1
|
Wang Y, Huang D, Li M, Yang M. MicroRNA-99 family in cancer: molecular mechanisms for clinical applications. PeerJ 2025; 13:e19188. [PMID: 40161350 PMCID: PMC11955196 DOI: 10.7717/peerj.19188] [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: 09/16/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
MicroRNAs (miRNAs) are a class of non-coding RNA sequences that regulate gene expression post-transcriptionally. The miR-99 family, which is highly evolutionarily conserved, comprises three homologs: miR-99a, miR-99b, and miR-100. Its members are under-expressed in most cancerous tissues, suggesting their cancer-repressing properties in multiple cancers; however, in some contexts, they also promote malignant lesion progression. MiR-99 family members target numerous genes involved in various tumor-related processes such as tumorigenesis, proliferation, cell-cycle regulation, apoptosis, invasion, and metastasis. We review the recent research on this family, summarize its implications in cancer, and explore its potential as a biomarker and cancer therapeutic target. This review contributes to the clinical translation of the miR-99 family members.
Collapse
Affiliation(s)
- Yueyuan Wang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, ChangChun, Jilin, China
| | - Dan Huang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, ChangChun, Jilin, China
| | - Mingxi Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, ChangChun, Jilin, China
| | - Ming Yang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, ChangChun, Jilin, China
| |
Collapse
|
2
|
Shaik Syed Ali P, Ahmad MP, Parveen KMH. Lin28/let-7 axis in breast cancer. Mol Biol Rep 2025; 52:311. [PMID: 40085362 DOI: 10.1007/s11033-025-10413-6] [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: 09/19/2024] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Let-7 microRNAs are tumor suppressor microRNAs, and their reduced expression frequently occurs in various types of cancers, including breast cancer. A notable correlation exists between decreased let-7 microRNA levels and the overexpression of Lin28A and Lin28B, particularly in breast cancer cases with poor prognoses. Dysregulation of Wnt signaling significantly contributes to the upregulation of Lin28A and Lin28B in breast cancer. Both Lin28A and Lin28B operate from different cellular compartments to inhibit the biogenesis of let-7 microRNAs, which are essential for the post-transcriptional regulation of genes involved in key cellular functions such as proliferation, differentiation, and apoptosis. Decreased expression of let-7 microRNAs leads to the overexpression of oncogenes such as K-ras, C-myc, and SOX-2 in breast cancer. Overexpression of Lin28A associated with reduced let-7 microRNA levels is observed in estrogen receptor positive, estrogen receptor negative, and human epidermal growth factor receptor 2 positive breast cancers, whereas Lin28B overexpression with reduced let-7 microRNA levels occurs specifically in triple negative breast cancer. This review aims to dissect the molecular interplay between Lin28A, Lin28B, and let-7 microRNAs, elucidating their roles in breast carcinogenesis, metastasis, and the development of resistance to conventional treatments like radiation and chemotherapy. Additionally, the review addresses potential therapeutic avenues offered by let-7 microRNAs or their mimics, as well as Lin28A and Lin28B inhibitors, in the treatment of breast cancer.
Collapse
Affiliation(s)
- P Shaik Syed Ali
- School of Medicine, The Maldives National University, Malé, Maldives.
| | - Md Parwez Ahmad
- School of Medicine, The Maldives National University, Malé, Maldives
| | - K M Huria Parveen
- School of Medicine, The Maldives National University, Malé, Maldives
| |
Collapse
|
3
|
Chen J, Yin Q, Xu S, Tan X, Liang Y, Chen C, Li L, Zhang T, Shen T. IFN-treated macrophage-derived exosomes prevents HBV-HCC migration and invasion via regulating miR-106b-3p/PCGF3/PI3K/AKT signaling axis. Front Cell Infect Microbiol 2024; 14:1421195. [PMID: 39529637 PMCID: PMC11551115 DOI: 10.3389/fcimb.2024.1421195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/05/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Studies revealed that exosomes from IFN-α-treated liver non-parenchymal cells (IFN-exo) mediate antiviral activity. MiR-106b-3p has been shown to play a paradoxical role in disease progressing from different studies. However, its specific role in HBV-related hepatocellular carcinoma (HBV-HCC) and the underlying mechanism remains unclear. METHOD Huh7 cells transient transfected with plasmids of HBV-C2 and B3 were co-cultured with IFN-exo. Cell supernatants were collected to detect miR-106b-3p, HBsAg, HBeAg and HBV DNA levels. Cell proliferation, apoptosis, migration and invasion were analyzed. The putative targets of miR-106b-3p were identified by a dual-luciferase reporter system. The expression of PCGF3, migratory proteins(MMP2/9), and the PI3K/AKT signaling pathway-related proteins were assessed by western blot. The expression of PCGF3 mRNA was quantitative analyzed by using 52 pairs of paraffin-embedded tissues from HCC patients. siRNAs-PCGF3 were used to knocked-down PCGF3 expression. RESULTS The expression of miR-106b-3p was significantly higher in THP-1 cells and supernatants treated with IFN-exo than those untreated. Significantly increased expression of miR-106b-3p and decreased expression of HBsAg and HBV DNA were observed in Huh7-C2/B3 cells treated with IFN-exo. In addition, miR-106b-3p was directly target to PCGF3. Scratch healing assay and transwell assay showed that either IFN-exo or miRNA-106-3p over-expression, or siRNAs-PCGF3 inhibited migration and invasion of Huh7-C2/B3 cells, and subsequently resulted in suppression of p-AKT/AKT and p-PI3K/PI3K. Notably, the expression level of PCGF3 was significantly lower in HBeAg (+)-HCC tumor tissues than HBeAg (-)-HCC tumor. CONCLUSION IFN-α-induced macrophage-derived miR-106b-3p inhibits HBV replication, HBV- Huh7 cells migration and invasion via regulating PCGF3/PI3K/AKT signaling axis. miR-106b-3p and PCGF3 were potential biomarkers in the prevention and treatment of HBV-HCC.
Collapse
Affiliation(s)
- Jing Chen
- Department of Pulmonary and Critical Care Medicine, Yunnan Provincial Key Laboratory for Clinical Virology, Institute of Basic and Clinical Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
- Medical School, Kunming University of Science and Technology, Kunming, China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Qi Yin
- Medical School, Kunming University of Science and Technology, Kunming, China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Shiheng Xu
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Xiaoqing Tan
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Yu Liang
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Chaohui Chen
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Li Li
- Department of Pulmonary and Critical Care Medicine, Yunnan Provincial Key Laboratory for Clinical Virology, Institute of Basic and Clinical Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
- Department of Infectious Diseases and Hepatic Disease, Yunnan Province Innovation Team of Intestinal Microecology Related Disease Research and Technological Transformation, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Tao Zhang
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Tao Shen
- Department of Pulmonary and Critical Care Medicine, Yunnan Provincial Key Laboratory for Clinical Virology, Institute of Basic and Clinical Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
- Medical School, Kunming University of Science and Technology, Kunming, China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
- Department of Infectious Diseases and Hepatic Disease, Yunnan Province Innovation Team of Intestinal Microecology Related Disease Research and Technological Transformation, The First People’s Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
4
|
Dong W, Luo Y, He D, Zhang M, Zeng J, Chen Y. Oncolytic virotherapy against lung cancer: key receptors and signaling pathways of viral entry. Front Immunol 2024; 15:1473288. [PMID: 39430750 PMCID: PMC11486668 DOI: 10.3389/fimmu.2024.1473288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
Lung cancer accounts for the highest cancer-related mortality worldwide. While immunotherapies targeting anti-tumor immune responses have demonstrated efficacy in clinical practice, the demand for novel treatment modalities remains urgent. Oncolytic viruses (OVs), which selectively kill tumor cells while stimulating an anti-tumor immune response, represent a potential breakthrough in lung cancer therapy. The induction of anti-tumor immunity by OVs is central to their overall therapeutic effectiveness. Many natural receptors on the surface of cancer cells are dysregulated, providing potential entry points for OVs. Furthermore, the inherent dysregulation of some key signaling pathways in lung cancer cells promotes proliferation, progression and metastasis, which may facilitate selective viral replication. In this review, we explore the application of OVs in lung cancer by analyzing several major OVs and their corresponding entry receptors. Then, we also examine the key signaling pathways and molecules with the potential to synergize with OVs in modulating the immune tumor microenvironment. Finally, we discuss the combination and administration strategies that warrant further clinical trials for validation. Despite certain limitations, the tolerability of OVs positions virotherapy as a promising avenue in the future of lung cancer treatment.
Collapse
Affiliation(s)
- Wenxun Dong
- Department of Thoracic Surgery I, Peking University Cancer Hospital Yunnan, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Ying Luo
- Department of Thoracic Surgery I, Peking University Cancer Hospital Yunnan, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Daqian He
- Department of Thoracic Surgery I, Peking University Cancer Hospital Yunnan, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Ming Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming, China
| | - Jingtong Zeng
- Department of Thoracic Surgery I, Peking University Cancer Hospital Yunnan, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Ying Chen
- Department of Thoracic Surgery I, Peking University Cancer Hospital Yunnan, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| |
Collapse
|
5
|
Manoleras AV, Sloan EK, Chang A. The sympathetic nervous system shapes the tumor microenvironment to impair chemotherapy response. Front Oncol 2024; 14:1460493. [PMID: 39381049 PMCID: PMC11458372 DOI: 10.3389/fonc.2024.1460493] [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: 07/06/2024] [Accepted: 08/30/2024] [Indexed: 10/10/2024] Open
Abstract
The tumor microenvironment influences cancer progression and response to treatments, which ultimately impacts the survival of patients with cancer. The sympathetic nervous system (SNS) is a core component of solid tumors that arise in the body. In addition to influencing cancer progression, a role for the SNS in the effectiveness of cancer treatments is beginning to emerge. This review explores evidence that the SNS impairs chemotherapy efficacy. We review findings of studies that evaluated the impact of neural ablation on chemotherapy outcomes and discuss plausible mechanisms for the impact of neural signaling on chemotherapy efficacy. We then discuss implications for clinical practice, including opportunities to block neural signaling to improve response to chemotherapy.
Collapse
Affiliation(s)
| | | | - Aeson Chang
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| |
Collapse
|
6
|
Maas RJA, Hoogstad-van Evert JS, Hagemans IM, Brummelman J, van Ens D, de Jonge PKJD, Hooijmaijers L, Mahajan S, van der Waart AB, Hermans CKJC, de Klein J, Woestenenk R, van Herwaarden AE, Schaap NPM, Rezaeifard S, Tauriello DVF, Zusterzeel PLM, Ottevanger N, Jansen JH, Hobo W, Dolstra H. Increased peritoneal TGF-β1 is associated with ascites-induced NK-cell dysfunction and reduced survival in high-grade epithelial ovarian cancer. Front Immunol 2024; 15:1448041. [PMID: 39376560 PMCID: PMC11456434 DOI: 10.3389/fimmu.2024.1448041] [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: 06/12/2024] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
Natural killer (NK) cell therapy represents an attractive immunotherapy approach against recurrent epithelial ovarian cancer (EOC), as EOC is sensitive to NK cell-mediated cytotoxicity. However, NK cell antitumor activity is dampened by suppressive factors in EOC patient ascites. Here, we integrated functional assays, soluble factor analysis, high-dimensional flow cytometry cellular component data and clinical parameters of advanced EOC patients to study the mechanisms of ascites-induced inhibition of NK cells. Using a suppression assay, we found that ascites from EOC patients strongly inhibits peripheral blood-derived NK cells and CD34+ progenitor-derived NK cells, albeit the latter were more resistant. Interestingly, we found that higher ascites-induced NK cell inhibition correlated with reduced progression-free and overall survival in EOC patients. Furthermore, we identified transforming growth factor (TGF)-β1 to correlate with ascites-induced NK cell dysfunction and reduced patient survival. In functional assays, we showed that proliferation and anti-tumor reactivity of CD34+ progenitor-derived NK cells are significantly affected by TGF-β1 exposure. Moreover, inhibition of TGF-β1 signaling with galunisertib partly restored NK cell functionality in some donors. For the cellular components, we showed that the secretome is associated with a different composition of CD45+ cells between ascites of EOC and benign reference samples with higher proportions of macrophages in the EOC patient samples. Furthermore, we revealed that higher TGF-β1 levels are associated with the presence of M2-like macrophages, B cell populations and T-regulatory cells in EOC patient ascites. These findings reveal that targeting TGF-β1 signaling could increase NK cell immune responses in high-grade EOC patients.
Collapse
Affiliation(s)
- Ralph J. A. Maas
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Janneke S. Hoogstad-van Evert
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Obstetrics and Gynecology Amphia, Breda, Netherlands
| | - Iris M. Hagemans
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jolanda Brummelman
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Diede van Ens
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Paul K. J. D. de Jonge
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Laura Hooijmaijers
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Shweta Mahajan
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Anniek B. van der Waart
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Charlotte K. J. C. Hermans
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Janne de Klein
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rob Woestenenk
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Antonius E. van Herwaarden
- Diagnostic Laboratory – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Somayeh Rezaeifard
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Cell Biology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Petra L. M. Zusterzeel
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nelleke Ottevanger
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Joop H. Jansen
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Willemijn Hobo
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Harry Dolstra
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
7
|
Ji L, He X, Min X, Yang H, Wu W, Xu H, Chen J, Mei A. Glucagon-like peptide-1 receptor agonists in neoplastic diseases. Front Endocrinol (Lausanne) 2024; 15:1465881. [PMID: 39371922 PMCID: PMC11449759 DOI: 10.3389/fendo.2024.1465881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/28/2024] [Indexed: 10/08/2024] Open
Abstract
Glucagon-like peptide-1 receptor agonist (GLP-1RA), a novel hypoglycemic agent for the treatment of type 2 diabetes, has well-known effects such as lowering blood sugar, ameliorating inflammation, reducing weight, and lowering blood lipids. It has also been shown that it can influence the proliferation and survival of cells and has a certain effect on the prognosis of some neoplastic diseases. In this study, the potential effects of GLP-1RAs on the occurrence and development of tumors were reviewed to provide new ideas for the prevention and treatment of tumors in patients.
Collapse
Affiliation(s)
- Lisan Ji
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Xianzhen He
- Children’s Medical Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xinwen Min
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Handong Yang
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Wenwen Wu
- School of Public Health, Hubei University of Medicine, Shiyan, Hubei, China
| | - Hao Xu
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| | - Jun Chen
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
- Virology Key Laboratory of Shiyan City, Hubei University of Medicine, Shiyan, China
| | - Aihua Mei
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
8
|
Inocencio JF, Mitrasinovic S, Asad M, Parney IF, Zang X, Himes BT. Immune checkpoint pathways in glioblastoma: a diverse and evolving landscape. Front Immunol 2024; 15:1424396. [PMID: 39346924 PMCID: PMC11427296 DOI: 10.3389/fimmu.2024.1424396] [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: 04/28/2024] [Accepted: 08/27/2024] [Indexed: 10/01/2024] Open
Abstract
Immune checkpoint (IC) inhibition in glioblastoma (GBM) has not shown promising results in the last decade compared to other solid tumors. Several factors contributing to the lack of immunotherapy response include the profound immunosuppressive nature of GBM, highly redundant signaling pathways underlying immune checkpoints, and the negative immunogenic impact of current standard of care on the tumor microenvironment. In this review, we will discuss various ICs in the context of GBM, their interplay with the tumor immune microenvironment, relevant pre-clinical and clinical studies, and the impact of current treatment modalities on GBM IC blockade therapy. Understanding the molecular mechanisms that drive ICs, and how they contribute to an immunosuppressive tumor microenvironment is critical in advancing IC inhibition therapy in GBM. Furthermore, revisiting current treatment modalities and their impact on the immune landscape is instrumental in designing future combinatorial therapies that may overcome treatment resistance.
Collapse
Affiliation(s)
- Julio F Inocencio
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
| | - Stefan Mitrasinovic
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
| | - Mohammad Asad
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ian F Parney
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Benjamin T Himes
- Department of Neurological Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
9
|
Ma P, Yuan L, Jia S, Zhou Z, Xu D, Huang S, Meng F, Zhang Z, Nan Y. Lonicerae Japonicae Flos with the homology of medicine and food: a review of active ingredients, anticancer mechanisms, pharmacokinetics, quality control, toxicity and applications. Front Oncol 2024; 14:1446328. [PMID: 39314630 PMCID: PMC11417411 DOI: 10.3389/fonc.2024.1446328] [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: 06/09/2024] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
Lonicerae Japonicae Flos (LJF, called Jinyinhua in China), comes from the dried flower buds or flowers to be opened of Lonicera japonica Thunb. in the Lonicera family. It has a long history of medicinal use and has a wide range of application prospects. As modern research advances, an increasing number of scientific experiments have demonstrated the anticancer potential of LJF. However, there is a notable absence of systematic reports detailing the anti-tumor effects of LJF. This review integrates the principles of Traditional Chinese Medicine (TCM) with contemporary pharmacological techniques, drawing upon literature from authoritative databases such as PubMed, CNKI, and WanFang to conduct a comprehensive study of LJF. Notably, a total of 507 compounds have been isolated and characterized from the plant to date, which include volatile oils, organic acids, flavonoids, iridoids, triterpenes and triterpenoid saponins. Pharmacological studies have demonstrated that LJF extract, along with components such as chlorogenic acid, luteolin, rutin, luteoloside, hyperoside and isochlorogenic acid, exhibits potential anticancer activities. Consequently, we have conducted a comprehensive review and summary of the mechanisms of action and clinical applications of these components. Furthermore, we have detailed the pharmacokinetics, quality control, and toxicity of LJF, while also discussing its prospective applications in the fields of biomedicine and preventive healthcare. It is hoped that these studies will provide valuable reference for the clinical research, development, and application of LJF.
Collapse
Affiliation(s)
- Ping Ma
- Pharmacy Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Shumin Jia
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Ziying Zhou
- Pharmacy Department, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Duojie Xu
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Shicong Huang
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Fandi Meng
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Zhe Zhang
- Department of Chinese Medical Gastrointestinal, China-Japan Friendship Hospital, Beijing, China
| | - Yi Nan
- Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| |
Collapse
|
10
|
Chen Q. Efficacy of different medications in the treatment of gynaecological tumours: a clinical meta-analysis. Front Oncol 2024; 14:1428937. [PMID: 39314635 PMCID: PMC11416918 DOI: 10.3389/fonc.2024.1428937] [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: 05/07/2024] [Accepted: 06/26/2024] [Indexed: 09/25/2024] Open
Abstract
Background A gynaecological tumour is one of the world's leading causes of death for women globally. Among women, cancer is the 8th most common cause of death. Since there are no such programmes, the majority of women who are diagnosed with the condition are either in advanced stages or do not respond well to current treatments. Even if patients react to the treatments, they still risk having the cancer return, at which point any further medical intervention is met with resistance. Method For this study, we selected the systemic reviews and articles that have the use of different medications used for the treatment of gynaecological tumours. Results Regarding metformin use, this study found a positive relationship between higher survival and metformin use. Five of the studies that examined the use of statins revealed a link between statin use and higher overall and/or progression-free survival rates. Individuals on lipophilic and hydrophilic statins would do better. Research evaluating beta-blocker use during neoadjuvant treatment revealed a time-varying effect, with improved survival seen across all users early in the follow-up period. However, only non-selective beta-blocker users demonstrated a correlation with higher survival after five years. One study found that the benefits of aspirin use were significant, but the advantage for continuous users (both before and after diagnosis) was minimal. Conclusion Conclusions on the association between gynaecological tumour survival and NA-NSAIDs, metformin, beta-blockers, and aspirin cannot be drawn due to insufficient evidence. However, the vast majority of statin studies have demonstrated that users had higher rates of survival. Bias, however, bias may affect the results of the studies.
Collapse
Affiliation(s)
- Qiaoying Chen
- Department of Gynecology, Women’s and Children’s Hospital of Ningbo
University, Ningbo, Zhejiang, China
| |
Collapse
|
11
|
Yao Z, Wang T, Liu J, Zhou Z, Zhang Y. Diagnostic accuracy of cytology and urine methylation test in patients with non-muscle invasive bladder cancer: a systematic review and meta-analysis. Front Oncol 2024; 14:1412346. [PMID: 39290246 PMCID: PMC11405155 DOI: 10.3389/fonc.2024.1412346] [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: 05/21/2024] [Accepted: 08/13/2024] [Indexed: 09/19/2024] Open
Abstract
Background Multiple clinical studies have demonstrated the numerous advantages of urine methylation test over cytology for monitoring patients with non-muscle invasive bladder cancer (NMIBC) following surgery. This research aims to provide a systematic review and meta-analysis to evaluate the efficacy and limits of urine methylation test in the clinical management of NMIBC. Methods This research was carried out by conducting a comprehensive search of clinical trials comparing cytology and urine methylation test for NMIBC follow-up using databases such as PubMed, Embase, Web of Science, and the Cochrane Library up until May 2023, including references from relevant articles. The study is registered on PROSPERO with ID CRD42023398969. Result This study comprised six studies with a total of 1676 patients. The analysis revealed that the AUC of urine methylation test had a greater AUC than that of the cytology examination (0.89 vs 0.71). In post-operative follow-up of patients with NMIBC, the urine methylation test demonstrated a significant sensitivity (0.69 vs 0.52), but with lower specificity (0.87 vs 0.93) than cytology examination. Conclusion The urine methylation test and cytology examination have both shown strong diagnostic performance in screening for NMIBC patients. However, urine methylation test outperforms cytology examination in terms of accuracy and sensitivity. Systematic review registration PROSPERO, identifier CRD42023398969.
Collapse
Affiliation(s)
- Zhuoyue Yao
- Department of Urology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Wang
- Department of Urology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jingpeng Liu
- Department of Urology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhongbao Zhou
- Department of Urology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yong Zhang
- Department of Urology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Wang S, Mao S, Li X, Yang D, Zhou Y, Yue H, Li B, Li W, Li C, Zhang X. Identification and validation of potential prognostic biomarkers in glioblastoma via the mesenchymal stem cell infiltration level. Front Oncol 2024; 14:1406186. [PMID: 39286023 PMCID: PMC11403407 DOI: 10.3389/fonc.2024.1406186] [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: 03/24/2024] [Accepted: 08/06/2024] [Indexed: 09/19/2024] Open
Abstract
Aims Mesenchymal stem cells (MSCs) are key components in promoting glioblastoma (GBM) progression. This study aimed to explore new therapeutic targets and related pathogenic mechanisms based on different MSCs infiltration levels in GBM patients. Methods We estimated the relationship between cell infiltration and prognosis of GBM. Subsequently, key risk genes were identified and prognostic models were constructed by LASSO-Cox analysis. The risk genes were validated by five independent external cohorts, single-cell RNA analysis, and immunohistochemistry of human GBM tissues. TIDE analysis predicted responsiveness to immune checkpoint inhibitors in different risk groups. Results The MSCs infiltration level was negatively associated with survival in GBM patients. LOXL1, LOXL4, and GUCA1A are key risk genes that promote GBM progression and may act through complex intercellular communication. Conclusion This research has provided a comprehensive study for exploring the MSCs infiltration environment on GBM progression, which could shed light on novel biomarkers and mechanisms involved in GBM progression.
Collapse
Affiliation(s)
- Shengyu Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Senlin Mao
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaofu Li
- Department of Magnetic Resonance Imaging, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dan Yang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yinglian Zhou
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hui Yue
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bing Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Li
- Department of Neurology, Heilongjiang Hospital, Harbin, China
| | - Chengyun Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuemei Zhang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
13
|
Gan L, Wang W, Jiang J, Tian K, Liu W, Cao Z. Dual role of Nrf2 signaling in hepatocellular carcinoma: promoting development, immune evasion, and therapeutic challenges. Front Immunol 2024; 15:1429836. [PMID: 39286246 PMCID: PMC11402828 DOI: 10.3389/fimmu.2024.1429836] [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: 05/08/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the predominant form of liver cancer and ranks as the third leading cause of cancer-related mortality globally. The liver performs a wide range of tasks and is the primary organ responsible for metabolizing harmful substances and foreign compounds. Oxidative stress has a crucial role in growth and improvement of hepatocellular carcinoma (HCC). Nuclear factor erythroid 2 (1)-related factor 2 (Nrf2) is an element that regulates transcription located in the cytoplasm. It controls the balance of redox reactions by stimulating the expression of many genes that depend on antioxidant response elements. Nrf2 has contrasting functions in the normal, healthy liver and HCC. In the normal liver, Nrf2 provides advantageous benefits, while in HCC it promotes harmful effects that support the growth and survival of HCC. Continuous activation of Nrf2 has been detected in HCC and promotes its advancement and aggressiveness. In addition, Activation of Nrf2 may lead to immune evasion, weakening the immune cells' ability to attack tumors and thereby promoting tumor development. Furthermore, chemoresistance in HCC, which is considered a form of stress response to chemotherapy medications, significantly impedes the effectiveness of HCC treatment. Stress management is typically accomplished by activating specific signal pathways and chemical variables. One important element in the creation of chemoresistance in HCC is nuclear factor-E2-related factor 2 (Nrf2). Nrf2 is a transcription factor that regulates the activation and production of a group of genes that encode proteins responsible for protecting cells from damage. This occurs through the Nrf2/ARE pathway, which is a crucial mechanism for combating oxidative stress within cells.
Collapse
Affiliation(s)
- Lin Gan
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Wei Wang
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Jinxiu Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Ke Tian
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Wei Liu
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Zhumin Cao
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| |
Collapse
|
14
|
Hajimohammadebrahim-Ketabforoush M, Zali A, Shahmohammadi M, Hamidieh AA. Metformin and its potential influence on cell fate decision between apoptosis and senescence in cancer, with a special emphasis on glioblastoma. Front Oncol 2024; 14:1455492. [PMID: 39267853 PMCID: PMC11390356 DOI: 10.3389/fonc.2024.1455492] [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: 06/26/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Despite reaching enormous achievements in therapeutic approaches worldwide, GBM still remains the most incurable malignancy among various cancers. It emphasizes the necessity of adjuvant therapies from the perspectives of both patients and healthcare providers. Therefore, most emerging studies have focused on various complementary and adjuvant therapies. Among them, metabolic therapy has received special attention, and metformin has been considered as a treatment in various types of cancer, including GBM. It is clearly evident that reaching efficient approaches without a comprehensive evaluation of the key mechanisms is not possible. Among the studied mechanisms, one of the more challenging ones is the effect of metformin on apoptosis and senescence. Moreover, metformin is well known as an insulin sensitizer. However, if insulin signaling is facilitated in the tumor microenvironment, it may result in tumor growth. Therefore, to partially resolve some paradoxical issues, we conducted a narrative review of related studies to address the following questions as comprehensively as possible: 1) Does the improvement of cellular insulin function resulting from metformin have detrimental or beneficial effects on GBM cells? 2) If these effects are detrimental to GBM cells, which is more important: apoptosis or senescence? 3) What determines the cellular decision between apoptosis and senescence?
Collapse
Affiliation(s)
- Melika Hajimohammadebrahim-Ketabforoush
- Student Research Committee, Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Shahmohammadi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Jurić Čuljak Ž, Tomić S, Šitum K, Boraska Jelavić T. Case report: complete response and long-term survival on third-line immunotherapy in patient with pleural mesothelioma. Front Oncol 2024; 14:1388829. [PMID: 39267830 PMCID: PMC11391104 DOI: 10.3389/fonc.2024.1388829] [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: 02/20/2024] [Accepted: 08/13/2024] [Indexed: 09/15/2024] Open
Abstract
Introduction Pleural mesothelioma (PM) is a rare neoplasm with median survival time range from 8 to 14 months from diagnosis, and the 5-year survival rate less than 10%, indicating a poor prognosis. The standard treatment for unresectable PM for a long time has been polychemotherapy with pemetrexed and cisplatin for fit patients. Currently, the combination of the anti PD-1 inhibitor nivolumab and the anti-CTLA4 inhibitor ipilimumab has been recognized as the best possible frontline therapy (especially in the sarcomatoid or biphasic type) due to improved outcomes compared to the standard chemotherapy combination. There are still no established predictive biomarkers for any type of systemic therapy in this disease. Case presentation Patient who presented with cough and dyspnea has been diagnosed with advanced epithelioid type PM in May 2016. He was treated with three lines of therapy, including an antiangiogenic agent and immunotherapy with pembrolizumab in the third line. Immunotherapy with the PD-1 inhibitor pembrolizumab achieved a complete and prolonged response that transferred to long- term survival. Seven years from diagnosis, the patient is still alive. Histological findings showed an unusually immune-inflamed tumor microenvironment possibly leading to excellent response on immunotherapy. Conclusions The course of the disease in our patient points out that we need better predictive biomarkers to direct the treatment algorithm, as some of the patients, although chemorefractory to the best chemotherapy option, can sustain great benefit of second-line chemotherapy in combination with antiangiogenic agent, and especially immunotherapy, even in late lines of therapy.
Collapse
Affiliation(s)
- Željka Jurić Čuljak
- Department of Pulmonary Diseases, University Hospital of Split, Split, Croatia
| | - Snježana Tomić
- Department of Pathology, Forensic Medicine and Cytology, University Hospital of Split, School of Medicine, University of Split, Split, Croatia
| | - Kristina Šitum
- Department of Radiology, University Hospital of Split, Split, Croatia
| | - Tihana Boraska Jelavić
- Department of Oncology, General Hospital Dubrovnik, Dubrovnik, Croatia
- University Department for Health Studies, University of Split, Split, Croatia
| |
Collapse
|
16
|
García-Gutiérrez V, Gómez-Casares MT, Xicoy B, Casado-Montero F, Orti G, Giraldo P, Hernández-Boluda JC. Critical review of clinical data and expert-based recommendations for the use of bosutinib in the treatment of chronic myeloid leukemia. Front Oncol 2024; 14:1405467. [PMID: 39252937 PMCID: PMC11381280 DOI: 10.3389/fonc.2024.1405467] [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: 03/22/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
Chronic myeloid leukemia (CML), characterized by the presence of the BCR::ABL1 fusion gene, has undergone a transformative shift with the introduction of tyrosine kinase inhibitors (TKIs). The current availability of six different TKIs (imatinib, dasatinib, nilotinib, bosutinib, ponatinib, and asciminib) in clinical practice makes it important to know their efficacy and toxicity profile for treatment optimization. This review examines the latest insights regarding the use of bosutinib in CML treatment. Clinical trials have demonstrated the effectiveness of bosutinib, positioning it as a first-line treatment that can induce sustained molecular responses. Importantly, it can also be effective in patients who have experienced treatment failure or intolerance with prior TKIs, revealing the potential of bosutinib also in second- and later-line settings. Even in the advanced phase of CML, bosutinib has demonstrated its capacity to achieve molecular responses, expanding its usefulness. Real-world evidence studies echo these findings, emphasizing bosutinib's effectiveness in achieving deep molecular responses, maintaining remissions, and serving as an alternative for patients intolerant or resistant to other TKIs as a second-line therapy. Notably, one of the greatest strengths of bosutinib is its favorable safety profile, in particular the low incidence of vascular complications with its use, which is undoubtedly a comparative advantage over other TKIs. In summary, the latest research highlights the versatility of bosutinib in CML treatment and underscores its pivotal role in optimizing patient management in challenging cases. Continuing research and investigation will further establish bosutinib's place in the evolving landscape of CML therapy, offering an alternative for CML patients across different treatment stages.
Collapse
Affiliation(s)
| | - María Teresa Gómez-Casares
- Servicio de Hematología, Hospital Universitario de Gran Canaria Dr. Negrin. Profesor asociado de la ULPGC, Las Palmas de Gran Canaria, Spain
| | - Blanca Xicoy
- Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Josep Carreras Leukemia Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Felipe Casado-Montero
- Servicio de Hematología y Hemoterapia, Hospital General Universitario de Toledo, Toledo, Spain
| | - Guillermo Orti
- Servicio de Hematologia, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Pilar Giraldo
- Hematologia, Hospital Quironsalud, Fundación ZeroLMC, Zaragoza, Spain
| | | |
Collapse
|
17
|
Luo Y, Cai X, Yang B, Lu F, Yi C, Wu G. Advances in understanding the role of immune checkpoint LAG-3 in tumor immunity: a comprehensive review. Front Oncol 2024; 14:1402837. [PMID: 39252941 PMCID: PMC11381248 DOI: 10.3389/fonc.2024.1402837] [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: 03/18/2024] [Accepted: 07/26/2024] [Indexed: 09/11/2024] Open
Abstract
Lymphocyte activation gene 3 (LAG-3), also known as CD223, is an emerging immune checkpoint that follows PD-1 and CTLA-4. Several LAG-3 targeting inhibitors in clinical trials and the combination of relatlimab (anti-LAG-3) and nivolumab (anti-PD-1) have been approved for treating - unresectable or metastatic melanoma. Despite the encouraging clinical potential of LAG-3, the physiological function and mechanism of action in tumors are still not well understood. In this review, we systematically summarized the structure of LAG-3, ligands of LAG-3, cell-specific functions and signaling of LAG-3, and the current status of LAG-3 inhibitors under development.
Collapse
Affiliation(s)
- Yingzhe Luo
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xuebin Cai
- Department of Abdominal Oncology, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Biao Yang
- Department of Abdominal Oncology, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Facheng Lu
- Department of Abdominal Oncology, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Yi
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Guoyu Wu
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
18
|
Xue C, Zhu D, Wang X, Jiao L, Lu Y, Zhang S, Lv J, Cui L, Ruan M, Xu D, Liu Q, Feng Y, Mei S. Durable response to pembrolizumab in hepatic metastasis from colonic carcinoma with Lynch syndrome: a case report. Front Immunol 2024; 15:1455907. [PMID: 39247185 PMCID: PMC11377275 DOI: 10.3389/fimmu.2024.1455907] [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: 06/28/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Pembrolizumab and other immunotherapies have become central in treating metastatic colon cancer, particularly effective in patients with mismatch repair deficiencies. We report a case involving a man who initially underwent radical surgery for sigmoid colon cancer on April 27, 2011, followed by hepatic tumor resection on September 21, 2017. Post-surgery, he received eight cycles of adjuvant chemotherapy with the CAPEOX regimen and was regularly monitored through CT and MRI scans. On August 24, 2022, liver metastases were detected, and he was diagnosed with Lynch syndrome (LS) due to germline mutation in the MSH2 and EPCAM genes. He commenced treatment with 200mg of pembrolizumab intravenously every three weeks on September 2, 2022, and demonstrated a sustained response. However, after 17 cycles, he developed a treatment related adverse event (TRAE) of pancreatic endocrine dysfunction, leading to type 1 diabetes, managed with subcutaneous insulin injections. After 30 cycles of treatment, no evidence of disease was observed. This case underscores the significant clinical benefits of first-line pembrolizumab in managing hepatic metastasis in colonic carcinoma associated with LS, despite the occurrence of TRAEs. It raises critical questions regarding the optimal duration of immunotherapy following a complete or partial response and whether treatment should be discontinued upon the emergency of TRAEs. Continued research and forthcoming clinical trials with checkpoint inhibitors are expected to refine treatment protocols for LS-associated carcinoma.
Collapse
Affiliation(s)
- Cheng Xue
- Kidney Institute, Department of Nephrology, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Dongqing Zhu
- Department of Radiology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xin Wang
- Department of Health Management Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Lina Jiao
- Kidney Institute, Department of Nephrology, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yunhui Lu
- Kidney Institute, Department of Nephrology, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Sanli Zhang
- Kidney Institute, Department of Nephrology, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Jiayi Lv
- Kidney Institute, Department of Nephrology, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Linlin Cui
- Kidney Institute, Department of Nephrology, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Mengna Ruan
- Kidney Institute, Department of Nephrology, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Dechao Xu
- Kidney Institute, Department of Nephrology, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Qingyang Liu
- Department of Radiology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yun Feng
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shuqin Mei
- Kidney Institute, Department of Nephrology, Shanghai Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
19
|
Vanthienen W, Fernández-García J, Baietti MF, Claeys E, Van Leemputte F, Nguyen L, Goossens V, Deparis Q, Broekaert D, Vlayen S, Audenaert D, Delforge M, D'Amuri A, Van Zeebroeck G, Leucci E, Fendt SM, Thevelein JM. The novel family of Warbicin ® compounds inhibits glucose uptake both in yeast and human cells and restrains cancer cell proliferation. Front Oncol 2024; 14:1411983. [PMID: 39239276 PMCID: PMC11374660 DOI: 10.3389/fonc.2024.1411983] [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: 04/03/2024] [Accepted: 07/29/2024] [Indexed: 09/07/2024] Open
Abstract
Many cancer cells share with yeast a preference for fermentation over respiration, which is associated with overactive glucose uptake and breakdown, a phenomenon called the Warburg effect in cancer cells. The yeast tps1Δ mutant shows even more pronounced hyperactive glucose uptake and phosphorylation causing glycolysis to stall at GAPDH, initiation of apoptosis through overactivation of Ras and absence of growth on glucose. The goal of the present work was to use the yeast tps1Δ strain to screen for novel compounds that would preferentially inhibit overactive glucose influx into glycolysis, while maintaining basal glucose catabolism. This is based on the assumption that the overactive glucose catabolism of the tps1Δ strain might have a similar molecular cause as the Warburg effect in cancer cells. We have isolated Warbicin ® A as a compound restoring growth on glucose of the yeast tps1Δ mutant, showed that it inhibits the proliferation of cancer cells and isolated structural analogs by screening directly for cancer cell inhibition. The Warbicin ® compounds are the first drugs that inhibit glucose uptake by both yeast Hxt and mammalian GLUT carriers. Specific concentrations did not evoke any major toxicity in mice but increase the amount of adipose tissue likely due to reduced systemic glucose uptake. Surprisingly, Warbicin ® A inhibition of yeast sugar uptake depends on sugar phosphorylation, suggesting transport-associated phosphorylation as a target. In vivo and in vitro evidence confirms physical interaction between yeast Hxt7 and hexokinase. We suggest that reversible transport-associated phosphorylation by hexokinase controls the rate of glucose uptake through hydrolysis of the inhibitory ATP molecule in the cytosolic domain of glucose carriers and that in yeast tps1Δ cells and cancer cells reversibility is compromised, causing constitutively hyperactive glucose uptake and phosphorylation. Based on their chemical structure and properties, we suggest that Warbicin ® compounds replace the inhibitory ATP molecule in the cytosolic domain of the glucose carriers, preventing hexokinase to cause hyperactive glucose uptake and catabolism.
Collapse
Affiliation(s)
- Ward Vanthienen
- Center for Microbiology, VIB, Leuven-Heverlee, Belgium
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium
| | - Juan Fernández-García
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Maria Francesca Baietti
- TRACE PDX Platform, Laboratory of RNA Cancer Biology, LKI Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Elisa Claeys
- TRACE PDX Platform, Laboratory of RNA Cancer Biology, LKI Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Frederik Van Leemputte
- Center for Microbiology, VIB, Leuven-Heverlee, Belgium
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium
| | - Long Nguyen
- Screening Core, VIB, Ghent, Belgium
- Centre for Bioassay Development and Screening (C-BIOS), Ghent University, Ghent, Belgium
| | - Vera Goossens
- Screening Core, VIB, Ghent, Belgium
- Centre for Bioassay Development and Screening (C-BIOS), Ghent University, Ghent, Belgium
| | - Quinten Deparis
- Center for Microbiology, VIB, Leuven-Heverlee, Belgium
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium
| | - Dorien Broekaert
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Sophie Vlayen
- LKI Leuven Cancer Institute Leuven, KU Leuven, Leuven, Belgium
| | - Dominique Audenaert
- Screening Core, VIB, Ghent, Belgium
- Centre for Bioassay Development and Screening (C-BIOS), Ghent University, Ghent, Belgium
| | - Michel Delforge
- LKI Leuven Cancer Institute Leuven, KU Leuven, Leuven, Belgium
| | | | - Griet Van Zeebroeck
- Center for Microbiology, VIB, Leuven-Heverlee, Belgium
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium
| | - Eleonora Leucci
- TRACE PDX Platform, Laboratory of RNA Cancer Biology, LKI Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Sarah-Maria Fendt
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Johan M Thevelein
- Center for Microbiology, VIB, Leuven-Heverlee, Belgium
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium
- NovelYeast bv, Bio-Incubator, BIO4, Leuven-Heverlee, Belgium
| |
Collapse
|
20
|
Dong S, Zhao M, Zhu J, Li T, Yan M, Xing K, Liu P, Yu S, Ma J, He H. Natural killer cells: a future star for immunotherapy of head and neck squamous cell carcinoma. Front Immunol 2024; 15:1442673. [PMID: 39234249 PMCID: PMC11371580 DOI: 10.3389/fimmu.2024.1442673] [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: 06/02/2024] [Accepted: 07/15/2024] [Indexed: 09/06/2024] Open
Abstract
The interplay between immune components and the epithelium plays a crucial role in the development and progression of head and neck squamous cell carcinoma (HNSCC). Natural killer (NK) cells, one of the main tumor-killing immune cell populations, have received increasing attention in HNSCC immunotherapy. In this review, we explore the mechanism underlying the interplay between NK cells and HNSCC. A series of immune evasion strategies utilized by cancer cells restrict HNSCC infiltration of NK cells. Overcoming these limitations can fully exploit the antineoplastic potential of NK cells. We also investigated the tumor-killing efficacy of NK cell-based immunotherapies, immunotherapeutic strategies, and new results from clinical trials. Notably, cetuximab, the most essential component of NK cell-based immunotherapy, inhibits the epidermal growth factor receptor (EGFR) signaling pathway and activates the immune system in conjunction with NK cells, inducing innate effector functions and improving patient prognosis. In addition, we compiled information on other areas for the improvement of patient prognosis using anti-EGFR receptor-based monoclonal antibody drugs and the underlying mechanisms and prognoses of new immunotherapeutic strategies for the treatment of HNSCC.
Collapse
Affiliation(s)
- Shuyan Dong
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ming Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jin Zhu
- Department of Pathology, Xi’an Daxing Hospital, Xi’an, China
| | - Ting Li
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Mingze Yan
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Kaixun Xing
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Peng Liu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Shan Yu
- Department of Pathology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jian Ma
- Department of General Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Immunology, Harbin Medical University, Harbin, China
| | - Hongjiang He
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
21
|
Zhang K, He R, Tang F, Zhou L, Zhang X, Li J, Wei Z, Li Y. A novel x-Ray and γ-Ray combination strategy for radiotherapy after breast-conserving surgery in patients with right breast cancer. Front Oncol 2024; 14:1397273. [PMID: 39286018 PMCID: PMC11404321 DOI: 10.3389/fonc.2024.1397273] [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: 03/07/2024] [Accepted: 07/29/2024] [Indexed: 09/19/2024] Open
Abstract
Background and purpose Radiotherapy is a primary therapeutic approach for breast cancer following breast-conserving surgery. The TaiChiB dual-modality radiotherapy system combining X-ray and focused γ-ray, offers a new approach to reduce the radiation dose of organs at risk (OARs) and has the potential to mitigate the adverse effects of radiotherapy. Currently, there are few studies on the dosimetric characteristics of the TaiChiB dual-modality system for actual treatment plans for specific diseases. The purpose of this work is to study the dosimetric advantages of dual-modal systems for right breast patients after breast-conserving surgery. Material and methods Treatment plans for 20 patients with right breast cancer were generated for a linear accelerator (LINAC) based system and the TaiChiB dual-modality system, respectively. Volumetric modulated arc therapy plans with simultaneous integrated boost (VMAT-SIB) were made for the LINAC. Focused γ-ray was used to deliver the boost dose with the dual-modality system. The dosimetric parameters of the target and OARs were evaluated and compared between the treatment plans generated for the two systems. Results The TaiChiB dual-modality plans exhibit a higher conformal index (CI) and lower gradient index (GI) for the PGTV and PTV compared with the LINAC-based VMAT-SIB plans. Compared to VMAT-SIB plans, the PTV Dmax, PTV Dmean, PTV V110, PGTV Dmax, and PGTV Dmean of the TaiChiB dual-modality plans are significantly lower. Meanwhile, the dose to OARs, such as the Dmean of the heart, the V5 of liver, the Dmean of ipsilateral lung, the V30 of ipsilateral lung, the V20 of ipsilateral lung, the V5 of ipsilateral lung, the Dmean of contralateral lung, Dmax of contralateral breast and the Dmean of contralateral breast are significantly reduced. Conclusions Our study demonstrates the dosimetric advantages of the novel TaiChiB dual-modality radiotherapy system for the treatment of right-sided breast cancer. Overall, for the TaiChiB dual-modality radiotherapy system, the radiation dose outside the target region decreases rapidly, thereby minimizing radiation exposure to neighboring organs and ensuring the conformity of the target area. Our research confirms the potential of the TaiChiB dual-modality system for future radiotherapy.
Collapse
Affiliation(s)
- Kunpeng Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ruixin He
- Department of Radiotherapy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fenwen Tang
- Department of Radiotherapy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Luping Zhou
- Department of Radiotherapy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiaozhi Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | | | | | - Yi Li
- Department of Radiotherapy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
22
|
Jiang Y, Zhu Y, Ding Y, Lu X. Nomograms to predict lung metastasis in malignant primary osseous spinal neoplasms and cancer-specific survival in lung metastasis subgroup. Front Oncol 2024; 14:1393990. [PMID: 39228988 PMCID: PMC11368787 DOI: 10.3389/fonc.2024.1393990] [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: 03/31/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
Purpose To construct and validate nomograms for predicting lung metastasis probability in patients with malignant primary osseous spinal neoplasms (MPOSN) at initial diagnosis and predicting cancer-specific survival (CSS) in the lung metastasis subgroup. Methods A total of 1,298 patients with spinal primary osteosarcoma, chondrosarcoma, Ewing sarcoma, and chordoma were retrospectively collected. Least absolute shrinkage and selection operator (LASSO) and multivariate logistic analysis were used to identify the predictors for lung metastasis. LASSO and multivariate Cox analysis were used to identify the prognostic factors for 3- and 5-year CSS in the lung metastasis subgroup. Receiver operating characteristic (ROC) curves, calibration curves, and decision curve analyses (DCA) were used to estimate the accuracy and net benefits of nomograms. Results Histologic type, grade, lymph node involvement, tumor size, tumor extension, and other site metastasis were identified as predictors for lung metastasis. The area under the curve (AUC) for the training and validating cohorts were 0.825 and 0.827, respectively. Age, histologic type, surgery at primary site, and grade were identified as the prognostic factors for the CSS. The AUC for the 3- and 5-year CSS were 0.790 and 0.740, respectively. Calibration curves revealed good agreements, and the Hosmer and Lemeshow test identified the models to be well fitted. DCA curves demonstrated that nomograms were clinically useful. Conclusion The nomograms constructed and validated by us could provide clinicians with a rapid and user-friendly tool to predict lung metastasis probability in patients with MPOSN at initial diagnosis and make a personalized CSS evaluation for the lung metastasis subgroup.
Collapse
Affiliation(s)
- Yong Jiang
- Orthopaedic Department, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yapeng Zhu
- Orthopaedic Department, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yongli Ding
- Orthopaedic Department, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xinchang Lu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
23
|
Zhou C, Wang W, Mu Y, Meng M. Efficacy and safety of a novel TKI (anlotinib) for the treatment of advanced digestive system neoplasms: a systematic review and meta-analysis. Front Immunol 2024; 15:1393404. [PMID: 39206183 PMCID: PMC11349560 DOI: 10.3389/fimmu.2024.1393404] [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: 02/29/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Objective To systematically evaluate the efficacy and safety of anlotinib targeted therapy for the treatment of patients with advanced digestive system neoplasms (DSNs). Methods Clinical trials were extracted from PubMed, the Cochrane Library, Web of Science, Embase, China National Knowledge Infrastructure (CNKI) and the Wanfang database up to October 2023. Outcome measures, including therapeutic efficacy, quality of life (QOL) and adverse events, were extracted and evaluated. Results Twenty trials, including 1,613 advanced DSNs patients, were included. The results indicated that, compared with conventional treatment alone, the combination of anlotinib targeted therapy with conventional treatment significantly improved the patients' 6-months overall survival (OS, OR=1.76, CI=1.53 to 2.02, P<0.00001), overall response (ORR, OR=1.76, CI=1.53 to 2.02, P<0.00001) and disease control rate (DCR, OR=1.51, 95% CI=1.25 to 1.84, P<0.0001). Moreover, the group that received the combined therapy had higher rates of hypertension (P<0.00001), proteinuria (P<0.00001), fatigue (P<0.00001), diarrhea (P<0.00001), hypertriglyceridemia (P=0.02), alanine aminotransfease (ALT)increased (P=0.004), aspartate transaminase (AST) increased (P=0.006), anorexia (P<0.00001), weight loss (P=0.002), abdominal pain (P=0.0006), hypothyroidism (P=0.02), prolonged QT interval (P=0.04). Analyses of other adverse events, such as gastrointestinal reaction, leukopenia, and neutropenia, did not reveal significant differences (P>0.05). Conclusion The combination of anlotinib targeted therapy and conventional treatment is more effective for DSNs treatment than conventional treatment alone. However, this combined treatment could lead to greater rates of hypertension, albuminuria and hand-foot syndrome. Therefore, the benefits and risks should be considered before treatment.
Collapse
Affiliation(s)
- Changhui Zhou
- Department of Central Laboratory, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Weihua Wang
- Department of Central Laboratory, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Ying Mu
- Department of Gastroenterology, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Min Meng
- Department of Central Laboratory, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| |
Collapse
|
24
|
Fattahi MR, Dehghani M, Paknahad S, Rahiminia S, Zareie D, Hoseini B, Oroomi TR, Motedayyen H, Arefnezhad R. Clinical insights into nanomedicine and biosafety: advanced therapeutic approaches for common urological cancers. Front Oncol 2024; 14:1438297. [PMID: 39193389 PMCID: PMC11347329 DOI: 10.3389/fonc.2024.1438297] [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: 05/25/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Urological cancers including those of the prostate, bladder, and kidney, are prevalent and often lethal malignancies besides other less common ones like testicular and penile cancers. Current treatments have major limitations like side effects, recurrence, resistance, high costs, and poor quality of life. Nanotechnology offers promising solutions through enhanced diagnostic accuracy, targeted drug delivery, controlled release, and multimodal imaging. This review reflects clinical challenges and nanomedical advances across major urological cancers. In prostate cancer, nanoparticles improve delineation and radiosensitization in radiation therapy, enable fluorescent guidance in surgery, and enhance chemotherapy penetration in metastatic disease. Nanoparticles also overcome bladder permeability barriers to increase the residence time of intravesical therapy and chemotherapy agents. In renal cancer, nanocarriers potentiate tyrosine kinase inhibitors and immunotherapy while gene vectors and zinc oxide nanoparticles demonstrate antiproliferative effects. Across modalities, urological applications of nanomedicine include polymeric, liposomal, and metal nanoparticles for targeted therapy, prodrug delivery, photodynamic therapy, and thermal ablation. Biosafety assessments reveal favorable profiles but clinical translation remains limited, necessitating further trials. In conclusion, nanotechnology holds significant potential for earlier detection, precise intervention, and tailored treatment of urological malignancies, warranting expanded research to transform patient outcomes.
Collapse
Affiliation(s)
- Mohammad Reza Fattahi
- Student Research Committee, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Shafa Rahiminia
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Deniz Zareie
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Behzad Hoseini
- School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Hossein Motedayyen
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Reza Arefnezhad
- Coenzyme R Research Institute, Tehran, Iran
- Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
25
|
Zhang Y, Xie J, Wu H, Huang J, Zheng D, Wang S, Jia X, He Z, Gong Y, Ju L, Sun Q. NK cell based immunotherapy against oral squamous cell carcinoma. Front Immunol 2024; 15:1440764. [PMID: 39192980 PMCID: PMC11347299 DOI: 10.3389/fimmu.2024.1440764] [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: 05/30/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC), a major subtype of head and neck cancers, presents significant challenges due to its aggressive feature and limited therapeutic efficacy of conventional treatments. In response to these challenges, Natural Killer (NK) cells, a vital component of the innate immune system, are being explored for their therapeutic potential in OSCC due to their inherent ability to target and eliminate cancer cells without prior sensitization. This review uniquely focuses on the evolving role of NK cells specifically in OSCC, incorporating recent advancements in CAR-NK cell engineering and personalized therapy approaches that have not been comprehensively covered in previous reviews. The mechanisms through which NK cells exert cytotoxic effects on tumor cells include direct killing through the engagement of natural cytotoxic receptors and antibody-dependent cellular cytotoxicity (ADCC), making them promising agents in cancer immunotherapy. Additionally, the article explores recent advancements in engineering NK cells to enhance their antitumor activity, such as the modification with chimeric antigen receptors (CARs) to target specific tumor antigens. Clinical implications of NK cell-based therapies, including the challenges of integrating these treatments with existing protocols and the potential for personalized therapy, are examined. The review highlights the promise of NK cell therapies in improving outcomes for OSCC patients and outlines future directions for research in this dynamic field of oncological immunotherapy.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Stomatology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jianming Xie
- Department of Otolaryngology & Head and Neck Surgery, Anyuan People’s hospital, Ganzhou, China
| | - Haoran Wu
- Southern Medical University, Guangzhou, Guangdong, China
| | - Jinhui Huang
- Southern Medical University, Guangzhou, Guangdong, China
| | - Danna Zheng
- Southern Medical University, Guangzhou, Guangdong, China
| | - Shaotong Wang
- Southern Medical University, Guangzhou, Guangdong, China
| | - Xueqiang Jia
- Southern Medical University, Guangzhou, Guangdong, China
| | - Zongzhong He
- Department of Transfusion Medicine, General Hospital of Southern Theatre Command, Guangzhou, Guangdong, China
| | - Ying Gong
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Linling Ju
- Medical School of Nantong University, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Qiurong Sun
- Department of Stomatology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
26
|
Yang Z, Chen J, Wen M, Lei J, Zeng M, Li S, Long Y, Zhou Z, Wang C. Genetic association of lipids and lipid-lowering drug target genes with Endometrial carcinoma: a drug target Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1446457. [PMID: 39193372 PMCID: PMC11347345 DOI: 10.3389/fendo.2024.1446457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Aberrant lipid metabolism is intricately linked to the development of endometrial cancer, and statin lipid-lowering medications are regarded as promising adjunctive therapies for future management of this malignancy. This study employed Mendelian randomization (MR) to explore the causal association between lipid traits and endometrial cancer while assessing the potential impact of drug targets on lower lipids on endometrial cancer. METHOD Two-sample Mendelian randomization was employed to probe the causal association between lipid traits and endometrial carcinoma. Drug-target Mendelian randomization was also utilized to identify potential drug-target genes for managing endometrial carcinoma. In instances where lipid-mediated effects through particular drug targets were notable, the impacts of these drug targets on endometrial carcinoma risk factors were investigated to bolster the findings. RESULT No causal association between genetically predicted lipid traits (LDL-C, TG, TC, and HDL-C) and EC was found in two-sample Mendelian randomization. In drug target Mendelian randomization, genetic modeling of apolipoprotein B (APOB) (OR [95%CI]=0.31, [0.16-0.60]; p=4.73e-04) and cholesteryl ester transfer protein (CETP) (OR [95%CI]=1.83, [1.38-2.43]; p=2.91e-05) genetic mimicry was associated with non-endometrioid carcinoma. CONCLUSION The results of our MR study revealed no causal association between genetically predicted lipid traits (LDL-C, TG, TC, and HDL-C) and EC. Among the six lipid-lowering drug targets, we observed a significant association between lower predicted APOB levels and higher CETP levels with an increased risk of endometrioid carcinoma. These findings provide novel insights into the importance of lipid regulation in individuals with endometrial carcinoma, warranting further clinical validation and mechanistic investigations.
Collapse
Affiliation(s)
- Zhehan Yang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Junpan Chen
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Minghao Wen
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Jiayuan Lei
- The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
| | - Ming Zeng
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Sichen Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yao Long
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiyi Zhou
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Chunyan Wang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
27
|
Coelho KBCA, Wosniaki DK, Marin AM, Fabris L, Borges dos Reis R, Aoki MN, Zanette DL. Urinary mRNA-based biomarkers for non-muscle-invasive bladder cancer: a mini-review. Front Oncol 2024; 14:1441883. [PMID: 39184050 PMCID: PMC11341357 DOI: 10.3389/fonc.2024.1441883] [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: 05/31/2024] [Accepted: 07/16/2024] [Indexed: 08/27/2024] Open
Abstract
Bladder cancer (BC) is the second most common type of cancer of the urinary system. Approximately 75% of the cases are non-muscle invasive bladder cancer (NMIBC), which has a high recurrence and progression rate. Current diagnosis and surveillance methods present challenges, including risks to the patients. For this reason, urinary biomarkers have been proposed as alternatives to the methods. The goal of this mini-review is to describe urinary mRNA-based biomarkers available in current literature for NMIBC tumors, using the PubMed database. The search included the following keywords: "biomarkers" AND "bladder cancer" AND "urine" and "RNA" and "non-muscle". The search yielded 11 original researchers utilizing mRNA-based urinary biomarkers. Although there is a wide variety of biomarkers described, the cohorts of the studies were not exclusively NMIBC, which is the subtype of BC that would mostly benefit from the introduction of a good follow-up biomarker, highlighting the need for randomized interventional trials for NMIBC.
Collapse
Affiliation(s)
| | - Denise Kusma Wosniaki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Paraná, Brazil
| | - Anelis Maria Marin
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Paraná, Brazil
| | - Laura Fabris
- Department of Applied Science and Technology, Politecnico di Torino, Torino, Italy
| | - Rodolfo Borges dos Reis
- Uro-Oncology Laboratory, Surgery and Anatomy Department, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Mateus Nóbrega Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Paraná, Brazil
| | - Dalila Lucíola Zanette
- Uro-Oncology Laboratory, Surgery and Anatomy Department, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto, São Paulo, Brazil
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Paraná, Brazil
| |
Collapse
|
28
|
Luo M, Luan X, Yang C, Chen X, Yuan S, Cao Y, Zhang J, Xie J, Luo Q, Chen L, Li S, Xiang W, Zhou J. Revisiting the potential of regulated cell death in glioma treatment: a focus on autophagy-dependent cell death, anoikis, ferroptosis, cuproptosis, pyroptosis, immunogenic cell death, and the crosstalk between them. Front Oncol 2024; 14:1397863. [PMID: 39184045 PMCID: PMC11341384 DOI: 10.3389/fonc.2024.1397863] [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: 03/08/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
Gliomas are primary tumors that originate in the central nervous system. The conventional treatment options for gliomas typically encompass surgical resection and temozolomide (TMZ) chemotherapy. However, despite aggressive interventions, the median survival for glioma patients is merely about 14.6 months. Consequently, there is an urgent necessity to explore innovative therapeutic strategies for treating glioma. The foundational study of regulated cell death (RCD) can be traced back to Karl Vogt's seminal observations of cellular demise in toads, which were documented in 1842. In the past decade, the Nomenclature Committee on Cell Death (NCCD) has systematically classified and delineated various forms and mechanisms of cell death, synthesizing morphological, biochemical, and functional characteristics. Cell death primarily manifests in two forms: accidental cell death (ACD), which is caused by external factors such as physical, chemical, or mechanical disruptions; and RCD, a gene-directed intrinsic process that coordinates an orderly cellular demise in response to both physiological and pathological cues. Advancements in our understanding of RCD have shed light on the manipulation of cell death modulation - either through induction or suppression - as a potentially groundbreaking approach in oncology, holding significant promise. However, obstacles persist at the interface of research and clinical application, with significant impediments encountered in translating to therapeutic modalities. It is increasingly apparent that an integrative examination of the molecular underpinnings of cell death is imperative for advancing the field, particularly within the framework of inter-pathway functional synergy. In this review, we provide an overview of various forms of RCD, including autophagy-dependent cell death, anoikis, ferroptosis, cuproptosis, pyroptosis and immunogenic cell death. We summarize the latest advancements in understanding the molecular mechanisms that regulate RCD in glioma and explore the interconnections between different cell death processes. By comprehending these connections and developing targeted strategies, we have the potential to enhance glioma therapy through manipulation of RCD.
Collapse
Affiliation(s)
- Maowen Luo
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Xingzhao Luan
- Department of Neurosurgery, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Chaoge Yang
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Xiaofan Chen
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Suxin Yuan
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Youlin Cao
- Department of Neurosurgery, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Jing Zhang
- School of Clinical Medicine, the Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, China
| | - Jiaying Xie
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Qinglian Luo
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Ligang Chen
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Shenjie Li
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Wei Xiang
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Jie Zhou
- Department of Neurosurgery, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
- School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| |
Collapse
|
29
|
Zhu X, Ye R, Jiang X, Zhang J. Smoking as a mediator in the association between major depressive disorder and schizophrenia on lung cancer risk: a bidirectional/multivariable and mediation Mendelian randomization study. Front Psychiatry 2024; 15:1367858. [PMID: 39176232 PMCID: PMC11338888 DOI: 10.3389/fpsyt.2024.1367858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/17/2024] [Indexed: 08/24/2024] Open
Abstract
Background & Aims Major depressive disorder and schizophrenia have been hypothesized to be closely associated with cancer. However, the associations between these psychiatric conditions and the development of lung cancer remain uncertain. This study aimed to explore the causal relationship among major depressive disorder, schizophrenia, and the risk of lung cancer. Methods Two-sample bidirectional/multivariable and mediation Mendelian randomization (MR) analyses were conducted. Genome-wide summary data on major depressive disorder (N=500,199) and schizophrenia (N=127,906) were utilized. Data on the risk of lung cancer (overall, adenocarcinoma, and squamous cell) were collected from a cohort of individuals of European ancestry (N=27,209). Three smoking-related behaviors (smoking initiation, pack years of smoking, and cigarettes smoked per day) were included in the multivariable and mediation MR analyses. Results Patients with schizophrenia had a significantly greater risk of developing lung cancer (odds ratio (OR) = 1.144, 95% confidence interval (95% CI): 1.048-1.248, P = 0.003). The number of cigarettes smoked per day partially mediated the relationship between schizophrenia and the overall risk of lung cancer (OR = 1.185, 95% CI: 1.112-1.264, P = 0.021, proportion of mediation effect: 61.033%). However, there is no reliable evidence indicating an association between major depressive disorder and the risk of lung cancer (overall, adenocarcinoma, and squamous cell cancer). Conclusions The findings indicated an association between schizophrenia and an increased risk of lung cancer, with smoking served as a partial mediator. When smoking was included in the regression analysis, the explanatory power of schizophrenia diagnosis was reduced, suggesting that smoking may be an important causal contributor to lung cancer in this population. Given the high prevalence of smoking among individuals with schizophrenia, these results underscore the need for further research to explore the underlying mechanisms of smoking's impact. Consequently, greater emphasis should be placed on monitoring the respiratory health of individuals with schizophrenia and implementing early interventions to address smoking-related behaviors.
Collapse
Affiliation(s)
- Xirong Zhu
- Taizhou Cancer Hospital, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou, China
| | - Ruizhi Ye
- Taizhou Cancer Hospital, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou, China
| | - Xianming Jiang
- Taizhou Cancer Hospital, Taizhou, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou, China
| | - Jing Zhang
- Wenling Hospital of Traditional Chinese Medicine, Wenling, China
- Department of General Practice, Wenling, China
| |
Collapse
|
30
|
Zhang Y, Han G, Gu J, Chen Z, Wu J. Role of tumor-associated macrophages in hepatocellular carcinoma: impact, mechanism, and therapy. Front Immunol 2024; 15:1429812. [PMID: 39170620 PMCID: PMC11335564 DOI: 10.3389/fimmu.2024.1429812] [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: 05/08/2024] [Accepted: 07/08/2024] [Indexed: 08/23/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly frequent malignancy worldwide. The occurrence and progression of HCC is a complex process closely related to the polarization of tumor-associated macrophages (TAMs) in the tumor microenvironment (TME). The polarization of TAMs is affected by a variety of signaling pathways and surrounding cells. Evidence has shown that TAMs play a crucial role in HCC, through its interaction with other immune cells in the TME. This review summarizes the origin and phenotypic polarization of TAMs, their potential impacts on HCC, and their mechanisms and potential targets for HCC immunotherapy.
Collapse
Affiliation(s)
- Yinqi Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
- National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Guoyong Han
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
- National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Jian Gu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
- National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Zhiqiang Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
- National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Jindao Wu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
- National Health Commission (NHC) Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| |
Collapse
|
31
|
Ieni A, Pizzimenti C, Fiorentino V, Franchina M, Germanò A, Raffa G, Martini M, Fadda G, Tuccari G. Immunohistochemical Profile of p62/SQSTM1/Sequestosome-1 in Human Low- and High-Grade Intracranial Meningiomas. Anal Cell Pathol (Amst) 2024; 2024:5573892. [PMID: 39131899 PMCID: PMC11315968 DOI: 10.1155/2024/5573892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 06/25/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024] Open
Abstract
Among autophagic-related proteins, p62/SQSTM1/Sequestosome-1 represents a relevant actor in cellular proliferation and neoplastic growth. Although, recently, p62 expression has been analyzed in different neurodegenerative and glial neoplastic diseases, no available information have been reported in meningiomas, which have an high epidemiological relevance being the second most common category of intracranial tumors after gliomas. Generally meningiomas have a benign behavior, but their recurrence is not uncommon mainly when atypical or anaplastic varieties occur. However, intranuclear vacuoles have been ultrastructurally observed in meningiomas, and they were labelled by p62 antibodies. Therefore, in the present study, we have investigated p62 immunohistochemical pattern in a cohort of 133 cases representative of low- and high-grade meningiomas, to verify if p62 expression may be related to clinicopathological data, thus achieving a potential prognostic role. The p62 immunoexpression was frequently found in the nucleus and cytoplasm of neoplastic elements, and utilizing an intensity-distribution score, 55 (41.3%) cases were considered as high expressors while 78 (58.7%) cases were instead recorded as low expressors. Fifteen cases exhibited recurrences of the disease, 14 of which were codified as high expressors. Moreover, a direct relationship between p62 and Mib-1 immunoexpression as well as between p62 and neoplastic grade have been documented. Finally, we suggest that impaired autophagic flux with an increase in p62 expression may be involved in the activation of NRF2 also contributing in the development of recurrence in meningioma patients.
Collapse
Affiliation(s)
- Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”Section of PathologyUniversity of Messina, Messina 98125, Italy
| | - Cristina Pizzimenti
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, NeurosurgeryUniversity of Messina, Viale Gazzi, Messina 98125, Italy
| | - Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”Section of PathologyUniversity of Messina, Messina 98125, Italy
| | - Mariausilia Franchina
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”Section of PathologyUniversity of Messina, Messina 98125, Italy
| | - Antonino Germanò
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, NeurosurgeryUniversity of Messina, Viale Gazzi, Messina 98125, Italy
| | - Giovanni Raffa
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, NeurosurgeryUniversity of Messina, Viale Gazzi, Messina 98125, Italy
| | - Maurizio Martini
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”Section of PathologyUniversity of Messina, Messina 98125, Italy
| | - Guido Fadda
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”Section of PathologyUniversity of Messina, Messina 98125, Italy
| | - Giovanni Tuccari
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”Section of PathologyUniversity of Messina, Messina 98125, Italy
| |
Collapse
|
32
|
Fan W, Zhu B, Chen S, Wu Y, Zhao X, Qiao L, Huang Z, Tang R, Chen J, Lau WY, Chen M, Li J, Kuang M, Peng Z. Survival in Patients With Recurrent Intermediate-Stage Hepatocellular Carcinoma: Sorafenib Plus TACE vs TACE Alone Randomized Clinical Trial. JAMA Oncol 2024; 10:1047-1054. [PMID: 38900435 PMCID: PMC11190833 DOI: 10.1001/jamaoncol.2024.1831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/29/2023] [Indexed: 06/21/2024]
Abstract
Importance Transarterial chemoembolization (TACE) is commonly used to treat patients with recurrent intermediate-stage hepatocellular carcinoma (HCC) and positive microvascular invasion (MVI); however, TACE alone has demonstrated unsatisfactory survival benefits. A previous retrospective study suggested that TACE plus sorafenib (SOR-TACE) may be a better therapeutic option compared with TACE alone. Objective To investigate the clinical outcomes of SOR-TACE vs TACE alone for patients with recurrent intermediate-stage HCC after R0 hepatectomy with positive MVI. Design, Setting, and Participants In this phase 3, open-label, multicenter randomized clinical trial, patients with recurrent intermediate-stage HCC and positive MVI were randomly assigned in a 1:1 ratio via a computerized minimization technique to either SOR-TACE treatment or TACE alone. This trial was conducted at 5 hospitals in China, and enrolled patients from October 2019 to December 2021, with a follow-up period of 24 months. Data were analyzed from June 2023 to September 2023. Interventions Randomization to on-demand TACE (conventional TACE: doxorubicin, 50 mg, mixed with lipiodol and gelatin sponge particles [diameter: 150-350 μm]; drug-eluting bead TACE: doxorubicin, 75 mg, mixed with drug-eluting particles [diameter: 100-300 μm or 300-500 μm]) (TACE group) or sorafenib, 400 mg, twice daily plus on-demand TACE (SOR-TACE group) (conventional TACE: doxorubicin, 50 mg, mixed with lipiodol and gelatin sponge particles [diameter, 150-350 μm]; drug-eluting bead TACE: doxorubicin, 75 mg, mixed with drug-eluting particles [diameter: 100-300 μm or 300-500 μm]). Main Outcomes and Measures The primary end point was overall survival by intention-to-treat analysis. Safety was assessed in patients who received at least 1 dose of study treatment. Results A total of 162 patients (median [range] age, 55 [28-75] years; 151 males [93.2%]), were randomly assigned to be treated with either SOR-TACE (n = 81) or TACE alone (n = 81). The median overall survival was significantly longer in the SOR-TACE group than in the TACE group (22.2 months vs 15.1 months; hazard ratio [HR], 0.55; P < .001). SOR-TACE also prolonged progression-free survival (16.2 months vs 11.8 months; HR, 0.54; P < .001), and improved the objective response rate when compared with TACE alone based on the modified Response Evaluation Criteria in Solid Tumors criteria (80.2% vs 58.0%; P = .002). Any grade adverse events were more common in the SOR-TACE group, but all adverse events responded well to treatment. No unexpected adverse events or treatment-related deaths occurred in this study. Conclusions and Relevance The results of this randomized clinical trial demonstrated that SOR-TACE achieved better clinical outcomes than TACE alone. These findings suggest that combined treatment should be used for patients with recurrent intermediate-stage HCC after R0 hepatectomy with positive MVI. Trial Registration ClinicalTrials.gov Identifier: NCT04103398.
Collapse
Affiliation(s)
- Wenzhe Fan
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bowen Zhu
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuling Chen
- Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanqin Wu
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiao Zhao
- Cancer Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liangliang Qiao
- Department of Interventional Oncology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhen Huang
- Department of Interventional Angiology, Huizhou First People’s Hospital, Huizhou, China
| | - Rong Tang
- Department of Hepatopancreatobiliary Surgery, Hainan General Hospital, Haikou, China
| | - Jinghua Chen
- Cancer Center, Guangzhou Twelfth People’s Hospital, Guangzhou, China
| | - Wan Yee Lau
- Faculty of Medicine, the Chinese University of Hong Kong, Prince of Wale Hospital, Shatin, New Territories, Hongkong, SAR, China
| | - Minshan Chen
- Department of Liver Surgery, Cancer Center of Sun Yat-sen University, Guangzhou, China
| | - Jiaping Li
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ming Kuang
- Center of Hepato-PancreatoBiliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhenwei Peng
- Cancer Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
33
|
Blancas-Zugarazo SS, Langley E, Hidalgo-Miranda A. Exosomal lncRNAs as regulators of breast cancer chemoresistance and metastasis and their potential use as biomarkers. Front Oncol 2024; 14:1419808. [PMID: 39148900 PMCID: PMC11324554 DOI: 10.3389/fonc.2024.1419808] [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: 04/18/2024] [Accepted: 07/16/2024] [Indexed: 08/17/2024] Open
Abstract
Breast cancer is the most common cancer in women and the leading cause of female deaths by cancer in the world worldwide. Hence, understanding the molecular mechanisms associated with breast cancer development and progression, including drug resistance and breast cancer metastasis, is essential for achieving the best management of breast cancer patients. Cancer-related long noncoding RNAs have been shown to be involved in the regulation of each stage of breast cancer progression. Additionally, exosomes are extracellular microvesicles that are central to intercellular communication and play an important role in tumorigenesis. Exosomes can be released from primary tumor cells into the bloodstream and transmit cellular signals to distant body sites. In this work, we review the findings regarding the cellular mechanisms regulated by exosomal lncRNAs that are essentials to chemoresistance development and metastasis of breast cancer. Likewise, we evaluate the outcomes of the potential clinical use of exosomal lncRNAs as breast cancer biomarkers to achieve personalized management of the patients. This finding highlights the importance of transcriptomic analysis of exosomal lncRNAs to understand the breast cancer tumorigenesis as well as to improve the clinical tests available for this disease.
Collapse
Affiliation(s)
- Sugela Susana Blancas-Zugarazo
- Cátedras CONAHCYT (Consejo Nacional de Humanidades Ciencia y Tecnología) - Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Elizabeth Langley
- Laboratorio de Cáncer Hormono Regulado, Instituto Nacional de Cancerología (INCAN), Mexico City, Mexico
| | - Alfredo Hidalgo-Miranda
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| |
Collapse
|
34
|
Nymoen HM, Alver TN, Horndalsveen H, Eide HA, Bjaanæs MM, Brustugun OT, Grønberg BH, Haakensen VD, Helland Å. Thoracic radiation in combination with erlotinib-results from a phase 2 randomized trial. Front Oncol 2024; 14:1412716. [PMID: 39148905 PMCID: PMC11324589 DOI: 10.3389/fonc.2024.1412716] [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: 04/05/2024] [Accepted: 07/09/2024] [Indexed: 08/17/2024] Open
Abstract
Background Radiotherapy (RT) can be used to reduce symptoms and maintain open airways for patients with non-small cell lung cancer when systemic treatment is not sufficient. For some patients, tumor control is not achieved due to radioresistance. Concurrent inhibition of epidermal growth factor receptors has been proposed as a strategy to overcome radioresistance but may increase toxicity. We performed a randomized trial to assess the efficacy, tolerance, and quality of life of concurrent erlotinib and palliative thoracic RT for patients with advanced non-small cell lung cancer. Methods Patients were randomized 1:1 to RT alone (arm A) or in combination with erlotinib (arm B). A computed tomography (CT) scan at baseline and one at 4-12 weeks after inclusion was used to evaluate treatment response. Adverse events were registered during treatment and the subsequent 30 days. Health-related quality-of-life questionnaires were completed by the patients at baseline, weeks 2, 6, and 20. Results A total of 114 patients were included. Of the 74 patients with CT scans available for evaluation of treatment effect, there were no significant differences in tumor size reduction between the two groups: median 14.5% reduction in the control arm A and 17.0% in the erlotinib arm B (p = 0.68). Overall survival was not significantly different between the two treatment arms: 7.0 and 7.8 months in arm A and arm B, respectively (log-rank p = 0.32). There was no significant increase in adverse events in the experimental arm, other than what is expected from erlotinib treatment alone. Overall, patients reported similar quality of life in both treatment arms. Conclusion Concurrent erlotinib and palliative thoracic RT for patients with advanced non-small cell lung cancer was well tolerated but did not improve the efficacy of the RT. Clinical trial registration ClinicalTrials.gov, identifier NCT02714530.
Collapse
Affiliation(s)
- Hanne Marte Nymoen
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- Department of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Tine Norman Alver
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- Department of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Henrik Horndalsveen
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- Department of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | | | | | - Odd Terje Brustugun
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- Department of Clinical Medicine, University of Oslo, Oslo, Norway
- Section of Oncology, Drammen Hospital, Vestre Viken Health Trust, Drammen, Norway
| | - Bjørn Henning Grønberg
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Oncology, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Vilde Drageset Haakensen
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- Department of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Åslaug Helland
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- Department of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
35
|
Huang LW, Shi Y, Boscardin WJ, Steinman MA. Cognitive Trajectories in Older Adults Diagnosed With Hematologic Malignant Neoplasms. JAMA Netw Open 2024; 7:e2431057. [PMID: 39212987 PMCID: PMC11365001 DOI: 10.1001/jamanetworkopen.2024.31057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024] Open
Abstract
Importance More people are surviving long-term after diagnosis with hematologic malignant neoplasm (HMN), yet there are limited data on cancer-related cognitive impairment in people with HMN. Better understanding cognitive outcomes after HMN in older adults is important for patient counseling and management. Objective To model cognitive trajectories and rates of cognitive decline before and after HMN diagnosis in older adults compared with a matched noncancer cohort. Design, Setting, and Participants In this population-based cohort study, older adults from the Health and Retirement Study (HRS) diagnosed with HMN between 1998 and 2016 after age 65 years were matched 1:3 to participants without cancer from the same HRS wave using propensity scores incorporating variables relevant to cognition. Cognitive trajectories were modeled with piecewise linear splines, and rates of cognitive decline before, during, and after diagnosis were compared in the 2 groups. Data were analyzed from April 2022 to April 2024. Exposures HMN diagnosis by Medicare diagnosis codes. Main Outcomes and Measures Cognitive function was assessed by the Langa-Weir cognitive summary score from 1992 to 2020. Sociodemographic and health-related variables relevant to cognition were incorporated into propensity scores. Results At baseline, there were 668 participants in the HMN cohort (mean [SD] age, 76.8 [7.6] years; 343 [51.3%] male; 72 [10.8%] Black, 33 [4.9%] Hispanic, and 585 [87.6%] White) and 1994 participants in the control cohort (mean [SD] age, 76.5 [7.3] years; 1020 [51.2%] male; 226 [11.3%] Black, 91 [4.6%] Hispanic, and 1726 [86.6%] White). The HMN cohort consisted predominantly of more indolent diagnoses, and only 96 patients (14.4%) received chemotherapy. Before and in the 2 years around the time of diagnosis, the HMN and control cohorts had similar rates of cognitive decline. At 1 year postdiagnosis and beyond, the rate of cognitive decline was slower in the HMN cohort (-0.18; 95% CI, -0.23 to -0.14) than in the control group (-0.24; 95% CI, -0.26 to -0.23) (P = .02), but this difference was no longer significant after accounting for the competing risk of death (HMN group, -0.27; 95% CI, -0.34 to -0.19; control group, -0.30; 95% CI, -0.33 to -0.27; P = .48). Conclusions and Relevance In this cohort study of older adults, the HMN and matched noncancer control cohorts had similar rates of cognitive decline before, during, and after diagnosis after accounting for the competing risk of death.
Collapse
Affiliation(s)
- Li-Wen Huang
- Division of Hematology/Oncology, Department of Medicine, San Francisco Veterans Affairs Medical Center, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco
| | - Ying Shi
- Division of Geriatrics, University of California San Francisco
- San Francisco Veterans Affairs Health Care System, San Francisco, California
| | - W. John Boscardin
- Division of Geriatrics, University of California San Francisco
- San Francisco Veterans Affairs Health Care System, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California San Francisco
| | - Michael A. Steinman
- Division of Geriatrics, University of California San Francisco
- San Francisco Veterans Affairs Health Care System, San Francisco, California
| |
Collapse
|
36
|
Esmaeili N, Bakheet A, Tse W, Liu S, Han X. Interaction of the intestinal cytokines-JAKs-STAT3 and 5 axes with RNA N6-methyladenosine to promote chronic inflammation-induced colorectal cancer. Front Oncol 2024; 14:1352845. [PMID: 39136000 PMCID: PMC11317299 DOI: 10.3389/fonc.2024.1352845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 06/25/2024] [Indexed: 08/15/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers, with a high mortality rate worldwide. Mounting evidence indicates that mRNA modifications are crucial in RNA metabolism, transcription, processing, splicing, degradation, and translation. Studies show that N6-methyladenosine (m6A) is mammalians' most common epi-transcriptomic modification. It has been demonstrated that m6A is involved in cancer formation, progression, invasion, and metastasis, suggesting it could be a potential biomarker for CRC diagnosis and developing therapeutics. Cytokines, growth factors, and hormones function in JAK/STAT3/5 signaling pathway, and they could regulate the intestinal response to infection, inflammation, and tumorigenesis. Reports show that the JAK/STAT3/5 pathway is involved in CRC development. However, the underlying mechanism is still unclear. Signal Transducer and Activator of Transcription 3/5 (STAT3, STAT5) can act as oncogenes or tumor suppressors in the context of tissue types. Also, epigenetic modifications and mutations could alter the balance between pro-oncogenic and tumor suppressor activities of the STAT3/5 signaling pathway. Thus, exploring the interaction of cytokines-JAKs-STAT3 and/or STAT5 with mRNA m6A is of great interest. This review provides a comprehensive overview of the characteristics and functions of m6A and JAKs-STAT3/5 and their relationship with gastrointestinal (GI) cancers.
Collapse
Affiliation(s)
- Nardana Esmaeili
- Division of Hematology and Oncology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
| | - Ahmed Bakheet
- Division of Hematology and Oncology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
| | - William Tse
- Division of Hematology and Oncology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
| | - Shujun Liu
- Division of Hematology and Oncology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
| | - Xiaonan Han
- Division of Hematology and Oncology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
- Cancer Genomics and Epigenomics Program, Case Comprehensive Cancer Center, Case Western Reserve University (CWRU), Cleveland, OH, United States
| |
Collapse
|
37
|
Li L, Jin T, Hu L, Ding J. Alternative splicing regulation and its therapeutic potential in bladder cancer. Front Oncol 2024; 14:1402350. [PMID: 39132499 PMCID: PMC11310127 DOI: 10.3389/fonc.2024.1402350] [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: 03/17/2024] [Accepted: 07/05/2024] [Indexed: 08/13/2024] Open
Abstract
Bladder cancer is one of the leading causes of mortality globally. The development of bladder cancer is closely associated with alternative splicing, which regulates human gene expression and enhances the diversity of functional proteins. Alternative splicing is a distinctive feature of bladder cancer, and as such, it may hold promise as a therapeutic target. This review aims to comprehensively discuss the current knowledge of alternative splicing in the context of bladder cancer. We review the process of alternative splicing and its regulation in bladder cancer. Moreover, we emphasize the significance of abnormal alternative splicing and splicing factor irregularities during bladder cancer progression. Finally, we explore the impact of alternative splicing on bladder cancer drug resistance and the potential of alternative splicing as a therapeutic target.
Collapse
Affiliation(s)
- Lina Li
- College of Medicine, Jinhua University of Vocational Technology, Jinhua, Zhejiang, China
| | - Ting Jin
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Liang Hu
- Department of Urology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Jin Ding
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| |
Collapse
|
38
|
Zakari S, Niels NK, Olagunju GV, Nnaji PC, Ogunniyi O, Tebamifor M, Israel EN, Atawodi SE, Ogunlana OO. Emerging biomarkers for non-invasive diagnosis and treatment of cancer: a systematic review. Front Oncol 2024; 14:1405267. [PMID: 39132504 PMCID: PMC11313249 DOI: 10.3389/fonc.2024.1405267] [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: 03/22/2024] [Accepted: 07/05/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a global health challenge, necessitating continuous advancements in diagnostic and treatment strategies. This review focuses on the utility of non-invasive biomarkers in cancer diagnosis and treatment, their role in early detection, disease monitoring, and personalized therapeutic interventions. Through a systematic review of the literature, we identified 45 relevant studies that highlight the potential of these biomarkers across various cancer types, such as breast, prostate, lung, and colorectal cancers. The non-invasive biomarkers discussed include liquid biopsies, epigenetic markers, non-coding RNAs, exosomal cargo, and metabolites. Notably, liquid biopsies, particularly those based on circulating tumour DNA (ctDNA), have emerged as the most promising method for early, non-invasive cancer detection due to their ability to provide comprehensive genetic and epigenetic information from easily accessible blood samples. This review demonstrates how non-invasive biomarkers can facilitate early cancer detection, accurate subtyping, and tailored treatment strategies, thereby improving patient outcomes. It underscores the transformative potential of non-invasive biomarkers in oncology, highlighting their application for enhancing early detection, survival rates, and treatment precision in cancer care. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023474749 PROSPERO, identifier CRD42023474749.
Collapse
Affiliation(s)
- Suleiman Zakari
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Ogun State, Nigeria
- Covenant Applied Informatics and Communication - Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Ogun State, Nigeria
- Department of Biochemistry, College of Medicine, Federal University of Health Sciences Otukpo, Otukpo, Benue State, Nigeria
| | - Nguedia K. Niels
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Ogun State, Nigeria
- Covenant Applied Informatics and Communication - Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Ogun State, Nigeria
- Biotechnology Centre, University of Yaounde I, Yaounde, Cameroon
| | - Grace V. Olagunju
- Department of Molecular Biology, New Mexico State University, Las Cruces, NM, United States
| | - Precious C. Nnaji
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Ogun State, Nigeria
| | - Oluwabusayo Ogunniyi
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Ogun State, Nigeria
| | - Mercy Tebamifor
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Ogun State, Nigeria
- Covenant Applied Informatics and Communication - Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Ogun State, Nigeria
| | - Emmanuel N. Israel
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Ogun State, Nigeria
- Covenant Applied Informatics and Communication - Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Ogun State, Nigeria
| | - Sunday E. Atawodi
- Department of Biochemistry, Federal University Lokoja, Lokoja, Kogi State, Nigeria
| | - Olubanke Olujoke Ogunlana
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Ogun State, Nigeria
- Covenant Applied Informatics and Communication - Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Ogun State, Nigeria
| |
Collapse
|
39
|
Nicolini A, Ferrari P. Involvement of tumor immune microenvironment metabolic reprogramming in colorectal cancer progression, immune escape, and response to immunotherapy. Front Immunol 2024; 15:1353787. [PMID: 39119332 PMCID: PMC11306065 DOI: 10.3389/fimmu.2024.1353787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/04/2024] [Indexed: 08/10/2024] Open
Abstract
Metabolic reprogramming is a k`ey hallmark of tumors, developed in response to hypoxia and nutrient deficiency during tumor progression. In both cancer and immune cells, there is a metabolic shift from oxidative phosphorylation (OXPHOS) to aerobic glycolysis, also known as the Warburg effect, which then leads to lactate acidification, increased lipid synthesis, and glutaminolysis. This reprogramming facilitates tumor immune evasion and, within the tumor microenvironment (TME), cancer and immune cells collaborate to create a suppressive tumor immune microenvironment (TIME). The growing interest in the metabolic reprogramming of the TME, particularly its significance in colorectal cancer (CRC)-one of the most prevalent cancers-has prompted us to explore this topic. CRC exhibits abnormal glycolysis, glutaminolysis, and increased lipid synthesis. Acidosis in CRC cells hampers the activity of anti-tumor immune cells and inhibits the phagocytosis of tumor-associated macrophages (TAMs), while nutrient deficiency promotes the development of regulatory T cells (Tregs) and M2-like macrophages. In CRC cells, activation of G-protein coupled receptor 81 (GPR81) signaling leads to overexpression of programmed death-ligand 1 (PD-L1) and reduces the antigen presentation capability of dendritic cells. Moreover, the genetic and epigenetic cell phenotype, along with the microbiota, significantly influence CRC metabolic reprogramming. Activating RAS mutations and overexpression of epidermal growth factor receptor (EGFR) occur in approximately 50% and 80% of patients, respectively, stimulating glycolysis and increasing levels of hypoxia-inducible factor 1 alpha (HIF-1α) and MYC proteins. Certain bacteria produce short-chain fatty acids (SCFAs), which activate CD8+ cells and genes involved in antigen processing and presentation, while other mechanisms support pro-tumor activities. The use of immune checkpoint inhibitors (ICIs) in selected CRC patients has shown promise, and the combination of these with drugs that inhibit aerobic glycolysis is currently being intensively researched to enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Andrea Nicolini
- Department of Oncology, Transplantations and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Paola Ferrari
- Unit of Oncology, Department of Medical and Oncological Area, Azienda Ospedaliera-Universitaria Pisana, Pisa, Italy
| |
Collapse
|
40
|
Calik J, Oślizło M, Słocka-Romaniuk B, Elsaftawy A, Sauer N. Case report: Sequential treatment strategy for advanced basal cell carcinoma in Gorlin-Goltz syndrome: integration of vismodegib, radiotherapy, surgery, and high-intensity focused ultrasound. Front Oncol 2024; 14:1428702. [PMID: 39091908 PMCID: PMC11291366 DOI: 10.3389/fonc.2024.1428702] [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: 05/06/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Managing advanced basal cell carcinoma (BCC) in patients with Gorlin-Goltz syndrome presents unique clinical challenges due to the tumor's aggressive nature and potential for widespread metastasis. This case study details a sequential treatment regimen for a 68-year-old female patient with an extensive, inoperable BCC. Employing a multimodal approach that integrates radiotherapy, the Hedgehog pathway inhibitor vismodegib, and High-Intensity Focused Ultrasound (HIFU), we demonstrate the potential for nearly complete remission in a patient with advanced BCC. Initial treatment with radiotherapy and vismodegib reduced tumor size significantly, but the largest mass displayed resistance over time, signifying the need for innovative therapies. Subsequent HIFU treatment targeted individual lesions, showcasing a non-invasive method that provided precise treatment while mitigating systemic side effects. The case emphasizes the necessity of continual adaptation in treatment plans to address the development of resistance and underscores the importance of incorporating new technologies and targeted therapies for complex BCC cases. The successful outcome of this integrated strategy suggests a promising direction for future research and highlights the importance of multidisciplinary approaches that tailor treatment to individual patient needs, tumor characteristics, and evolving therapeutic landscapes.
Collapse
Affiliation(s)
- Jacek Calik
- Department of Clinical Oncology, Wroclaw Medical University, Wrocław, Poland
- Old Town Clinic, Wrocław, Poland
| | | | - Beata Słocka-Romaniuk
- Department of Radiotherapy, Lower Silesian Center of Pulmonology, Oncology and Hematology in Wrocław, Wrocław, Poland
| | - Ahmed Elsaftawy
- Department of Plastic and Hand Surgery, St. Jadwiga Śląska Hospital, Trzebnica, Poland
| | - Natalia Sauer
- Old Town Clinic, Wrocław, Poland
- Department of Clinical Pharmacology, Faculty of Pharmacy, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
41
|
Mustafa M, Abbas K, Alam M, Habib S, Zulfareen, Hasan GM, Islam S, Shamsi A, Hassan I. Investigating underlying molecular mechanisms, signaling pathways, emerging therapeutic approaches in pancreatic cancer. Front Oncol 2024; 14:1427802. [PMID: 39087024 PMCID: PMC11288929 DOI: 10.3389/fonc.2024.1427802] [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: 05/04/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Pancreatic adenocarcinoma, a clinically challenging malignancy constitutes a significant contributor to cancer-related mortality, characterized by an inherently poor prognosis. This review aims to provide a comprehensive understanding of pancreatic adenocarcinoma by examining its multifaceted etiologies, including genetic mutations and environmental factors. The review explains the complex molecular mechanisms underlying its pathogenesis and summarizes current therapeutic strategies, including surgery, chemotherapy, and emerging modalities such as immunotherapy. Critical molecular pathways driving pancreatic cancer development, including KRAS, Notch, and Hedgehog, are discussed. Current therapeutic strategies, including surgery, chemotherapy, and radiation, are discussed, with an emphasis on their limitations, particularly in terms of postoperative relapse. Promising research areas, including liquid biopsies, personalized medicine, and gene editing, are explored, demonstrating the significant potential for enhancing diagnosis and treatment. While immunotherapy presents promising prospects, it faces challenges related to immune evasion mechanisms. Emerging research directions, encompassing liquid biopsies, personalized medicine, CRISPR/Cas9 genome editing, and computational intelligence applications, hold promise for refining diagnostic approaches and therapeutic interventions. By integrating insights from genetic, molecular, and clinical research, innovative strategies that improve patient outcomes can be developed. Ongoing research in these emerging fields holds significant promise for advancing the diagnosis and treatment of this formidable malignancy.
Collapse
Affiliation(s)
- Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Zulfareen
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sidra Islam
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates
| | - Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
42
|
Le Menn G, Pikkarainen K, Mennerich D, Miroszewska D, Kietzmann T, Chen Z. USP28 protects development of inflammation in mouse intestine by regulating STAT5 phosphorylation and IL22 production in T lymphocytes. Front Immunol 2024; 15:1401949. [PMID: 39076972 PMCID: PMC11284026 DOI: 10.3389/fimmu.2024.1401949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/27/2024] [Indexed: 07/31/2024] Open
Abstract
Introduction Ubiquitin-specific proteases (USPs), a large subset of more than 50 deubiquitinase proteins, have recently emerged as promising targets in cancer. However, their role in immune cell regulation, particularly in T cell activation, differentiation, and effector functions, remains largely unexplored. Methods We utilized a USP28 knockout mouse line to study the effect of USP28 on T cell activation and function, and its role in intestinal inflammation using the dextran sulfate sodium (DSS)-induced colitis model and a series of in vitro assays. Results Our results show that USP28 exerts protective effects in acute intestinal inflammation. Mechanistically, USP28 knockout mice (USP28-/-) exhibited an increase in total T cells mainly due to an increased CD8+ T cell content. Additionally, USP28 deficiency resulted in early defects in T cell activation and functional changes. Specifically, we observed a reduced expression of IL17 and an increase in inducible regulatory T (iTreg) suppressive functions. Importantly, activated T cells lacking USP28 showed increased STAT5 phosphorylation. Consistent with these findings, these mice exhibited increased susceptibility to acute DSS-induced intestinal inflammation, accompanied by elevated IL22 cytokine levels. Conclusions Our findings demonstrate that USP28 is essential for T cell functionality and protects mice from acute DSS-induced colitis by regulating STAT5 signaling and IL22 production. As a T cell regulator, USP28 plays a crucial role in immune responses and intestinal health.
Collapse
Affiliation(s)
- Gwenaëlle Le Menn
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Keela Pikkarainen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Daniela Mennerich
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Dominika Miroszewska
- Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, University of Gdańsk, Gdańsk, Poland
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Zhi Chen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
43
|
Qiao W, Li J, Wang P, Zhang Y, Jin R, Li J. Prognostic nomogram based on the gamma-glutamyl transpeptidase-to-platelet ratio for patients with compensated cirrhotic hepatocellular carcinoma after local ablation. Front Oncol 2024; 14:1406764. [PMID: 39055565 PMCID: PMC11269228 DOI: 10.3389/fonc.2024.1406764] [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: 03/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Background Hepatocellular carcinoma (HCC) patients with compensated cirrhosis typically face a high prevalence and unfavorable prognosis. However, there is currently a deficiency in prediction models to anticipate the prognosis of these patients. Therefore, our study included the Gamma-glutamyl transpeptidase-to-platelet ratio (GPR) in analysis and aimed to develop a nomogram for HCC patients with compensated cirrhosis after local ablation. Methods Enrolling 669 patients who underwent local ablation at Beijing You'an Hospital during the period from January 1, 2014, to December 31, 2022, this study focused on individuals with compensated cirrhotic HCC. In a ratio of 7:3, patients were allocated to the training cohort (n=468) and the validation cohort (n=201). Lasso-Cox regression was employed to identify independent prognostic factors for overall survival (OS). Subsequently, a nomogram was constructed using these factors and was validated through receiver operating characteristic (ROC) curves, calibration curves, and decision curve analysis (DCA). Results GPR, age, and hemoglobin were identified by Lasso-Cox regression as independent prognostic factors of the nomogram. The area under the ROC curves (AUCs) for 3-, 5-, and 8-year OS (0.701, 0.755, and 0.768 for the training cohort; 0.684, 0.707, and 0.778 for the validation cohort), and C-indices (0.695 for training cohort; 0.679 for validation cohort) exhibited the excellent predictive ability of the nomogram. Calibration curves and DCA curves indicated favorable calibration performance and clinical utility. Patients were further stratified into two risk groups according to the median nomogram score. There existed an obvious distinction between the two groups both in the training cohort and validation cohort. Conclusion In summary, this research established and validated a novel nomogram to predict OS, which had good predictive power for HCC patients with compensated cirrhosis after local ablation.
Collapse
Affiliation(s)
- Wenying Qiao
- Hepatic Disease and Oncology Minimally Invasive Interventional Center, Beijing You’an Hospital, Capital Medical University, Beijing, China
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Jiashuo Li
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Peiyi Wang
- Qingdao Agricultural University, Qingdao, China
| | - Yuanyuan Zhang
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Ronghua Jin
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Jianjun Li
- Hepatic Disease and Oncology Minimally Invasive Interventional Center, Beijing You’an Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
44
|
Zhang H, Zhang Y, Zhu Y, Dong T, Liu Z. Understanding the treatment response and resistance to targeted therapies in non-small cell lung cancer: clinical insights and perspectives. Front Oncol 2024; 14:1387345. [PMID: 39055566 PMCID: PMC11269125 DOI: 10.3389/fonc.2024.1387345] [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: 02/17/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Lung cancer remains the leading cause of mortality worldwide. Non-small cell lung cancer (NSCLC) is the most common subtype of lung cancer with a generally poor prognosis. In recent years, advances in targeted therapy and sequencing technology have brought significant improvement in the therapeutic outcomes of patients with advanced NSCLC. Targeted inhibitors directed against specific mutated or rearranged oncogenes, such as epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), and receptor tyrosine kinase ROS proto-oncogene 1(ROS1) among others, exhibit promising anti-tumor activity. Unfortunately, some patients develop acquired resistance and disease progression soon after initial remission. Despite the continuous development of new drugs and strategies to overcome drug resistance, it is still a major challenge in the treatment of NSCLC. The landscape of targeted therapy for NSCLC is evolving rapidly in response to the pace of scientific research. This study aimed to provide a comprehensive review of tumor target antigens and agents related to targeted therapy in NSCLC.
Collapse
Affiliation(s)
- Hang Zhang
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Yingying Zhang
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, China
| | - Yingying Zhu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Dong
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Zheng Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
45
|
Si Y, Song N, Ji Y. Construction of a nomogram model for predicting the outcome of debulking surgery for ovarian cancer on the basis of clinical indicators. Front Oncol 2024; 14:1421247. [PMID: 39050577 PMCID: PMC11266020 DOI: 10.3389/fonc.2024.1421247] [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: 04/22/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Objective This study aimed to investigate the risk factors affecting satisfaction with debulking surgery for ovarian cancer and establish a preoperative clinical predictive model. Methods Clinical data from 131 patients who underwent ovarian cancer debulking surgery at Jiangnan University Affiliated Hospital between 2016 and 2022 were collected. Patients were randomly separated into an experimental group and a control group in a 7:3 ratio. On the basis of intraoperative outcomes, patients were grouped as either surgery-satisfactory or surgery-unsatisfactory. Clinical indicators were compared through single-factor analysis between groups. Significantly different factors (p < 0.1) were further analyzed through multivariate logistic regression. A predictive nomogram model was developed and validated by receiver operating characteristic (ROC), calibration, and clinical decision curves. Results Single-factor analysis revealed the significance of factors such as albumin levels, alkaline phosphatase (ALP), ECOG scores, CA125, HE4, and lymph node metastasis. Multivariate regression analysis identified albumin levels, ALP, ECOG scores, HE4, and lymph node metastasis as independent risk factors for satisfactory surgical outcomes in patients with ovarian cancer undergoing debulking surgery as (p < 0.05). A clinical predictive model was successfully constructed. ROC curves showed AUC values of 0.818 and 0.796 for the experimental and validation groups, respectively. Internal validation through the bootstrap method confirmed the model's fit in both groups. Meanwhile, the clinical decision curve demonstrated the model's high utility. Conclusion Independent risk factors associated with satisfactory tumor reduction in patients with ovarian cancer undergoing debulking surgery included decreased albumin levels, ALP > 137 U/L, ECOG = 1 score, HE4 > 140 pmol/L, and lymph node metastasis. Constructing a clinical predictive model through logistic regression analysis enables individualized testing and maximizes clinical benefits.
Collapse
Affiliation(s)
- Yuanyuan Si
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Ningjia Song
- Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Yong Ji
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
46
|
Chen W, Cao K, Zhang L, Zhao X, Chen B, Li W, Shang R, Sun L, Jiang Z, Wang J, Xue W. Efficacy and safety evaluation of frontline immunotherapy combinations in advanced esophageal squamous cell carcinoma: a network meta-analysis highlighting the value of PD-L1 expression positivity scores. Front Immunol 2024; 15:1414753. [PMID: 39050848 PMCID: PMC11266001 DOI: 10.3389/fimmu.2024.1414753] [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: 04/09/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction The systematic review and network meta-analysis (NMA) consolidate all relevant randomized controlled trials (RCTs) related to initial immunotherapy treatments for advanced esophageal squamous cell carcinoma (ESCC). Our goal is to thoroughly assess the effectiveness and safety of various immunotherapy methods, focusing on overall survival (OS) and progression-free survival (PFS) among patients with advanced ESCC positive for PD-L1. Methods We conducted a systematic search of the PubMed, Embase, Cochrane Library, and Web of Science databases, covering all records from their inception until January 22, 2024. The inclusion criteria targeted patients with advanced ESCC undergoing first-line immunotherapy or chemotherapy, limiting the study selection to randomized controlled trials (RCTs) exclusively. The study upholds the values of openness, originality, and dependability, as evidenced by its enrollment in the Prospective Register of Systematic Reviews (CRD42024504992). Results Our analysis encompasses 7 RCTs, totaling 4688 patients, and evaluates 8 distinct immunotherapy combinations. In advanced ESCC patients irrespective of PD-L1 expression, both sintilimab-chemotherapy and toripalimab-chemotherapy regimens demonstrated comparable OS benefits (HR=0.92, 95% CI: 0.64-1.33). The most pronounced PFS advantages were seen with sintilimab-chemotherapy and camrelizumab-chemotherapy as compared to standard chemotherapy (HR=0.56, 95% CI: 0.46-0.58). Notably, camrelizumab-chemotherapy (HR=0.83, 95% CI: 0.59-1.16) and nivolumab-ipilimumab (HR=0.84, 95% CI: 0.60-1.17) demonstrated significant safety profiles over chemotherapy alone. Subgroup analysis based on PD-L1 expression revealed nivolumab-chemotherapy to yield the highest OS benefit (HR=0.54, 95% CI: 0.37-0.79) in ESCC patients with PD-L1 expression ≥1%. Furthermore, camrelizumab-chemotherapy (HR=0.51, 95% CI: 0.39-0.67) exhibited superior PFS benefits. Among patients with PD-L1 expression ≥10%, camrelizumab-chemotherapy (HR=0.52, 95% CI: 0.35-0.78) emerged as the most efficacious in improving OS, while serplulimab-chemotherapy (HR=0.48, 95% CI: 0.34-0.68) was associated with the longest PFS benefit. Conclusion The integration of immune checkpoint inhibitors (ICIs) with chemotherapy appears to significantly enhance survival outcomes in patients with advanced ESCC compared to chemotherapy alone. Sintilimab-chemotherapy is potentially the optimal regimen for patients without PD-L1 expression. In contrast, nivolumab-chemotherapy and camrelizumab-chemotherapy are likely to offer the best OS and PFS benefits, respectively, in patients with PD-L1 expression ≥1%. Among those with PD-L1 expression ≥10%, camrelizumab-chemotherapy is projected to provide the greatest OS advantage, whereas serplulimab-chemotherapy is anticipated to offer the most prolonged PFS benefit. Since most of the patients in this study originated from Asia, the above findings are more applicable to the Asian population. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024504992.
Collapse
Affiliation(s)
- Wei Chen
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| | - Keming Cao
- School of Pharmacy, North China University of Science and Technology, Heibei, China
| | - Lili Zhang
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| | - Xingyu Zhao
- School of Pharmaceutical Sciences, Capital Medical University School, Beijing, China
| | - Bixiao Chen
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Wei Li
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| | - Rongguo Shang
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| | - Lichaoyue Sun
- Pharmacy Department, Aerospace Center Hospital, Beijing, China
| | - Ze Jiang
- Pharmaceutical Department, TongRen Hospital, Capital Medical University, Beijing, China
| | - Jingxin Wang
- School of Pharmacy, North China University of Science and Technology, Heibei, China
| | - Wenxin Xue
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| |
Collapse
|
47
|
Chen Y, Zhang W, Xu X, Xu B, Yang Y, Yu H, Li K, Liu M, Qi L, Jiao X. Gene signatures of copper metabolism related genes may predict prognosis and immunity status in Ewing's sarcoma. Front Oncol 2024; 14:1388868. [PMID: 39050579 PMCID: PMC11267503 DOI: 10.3389/fonc.2024.1388868] [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: 02/20/2024] [Accepted: 06/12/2024] [Indexed: 07/27/2024] Open
Abstract
Background Cuproptosis is copper-induced cell death. Copper metabolism related genes (CMRGs) were demonstrated that used to assess the prognosis out of tumors. In the study, CMRGs were tested for their effect on TME cell infiltration in Ewing's sarcoma (ES). Methods The GEO and ICGC databases provided the mRNA expression profiles and clinical features for downloading. In the GSE17674 dataset, 22prognostic-related copper metabolism related genes (PR-CMRGs) was identified by using univariate regression analysis. Subsequently, in order to compare the survival rates of groups with high and low expression of these PR-CMRGs,Kaplan-Meier analysis was implemented. Additionally, correlations among them were examined. The study employed functional enrichment analysis to investigate probable underlying pathways, while GSVA was applied to evaluate enriched pathways in the ES (Expression Set). Through an unsupervised clustering algorithm, samples were classified into two clusters, revealing significant differences in survival rates and levels of immune infiltration. Results Using Lasso and step regression methods, five genes (TFRC, SORD, SLC11A2, FKBP4, and AANAT) were selected as risk signatures. According to the Kaplan-Meier survival analysis, the high-risk group had considerably lower survival rates than the low-risk group(p=6.013e-09). The area under the curve (AUC) values for the receiver operating characteristic (ROC) curve were 0.876, 0.883, and 0.979 for 1, 3, and 5 years, respectively. The risk model was further validated in additional datasets, namely GSE63155, GSE63156, and the ICGC datasets. To aid in outcome prediction, a nomogram was developed that incorporated risk levels and clinical features. This nomogram's performance was effectively validated through calibration curves.Additionally, the study evaluated the variations in immune infiltration across different risk groups, as well as high-expression and low-expression groups. Importantly, several drugs were identified that displayed sensitivity, offering potential therapeutic options for ES. Conclusion The findings above strongly indicate that CMRGs play crucial roles in predicting prognosis and immune status in ES.
Collapse
Affiliation(s)
- Yongqin Chen
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wencan Zhang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiao Xu
- Sterile Supply Department, The First People Hospital of Jinan, Jinan, Shandong, China
| | - Biteng Xu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yuxuan Yang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Haozhi Yu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ke Li
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Mingshan Liu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lei Qi
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiejia Jiao
- Department of Orthopedics, The Second Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
48
|
Zhang L, Yu F, Zhang Y, Li P. Implications of lncRNAs in Helicobacter pylori-associated gastrointestinal cancers: underlying mechanisms and future perspectives. Front Cell Infect Microbiol 2024; 14:1392129. [PMID: 39035354 PMCID: PMC11257847 DOI: 10.3389/fcimb.2024.1392129] [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: 02/27/2024] [Accepted: 06/19/2024] [Indexed: 07/23/2024] Open
Abstract
Helicobacter pylori (H. pylori) is a harmful bacterium that is difficult to conveniently diagnose and effectively eradicate. Chronic H. pylori infection increases the risk of gastrointestinal diseases, even cancers. Despite the known findings, more underlying mechanisms are to be deeply explored to facilitate the development of novel prevention and treatment strategies of H. pylori infection. Long noncoding RNAs (lncRNAs) are RNAs with more than 200 nucleotides. They may be implicated in cell proliferation, inflammation and many other signaling pathways of gastrointestinal cancer progression. The dynamic expression of lncRNAs indicates their potential to be diagnostic or prognostic biomarkers. In this paper, we comprehensively summarize the processes of H. pylori infection and the treatment methods, review the known findings of lncRNA classification and functional mechanisms, elucidate the roles of lncRNAs in H. pylori-related gastrointestinal cancer, and discuss the clinical perspectives of lncRNAs.
Collapse
Affiliation(s)
- Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | | | | | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
49
|
Xia X, Du W, Zhang Y, Li Y, Yu M, Liu Y. Efficacy of epidermal growth factor receptor-tyrosine kinase inhibitor for lung adenosquamous cell carcinoma harboring EGFR mutation: a retrospective study and pooled analysis. Front Oncol 2024; 14:1354854. [PMID: 39026979 PMCID: PMC11254804 DOI: 10.3389/fonc.2024.1354854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
Objectives To explore the efficacy of epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) on lung adenosquamous cell carcinoma (ASC) with EGFR mutation. Methods Efficacy of EGFR-TKIs in the treatment of advanced or recurrent lung ASC with EGFR mutations was assessed retrospectively in 44 patients. Pooled analysis of 74 patients using EGFR-TKIs, including 30 patients selected from 11 publications, was conducted. Results In our retrospective research, patients treated with EGFR-TKI in ASC with EGFR mutations had objective response rate (ORR) of 54.5%, disease control rate (DCR) of 79.5%, median progression free survival (mPFS) of 8.8 months, and median overall survival (mOS) of 19.43 months, respectively. A pooled analysis reveals ORR, DCR, mPFS, and mOS are, respectively, 63.4%, 85.9%, 10.00 months, and 21.37 months for ASC patients. In patients with deletions in exon 19 and exon 21 L858R mutations, mPFS (11.0 versus 10.0 months, P=0.771) and mOS (23.67 versus 20.33 months, P=0.973) were similar. Erlotinib or gefitinib-treated patients had an overall survival trend that was superior to that of icotinib-treated patients. Conclusions ASC harboring EGFR mutations can be treated with EGFR-TKI in a similar manner to Adenocarcinoma (ADC) harboring EGFR mutations. There is still a need for further investigation to identify the separate roles of ASC's two components in treating EGFR.
Collapse
Affiliation(s)
- Xueming Xia
- Division of Head & Neck Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Du
- Department of Targeting Therapy & Immunology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yanying Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Min Yu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yongmei Liu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
50
|
Barbosa LC, Machado GC, Heringer M, Ferrer VP. Identification of established and novel extracellular matrix components in glioblastoma as targets for angiogenesis and prognosis. Neurogenetics 2024; 25:249-262. [PMID: 38775886 DOI: 10.1007/s10048-024-00763-x] [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: 03/06/2024] [Accepted: 05/10/2024] [Indexed: 07/16/2024]
Abstract
Glioblastomas (GBM) are aggressive tumors known for their heterogeneity, rapid proliferation, treatment resistance, and extensive vasculature. Angiogenesis, the formation of new vessels, involves endothelial cell (EC) migration and proliferation. Various extracellular matrix (ECM) molecules regulate EC survival, migration, and proliferation. Culturing human brain EC (HBMEC) on GBM-derived ECM revealed a decrease in EC numbers compared to controls. Through in silico analysis, we explored ECM gene expression differences between GBM and brain normal glia cells and the impact of GBM microenvironment on EC ECM transcripts. ECM molecules such as collagen alpha chains (COL4A1, COL4A2, p < 0.0001); laminin alpha (LAMA4), beta (LAMB2), and gamma (LAMC1) chains (p < 0.0005); neurocan (NCAN), brevican (BCAN) and versican (VCAN) (p < 0.0005); hyaluronan synthase (HAS) 2 and metalloprotease (MMP) 2 (p < 0.005); MMP inhibitors (TIMP1-4, p < 0.0005), transforming growth factor beta-1 (TGFB1) and integrin alpha (ITGA3/5) (p < 0.05) and beta (ITGB1, p < 0.0005) chains showed increased expression in GBM. Additionally, GBM-influenced EC exhibited elevated expression of COL5A3, COL6A1, COL22A1 and COL27A1 (p < 0.01); LAMA1, LAMB1 (p < 0.001); fibulins (FBLN1/2, p < 0.01); MMP9, HAS1, ITGA3, TGFB1, and wingless-related integration site 9B (WNT9B) (p < 0.01) compared to normal EC. Some of these molecules: COL5A1/3, COL6A1, COL22/27A1, FBLN1/2, ITGA3/5, ITGB1 and LAMA1/B1 (p < 0.01); NCAN, HAS1, MMP2/9, TIMP1/2 and TGFB1 (p < 0.05) correlated with GBM patient survival. In conclusion, this study identified both established and novel ECM molecules regulating GBM angiogenesis, suggesting NCAN and COL27A1 are new potential prognostic biomarkers for GBM.
Collapse
Affiliation(s)
- Lucas Cunha Barbosa
- Graduation Program of Pathological Anatomy, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory of Cellular and Molecular Biology of Tumors, Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, Brazil
| | - Gabriel Cardoso Machado
- Graduation Program of Pathological Anatomy, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory of Cellular and Molecular Biology of Tumors, Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, Brazil
| | - Manoela Heringer
- Brain's Biomedicine Lab, Paulo Niemeyer State Brain Institute, Rio de Janeiro, Brazil
| | - Valéria Pereira Ferrer
- Graduation Program of Pathological Anatomy, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- Laboratory of Cellular and Molecular Biology of Tumors, Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, Brazil.
| |
Collapse
|