Pub Date : 2025-03-29eCollection Date: 2025-01-01DOI: 10.2147/CMAR.S508567
Zhejing Wu, Jingdong Li
Fatty pancreas disease (FPD) refers to excessive fat accumulation and fat infiltration in pancreatic tissue. Factors such as obesity, diabetes, non-alcoholic fatty liver disease (NAFLD), and alcohol consumption can contribute to the development of FPD. Patients with FPD typically lack obvious clinical symptoms or signs, and diagnosis primarily relies on imaging techniques. Currently, there is limited attention to this disease both domestically and internationally. FPD is closely associated with pancreatic-related diseases (eg, diabetes, pancreatitis, pancreatic cancer). Pancreatic cancer, characterized by high mortality and low survival rates, has been linked to FPD in terms of its occurrence, progression, and patient prognosis. FPD is considered a potential early clinical manifestation of pancreatic cancer and may promote distant metastasis. However, the mechanisms by which FPD contributes to pancreatic carcinogenesis remain unclear. Additionally, Studies have found that FPD can lead to perioperative complications (postoperative pancreatic fistula, postoperative nonalcoholic fatty liver, endoscopic retrograde cholangiopancreatography pancreatitis), which are closely related to the prognosis of patients after pancreatic surgery.Although FPD is challenging to diagnose, its instability allows for clinical management through early dietary interventions, oral medications, and, when necessary, bariatric surgery to alter disease progression. Whether targeting adipocytes, lipid metabolism, or adipocyte-related cytokines could serve as novel intervention strategies for pancreatic cancer remains a critical area for further investigation.
{"title":"Correlation Between Fatty Pancreas Disease and Pancreatic Diseases, Perioperative Complications of Pancreatic Surgery.","authors":"Zhejing Wu, Jingdong Li","doi":"10.2147/CMAR.S508567","DOIUrl":"10.2147/CMAR.S508567","url":null,"abstract":"<p><p>Fatty pancreas disease (FPD) refers to excessive fat accumulation and fat infiltration in pancreatic tissue. Factors such as obesity, diabetes, non-alcoholic fatty liver disease (NAFLD), and alcohol consumption can contribute to the development of FPD. Patients with FPD typically lack obvious clinical symptoms or signs, and diagnosis primarily relies on imaging techniques. Currently, there is limited attention to this disease both domestically and internationally. FPD is closely associated with pancreatic-related diseases (eg, diabetes, pancreatitis, pancreatic cancer). Pancreatic cancer, characterized by high mortality and low survival rates, has been linked to FPD in terms of its occurrence, progression, and patient prognosis. FPD is considered a potential early clinical manifestation of pancreatic cancer and may promote distant metastasis. However, the mechanisms by which FPD contributes to pancreatic carcinogenesis remain unclear. Additionally, Studies have found that FPD can lead to perioperative complications (postoperative pancreatic fistula, postoperative nonalcoholic fatty liver, endoscopic retrograde cholangiopancreatography pancreatitis), which are closely related to the prognosis of patients after pancreatic surgery.Although FPD is challenging to diagnose, its instability allows for clinical management through early dietary interventions, oral medications, and, when necessary, bariatric surgery to alter disease progression. Whether targeting adipocytes, lipid metabolism, or adipocyte-related cytokines could serve as novel intervention strategies for pancreatic cancer remains a critical area for further investigation.</p>","PeriodicalId":9479,"journal":{"name":"Cancer Management and Research","volume":"17 ","pages":"723-730"},"PeriodicalIF":2.5,"publicationDate":"2025-03-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11963821/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143771401","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-03-27eCollection Date: 2025-01-01DOI: 10.2147/CMAR.S508047
Ting Wang, Rui Liu
The S100 protein family comprises 25 known members that modulate variously basic biological behaviors of cells by binding Ca2+ and activating Ca2+-signaling pathways. As the primary cause of cancer-related death, lung cancer is closely associated with several S100 proteins, like S100A2, S100A4, S100A6, S100A8/9, etc. Notably, the functions and mechanisms of different S100 proteins vary in every sub-type of lung cancer. Overall speaking, the abnormal expression of S100 proteins is predominantly observed in lung adenocarcinoma, while their roles are limited in small-cell lung cancer. This review, which presents an update on our previously published review of S100 proteins in lung cancer (2021), aims to enable readers having a deeper understanding of the roles of different S100 proteins in three main sub-types of lung cancer, as well as to facilitate their future researches. It focuses on relevant studies examining the functions of S100 proteins in lung adenocarcinoma, squamous carcinoma, and small-cell lung cancer. This review was conducted based on this standard, and provides a comprehensive evaluation of the literature review on S100 proteins as well as enhances understanding of the relationship between S100 proteins and every sub-type of lung cancer from a new perspective.
{"title":"S100 Protein Family in Lung Cancer: an Updated Narrative Review.","authors":"Ting Wang, Rui Liu","doi":"10.2147/CMAR.S508047","DOIUrl":"10.2147/CMAR.S508047","url":null,"abstract":"<p><p>The S100 protein family comprises 25 known members that modulate variously basic biological behaviors of cells by binding Ca<sup>2+</sup> and activating Ca<sup>2+</sup>-signaling pathways. As the primary cause of cancer-related death, lung cancer is closely associated with several S100 proteins, like S100A2, S100A4, S100A6, S100A8/9, etc. Notably, the functions and mechanisms of different S100 proteins vary in every sub-type of lung cancer. Overall speaking, the abnormal expression of S100 proteins is predominantly observed in lung adenocarcinoma, while their roles are limited in small-cell lung cancer. This review, which presents an update on our previously published review of S100 proteins in lung cancer (2021), aims to enable readers having a deeper understanding of the roles of different S100 proteins in three main sub-types of lung cancer, as well as to facilitate their future researches. It focuses on relevant studies examining the functions of S100 proteins in lung adenocarcinoma, squamous carcinoma, and small-cell lung cancer. This review was conducted based on this standard, and provides a comprehensive evaluation of the literature review on S100 proteins as well as enhances understanding of the relationship between S100 proteins and every sub-type of lung cancer from a new perspective.</p>","PeriodicalId":9479,"journal":{"name":"Cancer Management and Research","volume":"17 ","pages":"713-722"},"PeriodicalIF":2.5,"publicationDate":"2025-03-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11956737/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143751261","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-03-27eCollection Date: 2025-01-01DOI: 10.2147/CMAR.S499771
Si Hao Lu, Kui Lui, Yue Qian, Wei Ye Zhou, Ying Ying Mu, Wei Zhang
Background: Clear cell renal cell carcinoma (ccRCC) is a relatively frequently diagnosed form of urological cancer that is highly malignant and associated with high rates of patient mortality. At present, there are few effective options for treating advanced cases of ccRCC, emphasizing the need to establish novel biomarkers and targets suitable for therapeutic intervention. SET domain bifurcated histone lysine methyltransferase 2 (SETDB2) belongs to the Su(var)3-9 subfamily of methyltransferases and has been linked to various forms of cancer, but the role it plays in ccRCC remains to be fully established.
Methods: Data on SETDB2 expression were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Functional enrichment analyses were then used to probe the putative role that SETDB2 plays in the onset of ccRCC. The Gene Set Cancer Analysis (GSCA) platform and molecular docking analysis were utilized to investigate the relationship between gene expression and drug sensitivity. In the end, the core target and the active molecule were both given the green light for a molecular docking investigation. Functional assays and Western blotting performed with ccRCC cell lines were employed for the validation of the findings from these predictive analyses.
Results: SETDB2 downregulation was observed in ccRCC, and lower levels were found to linked with poor patient outcomes. Lower SETDB2 levels were associated with worse overall, progression-free, and disease-specific survival. In Functional enrichment analyses, SETDB2 was predicted to regulate key ccRCC development-associated pathways. SETDB2 levels were also significantly associated with cuproptosis induction in KIRC tissues, while in immune cell infiltration analyses, SETDB2 expression was linked with immune responses within the tumor microenvironment. Functional experiments conducted with ccRCC cell lines unveiled molecular mechanisms through which SETDB2 appears to be capable of inhibiting the development of ccRCC.
Conclusion: Together, these analyses highlight the utility of SETDB2 as a prognostic biomarker in ccRCC. The interactions and associated pathways detected through these analyses provide unique insight into the potential functions of SETDB2 in this cancer type, providing an evidence base for future studies.
{"title":"Prognostic Role of SETDB2 in Clear Cell Renal Cell Carcinoma: Linking Immune Infiltration, Cuproptosis, and Tumor Suppression.","authors":"Si Hao Lu, Kui Lui, Yue Qian, Wei Ye Zhou, Ying Ying Mu, Wei Zhang","doi":"10.2147/CMAR.S499771","DOIUrl":"10.2147/CMAR.S499771","url":null,"abstract":"<p><strong>Background: </strong>Clear cell renal cell carcinoma (ccRCC) is a relatively frequently diagnosed form of urological cancer that is highly malignant and associated with high rates of patient mortality. At present, there are few effective options for treating advanced cases of ccRCC, emphasizing the need to establish novel biomarkers and targets suitable for therapeutic intervention. SET domain bifurcated histone lysine methyltransferase 2 (SETDB2) belongs to the Su(var)3-9 subfamily of methyltransferases and has been linked to various forms of cancer, but the role it plays in ccRCC remains to be fully established.</p><p><strong>Methods: </strong>Data on SETDB2 expression were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Functional enrichment analyses were then used to probe the putative role that SETDB2 plays in the onset of ccRCC. The Gene Set Cancer Analysis (GSCA) platform and molecular docking analysis were utilized to investigate the relationship between gene expression and drug sensitivity. In the end, the core target and the active molecule were both given the green light for a molecular docking investigation. Functional assays and Western blotting performed with ccRCC cell lines were employed for the validation of the findings from these predictive analyses.</p><p><strong>Results: </strong>SETDB2 downregulation was observed in ccRCC, and lower levels were found to linked with poor patient outcomes. Lower SETDB2 levels were associated with worse overall, progression-free, and disease-specific survival. In Functional enrichment analyses, SETDB2 was predicted to regulate key ccRCC development-associated pathways. SETDB2 levels were also significantly associated with cuproptosis induction in KIRC tissues, while in immune cell infiltration analyses, SETDB2 expression was linked with immune responses within the tumor microenvironment. Functional experiments conducted with ccRCC cell lines unveiled molecular mechanisms through which SETDB2 appears to be capable of inhibiting the development of ccRCC.</p><p><strong>Conclusion: </strong>Together, these analyses highlight the utility of SETDB2 as a prognostic biomarker in ccRCC. The interactions and associated pathways detected through these analyses provide unique insight into the potential functions of SETDB2 in this cancer type, providing an evidence base for future studies.</p>","PeriodicalId":9479,"journal":{"name":"Cancer Management and Research","volume":"17 ","pages":"675-692"},"PeriodicalIF":2.5,"publicationDate":"2025-03-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11956738/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143751260","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Although adenosylhomocysteinase (AHCY) is crucial to the oncogenesis and growth of some cancers, it is unknown how this affects bladder urothelial carcinoma (BLCA). Investigating the variations in AHCY expression in BLCA and examining the relationship between AHCY expression and BLCA patient prognosis were the goals of this investigation.
Methods: By leveraging The Cancer Genome Atlas (TCGA) database, we undertook a meticulous examination of AHCY expression levels, juxtaposing them between BLCA and normal tissues. Subsequently, Kaplan-Meier analysis and COX regression and nomogram was used to assess the effect of AHCY on the survival of BLCA patients. We further elaborated on the possible enriched pathways of AHCY and its immune relevance. In addition, we employed si-RNA technology to downregulate the AHCY gene expression and subsequently utilized quantitative real-time PCR (qRT-PCR), CCK-8, cell scratch assays, and Transwell migration assays to validate the pivotal role of AHCY in BLCA.
Results: The expression of AHCY was associated with various types of malignancies (including BCLA). In BLCA cancer tissues, there was an observed upregulation of AHCY expression in comparison to paracancerous tissues. Increased expression of AHCY was linked to decreased overall survival (OS), clinical stage, N stage, and T stage in individuals with BLCA. The functional enrichment of AHCY related genes mainly involves biological processes such as rRNA metabolic processes, proteasome activity, and cell cycle regulation, etc. Furthermore, AHCY showed significant associations with m6A related genes and infiltration of immune cells (Especially for Th2 cells and T-gd lymphocytes). In vitro functional experiments substantiated that the inhibition of AHCY effectively suppresses the growth, migration, and invasion of bladder cancer cells.
Conclusion: This study provides novel insights into the role of AHCY in BLCA, which holds significant potential to contribute towards advancing the diagnosis and treatment of BLCA in the future.
{"title":"The Role of AHCY Expression in Bladder Urothelial Carcinoma: A Bioinformatics and Experimental Analysis.","authors":"Shaorui Niu, Xiaozhe Zheng, Yuyang Yao, Yue Dong, Yupan Hu, Zhiyang Xiao, Jiaxue Yang, Chengli Jiang, Xin Zou, Zihao Zou, Pang Yang","doi":"10.2147/CMAR.S491044","DOIUrl":"10.2147/CMAR.S491044","url":null,"abstract":"<p><strong>Background: </strong>Although adenosylhomocysteinase (AHCY) is crucial to the oncogenesis and growth of some cancers, it is unknown how this affects bladder urothelial carcinoma (BLCA). Investigating the variations in AHCY expression in BLCA and examining the relationship between AHCY expression and BLCA patient prognosis were the goals of this investigation.</p><p><strong>Methods: </strong>By leveraging The Cancer Genome Atlas (TCGA) database, we undertook a meticulous examination of AHCY expression levels, juxtaposing them between BLCA and normal tissues. Subsequently, Kaplan-Meier analysis and COX regression and nomogram was used to assess the effect of AHCY on the survival of BLCA patients. We further elaborated on the possible enriched pathways of AHCY and its immune relevance. In addition, we employed si-RNA technology to downregulate the AHCY gene expression and subsequently utilized quantitative real-time PCR (qRT-PCR), CCK-8, cell scratch assays, and Transwell migration assays to validate the pivotal role of AHCY in BLCA.</p><p><strong>Results: </strong>The expression of AHCY was associated with various types of malignancies (including BCLA). In BLCA cancer tissues, there was an observed upregulation of AHCY expression in comparison to paracancerous tissues. Increased expression of AHCY was linked to decreased overall survival (OS), clinical stage, N stage, and T stage in individuals with BLCA. The functional enrichment of AHCY related genes mainly involves biological processes such as rRNA metabolic processes, proteasome activity, and cell cycle regulation, etc. Furthermore, AHCY showed significant associations with m6A related genes and infiltration of immune cells (Especially for Th2 cells and T-gd lymphocytes). In vitro functional experiments substantiated that the inhibition of AHCY effectively suppresses the growth, migration, and invasion of bladder cancer cells.</p><p><strong>Conclusion: </strong>This study provides novel insights into the role of AHCY in BLCA, which holds significant potential to contribute towards advancing the diagnosis and treatment of BLCA in the future.</p>","PeriodicalId":9479,"journal":{"name":"Cancer Management and Research","volume":"17 ","pages":"661-673"},"PeriodicalIF":2.5,"publicationDate":"2025-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11937648/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143718127","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-03-20eCollection Date: 2025-01-01DOI: 10.2147/CMAR.S505730
Xiaosong Sun, Zhengchao Wei, Yiqiang Luo, Ming Wang
Background: Thyroid cancer exhibits the highest cervical lymph node metastasis rate (20-50%) among head and neck malignancies, with occult metastasis occurring in 30-80% of papillary carcinoma cases. However, conventional single-modality imaging faces certain challenges: MRI has limited sensitivity for detecting micro-metastases (<2mm), while Doppler ultrasound may overlook metastases in isoechoic lymph nodes. Therefore, it is crucial to evaluate the diagnostic value of combining MRI and CDUS. This study aims to retrospectively analyze the diagnostic value of combining MRI and CDUS blood flow parameters in detecting cervical lymph node metastasis in thyroid cancer and to compare the diagnostic performance with MRI or CDUS alone.
Objective: To analyze the evaluation value of combining MRI and color Doppler ultrasound (CDUS) blood flow parameters in detecting cervical lymph node metastasis of thyroid cancer, particularly for occult metastases.
Methods: A retrospective analysis was conducted on 263 thyroid cancer patients (June 2022-June 2024). Diagnostic consistency between MRI, CDUS parameters (resistive index, pulsatility index, vascular patterns) and pathology were compared. Multimodal evaluation criteria were established: (1) MRI positive signs (lymph node diameter >8mm, cystic change, enhancement heterogeneity) (2) CDUS thresholds (RI≥0.75, PI≥1.25 with chaotic vascularity).
Results: Among 263 patients, 98 had pathologically confirmed metastases. CDUS showed higher consistency with pathology (Kappa=0.783) than MRI (Kappa=0.645). Combined modality achieved 94.9% sensitivity vs 86.7% (CDUS) and 78.6% (MRI), with accuracy improving from 82.1%/75.3% to 89.4% (P<0.05). Notably, 12/22 occult metastases (≤3mm) were only detected by combined approach.
Conclusion: The synergistic combination leverages MRI's structural characterization and CDUS's hemodynamic sensitivity, effectively overcoming single-modality limitations in detecting micro-metastases. This dual-assessment protocol addresses thyroid cancer's propensity for early lymphatic spread, providing critical preoperative staging guidance.
{"title":"Exploration of the Evaluation Value of Combined Magnetic Resonance Imaging and Ultrasound Blood Flow Parameters in Cervical Lymph Node Metastasis of Thyroid Cancer.","authors":"Xiaosong Sun, Zhengchao Wei, Yiqiang Luo, Ming Wang","doi":"10.2147/CMAR.S505730","DOIUrl":"10.2147/CMAR.S505730","url":null,"abstract":"<p><strong>Background: </strong>Thyroid cancer exhibits the highest cervical lymph node metastasis rate (20-50%) among head and neck malignancies, with occult metastasis occurring in 30-80% of papillary carcinoma cases. However, conventional single-modality imaging faces certain challenges: MRI has limited sensitivity for detecting micro-metastases (<2mm), while Doppler ultrasound may overlook metastases in isoechoic lymph nodes. Therefore, it is crucial to evaluate the diagnostic value of combining MRI and CDUS. This study aims to retrospectively analyze the diagnostic value of combining MRI and CDUS blood flow parameters in detecting cervical lymph node metastasis in thyroid cancer and to compare the diagnostic performance with MRI or CDUS alone.</p><p><strong>Objective: </strong>To analyze the evaluation value of combining MRI and color Doppler ultrasound (CDUS) blood flow parameters in detecting cervical lymph node metastasis of thyroid cancer, particularly for occult metastases.</p><p><strong>Methods: </strong>A retrospective analysis was conducted on 263 thyroid cancer patients (June 2022-June 2024). Diagnostic consistency between MRI, CDUS parameters (resistive index, pulsatility index, vascular patterns) and pathology were compared. Multimodal evaluation criteria were established: (1) MRI positive signs (lymph node diameter >8mm, cystic change, enhancement heterogeneity) (2) CDUS thresholds (RI≥0.75, PI≥1.25 with chaotic vascularity).</p><p><strong>Results: </strong>Among 263 patients, 98 had pathologically confirmed metastases. CDUS showed higher consistency with pathology (Kappa=0.783) than MRI (Kappa=0.645). Combined modality achieved 94.9% sensitivity vs 86.7% (CDUS) and 78.6% (MRI), with accuracy improving from 82.1%/75.3% to 89.4% (P<0.05). Notably, 12/22 occult metastases (≤3mm) were only detected by combined approach.</p><p><strong>Conclusion: </strong>The synergistic combination leverages MRI's structural characterization and CDUS's hemodynamic sensitivity, effectively overcoming single-modality limitations in detecting micro-metastases. This dual-assessment protocol addresses thyroid cancer's propensity for early lymphatic spread, providing critical preoperative staging guidance.</p>","PeriodicalId":9479,"journal":{"name":"Cancer Management and Research","volume":"17 ","pages":"651-659"},"PeriodicalIF":2.5,"publicationDate":"2025-03-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11932127/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143699794","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-03-19eCollection Date: 2025-01-01DOI: 10.2147/CMAR.S503932
Li-Kuan Tan, Jiaxing Liu, Cheng-Zhi Ma, Shaolong Huang, Feng-Hui He, Yang Long, Zhi-Sheng Zheng, Jia-Liang Liang, Nan Xu, Guanghui Wang, Yu-Fei Liu
Triple-negative breast cancer (TNBC) is characterized by aggressive behavior, high metastatic potential, and frequent relapses, presenting significant treatment challenges. Ferroptosis, a unique form of programmed cell death marked by iron-dependent lipid peroxidation, has emerged as a crucial factor in cancer biology. Recent studies indicate that TNBC cells possess a distinct metabolic profile linked to iron and glutathione, which may render them more susceptible to ferroptosis than other breast cancer subtypes. Moreover, ferroptosis plays a role in the interactions between immune cells and tumor cells, suggesting its potential to modulate the tumor microenvironment and influence the immune response against TNBC.Evidence reveals that ferroptosis not only affects TNBC cell viability but also alters the tumor microenvironment by promoting the release of damage-associated molecular patterns (DAMPs), which can recruit immune cells to the tumor site. Specific ferroptosis-related genes and biomarkers, such as ACSL4 and GPX4, demonstrate altered expression patterns in TNBC tissues, offering promising avenues for diagnostic and prognostic applications. Furthermore, in preclinical models, the induction of ferroptosis has been shown to enhance the efficacy of existing therapies, indicating a synergistic effect that could be harnessed for therapeutic benefit. The compelling link between ferroptosis and TNBC underscores its potential as a novel therapeutic target. Future research should focus on developing strategies that exploit ferroptosis in conjunction with traditional therapies, including the identification of natural compounds and efficacious ferroptosis inducers for personalized treatment regimens. This review elucidates the multifaceted implications of ferroptosis in TNBC, providing valuable insights for improving both diagnosis and treatment of this formidable breast cancer subtype.
{"title":"Iron-Dependent Cell Death: Exploring Ferroptosis as a Unique Target in Triple-Negative Breast Cancer Management.","authors":"Li-Kuan Tan, Jiaxing Liu, Cheng-Zhi Ma, Shaolong Huang, Feng-Hui He, Yang Long, Zhi-Sheng Zheng, Jia-Liang Liang, Nan Xu, Guanghui Wang, Yu-Fei Liu","doi":"10.2147/CMAR.S503932","DOIUrl":"10.2147/CMAR.S503932","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC) is characterized by aggressive behavior, high metastatic potential, and frequent relapses, presenting significant treatment challenges. Ferroptosis, a unique form of programmed cell death marked by iron-dependent lipid peroxidation, has emerged as a crucial factor in cancer biology. Recent studies indicate that TNBC cells possess a distinct metabolic profile linked to iron and glutathione, which may render them more susceptible to ferroptosis than other breast cancer subtypes. Moreover, ferroptosis plays a role in the interactions between immune cells and tumor cells, suggesting its potential to modulate the tumor microenvironment and influence the immune response against TNBC.Evidence reveals that ferroptosis not only affects TNBC cell viability but also alters the tumor microenvironment by promoting the release of damage-associated molecular patterns (DAMPs), which can recruit immune cells to the tumor site. Specific ferroptosis-related genes and biomarkers, such as ACSL4 and GPX4, demonstrate altered expression patterns in TNBC tissues, offering promising avenues for diagnostic and prognostic applications. Furthermore, in preclinical models, the induction of ferroptosis has been shown to enhance the efficacy of existing therapies, indicating a synergistic effect that could be harnessed for therapeutic benefit. The compelling link between ferroptosis and TNBC underscores its potential as a novel therapeutic target. Future research should focus on developing strategies that exploit ferroptosis in conjunction with traditional therapies, including the identification of natural compounds and efficacious ferroptosis inducers for personalized treatment regimens. This review elucidates the multifaceted implications of ferroptosis in TNBC, providing valuable insights for improving both diagnosis and treatment of this formidable breast cancer subtype.</p>","PeriodicalId":9479,"journal":{"name":"Cancer Management and Research","volume":"17 ","pages":"625-637"},"PeriodicalIF":2.5,"publicationDate":"2025-03-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11930262/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143691313","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-03-19eCollection Date: 2025-01-01DOI: 10.2147/CMAR.S503109
Zhan-Yi Li, Yuan Teng, Chen-Meng Long, Ren-Bin Liu, Yu Liu
Purpose: Special populations are not enrolled in randomized clinical trials, and their safety and efficacy of anticancer therapy are not well described. We aimed to assess the safety and efficacy of anticancer therapy in breast cancer (BC) patients with cirrhosis.
Patients and methods: We performed a retrospective case-control study (1:5) to assess the adverse events (AEs) morbidity and mortality of anticancer therapy in BC patients with cirrhosis based on a review of patients' medical records.
Results: We included 26 BC patients with cirrhosis and 130 matched BC patients without cirrhosis. Postoperative morbidity was higher in the group with cirrhosis (26.9% vs 6.9%, P = 0.007) when postoperative mortality was not significance (3.8% vs 0%, P = 0.167). Liver toxicity (73.1% vs 26.9%, P < 0.001) was more frequent in the group with cirrhosis. The incidence of disruption and mortality during chemotherapy was higher in the group with cirrhosis (46.2% vs 3.1%, P < 0.001 and 15.4% vs 0%, P = 0.001, respectively). The 2-year recurrence rate and 2-year metastasis rate were higher in the group with cirrhosis (19.0% vs 3.8%, P = 0.022 and 23.8% vs 6.9%, P = 0.028). Cirrhosis was the risk factor for liver metastasis (OR: 17.326, 95% CI: 2.164-138.707, P=0.007).
Conclusion: It is safe for BC patients with compensated cirrhosis to accept surgery. But they are vulnerable to AEs, disruptions and death during chemotherapy and have poor prognosis. Multidisciplinary cooperation before therapy and closely monitoring AEs during therapy are critical. Attention should be given to optimize the prognosis of special BC patients.
{"title":"The Safety and Efficacy of Anticancer Therapy in Breast Cancer Patients with Liver Cirrhosis.","authors":"Zhan-Yi Li, Yuan Teng, Chen-Meng Long, Ren-Bin Liu, Yu Liu","doi":"10.2147/CMAR.S503109","DOIUrl":"10.2147/CMAR.S503109","url":null,"abstract":"<p><strong>Purpose: </strong>Special populations are not enrolled in randomized clinical trials, and their safety and efficacy of anticancer therapy are not well described. We aimed to assess the safety and efficacy of anticancer therapy in breast cancer (BC) patients with cirrhosis.</p><p><strong>Patients and methods: </strong>We performed a retrospective case-control study (1:5) to assess the adverse events (AEs) morbidity and mortality of anticancer therapy in BC patients with cirrhosis based on a review of patients' medical records.</p><p><strong>Results: </strong>We included 26 BC patients with cirrhosis and 130 matched BC patients without cirrhosis. Postoperative morbidity was higher in the group with cirrhosis (26.9% vs 6.9%, P = 0.007) when postoperative mortality was not significance (3.8% vs 0%, P = 0.167). Liver toxicity (73.1% vs 26.9%, P < 0.001) was more frequent in the group with cirrhosis. The incidence of disruption and mortality during chemotherapy was higher in the group with cirrhosis (46.2% vs 3.1%, P < 0.001 and 15.4% vs 0%, P = 0.001, respectively). The 2-year recurrence rate and 2-year metastasis rate were higher in the group with cirrhosis (19.0% vs 3.8%, P = 0.022 and 23.8% vs 6.9%, P = 0.028). Cirrhosis was the risk factor for liver metastasis (OR: 17.326, 95% CI: 2.164-138.707, P=0.007).</p><p><strong>Conclusion: </strong>It is safe for BC patients with compensated cirrhosis to accept surgery. But they are vulnerable to AEs, disruptions and death during chemotherapy and have poor prognosis. Multidisciplinary cooperation before therapy and closely monitoring AEs during therapy are critical. Attention should be given to optimize the prognosis of special BC patients.</p>","PeriodicalId":9479,"journal":{"name":"Cancer Management and Research","volume":"17 ","pages":"639-650"},"PeriodicalIF":2.5,"publicationDate":"2025-03-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11930243/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143691332","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-03-17eCollection Date: 2025-01-01DOI: 10.2147/CMAR.S502155
Han Liu, Ruinian Zheng, Zhaowei Zhuang, Liwen Xue, Minggui Chen, Yuluo Wu, Yan Zeng
Purpose: Breast cancer, a predominant contributor to cancer-related mortality worldwide, is increasingly managed through the application of neoadjuvant chemotherapy (NAC). Analyzing the dynamic changes in peripheral blood lymphocyte subsets, granzyme B and perforin are crucial for investigating their roles in tumorigenesis, development and treatment; this study aimed to use these analyses to diagnose malignant breast tumor, assess the anti-tumor immunity and predict chemotherapy efficacy in breast cancer patients.
Patients and methods: To address this objective, a total of 582 peripheral blood samples were collected from healthy controls (n=47), benign breast disease patients (n=401) and breast cancer patients (n=134). Lymphocyte subsets, along with granzyme B and perforin expression, were assessed using flow cytometry. Changes before and after NAC were also monitored.
Results: Breast cancer patients exhibited reduced proportions and absolute counts of CD3+ and CD8+ T cells, increased NK cell percentage and CD4+/CD8+ ratio, and higher levels of granzyme B and perforin in CD3+, CD8+ T cells and NK cells. Post-NAC, the percentages of CD3+, CD4+, CD8+ T cells and NK cells increased, along with a higher CD4+/CD8+ ratio, while B cell percentages decreased compared to pre-NAC. Furthermore, the effective group showed higher percentages of CD3+, CD8+ T cells and lower percentages of B cells than the ineffective group post-NAC. Incidentally, Granzyme B and perforin expression in CD3+ and CD8+ T cells was elevated following postoperative chemotherapy.
Conclusion: These findings indicated that peripheral blood lymphocyte subsets, along with granzyme B and perforin levels, could serve as potential biomarkers for differentiating benign from malignant breast tumors, assessing anti-tumor immunity and predicting chemotherapy efficacy.
{"title":"Diagnostic Efficacy and Clinical Significance of Lymphocyte Subsets, Granzyme B and Perforin in the Peripheral Blood of Patients with Invasive Breast Cancer Following Neoadjuvant Chemotherapy.","authors":"Han Liu, Ruinian Zheng, Zhaowei Zhuang, Liwen Xue, Minggui Chen, Yuluo Wu, Yan Zeng","doi":"10.2147/CMAR.S502155","DOIUrl":"10.2147/CMAR.S502155","url":null,"abstract":"<p><strong>Purpose: </strong>Breast cancer, a predominant contributor to cancer-related mortality worldwide, is increasingly managed through the application of neoadjuvant chemotherapy (NAC). Analyzing the dynamic changes in peripheral blood lymphocyte subsets, granzyme B and perforin are crucial for investigating their roles in tumorigenesis, development and treatment; this study aimed to use these analyses to diagnose malignant breast tumor, assess the anti-tumor immunity and predict chemotherapy efficacy in breast cancer patients.</p><p><strong>Patients and methods: </strong>To address this objective, a total of 582 peripheral blood samples were collected from healthy controls (n=47), benign breast disease patients (n=401) and breast cancer patients (n=134). Lymphocyte subsets, along with granzyme B and perforin expression, were assessed using flow cytometry. Changes before and after NAC were also monitored.</p><p><strong>Results: </strong>Breast cancer patients exhibited reduced proportions and absolute counts of CD3<sup>+</sup> and CD8<sup>+</sup> T cells, increased NK cell percentage and CD4<sup>+</sup>/CD8<sup>+</sup> ratio, and higher levels of granzyme B and perforin in CD3<sup>+</sup>, CD8<sup>+</sup> T cells and NK cells. Post-NAC, the percentages of CD3<sup>+</sup>, CD4<sup>+</sup>, CD8<sup>+</sup> T cells and NK cells increased, along with a higher CD4<sup>+</sup>/CD8<sup>+</sup> ratio, while B cell percentages decreased compared to pre-NAC. Furthermore, the effective group showed higher percentages of CD3<sup>+</sup>, CD8<sup>+</sup> T cells and lower percentages of B cells than the ineffective group post-NAC. Incidentally, Granzyme B and perforin expression in CD3<sup>+</sup> and CD8<sup>+</sup> T cells was elevated following postoperative chemotherapy.</p><p><strong>Conclusion: </strong>These findings indicated that peripheral blood lymphocyte subsets, along with granzyme B and perforin levels, could serve as potential biomarkers for differentiating benign from malignant breast tumors, assessing anti-tumor immunity and predicting chemotherapy efficacy.</p>","PeriodicalId":9479,"journal":{"name":"Cancer Management and Research","volume":"17 ","pages":"589-602"},"PeriodicalIF":2.5,"publicationDate":"2025-03-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11928756/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143691311","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-03-17eCollection Date: 2025-01-01DOI: 10.2147/CMAR.S510811
Fan Xu, Xiaoli Zhong, Qiao Li, Xi Luo, Na Wang, Jing Wang, Shaoju Xie, Jiquan Zhang
Background: Improving the quality of life (QOL) of lung cancer patients undergoing chemotherapy is an indispensable part of cancer treatment, as it not only pertains to their physical health but also to their psychological and social well-being. Previous research has primarily focused on investigating health-related quality of life, while studies specifically addressing the QOL of lung cancer patients remain underrepresented and under researched.
Purpose: The study aims to investigate the current status of QOL among lung cancer patients and identify the predictive factors associated with QOL.
Patients and methods: From January 2024 to June 2024, lung cancer patients undergoing chemotherapy will be recruited from the outpatient clinics or wards of a tertiary A-level hospital in Deyang City as research subjects. They will be surveyed using the general information questionnaire, the Self-rating Depression Scale (SDS), the Multidimensional Scale of Perceived Social Support (MSPSS), and the Functional Assessment of Cancer Therapy-Lung (FACT-L) scale. Multiple linear regression analysis will be employed to determine the variables associated with QOL.
Results: A total of 390 lung cancer patients undergoing chemotherapy were recruited for this study, with a male predominance accounting for 72.31%. The mean age was (59.11±11.37) years. The overall QOL score was (66.43±23.67). Age, family monthly income per capita, cancer clinical stage, depression, and perceived social support (PSS) were identified as independent factors influencing the QOL of lung cancer patients, accounting for 19.4% of the total variance.
Conclusion: There is still considerable room for improvement in the overall QOL of lung cancer patients undergoing chemotherapy. Based on the analysis of influencing factors, targeted and personalized intervention measures should be implemented to enhance the QOL for these patients.
{"title":"Quality of Life Status and Its Influencing Factors Among Lung Cancer Chemotherapy Patients in China: A Cross-Sectional Study.","authors":"Fan Xu, Xiaoli Zhong, Qiao Li, Xi Luo, Na Wang, Jing Wang, Shaoju Xie, Jiquan Zhang","doi":"10.2147/CMAR.S510811","DOIUrl":"10.2147/CMAR.S510811","url":null,"abstract":"<p><strong>Background: </strong>Improving the quality of life (QOL) of lung cancer patients undergoing chemotherapy is an indispensable part of cancer treatment, as it not only pertains to their physical health but also to their psychological and social well-being. Previous research has primarily focused on investigating health-related quality of life, while studies specifically addressing the QOL of lung cancer patients remain underrepresented and under researched.</p><p><strong>Purpose: </strong>The study aims to investigate the current status of QOL among lung cancer patients and identify the predictive factors associated with QOL.</p><p><strong>Patients and methods: </strong>From January 2024 to June 2024, lung cancer patients undergoing chemotherapy will be recruited from the outpatient clinics or wards of a tertiary A-level hospital in Deyang City as research subjects. They will be surveyed using the general information questionnaire, the Self-rating Depression Scale (SDS), the Multidimensional Scale of Perceived Social Support (MSPSS), and the Functional Assessment of Cancer Therapy-Lung (FACT-L) scale. Multiple linear regression analysis will be employed to determine the variables associated with QOL.</p><p><strong>Results: </strong>A total of 390 lung cancer patients undergoing chemotherapy were recruited for this study, with a male predominance accounting for 72.31%. The mean age was (59.11±11.37) years. The overall QOL score was (66.43±23.67). Age, family monthly income per capita, cancer clinical stage, depression, and perceived social support (PSS) were identified as independent factors influencing the QOL of lung cancer patients, accounting for 19.4% of the total variance.</p><p><strong>Conclusion: </strong>There is still considerable room for improvement in the overall QOL of lung cancer patients undergoing chemotherapy. Based on the analysis of influencing factors, targeted and personalized intervention measures should be implemented to enhance the QOL for these patients.</p>","PeriodicalId":9479,"journal":{"name":"Cancer Management and Research","volume":"17 ","pages":"603-615"},"PeriodicalIF":2.5,"publicationDate":"2025-03-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11927569/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143691316","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-03-17eCollection Date: 2025-01-01DOI: 10.2147/CMAR.S494121
Tong Wei, Hongmin Quan, Rengui Wang, Xiaoli Sun
Background: Myoepithelial carcinoma is rare, and myoepithelial carcinoma occurring outside the head and neck is even rarer. We reported one case of retroperitoneal myoepithelial carcinoma.
Case summary: A 63-year-old woman who underwent computed tomography (CT) for progressive abdominal distension revealed a left retroperitoneal mass and subsequently underwent surgical treatment where the mass was completely removed with a postoperative diagnosis of retroperitoneal myoepithelial carcinoma. A follow-up CT review 40 days after surgery revealed a recurrence of the mass. After 8 months of chemotherapy and targeted immunotherapy, a follow-up review of the CT images revealed a gradual reduction in the mass. Four months after the cessation of chemotherapy and targeted drug combined immunotherapy, a follow-up review via CT revealed another recurrence and enlargement of the mass.
Conclusion: CT of retroperitoneal myoepithelial carcinoma revealed a massive cystic solid mass in the abdominal cavity and retroperitoneum. The solid region of the mass was significantly enhanced and the cystic region was without enhancement on enhanced CT; the mass involved the adjacent duodenum, partial jejunum, and left renal vein. PET‒CT imaging revealed hypermetabolism in the solid region of the mass and no hypermetabolism in the cystic region.
{"title":"Retroperitoneal Myoepithelial Carcinoma: A Case Report and Literature Review.","authors":"Tong Wei, Hongmin Quan, Rengui Wang, Xiaoli Sun","doi":"10.2147/CMAR.S494121","DOIUrl":"10.2147/CMAR.S494121","url":null,"abstract":"<p><strong>Background: </strong>Myoepithelial carcinoma is rare, and myoepithelial carcinoma occurring outside the head and neck is even rarer. We reported one case of retroperitoneal myoepithelial carcinoma.</p><p><strong>Case summary: </strong>A 63-year-old woman who underwent computed tomography (CT) for progressive abdominal distension revealed a left retroperitoneal mass and subsequently underwent surgical treatment where the mass was completely removed with a postoperative diagnosis of retroperitoneal myoepithelial carcinoma. A follow-up CT review 40 days after surgery revealed a recurrence of the mass. After 8 months of chemotherapy and targeted immunotherapy, a follow-up review of the CT images revealed a gradual reduction in the mass. Four months after the cessation of chemotherapy and targeted drug combined immunotherapy, a follow-up review via CT revealed another recurrence and enlargement of the mass.</p><p><strong>Conclusion: </strong>CT of retroperitoneal myoepithelial carcinoma revealed a massive cystic solid mass in the abdominal cavity and retroperitoneum. The solid region of the mass was significantly enhanced and the cystic region was without enhancement on enhanced CT; the mass involved the adjacent duodenum, partial jejunum, and left renal vein. PET‒CT imaging revealed hypermetabolism in the solid region of the mass and no hypermetabolism in the cystic region.</p>","PeriodicalId":9479,"journal":{"name":"Cancer Management and Research","volume":"17 ","pages":"617-624"},"PeriodicalIF":2.5,"publicationDate":"2025-03-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11928755/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143691330","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}