Justyna Pilch, Jakub Mizera, Maciej Tota, Piotr Donizy
Uveal melanoma (UM), recognized as the most prevalent primary intraocular malignancy in adults, is primarily driven by mutations in the GNAQ and GNA11 genes. These genetic alterations are also implicated in other conditions, which exhibit distinct morphological characteristics. In this article, we investigate the role of GNAQ and GNA11 mutations across varied disorders (e.g., UM, skin blue nevi, and hemangiomas), emphasizing the shared pathogenic mechanisms that connect them despite their differing clinical manifestations. By investigating the molecular pathways affected by these mutations, we provide insights into the potential for targeted therapies that could address not only UM but also other disorders associated with GNAQ/GNA11 mutations. Moreover, we discuss the role of SOX10-positive perivascular cells that may be implicated in the complex pathophysiology of GNAQ/GNA11-related entities. Understanding the common molecular foundation of these conditions opens new ways for research and treatment opportunities, potentially leading to more effective, personalized therapeutic strategies.
{"title":"GNAQ/GNA11-Related Benign and Malignant Entities-A Common Histoembriologic Origin or a Tissue-Dependent Coincidence.","authors":"Justyna Pilch, Jakub Mizera, Maciej Tota, Piotr Donizy","doi":"10.3390/cancers16213672","DOIUrl":"10.3390/cancers16213672","url":null,"abstract":"<p><p>Uveal melanoma (UM), recognized as the most prevalent primary intraocular malignancy in adults, is primarily driven by mutations in the <i>GNAQ</i> and <i>GNA11</i> genes. These genetic alterations are also implicated in other conditions, which exhibit distinct morphological characteristics. In this article, we investigate the role of <i>GNAQ</i> and <i>GNA11</i> mutations across varied disorders (e.g., UM, skin blue nevi, and hemangiomas), emphasizing the shared pathogenic mechanisms that connect them despite their differing clinical manifestations. By investigating the molecular pathways affected by these mutations, we provide insights into the potential for targeted therapies that could address not only UM but also other disorders associated with <i>GNAQ</i>/<i>GNA11</i> mutations. Moreover, we discuss the role of SOX10-positive perivascular cells that may be implicated in the complex pathophysiology of GNAQ/GNA11-related entities. Understanding the common molecular foundation of these conditions opens new ways for research and treatment opportunities, potentially leading to more effective, personalized therapeutic strategies.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"16 21","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11544895/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142635912","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Miguel Angel Jimenez Rios, Anna Scavuzzo, Nancy Reynoso Noverón, Caleb García Arango, Ivan Calvo Vazquez, Alonso Hurtado Vázquez, Oscar Gerardo Arrieta Rodriguez, Miguel Angel Jimenez Davila, Maria Chiara Sighinolfi, Bernardo Rocco
Introduction: Epidemiological data are crucial for adopting primary and secondary prevention strategies and to develop screening protocols against prostate cancer (PCa). Despite the comprehensive characterization of PCa across White and Black men, there is a lack of data from the Mexican population. This manuscript presents data from the Can.Prost registry that captures PCa trends over the past two decades in Mexico City; furthermore, we aimed to compare clinical differences and oncological outcomes before and after the promotion of early detection actions through a campaign against PCa that occurred in 2014.
Methods: A retrospective observational study on newly diagnosed Mexican PCa patients was carried out at the Instituto Nacional de Cancerología (INCan) in Mexico City. During 2014 and 2015, a project for the early diagnosis of PCa ("OPUS program") was launched in the aforementioned tertiary hospital. Starting at the age of 45 years, all men were invited for a PSA measurement and a specialist urologist consultation. All individuals with clinical or biochemical suspicion of PCa (PSA > 4 ng/mL), in the context of age and prostate volume, underwent ultrasound-guided transrectal prostate biopsy. Then, patients with pathologically confirmed prostate cancer were stratified according to the year of diagnosis: Group A accounted for those diagnosed between 2000 and 2014 and Group B for those patients diagnosed in the timeframe of 2015-2021. Comparisons of PCa characteristics, treatment modalities and oncologic outcomes between Group A and B were performed.
Results: Overall, we collected data from 2759 PCa patients from 2000 to 2021. The median PSA at baseline was 32 ng/mL, and 25% had a family history of PCa. Overall, 25.8% were asymptomatic and 46% had a non-metastatic presentation. After the OPUS campaign, PSA at diagnosis was significantly lower across all age groups. The incidence of PCa diagnosis in asymptomatic men was higher (31.4% vs. 19.9%) and a higher proportion of men were diagnosed with organ-confined, palpable disease (46% vs. 28%) (p < 0.001). The rate of patients eligible for active/radical treatment was higher after the OPUS campaign (patients who received surgery increased from 12.78% to 32.41%; patients who underwent radiation increased from 28.38% to 49.61%). The proportion of patients diagnosed with non-clinically significant disease was negligible and remained stable across time.
Conclusions: PCa in Mexican patients displays aggressive features at diagnosis, whereas the rate of non-significant disease is negligible. The introduction of early detection strategies may lead to lower symptomatic and metastatic PCa and higher opportunities for radical treatment. This emphasizes the need for public awareness and for adjustment of screening strategies to the peculiarities of the Mexican population.
{"title":"Lethal Prostate Cancer in Mexico: Data from the Can.Prost Mexican Registry and a Project for Early Detection.","authors":"Miguel Angel Jimenez Rios, Anna Scavuzzo, Nancy Reynoso Noverón, Caleb García Arango, Ivan Calvo Vazquez, Alonso Hurtado Vázquez, Oscar Gerardo Arrieta Rodriguez, Miguel Angel Jimenez Davila, Maria Chiara Sighinolfi, Bernardo Rocco","doi":"10.3390/cancers16213675","DOIUrl":"10.3390/cancers16213675","url":null,"abstract":"<p><strong>Introduction: </strong>Epidemiological data are crucial for adopting primary and secondary prevention strategies and to develop screening protocols against prostate cancer (PCa). Despite the comprehensive characterization of PCa across White and Black men, there is a lack of data from the Mexican population. This manuscript presents data from the Can.Prost registry that captures PCa trends over the past two decades in Mexico City; furthermore, we aimed to compare clinical differences and oncological outcomes before and after the promotion of early detection actions through a campaign against PCa that occurred in 2014.</p><p><strong>Methods: </strong>A retrospective observational study on newly diagnosed Mexican PCa patients was carried out at the Instituto Nacional de Cancerología (INCan) in Mexico City. During 2014 and 2015, a project for the early diagnosis of PCa (\"OPUS program\") was launched in the aforementioned tertiary hospital. Starting at the age of 45 years, all men were invited for a PSA measurement and a specialist urologist consultation. All individuals with clinical or biochemical suspicion of PCa (PSA > 4 ng/mL), in the context of age and prostate volume, underwent ultrasound-guided transrectal prostate biopsy. Then, patients with pathologically confirmed prostate cancer were stratified according to the year of diagnosis: Group A accounted for those diagnosed between 2000 and 2014 and Group B for those patients diagnosed in the timeframe of 2015-2021. Comparisons of PCa characteristics, treatment modalities and oncologic outcomes between Group A and B were performed.</p><p><strong>Results: </strong>Overall, we collected data from 2759 PCa patients from 2000 to 2021. The median PSA at baseline was 32 ng/mL, and 25% had a family history of PCa. Overall, 25.8% were asymptomatic and 46% had a non-metastatic presentation. After the OPUS campaign, PSA at diagnosis was significantly lower across all age groups. The incidence of PCa diagnosis in asymptomatic men was higher (31.4% vs. 19.9%) and a higher proportion of men were diagnosed with organ-confined, palpable disease (46% vs. 28%) (<i>p</i> < 0.001). The rate of patients eligible for active/radical treatment was higher after the OPUS campaign (patients who received surgery increased from 12.78% to 32.41%; patients who underwent radiation increased from 28.38% to 49.61%). The proportion of patients diagnosed with non-clinically significant disease was negligible and remained stable across time.</p><p><strong>Conclusions: </strong>PCa in Mexican patients displays aggressive features at diagnosis, whereas the rate of non-significant disease is negligible. The introduction of early detection strategies may lead to lower symptomatic and metastatic PCa and higher opportunities for radical treatment. This emphasizes the need for public awareness and for adjustment of screening strategies to the peculiarities of the Mexican population.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"16 21","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11544922/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615742","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Łukasz Zadka, Adam Ustaszewski, Natalia Glatzel-Plucińska, Agnieszka Rusak, Izabela Łaczmańska, Katarzyna Ratajczak-Wielgomas, Alicja Kmiecik, Aleksandra Piotrowska, Katarzyna Haczkiewicz-Leśniak, Agnieszka Gomułkiewicz, Magdalena Kostrzewska-Poczekaj, Piotr Dzięgiel
Background/objectives: The aim of this study was to examine the expression of TYK2 in colorectal cancer (CRC) and to determine the potential diagnostic and prognostic significance of this kinase.
Methods: Digital image analysis was performed to assess immunohistochemical TYK2 reactivity.
Results: There were significant differences for all positive pixels between CRC and normal colonic mucosa, with higher TYK2 expression levels observed in surgical margins than in adenocarcinomas (p = 0.0004). Paired t tests showed elevated immunoreactivity for overall TYK2 expression in matched pairs of CRC with adjacent surgical margins (p < 0.0001). Higher percentages of weak (p < 0.0001) and strong pixels (p = 0.0260) were detected in normal colonic mucosa than in cancer tissues. To distinguish cancer from normal intestinal mucosa, the following cutoffs for the TYK2 immune score were found: 29.5% for all cases and 31% for matched pairs. Tumor budding (Bd) was negatively correlated with the percentage of strong pixels for TYK2 (ρ = -0.270, p = 0.0096). The percentage of strong pixels was significantly elevated for the T parameter (p = 0.0428). There was a positive correlation between the number of involved lymph nodes and weak pixels (ρ = 0.239, p = 0.0242). Immunofluorescence staining showed significantly higher signal intensities in colonic mucosa than in CRC. The protein level of TYK2 was significantly higher in controls than in cancer tissues. TEM imaging showed lower levels of TYK2 in cancer than in ulcerative colitis.
Conclusions: TYK2 protein expression may bring diagnostic value in patients diagnosed with CRC.
{"title":"TYK2 Protein Expression and Its Potential as a Tissue-Based Biomarker for the Diagnosis of Colorectal Cancer.","authors":"Łukasz Zadka, Adam Ustaszewski, Natalia Glatzel-Plucińska, Agnieszka Rusak, Izabela Łaczmańska, Katarzyna Ratajczak-Wielgomas, Alicja Kmiecik, Aleksandra Piotrowska, Katarzyna Haczkiewicz-Leśniak, Agnieszka Gomułkiewicz, Magdalena Kostrzewska-Poczekaj, Piotr Dzięgiel","doi":"10.3390/cancers16213665","DOIUrl":"10.3390/cancers16213665","url":null,"abstract":"<p><strong>Background/objectives: </strong>The aim of this study was to examine the expression of TYK2 in colorectal cancer (CRC) and to determine the potential diagnostic and prognostic significance of this kinase.</p><p><strong>Methods: </strong>Digital image analysis was performed to assess immunohistochemical TYK2 reactivity.</p><p><strong>Results: </strong>There were significant differences for all positive pixels between CRC and normal colonic mucosa, with higher TYK2 expression levels observed in surgical margins than in adenocarcinomas (<i>p</i> = 0.0004). Paired <i>t</i> tests showed elevated immunoreactivity for overall TYK2 expression in matched pairs of CRC with adjacent surgical margins (<i>p</i> < 0.0001). Higher percentages of weak (<i>p</i> < 0.0001) and strong pixels (<i>p</i> = 0.0260) were detected in normal colonic mucosa than in cancer tissues. To distinguish cancer from normal intestinal mucosa, the following cutoffs for the TYK2 immune score were found: 29.5% for all cases and 31% for matched pairs. Tumor budding (Bd) was negatively correlated with the percentage of strong pixels for TYK2 (ρ = -0.270, <i>p</i> = 0.0096). The percentage of strong pixels was significantly elevated for the T parameter (<i>p</i> = 0.0428). There was a positive correlation between the number of involved lymph nodes and weak pixels (ρ = 0.239, <i>p</i> = 0.0242). Immunofluorescence staining showed significantly higher signal intensities in colonic mucosa than in CRC. The protein level of TYK2 was significantly higher in controls than in cancer tissues. TEM imaging showed lower levels of TYK2 in cancer than in ulcerative colitis.</p><p><strong>Conclusions: </strong>TYK2 protein expression may bring diagnostic value in patients diagnosed with CRC.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"16 21","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11545102/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142614278","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cesar Torres-Luna, Shuanzeng Wei, Sreenivas Bhattiprolu, George Tuszynski, Vicki L Rothman, Declan McNulty, Jeff Yang, Frank N Chang
Background/Objectives: Prostate cancer (PCa) is a prevalent malignancy, necessitating accurate diagnostic methods to distinguish it from benign conditions such as benign prostatic hyperplasia (BPH). Current diagnostic tools, relying primarily on serum prostate-specific antigen (PSA) levels, lack specificity, leading to an over-diagnosis and unnecessary treatment of patients with benign conditions. This study explores G-protein-coupled receptor-associated sorting protein 1 (GASP-1) as a more sensitive biomarker for PCa detection. Methods: Prostate tissue microarrays of healthy, BPH, and prostate cancer patients with different Gleason scores were studied. Polyclonal antibodies targeted against GASP-1 were used for routine immunohistochemistry (IHC) and enzyme-linked immunosorbent assay (ELISA) analyses. Results: The results indicated a 5-fold difference in serum GASP-1 levels between BPH and PCa, which was validated through GASP-1 IHC. Furthermore, a novel scoring system, the H-score, assesses GASP-1 granules' intensity and size, revealing a clear distinction between BPH and PCa. An additional analysis of GASP-1 expression between PCa cases with different Gleason scores reveals that GASP-1 overexpression correlates with PCa severity, providing insights into disease progression. Conclusions: The study supports GASP-1's role as a promising diagnostic marker, supplementing PSA testing, and offering improved risk stratification for PCa. Additionally, an open-source software system is introduced for an efficient GASP-1 granule color analysis, enhancing diagnostic accuracy.
{"title":"G-Protein-Coupled Receptor-Associated Sorting Protein 1 Overexpression Is Involved in the Progression of Benign Prostatic Hyperplasia, Early-Stage Prostatic Malignant Diseases, and Prostate Cancer.","authors":"Cesar Torres-Luna, Shuanzeng Wei, Sreenivas Bhattiprolu, George Tuszynski, Vicki L Rothman, Declan McNulty, Jeff Yang, Frank N Chang","doi":"10.3390/cancers16213659","DOIUrl":"10.3390/cancers16213659","url":null,"abstract":"<p><p><b>Background/Objectives:</b> Prostate cancer (PCa) is a prevalent malignancy, necessitating accurate diagnostic methods to distinguish it from benign conditions such as benign prostatic hyperplasia (BPH). Current diagnostic tools, relying primarily on serum prostate-specific antigen (PSA) levels, lack specificity, leading to an over-diagnosis and unnecessary treatment of patients with benign conditions. This study explores G-protein-coupled receptor-associated sorting protein 1 (GASP-1) as a more sensitive biomarker for PCa detection. <b>Methods:</b> Prostate tissue microarrays of healthy, BPH, and prostate cancer patients with different Gleason scores were studied. Polyclonal antibodies targeted against GASP-1 were used for routine immunohistochemistry (IHC) and enzyme-linked immunosorbent assay (ELISA) analyses. <b>Results:</b> The results indicated a 5-fold difference in serum GASP-1 levels between BPH and PCa, which was validated through GASP-1 IHC. Furthermore, a novel scoring system, the H-score, assesses GASP-1 granules' intensity and size, revealing a clear distinction between BPH and PCa. An additional analysis of GASP-1 expression between PCa cases with different Gleason scores reveals that GASP-1 overexpression correlates with PCa severity, providing insights into disease progression. <b>Conclusions</b>: The study supports GASP-1's role as a promising diagnostic marker, supplementing PSA testing, and offering improved risk stratification for PCa. Additionally, an open-source software system is introduced for an efficient GASP-1 granule color analysis, enhancing diagnostic accuracy.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"16 21","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11544983/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615689","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ab Majeed Ganai, Eirinaios I Vrettos, Stavroula G Kyrkou, Vasiliki Zoi, Tabasum Khan Pathan, Rajshekhar Karpoormath, Penelope Bouziotis, George A Alexiou, George A Kastis, Nicholas E Protonotarios, Andreas G Tzakos
Kinase inhibitors are potent therapeutic agents in cancer treatment, but their effectiveness is frequently restricted by the inability to image the tumor microenvironment. To address this constraint, kinase inhibitor-fluorophore conjugates have emerged as promising theranostic agents, allowing for simultaneous cancer diagnosis and treatment. These conjugates are gaining attention for their ability to visualize malignant tissues and concurrently enhance therapeutic interventions. This review explores the design principles governing the development of multimodal inhibitors, highlighting their potential as platforms for kinase tracking and inhibition via bioimaging. The structural aspects of constructing such theranostic agents are critically analyzed. This work could shed light on this intriguing field and provide adequate impetus for developing novel theranostic compounds based on small molecule inhibitors and fluorophores.
{"title":"Design Principles and Applications of Fluorescent Kinase Inhibitors for Simultaneous Cancer Bioimaging and Therapy.","authors":"Ab Majeed Ganai, Eirinaios I Vrettos, Stavroula G Kyrkou, Vasiliki Zoi, Tabasum Khan Pathan, Rajshekhar Karpoormath, Penelope Bouziotis, George A Alexiou, George A Kastis, Nicholas E Protonotarios, Andreas G Tzakos","doi":"10.3390/cancers16213667","DOIUrl":"10.3390/cancers16213667","url":null,"abstract":"<p><p>Kinase inhibitors are potent therapeutic agents in cancer treatment, but their effectiveness is frequently restricted by the inability to image the tumor microenvironment. To address this constraint, kinase inhibitor-fluorophore conjugates have emerged as promising theranostic agents, allowing for simultaneous cancer diagnosis and treatment. These conjugates are gaining attention for their ability to visualize malignant tissues and concurrently enhance therapeutic interventions. This review explores the design principles governing the development of multimodal inhibitors, highlighting their potential as platforms for kinase tracking and inhibition via bioimaging. The structural aspects of constructing such theranostic agents are critically analyzed. This work could shed light on this intriguing field and provide adequate impetus for developing novel theranostic compounds based on small molecule inhibitors and fluorophores.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"16 21","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11545566/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615604","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Owen P Leary, John P Zepecki, Mattia Pizzagalli, Steven A Toms, David D Liu, Yusuke Suita, Yao Ding, Jihong Wang, Renjie He, Caroline Chung, Clifton D Fuller, Jerrold L Boxerman, Nikos Tapinos, Richard J Gilbert
Background: The invasion of glioblastoma cells beyond the visible tumor margin depicted by conventional neuroimaging is believed to mediate recurrence and predict poor survival. Radiomic biomarkers that are associated with the direction and extent of tumor infiltration are, however, non-existent.
Methods: Patients from a single center with newly diagnosed glioblastoma (n = 7) underwent preoperative Q-space magnetic resonance imaging (QSI; 3T, 64 gradient directions, b = 1000 s/mm2) between 2018 and 2019. Tumors were manually segmented, and patterns of inter-voxel coherence spatially intersecting each segmentation were generated to represent tumor-associated tractography. One patient additionally underwent regional biopsy of diffusion tract- versus non-tract-associated tissue during tumor resection for RNA sequencing. Imaging data from this cohort were compared with a historical cohort of n = 66 glioblastoma patients who underwent similar QSI scans. Associations of tractography-derived metrics with survival were assessed using t-tests, linear regression, and Kaplan-Meier statistics. Patient-derived glioblastoma xenograft (PDX) mice generated with the sub-hippocampal injection of human-derived glioblastoma stem cells (GSCs) were scanned under high-field conditions (QSI, 7T, 512 gradient directions), and tumor-associated tractography was compared with the 3D microscopic reconstruction of immunostained GSCs.
Results: In the principal enrollment cohort of patients with glioblastoma, all cases displayed tractography patterns with tumor-intersecting tract bundles extending into brain parenchyma, a phenotype which was reproduced in PDX mice as well as in a larger comparison cohort of glioblastoma patients (n = 66), when applying similar methods. Reconstructed spatial patterns of GSCs in PDX mice closely mirrored tumor-associated tractography. On a Kaplan-Meier survival analysis of n = 66 patients, the calculated intra-tumoral mean diffusivity predicted the overall survival (p = 0.037), as did tractography-associated features including mean tract length (p = 0.039) and mean projecting tract length (p = 0.022). The RNA sequencing of human tissue samples (n = 13 tumor samples from a single patient) revealed the overexpression of transcripts which regulate cell motility in tract-associated samples.
Conclusions: QSI discriminates tumor-specific patterns of inter-voxel coherence believed to represent white matter pathways which may be susceptible to glioblastoma invasion. These findings may lay the groundwork for future work on therapeutic targeting, patient stratification, and prognosis in glioblastoma.
{"title":"Tumor-Associated Tractography Derived from High-Angular-Resolution Q-Space MRI May Predict Patterns of Cellular Invasion in Glioblastoma.","authors":"Owen P Leary, John P Zepecki, Mattia Pizzagalli, Steven A Toms, David D Liu, Yusuke Suita, Yao Ding, Jihong Wang, Renjie He, Caroline Chung, Clifton D Fuller, Jerrold L Boxerman, Nikos Tapinos, Richard J Gilbert","doi":"10.3390/cancers16213669","DOIUrl":"10.3390/cancers16213669","url":null,"abstract":"<p><strong>Background: </strong>The invasion of glioblastoma cells beyond the visible tumor margin depicted by conventional neuroimaging is believed to mediate recurrence and predict poor survival. Radiomic biomarkers that are associated with the direction and extent of tumor infiltration are, however, non-existent.</p><p><strong>Methods: </strong>Patients from a single center with newly diagnosed glioblastoma (<i>n</i> = 7) underwent preoperative Q-space magnetic resonance imaging (QSI; 3T, 64 gradient directions, b = 1000 s/mm<sup>2</sup>) between 2018 and 2019. Tumors were manually segmented, and patterns of inter-voxel coherence spatially intersecting each segmentation were generated to represent tumor-associated tractography. One patient additionally underwent regional biopsy of diffusion tract- versus non-tract-associated tissue during tumor resection for RNA sequencing. Imaging data from this cohort were compared with a historical cohort of <i>n</i> = 66 glioblastoma patients who underwent similar QSI scans. Associations of tractography-derived metrics with survival were assessed using <i>t</i>-tests, linear regression, and Kaplan-Meier statistics. Patient-derived glioblastoma xenograft (PDX) mice generated with the sub-hippocampal injection of human-derived glioblastoma stem cells (GSCs) were scanned under high-field conditions (QSI, 7T, 512 gradient directions), and tumor-associated tractography was compared with the 3D microscopic reconstruction of immunostained GSCs.</p><p><strong>Results: </strong>In the principal enrollment cohort of patients with glioblastoma, all cases displayed tractography patterns with tumor-intersecting tract bundles extending into brain parenchyma, a phenotype which was reproduced in PDX mice as well as in a larger comparison cohort of glioblastoma patients (<i>n</i> = 66), when applying similar methods. Reconstructed spatial patterns of GSCs in PDX mice closely mirrored tumor-associated tractography. On a Kaplan-Meier survival analysis of <i>n</i> = 66 patients, the calculated intra-tumoral mean diffusivity predicted the overall survival (<i>p</i> = 0.037), as did tractography-associated features including mean tract length (<i>p</i> = 0.039) and mean projecting tract length (<i>p</i> = 0.022). The RNA sequencing of human tissue samples (<i>n</i> = 13 tumor samples from a single patient) revealed the overexpression of transcripts which regulate cell motility in tract-associated samples.</p><p><strong>Conclusions: </strong>QSI discriminates tumor-specific patterns of inter-voxel coherence believed to represent white matter pathways which may be susceptible to glioblastoma invasion. These findings may lay the groundwork for future work on therapeutic targeting, patient stratification, and prognosis in glioblastoma.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"16 21","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11544887/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142613651","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Seo Hee Choi, Jong Won Park, Yeona Cho, Gowoon Yang, Hong In Yoon
Background: Accurate delineation of tumors and organs at risk (OARs) is crucial for intensity-modulated radiation therapy. This study aimed to evaluate the performance of OncoStudio, an AI-based auto-segmentation tool developed for Korean patients, compared with Protégé AI, a globally developed tool that uses data from Korean cancer patients.
Methods: A retrospective analysis of 1200 Korean cancer patients treated with radiotherapy was conducted. Auto-contours generated via OncoStudio and Protégé AI were compared with manual contours across the head and neck and thoracic, abdominal, and pelvic organs. Accuracy was assessed using the Dice similarity coefficient (DSC), mean surface distance (MSD), and 95% Hausdorff distance (HD). Feedback was obtained from 10 participants, including radiation oncologists, residents, and radiation therapists, via an online survey with a Turing test component.
Results: OncoStudio outperformed Protégé AI in 85% of the evaluated OARs (p < 0.001). For head and neck organs, OncoStudio achieved a similar DSC (0.70 vs. 0.70, p = 0.637) but significantly lower MSD and 95% HD values (p < 0.001). In thoracic organs, OncoStudio performed excellently in 90% of cases, with a significantly greater DSC (male: 0.87 vs. 0.82, p < 0.001; female: 0.95 vs. 0.87, p < 0.001). OncoStudio also demonstrated superior accuracy in abdominal (DSC 0.88 vs. 0.81, p < 0.001) and pelvic organs (male: DSC 0.95 vs. 0.85, p < 0.001; female: DSC 0.82 vs. 0.73, p < 0.001). Clinicians favored OncoStudio in 70% of cases, with 90% endorsing its clinical suitability for Korean patients.
Conclusions: OncoStudio, which is tailored for Korean patients, demonstrated superior segmentation accuracy across multiple anatomical regions, suggesting its suitability for radiotherapy planning in this population.
{"title":"Automated Organ Segmentation for Radiation Therapy: A Comparative Analysis of AI-Based Tools Versus Manual Contouring in Korean Cancer Patients.","authors":"Seo Hee Choi, Jong Won Park, Yeona Cho, Gowoon Yang, Hong In Yoon","doi":"10.3390/cancers16213670","DOIUrl":"10.3390/cancers16213670","url":null,"abstract":"<p><strong>Background: </strong>Accurate delineation of tumors and organs at risk (OARs) is crucial for intensity-modulated radiation therapy. This study aimed to evaluate the performance of OncoStudio, an AI-based auto-segmentation tool developed for Korean patients, compared with Protégé AI, a globally developed tool that uses data from Korean cancer patients.</p><p><strong>Methods: </strong>A retrospective analysis of 1200 Korean cancer patients treated with radiotherapy was conducted. Auto-contours generated via OncoStudio and Protégé AI were compared with manual contours across the head and neck and thoracic, abdominal, and pelvic organs. Accuracy was assessed using the Dice similarity coefficient (DSC), mean surface distance (MSD), and 95% Hausdorff distance (HD). Feedback was obtained from 10 participants, including radiation oncologists, residents, and radiation therapists, via an online survey with a Turing test component.</p><p><strong>Results: </strong>OncoStudio outperformed Protégé AI in 85% of the evaluated OARs (<i>p</i> < 0.001). For head and neck organs, OncoStudio achieved a similar DSC (0.70 vs. 0.70, <i>p</i> = 0.637) but significantly lower MSD and 95% HD values (<i>p</i> < 0.001). In thoracic organs, OncoStudio performed excellently in 90% of cases, with a significantly greater DSC (male: 0.87 vs. 0.82, <i>p</i> < 0.001; female: 0.95 vs. 0.87, <i>p</i> < 0.001). OncoStudio also demonstrated superior accuracy in abdominal (DSC 0.88 vs. 0.81, <i>p</i> < 0.001) and pelvic organs (male: DSC 0.95 vs. 0.85, <i>p</i> < 0.001; female: DSC 0.82 vs. 0.73, <i>p</i> < 0.001). Clinicians favored OncoStudio in 70% of cases, with 90% endorsing its clinical suitability for Korean patients.</p><p><strong>Conclusions: </strong>OncoStudio, which is tailored for Korean patients, demonstrated superior segmentation accuracy across multiple anatomical regions, suggesting its suitability for radiotherapy planning in this population.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"16 21","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11544936/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615442","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Artificial intelligence (AI), encompassing several tools and platforms such as artificial "general" intelligence (AGI) and generative artificial intelligence (GenAI), has facilitated cancer research, enhancing productivity in terms of research publications and translational value for cancer patients. AGI tools, such as ChatGPT, assist preclinical and clinical scientists in identifying tumor heterogeneity, predicting therapy outcomes, and streamlining research publications. However, this perspective review also explores the potential of AI's influence on cancer research with regard to its impact on disruptive sciences and discoveries by preclinical and clinical scientists. The increasing reliance on AI tools may compromise biological intelligence, disrupting abstraction, creativity, and critical thinking. This could contribute to the declining trend of disruptive sciences, hindering landmark discoveries and innovations. This perspective review narrates the role of different forms of AI in the potentiation of productive cancer research and the potential disruption of disruptive sciences due to AI's influence.
{"title":"Evolving Artificial Intelligence (AI) at the Crossroads: Potentiating Productive vs. Declining Disruptive Cancer Research.","authors":"Nilesh Kumar Sharma, Sachin C Sarode","doi":"10.3390/cancers16213646","DOIUrl":"10.3390/cancers16213646","url":null,"abstract":"<p><p>Artificial intelligence (AI), encompassing several tools and platforms such as artificial \"general\" intelligence (AGI) and generative artificial intelligence (GenAI), has facilitated cancer research, enhancing productivity in terms of research publications and translational value for cancer patients. AGI tools, such as ChatGPT, assist preclinical and clinical scientists in identifying tumor heterogeneity, predicting therapy outcomes, and streamlining research publications. However, this perspective review also explores the potential of AI's influence on cancer research with regard to its impact on disruptive sciences and discoveries by preclinical and clinical scientists. The increasing reliance on AI tools may compromise biological intelligence, disrupting abstraction, creativity, and critical thinking. This could contribute to the declining trend of disruptive sciences, hindering landmark discoveries and innovations. This perspective review narrates the role of different forms of AI in the potentiation of productive cancer research and the potential disruption of disruptive sciences due to AI's influence.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"16 21","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11544874/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615651","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Debora Maffeo, Angela Rina, Viola Bianca Serio, Athina Markou, Tomasz Powrózek, Vera Constâncio, Sandra P Nunes, Carmen Jerónimo, Alfonso Calvo, Francesca Mari, Elisa Frullanti, Diletta Rosati, Maria Palmieri
Background: Non-Small Cell Lung Cancer (NSCLC) remains a challenging disease to manage with effectiveness. Early detection and precise monitoring are crucial for improving patient outcomes. Circulating tumor DNA (ctDNA) offers a non-invasive cancer detection and monitoring method. Emerging biomarkers, such as ctDNA methylation, have shown promise in enhancing diagnostic accuracy and prognostic assessment in NSCLC. In this review, we examined the current evidence regarding ctDNA methylation's role in NSCLC detection through a systematic review of the existing literature and meta-analysis. Methods: We systematically searched PubMed, Medline, Embase, and Web of Science databases up to 26 June 2024 for studies on the role of ctDNA methylation analysis in NSCLC patients. We included studies from 2010 to 2024 on NSCLC patients. We excluded case reports, non-English articles, studies on cell lines or artificial samples, those without cfDNA detection, prognostic studies, and studies with non-extractable data or mixed cancer types. Funnel plots were visually examined for potential publication bias, with a p value < 0.05 indicating bias. Meta-analysis was conducted using R packages (meta, forestplot, and mada). Combined sensitivity, specificity, positive likelihood ratio (LR+), negative likelihood ratio (LR-), positive and negative predictive values, diagnostic odds ratio (DOR), and 95% confidence intervals (95% CI) were calculated. A summary receiver operating characteristic curve (SROC) and area under the curve (AUC) with related Standard Error (SE) were used to evaluate the overall diagnostic performance. Additionally, RASSF1A, APC, SOX17, SEPT9, and RARβ2 were analyzed, since their methylation was assessed in two or more studies. Results: From 38 candidate papers, we finally identified 12 studies, including 472 NSCLC patients. The pooled sensitivity was 0.62 (0.47-0.77) and the specificity was 0.90 (0.85-0.94). The diagnostic odds ratio was 15.6 (95% CI 9.36-26.09) and the area under the curve was 0.249 (SE = 0.138). The positive and negative predictive values were 5.38 (95% CI 3.89-7.44) and 0.34 (95% CI 0.22-0.54), respectively. For single genes, the specificity reached 0.83~0.96, except for RARβ2, but the sensitivity was relatively low for each gene. Significant heterogeneity across the included studies, the potential publication bias for specificity (p = 0.0231), and the need to validate the clinical utility of ctDNA methylation for monitoring treatment response and predicting outcomes in NSCLC patients represent the main limitations of this study. Conclusions: These results provide evidence of the significant potential of ctDNA methylation as a valuable biomarker for improving the diagnosis of NSCLC, advocating for its integration into clinical practice to enhance patient management.
背景:非小细胞肺癌(NSCLC非小细胞肺癌(NSCLC)仍然是一种难以有效控制的疾病。早期检测和精确监测对改善患者预后至关重要。循环肿瘤 DNA(ctDNA)提供了一种非侵入性癌症检测和监测方法。ctDNA甲基化等新兴生物标志物有望提高NSCLC的诊断准确性和预后评估。在这篇综述中,我们通过对现有文献的系统综述和荟萃分析,研究了有关ctDNA甲基化在NSCLC检测中作用的现有证据。研究方法我们系统检索了截至 2024 年 6 月 26 日的 PubMed、Medline、Embase 和 Web of Science 数据库中有关ctDNA 甲基化分析在 NSCLC 患者中作用的研究。我们纳入了 2010 年至 2024 年有关 NSCLC 患者的研究。我们排除了病例报告、非英文文章、细胞系或人工样本研究、未检测 cfDNA 的研究、预后研究以及无法提取数据或混合癌症类型的研究。对漏斗图进行直观检查,以确定是否存在潜在的发表偏倚,P 值小于 0.05 则表明存在偏倚。使用 R 软件包(meta、forestplot 和 mada)进行 Meta 分析。计算了综合灵敏度、特异性、阳性似然比(LR+)、阴性似然比(LR-)、阳性和阴性预测值、诊断几率比(DOR)和 95% 置信区间(95% CI)。受试者操作特征曲线(SROC)和曲线下面积(AUC)及相关标准误差(SE)用于评估总体诊断性能。此外,还对 RASSF1A、APC、SOX17、SEPT9 和 RARβ2 进行了分析,因为它们的甲基化在两项或多项研究中进行了评估。结果从 38 篇候选论文中,我们最终确定了 12 项研究,包括 472 名 NSCLC 患者。汇总灵敏度为 0.62(0.47-0.77),特异性为 0.90(0.85-0.94)。诊断几率比为 15.6(95% CI 9.36-26.09),曲线下面积为 0.249(SE = 0.138)。阳性和阴性预测值分别为 5.38(95% CI 3.89-7.44)和 0.34(95% CI 0.22-0.54)。除 RARβ2 外,单个基因的特异性达到 0.83~0.96,但每个基因的敏感性相对较低。本研究的主要局限性包括:纳入研究之间存在显著异质性、特异性可能存在发表偏倚(p = 0.0231),以及需要验证ctDNA甲基化在监测NSCLC患者治疗反应和预测预后方面的临床实用性。结论这些结果证明了ctDNA甲基化作为一种有价值的生物标记物在改善NSCLC诊断方面的巨大潜力,并倡导将其纳入临床实践以加强对患者的管理。
{"title":"The Evidence Base for Circulating Tumor DNA-Methylation in Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis.","authors":"Debora Maffeo, Angela Rina, Viola Bianca Serio, Athina Markou, Tomasz Powrózek, Vera Constâncio, Sandra P Nunes, Carmen Jerónimo, Alfonso Calvo, Francesca Mari, Elisa Frullanti, Diletta Rosati, Maria Palmieri","doi":"10.3390/cancers16213641","DOIUrl":"10.3390/cancers16213641","url":null,"abstract":"<p><p><i>Background</i>: Non-Small Cell Lung Cancer (NSCLC) remains a challenging disease to manage with effectiveness. Early detection and precise monitoring are crucial for improving patient outcomes. Circulating tumor DNA (ctDNA) offers a non-invasive cancer detection and monitoring method. Emerging biomarkers, such as ctDNA methylation, have shown promise in enhancing diagnostic accuracy and prognostic assessment in NSCLC. In this review, we examined the current evidence regarding ctDNA methylation's role in NSCLC detection through a systematic review of the existing literature and meta-analysis. <i>Methods</i>: We systematically searched PubMed, Medline, Embase, and Web of Science databases up to 26 June 2024 for studies on the role of ctDNA methylation analysis in NSCLC patients. We included studies from 2010 to 2024 on NSCLC patients. We excluded case reports, non-English articles, studies on cell lines or artificial samples, those without cfDNA detection, prognostic studies, and studies with non-extractable data or mixed cancer types. Funnel plots were visually examined for potential publication bias, with a <i>p</i> value < 0.05 indicating bias. Meta-analysis was conducted using R packages (meta, forestplot, and mada). Combined sensitivity, specificity, positive likelihood ratio (LR+), negative likelihood ratio (LR-), positive and negative predictive values, diagnostic odds ratio (DOR), and 95% confidence intervals (95% CI) were calculated. A summary receiver operating characteristic curve (SROC) and area under the curve (AUC) with related Standard Error (SE) were used to evaluate the overall diagnostic performance. Additionally, <i>RASSF1A</i>, <i>APC</i>, <i>SOX17</i>, <i>SEPT9,</i> and <i>RARβ2</i> were analyzed, since their methylation was assessed in two or more studies. <i>Results</i>: From 38 candidate papers, we finally identified 12 studies, including 472 NSCLC patients. The pooled sensitivity was 0.62 (0.47-0.77) and the specificity was 0.90 (0.85-0.94). The diagnostic odds ratio was 15.6 (95% CI 9.36-26.09) and the area under the curve was 0.249 (SE = 0.138). The positive and negative predictive values were 5.38 (95% CI 3.89-7.44) and 0.34 (95% CI 0.22-0.54), respectively. For single genes, the specificity reached 0.83~0.96, except for <i>RARβ2</i>, but the sensitivity was relatively low for each gene. Significant heterogeneity across the included studies, the potential publication bias for specificity (<i>p</i> = 0.0231), and the need to validate the clinical utility of ctDNA methylation for monitoring treatment response and predicting outcomes in NSCLC patients represent the main limitations of this study. <i>Conclusions:</i> These results provide evidence of the significant potential of ctDNA methylation as a valuable biomarker for improving the diagnosis of NSCLC, advocating for its integration into clinical practice to enhance patient management.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"16 21","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11544801/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615746","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kevin Nguyen, Karina Fama, Guadalupe Mercado, Yin Myat, Kyaw Thein
Recent advancements in oncology have led to the development of histology-agnostic therapies, which target genetic alterations irrespective of the tumor's tissue of origin. This review aimed to provide a comprehensive update on the current state of histology-agnostic drug development, focusing on key therapies, including pembrolizumab, larotrectinib, entrectinib, dostarlimab, dabrafenib plus trametinib, selpercatinib, trastuzumab deruxtecan, and reprotrectinib. We performed a detailed analysis of each therapy's mechanism of action, clinical trial outcomes, and associated biomarkers. The review further explores challenges in drug resistance, such as adaptive signaling pathways and neoantigen variability, as well as diagnostic limitations in identifying optimal patient populations. While these therapies have demonstrated efficacy in various malignancies, significant hurdles remain, including intratumoral heterogeneity and resistance mechanisms that diminish treatment effectiveness. We propose considerations for refining trial designs and emerging biomarkers, such as tumor neoantigen burden, to enhance patient selection. These findings illustrate the transformative potential of histology-agnostic therapies in precision oncology but highlight the need for continued research to optimize their use and overcome existing barriers.
肿瘤学的最新进展推动了组织学诊断疗法的发展,这种疗法不考虑肿瘤的原发组织,而是以基因改变为靶点。本综述旨在全面更新组织学诊断药物的开发现状,重点关注主要疗法,包括pembrolizumab、larotrectinib、entrectinib、dostarlimab、dabrafenib plus trametinib、selpercatinib、曲妥珠单抗deruxtecan和reprotrectinib。我们对每种疗法的作用机制、临床试验结果和相关生物标记物进行了详细分析。综述进一步探讨了耐药性方面的挑战,如适应性信号通路和新抗原的可变性,以及在确定最佳患者人群方面的诊断局限性。虽然这些疗法已在各种恶性肿瘤中显示出疗效,但仍存在重大障碍,包括肿瘤内异质性和降低治疗效果的耐药机制。我们提出了完善试验设计和新兴生物标志物(如肿瘤新抗原负荷)的注意事项,以加强对患者的选择。这些发现说明了组织学诊断疗法在精准肿瘤学中的变革潜力,但也强调了继续研究以优化其使用并克服现有障碍的必要性。
{"title":"Histology Agnostic Drug Development: An Updated Review.","authors":"Kevin Nguyen, Karina Fama, Guadalupe Mercado, Yin Myat, Kyaw Thein","doi":"10.3390/cancers16213642","DOIUrl":"10.3390/cancers16213642","url":null,"abstract":"<p><p>Recent advancements in oncology have led to the development of histology-agnostic therapies, which target genetic alterations irrespective of the tumor's tissue of origin. This review aimed to provide a comprehensive update on the current state of histology-agnostic drug development, focusing on key therapies, including pembrolizumab, larotrectinib, entrectinib, dostarlimab, dabrafenib plus trametinib, selpercatinib, trastuzumab deruxtecan, and reprotrectinib. We performed a detailed analysis of each therapy's mechanism of action, clinical trial outcomes, and associated biomarkers. The review further explores challenges in drug resistance, such as adaptive signaling pathways and neoantigen variability, as well as diagnostic limitations in identifying optimal patient populations. While these therapies have demonstrated efficacy in various malignancies, significant hurdles remain, including intratumoral heterogeneity and resistance mechanisms that diminish treatment effectiveness. We propose considerations for refining trial designs and emerging biomarkers, such as tumor neoantigen burden, to enhance patient selection. These findings illustrate the transformative potential of histology-agnostic therapies in precision oncology but highlight the need for continued research to optimize their use and overcome existing barriers.</p>","PeriodicalId":9681,"journal":{"name":"Cancers","volume":"16 21","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11544807/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615710","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}