidad de La Rioja (IQUR), Madre de Dios, 53, 26006, Logroño, Spain
| | - Gonçalo J L Bernardes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
- Basinnov Lifesciences, Av. José Malhoa 2, Escritório 3.7, 1070-325, Lisboa, Portugal
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW, Cambridge, UK
| |
Collapse
|
37
|
Thapa R, Gupta S, Gupta G, Bhat AA, Smriti, Singla M, Ali H, Singh SK, Dua K, Kashyap MK. Epithelial-mesenchymal transition to mitigate age-related progression in lung cancer. Ageing Res Rev 2024; 102:102576. [PMID: 39515620 DOI: 10.1016/j.arr.2024.102576] [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: 09/05/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Epithelial-Mesenchymal Transition (EMT) is a fundamental biological process involved in embryonic development, wound healing, and cancer progression. In lung cancer, EMT is a key regulator of invasion and metastasis, significantly contributing to the fatal progression of the disease. Age-related factors such as cellular senescence, chronic inflammation, and epigenetic alterations exacerbate EMT, accelerating lung cancer development in the elderly. This review describes the complex mechanism among EMT and age-related pathways, highlighting key regulators such as TGF-β, WNT/β-catenin, NOTCH, and Hedgehog signalling. We also discuss the mechanisms by which oxidative stress, mediated through pathways involving NRF2 and ROS, telomere attrition, regulated by telomerase activity and shelterin complex, and immune system dysregulation, driven by alterations in cytokine profiles and immune cell senescence, upregulate or downregulate EMT induction. Additionally, we highlighted pathways of transcription such as SNAIL, TWIST, ZEB, SIRT1, TP53, NF-κB, and miRNAs regulating these processes. Understanding these mechanisms, we highlight potential therapeutic interventions targeting these critical molecules and pathways.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Smriti
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhav Singla
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
38
|
Cui H, Yu Q, Xu Q, Lin C, Zhang L, Ye W, Yang Y, Tian S, Zhou Y, Sun R, Meng Y, Yao N, Wang H, Cao F, Liu M, Ma J, Liao C, Sun R. EGFR and MUC1 as dual-TAA drug targets for lung cancer and colorectal cancer. Front Oncol 2024; 14:1433033. [PMID: 39664199 PMCID: PMC11631732 DOI: 10.3389/fonc.2024.1433033] [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: 05/15/2024] [Accepted: 10/23/2024] [Indexed: 12/13/2024] Open
Abstract
Background Epidermal growth factor receptor (EGFR) is a key protein in cellular signaling that is overexpressed in many human cancers, making it a compelling therapeutic target. On-target severe skin toxicity has limited its clinical application. Dual-targeting therapy represents a novel approach to overcome the challenges of EGFR-targeted therapies. Methods A single-cell tumor-normal RNA transcriptomic meta-atlas of lung adenocarcinoma (LUAD) and normal lung tissues was constructed from published data. Tumor associated antigens (TAAs) were screened from the genes which were expressed on cell surface and could distinguish cancer cells from normal cells. Expression of MUC1 and EGFR in tumors and normal tissues was detected by immunohistochemistry (IHC), bulk transcriptomic and single-cell transcriptomic analyses. RNA cut-off values were calculated using paired analysis of RNA sequencing and IHC in patient-derived tumor xenograft samples. They were used to estimate the abundance of EGFR- and MUC-positive subjects in The Cancer Genome Atlas Program (TCGA) database. Survival analysis of EGFR and MUC1 expression was carried out using the transcription and clinical data from TCGA. Results A candidate TAA target, transmembrane glycoprotein mucin 1 (MUC1), showed strong expression in cancer cells and low expression in normal cells. Single-cell analysis suggested EGFR and MUC1 together had better tumor specificity than the combination of EGFR with other drug targets. IHC data confirmed that EGFR and MUC1 were highly expressed on LUAD and colorectal cancer (CRC) clinical samples but not on various normal tissues. Notably, co-expression of EGFR and MUC1 was observed in 98.4% (n=64) of patients with LUAD and in 91.6% (n=83) of patients with CRC. It was estimated that EGFR and MUC1 were expressed in 97.5% of LUAD samples in the TCGA dataset. Besides, high expression of EGFR and MUC1 was significantly associated with poor prognosis of LUAD and CRC patients. Conclusions Single-cell RNA, bulk RNA and IHC data demonstrated the high expression levels and co-expression patterns of EGFR and MUC1 in tumors but not normal tissues. Therefore, it is a promising TAA combination for therapeutic targeting which could enhance on-tumor efficacy while reducing off-tumor toxicity.
Collapse
Affiliation(s)
- Huilin Cui
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qianqian Yu
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Qumiao Xu
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Chen Lin
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Long Zhang
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Wei Ye
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Yifei Yang
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Sijia Tian
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Yilu Zhou
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Runzhe Sun
- School of Basic Medicine, Shanxi Medical University, Jinzhong, China
| | - Yongsheng Meng
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Ningning Yao
- Department of Radiobiology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Haizhen Wang
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Feilin Cao
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Meilin Liu
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Jinfeng Ma
- Department of Hepatobiliary and Pancreatogastric Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Cheng Liao
- Department of Translational Medicine, Shanghai Shengdi Medicine Co. Ltd., Shanghai, China
| | - Ruifang Sun
- Department of Tumor Biobank, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
39
|
Bao X, Yu H, Chen Z, Chen W, Xiao Y, Wu X, Li Z. C1GALT1-mediated O-glycan T antigen increase enhances the migration and invasion ability of gastric cancer cells. Biochem Biophys Res Commun 2024; 734:150641. [PMID: 39243676 DOI: 10.1016/j.bbrc.2024.150641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/07/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
Gastric cancer (GC) is one of the most aggressive and lethal diseases in the world. Cancer metastasis is the mainly leading cause of death in GC patients. Aberrant Protein O-glycosylation is closely associated with tumor occurrence and metastasis. However, the effect of aberrant O-glycosylation on the progress of GC is not completely clear. This study aimed to investigate the biological function and its underlying effects mechanism of core 1 β 1, 3-galactosyltransferase 1 (C1GALT1) C1GALT1-mediated O-glycan T antigen on GC progress. We conducted data mining analysis that C1GALT1 was obviously up-regulated in GC tissues than in para-carcinoma tissues. Elevated expression of C1GALT1 was closely associated with advanced TNM stage, lymph node metastasis, histological grade, and poor overall survival. In addition, C1GALT1 overexpression could promote GC cell proliferation, migration, and invasion, which was due to C1GALT1 overexpression-mediated O-glycan T antigen increase. Moreover, MUC1 was predicted to be a new downstream target of C1GALT1, which may be abnormally O-glycosylated by C1GALT1 thereby activating the cell adhesion signaling pathway. In conclusion, our studies proved that C1GALT1-mediated O-glycosylation increase could promote the metastasis of gastric cancer cells. These discoveries hint that C1GALT1 may serve as a novel therapeutic target for GC treatment.
Collapse
Affiliation(s)
- Xiaojuan Bao
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Hanjie Yu
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Zhuo Chen
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Wentian Chen
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Yaqing Xiao
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Xin Wu
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Zheng Li
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, 710069, China.
| |
Collapse
|
40
|
Gofron K, Berezowski A, Gofron M, Borówka M, Dziedzic M, Kazimierczak W, Kwiatkowski M, Gofron M, Nowaczyk Z, Małgorzewicz S. Akkermansia muciniphila - impact on the cardiovascular risk, the intestine inflammation and obesity. Acta Biochim Pol 2024; 71:13550. [PMID: 39611203 PMCID: PMC11602308 DOI: 10.3389/abp.2024.13550] [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: 07/16/2024] [Accepted: 11/05/2024] [Indexed: 11/30/2024]
Abstract
Contemporary scientific discussions are increasingly focusing on Akkermansia muciniphila due to its complex influence on intestinal physiology. This article provides a comprehensive analysis of the various effects Akkermansia muciniphila has on intestinal inflammation, while also exploring its potential associations with obesity and cardiovascular diseases. A systematic literature search was conducted using PubMed, Google Scholar, and ResearchGate with the following keywords: Akkermansia muciniphila, obesity, cardiovascular risk, and inflammatory bowel diseases. The aim of our mini-review was to examine the impact of Akkermansia bacteria on the intestines, cardiovascular system, and its relationship with obesity. Through a detailed review of current literature, the article seeks to elucidate the complex interactions of Akkermansia muciniphila within the human body, highlighting its potential contributions to health improvement and medical interventions. Research indicates that Akkermansia muciniphila positively correlates with maintaining intestinal health, modulating the cardiovascular system, and aiding in weight management. However, the number of studies available is small, and the effects of Akkermansia muciniphila on human health require further research.
Collapse
Affiliation(s)
- Krzysztof Gofron
- Students’ Circle of Clinical Nutrition, Medical University of Gdańsk, Gdańsk, Poland
| | - Adam Berezowski
- Department of Urology and Kidney Transplantation, Nikolay Pirogov Provincial Specialist Hospital, Łódź, Poland
| | - Maksymilian Gofron
- Urology Department, Municipal Teaching Hospital in Częstochowa, Częstochowa, Poland
| | - Małgorzata Borówka
- Department of Otolaryngology, Laryngological Oncology, Audiology and Phoniatrics, Medical University of Łódź, Łódź, Poland
| | - Michał Dziedzic
- Students’ Circle of Clinical Nutrition, Medical University of Gdańsk, Gdańsk, Poland
| | - Wojciech Kazimierczak
- Students’ Circle of Clinical Nutrition, Medical University of Gdańsk, Gdańsk, Poland
| | - Maciej Kwiatkowski
- Department of Orthopedics and Traumatology, Medical University of Warsaw, Warszawa, Poland
| | - Maria Gofron
- Students’ Circle of Clinical Nutrition, Medical University of Gdańsk, Gdańsk, Poland
| | - Zuzanna Nowaczyk
- Students’ Circle of Clinical Nutrition, Medical University of Gdańsk, Gdańsk, Poland
| | - Sylwia Małgorzewicz
- Department of Clinical Nutrition, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
41
|
Mao W, Zhang H, Wang K, Geng J, Wu J. Research progress of MUC1 in genitourinary cancers. Cell Mol Biol Lett 2024; 29:135. [PMID: 39491020 PMCID: PMC11533421 DOI: 10.1186/s11658-024-00654-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
MUC1 is a highly glycosylated transmembrane protein with a high molecular weight. It plays a role in lubricating and protecting mucosal epithelium, participates in epithelial cell renewal and differentiation, and regulates cell adhesion, signal transduction, and immune response. MUC1 is expressed in both normal and malignant epithelial cells, and plays an important role in the diagnosis, prognosis prediction and clinical monitoring of a variety of tumors and is expected to be a new therapeutic target. This article reviews the structural features, expression regulation mechanism, and research progress of MUC1 in the development of genitourinary cancers and its clinical applications.
Collapse
Affiliation(s)
- Weipu Mao
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, Jiangsu, China.
| | - Houliang Zhang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, Jiangsu, China
| | - Keyi Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| | - Jiang Geng
- Department of Urology, Bengbu First People's Hospital, Bengbu, People's Republic of China.
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China.
| | - Jianping Wu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, No. 87 Dingjiaqiao, Gulou District, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
42
|
Bates M, Mohamed BM, Lewis F, O'Toole S, O'Leary JJ. Biomarkers in high grade serous ovarian cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189224. [PMID: 39581234 DOI: 10.1016/j.bbcan.2024.189224] [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: 01/28/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024]
Abstract
High-grade serous ovarian cancer (HGSC) is the most common subtype of ovarian cancer. HGSC patients typically present with advanced disease, which is often resistant to chemotherapy and recurs despite initial responses to therapy, resulting in the poor prognosis associated with this disease. There is a need to utilise biomarkers to manage the various aspects of HGSC patient care. In this review we discuss the current state of biomarkers in HGSC, focusing on the various available immunohistochemical (IHC) and blood-based biomarkers, which have been examined for their diagnostic, prognostic and theranostic potential in HGSC. These include various routine clinical IHC biomarkers such as p53, WT1, keratins, PAX8, Ki67 and p16 and clinical blood-borne markers and algorithms such as CA125, HE4, ROMA, RMI, ROCA, and others. We also discuss various components of the liquid biopsy as well as a number of novel IHC biomarkers and non-routine blood-borne biomarkers, which have been examined in various ovarian cancer studies. We also discuss the future of ovarian cancer biomarker research and highlight some of the challenges currently facing the field.
Collapse
Affiliation(s)
- Mark Bates
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland.
| | - Bashir M Mohamed
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Faye Lewis
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Sharon O'Toole
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin, Ireland
| |
Collapse
|
43
|
Li S, Gao R, Han X, Wang K, Kang B, Ma X. MALAT1/miR-582-5p/GALNT1/MUC1 axis modulates progression of AML leukemia stem cells by regulating JAK2/STAT3 pathway. Ann Hematol 2024:10.1007/s00277-024-06043-w. [PMID: 39428449 DOI: 10.1007/s00277-024-06043-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: 04/14/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024]
Abstract
Acute myeloid leukemia (AML) is characterized by uncontrolled clonal expansion and differentiation block of immature myeloid cells. Some studies have shown that leukemia stem cells (LSC) are thought to be responsible for the initiation and development of leukemia. Moreover, abnormal O-glycosylation is a key modification in the process of cancer malignancy. In this study, GALNT1 expression was significantly upregulated in LSCs, while knockdown of GALNT1 inhibited cell viability and promoted apoptosis. Importantly, GALNT1 was the direct target of miR-582-5P, and MALAT1 directly interacted with miR-582-5P. In addition, Our investigation corroborated that MALAT1 functioned as an endogenous sponge of miR-582-5P to regulate mucin1 (MUC1) expression, catalyzed by GALNT1, which modulated the activity of JAK2/STAT3 pathway. MALAT1 and MUC1 were targets of transcription factor STAT3 and were regulated by STAT3. In general, these new findings indicated that MALAT1/miR-582-5P/GALNT1 axis is involved in the progression of LSCs, illuminating the possible mechanism mediated by O-glycosylated MUC1 via JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Si Li
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, Liaoning, 116011, China
| | - Rui Gao
- Department of Blood Transfusion, Dalian Municipal Central Hospital, Dalian, 116033, Liaoning, P.R. China
| | - Xu Han
- The Institute of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning, P.R. China
| | - Kai Wang
- The Institute of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning, P.R. China
| | - Bingyu Kang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, Liaoning, 116011, China
| | - Xiaolu Ma
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Dalian Medical University, Zhongshan Road 222, Dalian, Liaoning, 116011, China.
| |
Collapse
|
44
|
Wolters-Eisfeld G, Oliveira-Ferrer L. Glycan diversity in ovarian cancer: Unraveling the immune interplay and therapeutic prospects. Semin Immunopathol 2024; 46:16. [PMID: 39432076 PMCID: PMC11493797 DOI: 10.1007/s00281-024-01025-6] [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: 11/09/2023] [Accepted: 09/12/2024] [Indexed: 10/22/2024]
Abstract
Ovarian cancer remains a formidable challenge in oncology due to its late-stage diagnosis and limited treatment options. Recent research has revealed the intricate interplay between glycan diversity and the immune microenvironment within ovarian tumors, shedding new light on potential therapeutic strategies. This review seeks to investigate the complex role of glycans in ovarian cancer and their impact on the immune response. Glycans, complex sugar molecules decorating cell surfaces and secreted proteins, have emerged as key regulators of immune surveillance in ovarian cancer. Aberrant glycosylation patterns can promote immune evasion by shielding tumor cells from immune recognition, enabling disease progression. Conversely, certain glycan structures can modulate the immune response, leading to either antitumor immunity or immune tolerance. Understanding the intricate relationship between glycan diversity and immune interactions in ovarian cancer holds promise for the development of innovative therapeutic approaches. Immunotherapies that target glycan-mediated immune evasion, such as glycan-based vaccines or checkpoint inhibitors, are under investigation. Additionally, glycan profiling may serve as a diagnostic tool for patient stratification and treatment selection. This review underscores the emerging importance of glycan diversity in ovarian cancer, emphasizing the potential for unraveling immune interplay and advancing tailored therapeutic prospects for this devastating disease.
Collapse
Affiliation(s)
- Gerrit Wolters-Eisfeld
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | |
Collapse
|
45
|
Dyachenko EI, Bel’skaya LV. Salivary Transmembrane Mucins of the MUC1 Family (CA 15-3, CA 27.29, MCA) in Breast Cancer: The Effect of Human Epidermal Growth Factor Receptor 2 (HER2). Cancers (Basel) 2024; 16:3461. [PMID: 39456554 PMCID: PMC11506585 DOI: 10.3390/cancers16203461] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
The MUC1 family of transmembrane glycoproteins (CA 15-3, CA 27.29, MCA) is aberrantly expressed among patients with breast cancer. Objectives: to measure the level of degradation products of MUC1, including CA 15-3, CA 27.29, and MCA, in the saliva of breast cancer patients and to describe the biochemical processes that influence their expression and the regulation of their biological functions. Methods: The case-control study included three groups (breast cancer, fibroadenomas, and healthy controls). All study participants provided saliva samples strictly before starting treatment. The levels of MUC1, including CA 15-3, CA 27.29, and MCA, free progesterone and estradiol, cytokines (MCP-1, VEGF, TNF-α, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-18), and amino acids (Asp, Gln, Gly, His, Leu + Ile, Orn, Phe, Pro, Tyr) were determined. Results: It was shown that the levels of the MUC1 family in the saliva of patients with HER2-positive breast cancer were significantly lower compared to the control group. The level of pro-inflammatory cytokines and the level of free estradiol affected the expression of MUC1. We obtained a reliable relationship between the aggressive nature of tumor growth, an increased level of pro-inflammatory cytokines, a low level of free estradiol, and the suppressed expression of salivary MUC1. Conclusions: Among patients with aggressive breast cancer, a high level of pro-inflammatory cytokines, and a low level of free estradiol, there was an inhibition of the expression of pathologically unchanged glycoprotein MUC1 in saliva.
Collapse
Affiliation(s)
| | - Lyudmila V. Bel’skaya
- Biochemistry Research Laboratory, Omsk State Pedagogical University, 644099 Omsk, Russia;
| |
Collapse
|
46
|
McKeague ML, Lohmueller J, Dracz MT, Saadallah N, Ricci ED, Beckwith DM, Ayyalasomayajula R, Cudic M, Finn OJ. Preventative Cancer Vaccine-Elicited Human Anti-MUC1 Antibodies Have Multiple Effector Functions. Antibodies (Basel) 2024; 13:85. [PMID: 39449327 PMCID: PMC11503386 DOI: 10.3390/antib13040085] [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: 09/10/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND/OBJECTIVES Mucin-1 (MUC1) is a transmembrane glycoprotein that is overexpressed and hypoglycosylated in premalignant and malignant epithelial cells compared to normal cells, creating a target antigen for humoral and cellular immunity. Healthy individuals with a history of advanced colonic adenomas and at high risk for colon cancer were enrolled in a clinical trial to evaluate the feasibility of using a MUC1 peptide vaccine to prevent colon cancer. Anti-MUC1 antibodies elicited by this vaccine were cloned using peripheral blood B cells and sera collected two weeks after a one-year booster. Twelve of these fully human monoclonal antibodies (mAb) were tested for binding to MUC1+ target cells, and three with the highest binding were further evaluated for various effector functions important for tumor rejection. METHODS Immune cells were incubated together with target cells expressing variations in the number, distance, and membrane anchoring properties of the MUC1 epitope in the presence of each mAb. RESULTS All three mAbs mediated antibody-dependent cytokine release (ADCR), antibody-dependent cellular cytotoxicity (ADCC), and antibody-dependent cellular phagocytosis (ADCP). Two also mediated antibody-dependent trogocytosis/trogoptosis (ADCT). None were capable of complement-dependent cytotoxicity (CDC). CONCLUSIONS ADCP and ADCT functions were more efficient when antibodies bound epitopes proximal to and anchored to the membrane, providing insight for future therapeutic antibody validation strategies.
Collapse
Affiliation(s)
- Michelle L. McKeague
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.L.); (O.J.F.)
| | - Jason Lohmueller
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.L.); (O.J.F.)
- Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Matthew T. Dracz
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.L.); (O.J.F.)
| | - Najla Saadallah
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.L.); (O.J.F.)
| | - Eric D. Ricci
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.L.); (O.J.F.)
- Department of Psychology, Dietrich College of Humanities and Social Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Donella M. Beckwith
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Ramya Ayyalasomayajula
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Maré Cudic
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Olivera J. Finn
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA; (J.L.); (O.J.F.)
| |
Collapse
|
47
|
Yao Y, Fan D. Advances in MUC1 resistance to chemotherapy in pancreatic cancer. J Chemother 2024; 36:449-456. [PMID: 38006297 DOI: 10.1080/1120009x.2023.2282839] [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: 07/26/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/27/2023]
Abstract
The incidence of pancreatic cancer (PC), a highly fatal malignancy, is increasing every year. Chemotherapy is an important treatment for it in addition to surgery, yet most patients become resistant to chemotherapeutic agents within a few weeks of treatment initiation. MUC1 is a highly glycosylated transmembrane protein, and studies have shown that aberrantly glycosylated overexpression of MUC1 is involved in regulating the biology of chemoresistance in cancer cells. This article summarizes the mechanism of MUC1 in PC chemoresistance and reviews MUC1-based targeted therapies.
Collapse
Affiliation(s)
- Youhao Yao
- The Fifth Clinical Medical College of Shanxi Medical University, Shanxi, PR China
- Surgery Department, Shanxi Provincial People's Hospital, Taiyuan, PR China
| | - Daguang Fan
- Surgery Department, Shanxi Provincial People's Hospital, Taiyuan, PR China
| |
Collapse
|
48
|
Peng H, Jiang L, Yuan J, Wu X, Chen N, Liu D, Liang Y, Xie Y, Jia K, Li Y, Feng X, Li J, Zhang X, Shen L, Chen Y. Single-cell characterization of differentiation trajectories and drug resistance features in gastric cancer with peritoneal metastasis. Clin Transl Med 2024; 14:e70054. [PMID: 39422697 PMCID: PMC11488346 DOI: 10.1002/ctm2.70054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Gastric cancer patients with peritoneal metastasis (GCPM) experience a rapidly deteriorating clinical trajectory characterized by therapeutic resistance and dismal survival, particularly following the development of malignant ascites. However, the intricate dynamics within the peritoneal microenvironment (PME) during the treatment process remain largely unknown. METHODS Matched samples from primary tumours (PT), peritoneal metastases (PM), and paired pre-treatment and post-chemo/immunotherapy (anti-PD-1/PD-L1) progression malignant ascites samples, were collected from 48 patients. These samples were subjected to single-cell RNA sequencing (n = 30), multiplex immunofluorescence (n = 30), and spatial transcriptomics (n = 3). Furthermore, post hoc analyses of a phase 1 clinical trial (n = 20, NCT03710265) and an in-house immunotherapy cohort (n = 499) were conducted to validate the findings. RESULTS Tracing the evolutionary trajectory of epithelial cells unveiled the terminally differentially MUC1+ cancer cells with a high epithelial-to-mesenchymal transition potential, and they demonstrated spatial proximity with fibroblasts and endothelial cells, correlating with poor prognosis. A significant expansion of macrophage infiltrates, which exhibited the highest proangiogenic activity, was observed in the ascites compared with PT and PM. Besides, higher C1Q+ macrophage infiltrates correlated with significantly lower GZMA+ T-lymphocyte infiltrates in therapeutic failure cases, potentially mediated by the LGALS9-CD45 and SPP1-CD44 ligand-receptor interactions. In the chemoresistant group, intimate interactions between C1Q+ macrophages and fibroblasts through the complement activation pathway were found. In the group demonstrating immunoresistance, heightened TGF-β production activity was detected in MUC1+ cancer cells, and they were skewed to interplay with C1Q+ macrophages through the GDF15-TGF-βR2 axis. Ultimately, post hoc analyses indicated that co-targeting TGF-β and PDL1 pathways may confer superior clinical benefits than sole anti-PD-1/PD-L1 therapy for patients presenting with GCPM at the time of diagnosis. CONCLUSIONS Our findings elucidated the cellular differentiation trajectories and crucial drug resistance features within PME, facilitating the exploration of effective targets for GCPM treatment. HIGHLIGHTS MUC1+ cancer cells with a high epithelial-to-mesenchymal transition potential and exhibiting spatial proximity to fibroblasts and endothelial cells constitute the driving force of gastric cancer peritoneal metastasis (GCPM). Higher C1Q+ macrophage infiltrates correlated with significantly lower GZMA+ T-lymphocyte infiltrates within the peritoneal microenvironment in therapeutic failure cases. Co-targeting TGF-β and PDL1 pathways may confer superior clinical benefits than sole anti-PD-1/PD-L1 therapy for patients presenting with GCPM at diagnosis.
Collapse
Affiliation(s)
- Haoxin Peng
- Department of Gastrointestinal OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and InstituteBeijingChina
| | - Lei Jiang
- Department of Gastrointestinal OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and InstituteBeijingChina
| | - Jiajia Yuan
- Department of Gastrointestinal OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and InstituteBeijingChina
| | - Xiangrong Wu
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Nan Chen
- Department of Gastrointestinal Surgery IIIKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and InstituteBeijingChina
| | - Dan Liu
- Department of Gastrointestinal OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and InstituteBeijingChina
| | - Yueting Liang
- Department of Radiation OncologyPeking University Cancer Hospital and InstituteBeijingChina
| | - Yi Xie
- Department of Gastrointestinal OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and InstituteBeijingChina
| | - Keren Jia
- Department of Gastrointestinal OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and InstituteBeijingChina
| | - Yanyan Li
- Department of Gastrointestinal OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and InstituteBeijingChina
| | - Xujiao Feng
- Department of Gastrointestinal OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and InstituteBeijingChina
| | - Jian Li
- Department of Gastrointestinal OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and InstituteBeijingChina
| | - Xiaotian Zhang
- Department of Gastrointestinal OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and InstituteBeijingChina
| | - Lin Shen
- Department of Gastrointestinal OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and InstituteBeijingChina
| | - Yang Chen
- Department of Gastrointestinal OncologyKey Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and InstituteBeijingChina
- Department of Gastrointestinal CancerBeijing GoBroad HospitalBeijingChina
| |
Collapse
|
49
|
Sugiura S, Ikeda M. Supramolecular materials constructed from synthetic glycopeptides via aqueous self-assembly and their bioapplications in immunotherapy. Org Biomol Chem 2024; 22:7287-7306. [PMID: 39189690 DOI: 10.1039/d4ob01116c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Synthetic glycopeptides capable of self-assembly in aqueous environments form a range of supramolecular nanostructures, such as nanoparticles and nanofibers, owing to their amphiphilic nature and the diverse structures of the saccharides introduced. These glycopeptide-based supramolecular materials are promising for immunotherapy applications because of their biocompatibility and multivalent saccharide display, which enhances lectin-saccharide interactions. This review highlights recent advances in the molecular design of synthetic glycopeptide-based supramolecular materials and their use as immunomodulatory agents.
Collapse
Affiliation(s)
- Shintaro Sugiura
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Masato Ikeda
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.
- Institute for Glyco-core Research (iGCORE), Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Institute for Advanced Study, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| |
Collapse
|
50
|
Ayyalasomayajula R, Boneva I, Ormaza D, Whyte A, Farook K, Gorlin Z, Yancey E, André S, Kaltner H, Cudic M. Synthesis and Thermodynamic Evaluation of Sialyl-Tn MUC1 Glycopeptides Binding to Macrophage Galactose-Type Lectin. Chembiochem 2024; 25:e202400391. [PMID: 38877657 PMCID: PMC11560554 DOI: 10.1002/cbic.202400391] [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: 04/29/2024] [Revised: 05/30/2024] [Accepted: 06/14/2024] [Indexed: 06/16/2024]
Abstract
Interactions between the tumor-associated carbohydrate antigens of Mucin 1 (MUC1) and the carbohydrate-binding proteins, lectins, often lead to the creation of a pro-tumor microenvironment favoring tumor initiation, progression, metastasis, and immune evasion. Macrophage galactose binding lectin (MGL) is a C-type lectin receptor found on antigen-presenting cells that facilitates the uptake of carbohydrate antigens for antigen presentation, modulating the immune response homeostasis, autoimmunity, and cancer. Considering the crucial role of tumor-associated forms of MUC1 and MGL in tumor immunology, a thorough understanding of their binding interaction is essential for it to be exploited for cancer vaccine strategies. The synthesis of MUC1 glycopeptide models carrying a single or multiple Tn and/or sialyl-Tn antigen(s) is described. A novel approach for the sialyl-Tn threonine building block suitable for the solid phase peptide synthesis was developed. The thermodynamic profile of the binding interaction between the human MGL and MUC1 glycopeptide models was analyzed using isothermal titration calorimetry. The measured dissociation constants for the sialyl-Tn-bearing peptide epitopes were consistently lower compared to the Tn antigen and ranged from 10 μM for mono- to 1 μM for triglycosylated MUC1 peptide, respectively. All studied interactions, regardless of the glycan's site of attachment or density, exhibited enthalpy-driven thermodynamics.
Collapse
Affiliation(s)
- Ramya Ayyalasomayajula
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431
| | - Ivet Boneva
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431
| | - David Ormaza
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431
| | - Andrew Whyte
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431
| | - Kamran Farook
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431
| | - Zachary Gorlin
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431
| | - Evelyn Yancey
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431
| | - Sabine André
- Department of Veterinary Sciences, Physiological Chemistry, Ludwig-Maximilians-Universität München, Lena-Christ-Str. 48, 82152, Planegg-Martinsried
| | - Herbert Kaltner
- Department of Veterinary Sciences, Physiological Chemistry, Ludwig-Maximilians-Universität München, Lena-Christ-Str. 48, 82152, Planegg-Martinsried
| | - Maré Cudic
- Department of Chemistry and Biochemistry, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431
| |
Collapse
|