1
|
Gostomczyk K, Drozd M, Marsool Marsool MD, Pandey A, Tugas K, Chacon J, Tayyab H, Ullah A, Borowczak J, Szylberg Ł. Biomarkers for the detection of circulating tumor cells. Exp Cell Res 2025; 448:114555. [PMID: 40228709 DOI: 10.1016/j.yexcr.2025.114555] [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: 10/22/2024] [Revised: 04/05/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Circulating tumor cells (CTCs) have emerged as a key biomarker in cancer detection and prognosis, and their molecular profiling is gaining importance in precision oncology. Liquid biopsies, which allow the extraction of CTCs, circulating tumor DNA (ctDNA) or cell-free DNA (cfDNA), have measurable advantages over traditional tissue biopsies, especially when molecular material is difficult to obtain. However, this method is not without limitations. Difficulties in differentiating between primary and metastatic lesions, uncertain predictive values and the complexity of the biomarkers used can prove challenging. Recently, high cell heterogeneity has been identified as the main obstacle to achieving high diagnostic accuracy. Because not all cells undergo epithelial-mesenchymal transition (EMT) at the same time, there is a large population of hybrid CTCs that express both epithelial and mesenchymal markers. Since traditional diagnostic tools primarily detect epithelial markers, they are often unable to detect cells with a hybrid phenotype; therefore, additional markers may be required to avoid false negatives. In this review, we summarize recent reports on emerging CTCs markers, with particular emphasis on their use in cancer diagnosis. Most of them, including vimentin, TWIST1, SNAI1, ZEB1, cadherins, CD44, TGM2, PD-L1 and GATA, hold promise for the detection of CTCs, but are also implicated in cancer progression, metastasis, and therapeutic resistance. Therefore, understanding the nature and drivers of epithelial-mesenchymal plasticity (EMP) is critical to advancing our knowledge in this field.
Collapse
Affiliation(s)
- Karol Gostomczyk
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, Poland; Department of Tumor Pathology and Pathomorphology, Oncology Center - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland; Department of Pathology, Dr Jan Biziel Memorial University Hospital, Bydgoszcz, Poland.
| | - Magdalena Drozd
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, Poland; Department of Pathology, Dr Jan Biziel Memorial University Hospital, Bydgoszcz, Poland
| | | | - Anju Pandey
- Memorial Sloan Kettering Cancer Center, New York, USA
| | | | - Jose Chacon
- American University of Integrative Sciences, Saint Martin, Cole Bay, Barbados
| | | | - Ashraf Ullah
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jędrzej Borowczak
- Department of Clinical Oncology, Oncology Center - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland
| | - Łukasz Szylberg
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, Poland; Department of Tumor Pathology and Pathomorphology, Oncology Center - Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland; Department of Pathology, Dr Jan Biziel Memorial University Hospital, Bydgoszcz, Poland
| |
Collapse
|
2
|
Gama JM, Oliveira RC. CD44 and Its Role in Solid Cancers - A Review: From Tumor Progression to Prognosis and Targeted Therapy. FRONT BIOSCI-LANDMRK 2025; 30:24821. [PMID: 40152366 DOI: 10.31083/fbl24821] [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: 05/13/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 03/29/2025]
Abstract
Cluster of differentiation 44 (CD44) is a transmembrane protein expressed in normal cells but overexpressed in several types of cancer. CD44 plays a major role in tumor progression, both locally and systemically, by direct interaction with the extracellular matrix, inducing tissue remodeling, activation of different cellular pathways, such as Akt or mechanistic target of rapamycin (mTOR), and stimulation of angiogenesis. As a prognostic marker, CD44 has been identified as a major player in cancer stem cells (CSCs). CSCs with a CD44 phenotype are associated with chemoresistance, alone or in combination with other CSC markers, such as CD24 or aldehyde dehydrogenase 1 (ALDH1), and may be used for patient stratification. In the therapy setting, CD44 has been explored as a viable target, directly or indirectly. It has revealed promising potential, paving the way for its future use in the clinical setting. Immunohistochemistry effectively detects CD44 overexpression, enabling patients to be accurately selected for surgery and targeted anti-CD44 therapies. In this review, we highlight the properties of CD44, its expression in normal and tumoral tissues through immunohistochemistry and potential treatment options. We also discuss the clinical significance of this marker and its added value in therapeutic decision-making.
Collapse
Affiliation(s)
- João Martins Gama
- Serviço de Anatomia Patológica, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal
- Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), 4050-313 Porto, Portugal
| | - Rui Caetano Oliveira
- Centro de Investigação em Meio Ambiente, Genética e Oncobiologia-CIMAGO, Faculdade de Medicina, Universidade de Coimbra, 3004-535 Coimbra, Portugal
- Centro de Anatomia Patológica Germano de Sousa, 3000-377 Coimbra, Portugal
- Faculdade de Medicina, Universidade de Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
3
|
Wang Y, Qi P, Shi S, Pang C, Wang W, Fang D. Targeted therapy for glioblastoma utilizing hyaluronic acid-engineered liposomes for adriamycin delivery. NANOTECHNOLOGY 2025; 36:125102. [PMID: 39842037 DOI: 10.1088/1361-6528/adacef] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 01/22/2025] [Indexed: 01/24/2025]
Abstract
Glioblastoma (GBM) is a malignant tumor with highly heterogeneous and invasive characteristics leading to a poor prognosis. The CD44 molecule, which is highly expressed in GBM, has emerged as a highly sought-after biological marker. Therapeutic strategies targeting the cell membrane protein CD44 have emerged, demonstrating novel therapeutic potential. In this study, we constructed a nanodrug system (HA-Liposome@Dox) based on hyaluronic acid-engineered liposomes delivering adriamycin to target GBM. The system efficiently encapsulated Dox inside the liposomes through a hydrophilic-hydrophobic interaction mechanism, and the resulting HA-Liposome@Dox exhibited excellent loading efficacy, attributed to its uniform particle size distribution and negatively charged surface. Further evaluation revealed that HA-Liposome@Dox possessed excellent stability and safety and could promote the effective uptake of drug particles by CD44-overexpressing tumor cells, thus exerting a more potent cell-killing effect. Notably, in the treatment of GBM, HA-Liposome@Dox demonstrated significantly greater tumor growth inhibition compared to free Dox and prolonged the survival of tumor-bearing mice. Taken together, the present study not only verified the feasibility of HA-Liposome@Dox as an effective therapeutic tool against GBM and other CD44-positively expressing tumors, but also opened a promising new avenue for the clinical treatment of this type of refractory malignancies.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| | - Peiyan Qi
- Guangzhou International Travel Healthcare Center, Guangzhou Customs District P.R.China, Guangzhou, People's Republic of China
| | - Shenbao Shi
- Department of Neurosurgery, Zhujiang Hospital Southern Medical University, Guangzhou, People's Republic of China
| | - Cong Pang
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| | - Weijie Wang
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| | - Dazhao Fang
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, People's Republic of China
| |
Collapse
|
4
|
Pashkina E, Bykova M, Berishvili M, Lazarev Y, Kozlov V. Hyaluronic Acid-Based Drug Delivery Systems for Cancer Therapy. Cells 2025; 14:61. [PMID: 39851489 PMCID: PMC11764402 DOI: 10.3390/cells14020061] [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: 11/21/2024] [Revised: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 01/26/2025] Open
Abstract
In recent years, hyaluronic acid (HA) has attracted increasing attention as a promising biomaterial for the development of drug delivery systems. Due to its unique properties, such as high biocompatibility, low toxicity, and modifiability, HA is becoming a basis for the creation of targeted drug delivery systems, especially in the field of oncology. Receptors for HA overexpressed in subpopulations of cancer cells, and one of them, CD44, is recognized as a molecular marker for cancer stem cells. This review examines the role of HA and its receptors in health and tumors and analyzes existing HA-based delivery systems and their use in various types of cancer. The development of new HA-based drug delivery systems will bring new opportunities and challenges to anti-cancer therapy.
Collapse
Affiliation(s)
- Ekaterina Pashkina
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
- Department of Clinical Immunology, Novosibirsk State Medical University, 52, Krasny Prospect, 630091 Novosibirsk, Russia
| | - Maria Bykova
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
| | - Maria Berishvili
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
| | - Yaroslav Lazarev
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 2, Pirogova Street, 630090 Novosibirsk, Russia
| | - Vladimir Kozlov
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 2, Pirogova Street, 630090 Novosibirsk, Russia
| |
Collapse
|
5
|
Lopes J, Rodrigues CM, Godinho-Santos A, Coelho JMP, Cabaço LC, Barral DC, Faísca P, Catarino J, Nunes D, Fortunato E, Martins R, Rodrigues CMP, Gaspar MM, Reis CP. Combination of gold nanoparticles with near-infrared light as an alternative approach for melanoma management. Int J Pharm 2025; 668:124952. [PMID: 39547473 DOI: 10.1016/j.ijpharm.2024.124952] [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: 08/05/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Melanoma is the most aggressive type of skin cancer and recently approved drugs are often associated with resistance and significant adverse effects. Therefore, the design of more effective and safe options remains imperative. Photothermal therapy (PTT) using gold nanoparticles (AuNPs) presents a promising and innovative approach. In this work, the efficacy of combining a previously optimized formulation of AuNPs coated with a mixture of hyaluronic and oleic acids (HAOA-AuNPs) with near-infrared (NIR) laser irradiation in melanoma cell lines was explored. Coated and uncoated AuNPs formulations were characterized in physicochemical, morphological and elemental terms. Next, the cellular uptake efficiency as well as antiproliferative activity of the combination of each formulation with laser irradiation was evaluated. Subsequently, HAOA-AuNPs were selected to assess the underlying mechanism of combined therapy by cell cycle and Annexin V/PI assays. An in vivo syngeneic murine melanoma model was also conducted. In vitro studies demonstrated that 24 h after incubation and in the absence of laser, HAOA-AuNPs did not exhibit cytotoxic effects on the melanoma cell lines tested, similar to the laser alone. On the contrary, the combination therapy resulted in a large reduction in cell viability. Furthermore, it has been shown to promote S-phase cell cycle arrest and increase in the percentage of late apoptotic cells. Finally, the in vivo proof-of-concept showed that the intratumoral administration of HAOA-AuNPs followed by three laser irradiations impaired tumor progression. Collectively, AuNP-based PTT holds significant potential to improve treatment efficacy and safety, offering a versatile and potent tool against cancer.
Collapse
Affiliation(s)
- Joana Lopes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal
| | - Carla M Rodrigues
- REQUIMTE - LAQV, Chemistry Department, NOVA School of Science and Technology, NOVA University Lisbon, Campus da Caparica Caparica 2829-516, Portugal
| | - Ana Godinho-Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal
| | - João M P Coelho
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande 1749-016, Lisboa, Portugal
| | - Luís C Cabaço
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, Lisboa 1169-056, Portugal
| | - Duarte C Barral
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, Lisboa 1169-056, Portugal
| | - Pedro Faísca
- CECAV- Centro de Ciência Animal e Veterinária- Faculdade de Medicina, Veterinária de Lisboa- Universidade Lusófona-Centro Universitário de Lisboa, Portugal
| | - José Catarino
- Faculty of Veterinary Medicine, Universidade Lusófona-Centro Universitário de Lisboa, Portugal; School of Animal Health, Protection and Welfare, Lusophone Polytechnic Institute, Lisbon, Portugal
| | - Daniela Nunes
- Department of Materials Science, NOVA School of Science and Technology, Campus de Caparica, i3N/CENIMAT, Caparica 2829-516, Portugal
| | - Elvira Fortunato
- Department of Materials Science, NOVA School of Science and Technology, Campus de Caparica, i3N/CENIMAT, Caparica 2829-516, Portugal
| | - Rodrigo Martins
- Department of Materials Science, NOVA School of Science and Technology, Campus de Caparica, i3N/CENIMAT, Caparica 2829-516, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal; Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande 1749-016, Lisboa, Portugal.
| | - Catarina Pinto Reis
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Professor Gama Pinto, Lisboa 1649-003, Portugal; Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande 1749-016, Lisboa, Portugal.
| |
Collapse
|
6
|
Takagi Y, Kage M. Hyaluronan Tetrasaccharides Penetrate into the Skin by Passive Diffusion and Contribute to Skin Health. Chem Pharm Bull (Tokyo) 2025; 73:284-290. [PMID: 40175107 DOI: 10.1248/cpb.c23-00909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Hyaluronan (HA) is a commonly used material in cosmetics and pharmaceuticals because of its various pharmacological activities. However, because of its large molecular weight, HA penetrates the skin very poorly and most of it remains on the skin surface. Thus, topically applied HA could not be expected to function biologically in the skin. However, we have confirmed that HA tetrasaccharides (HA4), which is the smallest unit of HA, penetrate into the skin by passive diffusion and affect epidermal metabolism. Topical treatment of HA4 rescues the epidermal damage caused by long-term UVA irradiation. Furthermore, various biological functions of HA4 to maintain healthy skin was observed in cell culture studies. This review describes the skin permeability of HA4 and how it contributes to healthy skin.
Collapse
Affiliation(s)
- Yutaka Takagi
- Laboratory of Dermatological Physiology, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University
| | - Madoka Kage
- Laboratory of Dermatological Physiology, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University
| |
Collapse
|
7
|
Kim M, Jung MY, Lee DY, Ahn SM, Lee GM, Park CY. How to Fabricate Hyaluronic Acid for Ocular Drug Delivery. Pharmaceutics 2024; 16:1604. [PMID: 39771582 PMCID: PMC11680071 DOI: 10.3390/pharmaceutics16121604] [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: 11/26/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
This review aims to examine existing research on the development of ocular drug delivery devices utilizing hyaluronic acid (HA). Renowned for its exceptional biocompatibility, viscoelastic properties, and ability to enhance drug bioavailability, HA is a naturally occurring biopolymer. The review discussed specific mechanisms by which HA enhances drug delivery, including prolonging drug residence time on ocular surfaces, facilitating controlled drug release, and improving drug penetration through ocular tissues. By focusing on these unique functionalities, this review highlights the potential of HA-based systems to revolutionize ocular treatment. Various fabrication techniques for HA-based ocular drug delivery systems, including hydrogels, nanoparticles, and microneedles, are discussed, highlighting their respective advantages and limitations. Additionally, this review explores the clinical applications of HA-based devices in treating a range of ocular diseases, such as dry eye syndrome, glaucoma, retinal disorders, and ocular infections. By comparing the efficacy and safety profiles of these devices with traditional ocular drug delivery methods, this review aims to provide a comprehensive understanding of the potential benefits and challenges associated with HA-based systems. Moreover, this review discusses current limitations and future directions in the field, such as the need for standardized fabrication protocols, long-term biocompatibility studies, and large-scale clinical trials. The insights and advancements presented in this review aim to guide future research and development efforts, ultimately enhancing the effectiveness of ocular drug delivery and improving patient outcomes.
Collapse
Affiliation(s)
- Martha Kim
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea; (M.K.); (M.-Y.J.); (D.-Y.L.); (S.M.A.); (G.M.L.)
| | - Mi-Young Jung
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea; (M.K.); (M.-Y.J.); (D.-Y.L.); (S.M.A.); (G.M.L.)
| | - Do-Yeon Lee
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea; (M.K.); (M.-Y.J.); (D.-Y.L.); (S.M.A.); (G.M.L.)
| | - So Min Ahn
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea; (M.K.); (M.-Y.J.); (D.-Y.L.); (S.M.A.); (G.M.L.)
| | - Gyeong Min Lee
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea; (M.K.); (M.-Y.J.); (D.-Y.L.); (S.M.A.); (G.M.L.)
| | - Choul Yong Park
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| |
Collapse
|
8
|
Wu Q, Li L, Zhang Y, Ming X, Feng N. Measurement methods, influencing factors and applications of intercellular receptor-ligand binding kinetics in diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 194:43-54. [PMID: 39491758 DOI: 10.1016/j.pbiomolbio.2024.11.001] [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: 08/25/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Receptor-ligand binding on contacting cells dictates the extent of transmembrane signaling through membrane receptors during cell communication, influencing both the physiological and pathological activities of cells. This process is integral to fundamental biological mechanisms including signal transduction, cancer metastasis, immune responses, and inflammatory cascades, all of which are profoundly influenced by the cell microenvironment. This article provides an overview of the kinetic theory of receptor-ligand binding and examines methods for measuring this interaction, along with their respective advantages and disadvantages. Furthermore, it comprehensively explores the factors that impact receptor-ligand binding, encompassing protein-membrane interactions, the bioelectric microenvironment, auxiliary factors, hydrogen bond strength, pH levels, cis and trans interactions between ligands and receptors. The application of receptor-ligand binding kinetics in various diseases such as immunity, cancer, and inflammation are also discussed. Additionally, the investigation into how functional substances alter receptor-ligand binding dynamics within specific cellular microenvironments presents a promising new approach to treating related diseases.
Collapse
Affiliation(s)
- Qian Wu
- Hubei Key Laboratoy of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Research Center of Food Fermentation Engineering and Technology, Hubei University of Technology, Wuhan, 430068, Hubei, China.
| | - Liangchao Li
- Hubei Key Laboratoy of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Research Center of Food Fermentation Engineering and Technology, Hubei University of Technology, Wuhan, 430068, Hubei, China.
| | - Yuyan Zhang
- Hubei Key Laboratoy of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Research Center of Food Fermentation Engineering and Technology, Hubei University of Technology, Wuhan, 430068, Hubei, China.
| | - Xiaozhi Ming
- Hubei Key Laboratoy of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Research Center of Food Fermentation Engineering and Technology, Hubei University of Technology, Wuhan, 430068, Hubei, China.
| | - Nianjie Feng
- Hubei Key Laboratoy of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Research Center of Food Fermentation Engineering and Technology, Hubei University of Technology, Wuhan, 430068, Hubei, China.
| |
Collapse
|
9
|
Lin S, Zhang H, Zhao R, Wu Z, Zhang W, Yu M, Zhang B, Ma L, Li D, Peng L, Luo W. Single-cell multiomics reveals simvastatin inhibits pan-cancer epithelial-mesenchymal transition via the MEK/ERK pathway in XBP1+ mast cells. Sci Rep 2024; 14:29545. [PMID: 39604504 PMCID: PMC11603196 DOI: 10.1038/s41598-024-80858-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
Distant metastasis is the leading cause of cancer-related mortality, and achieving survival benefits through advancements in systemic therapy remains challenging. Mast cells play a dual role in shaping the tumor microenvironment (TME) and influencing distant metastasis, underscoring the significant research value of targeting mast cells for systemic therapy in advanced cancer. We investigated variations in mast cell infiltration levels in primary and metastatic malignancies using immunocyte infiltration analysis. Mast cell subsets were identified from pan-cancer distant metastasis single-cell sequencing data through dimensionality reduction clustering and cell type annotation, combined with cell trajectory and communication network analyses. A prognostic model was established using WGCNA and 12 machine learning algorithms to identify potential mast cell targets. Drug sensitivity and Mendelian randomization analyses were conducted to select potential drugs targeting mast cells, and their effects on epithelial-mesenchymal transition (EMT) were validated through in vitro experiments, including wound healing, transwell, and western blot assays. Results revealed that activated mast cells show increased infiltration in metastatic tumors, correlating with poor survival duration. XBP1+ mast cells were identified as key components of the inhibitory TME, potentially involved in EMT activation. Simvastatin was identified as a potential drug, reversing EMT induced by XBP1+ mast cells in pan-cancer. Aberrant activation of MEK/ERK signaling in XBP1+ mast cells can stimulate cancer cell EMT by modulating degranulation, while Simvastatin can inhibit EMT by suppressing degranulation.
Collapse
Affiliation(s)
- Sen Lin
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huimin Zhang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruiqi Zhao
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhulin Wu
- Department of Traditional Chinese Medicine, People's Hospital of Longhua, Shenzhen, China
| | - Weiqing Zhang
- Department of Traditional Chinese Medicine, People's Hospital of Longhua, Shenzhen, China
| | - Mengjiao Yu
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bei Zhang
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lanyue Ma
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Danfei Li
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lisheng Peng
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China.
| | - Weijun Luo
- Department of Traditional Chinese Medicine, People's Hospital of Longhua, Shenzhen, China.
| |
Collapse
|
10
|
Ishikawa K, Suzuki H, Ohishi T, Nakamura T, Yanaka M, Li G, Tanaka T, Ohkoshi A, Kawada M, Kaneko MK, Katori Y, Kato Y. Antitumor activities of anti‑CD44 monoclonal antibodies in mouse xenograft models of esophageal cancer. Oncol Rep 2024; 52:147. [PMID: 39219278 PMCID: PMC11391255 DOI: 10.3892/or.2024.8806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
CD44 is a type I transmembrane glycoprotein associated with poor prognosis in various solid tumors. Since CD44 plays a critical role in tumor development by regulating cell adhesion, survival, proliferation and stemness, it has been considered a target for tumor therapy. Anti‑CD44 monoclonal antibodies (mAbs) have been developed and applied to antibody‑drug conjugates and chimeric antigen receptor‑T cell therapy. Anti-pan‑CD44 mAbs, C44Mab‑5 and C44Mab‑46, which recognize both CD44 standard (CD44s) and variant isoforms were previously developed. The present study generated a mouse IgG2a version of the anti‑pan‑CD44 mAbs (5‑mG2a and C44Mab‑46‑mG2a) to evaluate the antitumor activities against CD44‑positive cells. Both 5‑mG2a and C44Mab‑46‑mG2a recognized CD44s‑overexpressed CHO‑K1 (CHO/CD44s) cells and esophageal tumor cell line (KYSE770) in flow cytometry. Furthermore, both 5‑mG2a and C44Mab‑46‑mG2a could activate effector cells in the presence of CHO/CD44s cells and exhibited complement-dependent cytotoxicity against both CHO/CD44s and KYSE770 cells. Furthermore, the administration of 5‑mG2a and C44Mab‑46‑mG2a significantly suppressed CHO/CD44s and KYSE770 xenograft tumor development compared with the control mouse IgG2a. These results indicate that 5‑mG2a and C44Mab‑46‑mG2a could exert antitumor activities against CD44‑positive cancers and be a promising therapeutic regimen for tumors.
Collapse
Affiliation(s)
- Kenichiro Ishikawa
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu, Shizuoka 410‑0301, Japan
| | - Takuro Nakamura
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Miyuki Yanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Guanjie Li
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Akira Ohkoshi
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Manabu Kawada
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, Shinagawa‑ku, Tokyo 141‑0021, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Yukio Katori
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Aoba‑ku, Sendai, Miyagi 980‑8575, Japan
| |
Collapse
|
11
|
Deng K, Wang W, Chi X, Yu Y, Zhang Y, Yuan J. Abnormal expression of serum miR-4746-5p in liver cancer patients after interventional chemotherapy and its possible mechanism. Biotechnol Appl Biochem 2024; 71:1154-1163. [PMID: 38809793 DOI: 10.1002/bab.2605] [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: 01/22/2024] [Accepted: 05/05/2024] [Indexed: 05/31/2024]
Abstract
Interventional chemotherapy is a common operation in the clinical treatment of liver cancer. The aim of this study was to investigate the expression and molecular mechanism of serum miR-4746-5p in liver cancer patients before and after interventional chemotherapy. The levels of miR-4746-5p and CDKN1C in serum samples from liver cancer patients were detected using real-time fluorescence quantitative polymerase chain reaction. Receiver operating characteristic curves revealed the diagnostic value of miR-4746-5p in tumors. Differences in clinical indicators between liver cancer patients and healthy controls were assessed using Pearson correlation analysis. Luciferase reporter gene assays confirmed the targeted interaction between miR-4746-5p and CDKN1C. In vitro cellular assays were validated by Cell Counting Kit-8, Transwell assay, and chemoresistance assay. Serum miR-4746-5p levels were increased in liver cancer patients but were downregulated after chemotherapy intervention. CDKN1C expression showed the opposite trend. Low levels of miR-4746-5p mediated cell growth and metastasis by targeting and negatively regulating CDKN1C expression, while silencing CDKN1C restored cell activity. Inhibition of miR-4746-5p reduced chemoresistance, while downregulation of CDKN1C affected cell sensitivity. miR-4746-5p may be a potential therapeutic factor for liver cancer diagnosis and interventional chemotherapy.
Collapse
Affiliation(s)
- Keping Deng
- Department of General Surgery, First Hospital of Changsha, Changsha, China
| | - Wei Wang
- Department of Pathology, Mianyang People's Hospital, Mianyang, China
| | - Xiaobin Chi
- Department of Hepatobiliary Surgery, 900 Hospital of the Joint Logistics Support Force, Fuzhou, China
| | - Yan Yu
- Department of Outpatient, Huai'an No. 4 People's Hospital, Huai'an, China
| | - Yichuan Zhang
- Minimally Invasive Endoscopy Center, Digestive Disease Center, The Affiliated Hospital of Panzhihua University, Panzhihua, China
| | - Jianming Yuan
- Department of Laboratory Medicine, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, China
| |
Collapse
|
12
|
Ishikawa K, Suzuki H, Ohishi T, Li G, Tanaka T, Kawada M, Ohkoshi A, Kaneko MK, Katori Y, Kato Y. Anti-CD44 Variant 10 Monoclonal Antibody Exerts Antitumor Activity in Mouse Xenograft Models of Oral Squamous Cell Carcinomas. Int J Mol Sci 2024; 25:9190. [PMID: 39273139 PMCID: PMC11395228 DOI: 10.3390/ijms25179190] [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: 08/02/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
CD44 regulates cell adhesion, proliferation, survival, and stemness and has been considered a tumor therapy target. CD44 possesses the shortest CD44 standard (CD44s) and a variety of CD44 variant (CD44v) isoforms. Since the expression of CD44v is restricted in epithelial cells and carcinomas compared to CD44s, CD44v has been considered a promising target for monoclonal antibody (mAb) therapy. We previously developed an anti-CD44v10 mAb, C44Mab-18 (IgM, kappa), to recognize the variant exon 10-encoded region. In the present study, a mouse IgG2a version of C44Mab-18 (C44Mab-18-mG2a) was generated to evaluate the antitumor activities against CD44-positive cells compared with the previously established anti-pan CD44 mAb, C44Mab-46-mG2a. C44Mab-18-mG2a exhibited higher reactivity compared with C44Mab-46-mG2a to CD44v3-10-overexpressed CHO-K1 (CHO/CD44v3-10) and oral squamous cell carcinoma cell lines (HSC-2 and SAS) in flow cytometry. C44Mab-18-mG2a exerted a superior antibody-dependent cellular cytotoxicity (ADCC) against CHO/CD44v3-10. In contrast, C44Mab-46-mG2a showed a superior complement-dependent cytotoxicity (CDC) against CHO/CD44v3-10. A similar tendency was observed in ADCC and CDC against HSC-2 and SAS. Furthermore, administering C44Mab-18-mG2a or C44Mab-46-mG2a significantly suppressed CHO/CD44v3-10, HSC-2, and SAS xenograft tumor growth compared with the control mouse IgG2a. These results indicate that C44Mab-18-mG2a could be a promising therapeutic regimen for CD44v10-positive tumors.
Collapse
Affiliation(s)
- Kenichiro Ishikawa
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (K.I.); (G.L.); (T.T.); (M.K.K.)
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (A.O.); (Y.K.)
| | - Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (K.I.); (G.L.); (T.T.); (M.K.K.)
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, 18-24 Miyamoto, Numazu-shi 410-0301, Shizuoka, Japan;
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan;
| | - Guanjie Li
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (K.I.); (G.L.); (T.T.); (M.K.K.)
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (K.I.); (G.L.); (T.T.); (M.K.K.)
| | - Manabu Kawada
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan;
| | - Akira Ohkoshi
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (A.O.); (Y.K.)
| | - Mika K. Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (K.I.); (G.L.); (T.T.); (M.K.K.)
| | - Yukio Katori
- Department of Otolaryngology, Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (A.O.); (Y.K.)
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Miyagi, Japan; (K.I.); (G.L.); (T.T.); (M.K.K.)
| |
Collapse
|
13
|
Buruiană A, Gheban BA, Gheban-Roșca IA, Georgiu C, Crișan D, Crișan M. The Tumor Stroma of Squamous Cell Carcinoma: A Complex Environment That Fuels Cancer Progression. Cancers (Basel) 2024; 16:1727. [PMID: 38730679 PMCID: PMC11083853 DOI: 10.3390/cancers16091727] [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: 04/08/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
The tumor microenvironment (TME), a complex assembly of cellular and extracellular matrix (ECM) components, plays a crucial role in driving tumor progression, shaping treatment responses, and influencing metastasis. This narrative review focuses on the cutaneous squamous cell carcinoma (cSCC) tumor stroma, highlighting its key constituents and their dynamic contributions. We examine how significant changes within the cSCC ECM-specifically, alterations in fibronectin, hyaluronic acid, laminins, proteoglycans, and collagens-promote cancer progression, metastasis, and drug resistance. The cellular composition of the cSCC TME is also explored, detailing the intricate interplay of cancer-associated fibroblasts (CAFs), mesenchymal stem cells (MSCs), endothelial cells, pericytes, adipocytes, and various immune cell populations. These diverse players modulate tumor development, angiogenesis, and immune responses. Finally, we emphasize the TME's potential as a therapeutic target. Emerging strategies discussed in this review include harnessing the immune system (adoptive cell transfer, checkpoint blockade), hindering tumor angiogenesis, disrupting CAF activity, and manipulating ECM components. These approaches underscore the vital role that deciphering TME interactions plays in advancing cSCC therapy. Further research illuminating these complex relationships will uncover new avenues for developing more effective treatments for cSCC.
Collapse
Affiliation(s)
- Alexandra Buruiană
- Department of Pathology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.B.); (C.G.); (D.C.)
| | - Bogdan-Alexandru Gheban
- Department of Histology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
- Emergency Clinical County Hospital, 400347 Cluj-Napoca, Romania
| | - Ioana-Andreea Gheban-Roșca
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400129 Cluj-Napoca, Romania;
| | - Carmen Georgiu
- Department of Pathology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.B.); (C.G.); (D.C.)
| | - Doința Crișan
- Department of Pathology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.B.); (C.G.); (D.C.)
| | - Maria Crișan
- Department of Histology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| |
Collapse
|
14
|
Wu Z, Lu J, Loo A, Ho N, Nguyen D, Cheng PY, Mohammed AI, Cirillo N. Role of CD44 in Chemotherapy Treatment Outcome: A Scoping Review of Clinical Studies. Int J Mol Sci 2024; 25:3141. [PMID: 38542115 PMCID: PMC10970610 DOI: 10.3390/ijms25063141] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 01/31/2025] Open
Abstract
Cluster of differentiation 44 (CD44), a cell surface adhesion molecule overexpressed in cancer stem cells, has been implicated in chemoresistance. This scoping review, following PRISMA-ScR guidelines, systematically identified and evaluated clinical studies on the impact of CD44 expression on chemotherapy treatment outcomes across various cancer types. The search encompassed PubMed (1985-2023) and SCOPUS (1936-2023) databases, yielding a total of 12,659 articles, of which 40 met the inclusion criteria and were included in the qualitative synthesis using a predefined data extraction table. Data collected included the cancer type, sample size, interventions, control, treatment outcome, study type, expression of CD44 variants and isoforms, and effect of CD44 on chemotherapy outcome. Most of the studies demonstrated an association between increased CD44 expression and negative chemotherapeutic outcomes such as shorter overall survival, increased tumor recurrence, and resistance to chemotherapy, indicating a potential role of CD44 upregulation in chemoresistance in cancer patients. However, a subset of studies also reported non-significant relationships or conflicting results. In summary, this scoping review highlighted the breadth of the available literature investigating the clinical association between CD44 and chemotherapeutic outcomes. Further research is required to elucidate this relationship to aid clinicians in managing CD44-positive cancer patients.
Collapse
Affiliation(s)
- Zihao Wu
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Jillian Lu
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Andrew Loo
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Nathan Ho
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Danny Nguyen
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Po Yueh Cheng
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Ali I. Mohammed
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
| | - Nicola Cirillo
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; (Z.W.); (A.I.M.)
- School of Dentistry, University of Jordan, Amman 11942, Jordan
| |
Collapse
|
15
|
Fan M, Huo S, Guo Y, Wang R, Hao W, Zhang Z, Wang L, Zhao Y. UDP-glucose dehydrogenase supports autophagy-deficient PDAC growth via increasing hyaluronic acid biosynthesis. Cell Rep 2024; 43:113808. [PMID: 38367236 DOI: 10.1016/j.celrep.2024.113808] [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: 09/19/2023] [Revised: 01/04/2024] [Accepted: 02/01/2024] [Indexed: 02/19/2024] Open
Abstract
Autophagy is an essential degradation and recycling process that maintains cellular homeostasis during stress or nutrient deprivation. However, certain types of tumors such as pancreatic cancers can circumvent autophagy inhibition to sustain growth. The mechanism that autophagy-deficient pancreatic ductal adenocarcinoma (PDAC) uses to grow under nutrient deprivation is poorly understood. Our data show that nutrient deprivation in PDAC results in UDP-glucose dehydrogenase (UGDH) degradation, which is dependent on autophagic cargo receptor sequestosome 1 (p62). Moreover, we demonstrate that accumulated UGDH is indispensable for autophagy-deficient PDAC cells proliferation by promoting hyaluronic acid (HA) synthesis upon energy deprivation. Using an orthotopic mouse model of PDAC, we find that inhibition of HA synthesis by targeting UGDH in PDAC reduces tumor weight. Thus, the combined inhibition of HA and autophagy might be an attractive strategy for PDAC treatment.
Collapse
Affiliation(s)
- Minghe Fan
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Sihan Huo
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Yuyao Guo
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Ruoxuan Wang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Wenqin Hao
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Ziyang Zhang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Lina Wang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China
| | - Ying Zhao
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, State Key Laboratory of Molecular Oncology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Peking University International Cancer Institute, Peking University Health Science Center, Beijing 100191, China; Peking University Cancer Hospital and Institute, Beijing 100142, China.
| |
Collapse
|
16
|
Zhou XQ, Li YP, Dang SS. Precision targeting in hepatocellular carcinoma: Exploring ligand-receptor mediated nanotherapy. World J Hepatol 2024; 16:164-176. [PMID: 38495282 PMCID: PMC10941735 DOI: 10.4254/wjh.v16.i2.164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 02/27/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer and poses a major challenge to global health due to its high morbidity and mortality. Conventional chemotherapy is usually targeted to patients with intermediate to advanced stages, but it is often ineffective and suffers from problems such as multidrug resistance, rapid drug clearance, nonspecific targeting, high side effects, and low drug accumulation in tumor cells. In response to these limitations, recent advances in nanoparticle-mediated targeted drug delivery technologies have emerged as breakthrough approaches for the treatment of HCC. This review focuses on recent advances in nanoparticle-based targeted drug delivery systems, with special attention to various receptors overexpressed on HCC cells. These receptors are key to enhancing the specificity and efficacy of nanoparticle delivery and represent a new paradigm for actively targeting and combating HCC. We comprehensively summarize the current understanding of these receptors, their role in nanoparticle targeting, and the impact of such targeted therapies on HCC. By gaining a deeper understanding of the receptor-mediated mechanisms of these innovative therapies, more effective and precise treatment of HCC can be achieved.
Collapse
Affiliation(s)
- Xia-Qing Zhou
- Department of Infectious Diseases, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Ya-Ping Li
- Department of Infectious Diseases, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Shuang-Suo Dang
- Department of Infectious Diseases, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China.
| |
Collapse
|
17
|
Kina Kilicaslan U, Aru B, Aydin Aksu S, Vardar Aker F, Yanikkaya Demirel G, Gurleyik MG. Relationship between immune checkpoint proteins and neoadjuvant chemotherapy response in breast cancer. Surg Oncol 2024; 52:102037. [PMID: 38290327 DOI: 10.1016/j.suronc.2024.102037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 01/02/2024] [Accepted: 01/19/2024] [Indexed: 02/01/2024]
Abstract
INTRODUCTION Following major developments in cancer immunotherapy, treatments targeting immune checkpoint proteins (ICP) gained interest in breast cancer, though studies mostly focus on patients with metastatic disease as well as patients nonresponsive to the conventional treatments. Herein, we aimed to investigate the levels of ICP in tumor stroma and tumor infiltrating lymphocytes, and tumor tissue prior to neoadjuvant chemotherapy administration to evaluate the relationship between ICP levels, clinicopathological parameters, and NAC response. MATERIALS AND METHODS This study was conducted with 51 patients where PD-1, PD-L1, CTLA-4, TIM-3, CD24 and CD44 levels were investigated in CD45+ cells while CD326, CD24, CD44 and PD-L1 protein expression levels were investigated in CD45- population. In addition, CD44 and CD24 levels were evaluated in the tumor stroma. TIL levels were investigated according to the TILS Working Group. Treatment responses after NAC were evaluated according to the MD Anderson RCB score. RESULTS Our results revealed positive correlation between CTLA-4 and CD44 expression in cases with high TIL levels as well as TIL levels and CTLA-4 expression in cases with partial response. Similarly, positive correlation was detected between TIM3 and PD-L1 levels in cases with good response. In addition, a negative correlation between TILs after NAC and PD-1/PD-L1 expression in lymphocytes in cases with partial complete response. CONCLUSIONS Our study provides preliminary data about the correlation between ICP and clinicopathological status and NAC response in breast cancer, in addition to underlining the requirement for further research to determine their potential as therapeutic targets.
Collapse
Affiliation(s)
- Umut Kina Kilicaslan
- Department of General Surgery, Istanbul Haydarpasa Numune Training and Research Hospital, University of Health Sciences Turkey, İstanbul, Turkey
| | - Basak Aru
- Department of Immunology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Sibel Aydin Aksu
- Department of Radiology, Istanbul Haydarpasa Numune Training and Research Hospital, University of Health Sciences Turkey, İstanbul, Turkey
| | - Fugen Vardar Aker
- Department of Pathology, Istanbul Haydarpasa Numune Training and Research Hospital, University of Health Sciences Turkey, İstanbul, Turkey
| | | | - Meryem Gunay Gurleyik
- Department of General Surgery, Istanbul Haydarpasa Numune Training and Research Hospital, University of Health Sciences Turkey, İstanbul, Turkey.
| |
Collapse
|