Loïc Van Dieren, Tom Quisenaerts, Mackenzie Licata, Arnaud Beddok, Alexandre G Lellouch, Dirk Ysebaert, Vera Saldien, Marc Peeters, Ivana Gorbaslieva
Introduction: The abscopal effect is a systemic immune response characterized by metastases regression at sites distant from the irradiated lesion. This systematic review aims to explore the immunological mechanisms of action underlying the abscopal effect and to investigate how hyperthermia (HT) can increase the chances of radiotherapy (RT) triggering systemic anti-tumor immune responses.
Methods: This review is created in accordance with the PRISMA guidelines.
Results and conclusion: HT and RT have both complementary and synergistic immunological effects. Both methods trigger danger signal release, promoting cytokine and chemokine secretion, which increases T-cell infiltration and facilitates cell death. Both treatments upregulate extracellular tumor HSP70, which could amplify DAMP recognition by macrophages and DCs, leading to stronger tumor antigen presentation and CTL-mediated immune responses. Additionally, the combined increase in cell adhesion molecules (VCAM-1, ICAM-1, E-selectin, L-selectin) could enhance leukocyte adhesion to tumors, improving lymphocyte trafficking and boosting systemic anti-tumor effects. Lastly, HT causes vasodilation and improves blood flow, which might exacerbate those distant effects. We suggest the combination of local radiotherapy with fever-range whole-body hyperthermia to optimally enhance the chances of triggering the abscopal effect mediated by the immune system.
简介缺席效应是一种全身性免疫反应,其特点是在远离照射病灶的部位出现转移消退。本系统性综述旨在探索缺席效应的免疫学作用机制,并研究热疗(HT)如何增加放疗(RT)引发全身抗肿瘤免疫反应的机会:本综述根据PRISMA指南撰写:热疗和放疗具有互补和协同的免疫效应。两种方法都会触发危险信号的释放,促进细胞因子和趋化因子的分泌,从而增加 T 细胞浸润并促进细胞死亡。这两种治疗方法都能上调细胞外肿瘤 HSP70,从而增强巨噬细胞和 DC 对 DAMP 的识别,导致更强的肿瘤抗原呈递和 CTL 介导的免疫反应。此外,细胞粘附分子(VCAM-1、ICAM-1、E-选择素、L-选择素)的联合增加可增强白细胞对肿瘤的粘附,改善淋巴细胞的迁移,增强全身抗肿瘤效应。最后,高温热疗会导致血管扩张,改善血流,这可能会加剧这些远处效应。我们建议将局部放疗与发热范围全身热疗相结合,以最大限度地提高免疫系统介导的脱灶效应的触发机会。
{"title":"Combined Radiotherapy and Hyperthermia: A Systematic Review of Immunological Synergies for Amplifying Radiation-Induced Abscopal Effects.","authors":"Loïc Van Dieren, Tom Quisenaerts, Mackenzie Licata, Arnaud Beddok, Alexandre G Lellouch, Dirk Ysebaert, Vera Saldien, Marc Peeters, Ivana Gorbaslieva","doi":"10.3390/cancers16213656","DOIUrl":"10.3390/cancers16213656","url":null,"abstract":"<p><strong>Introduction: </strong>The abscopal effect is a systemic immune response characterized by metastases regression at sites distant from the irradiated lesion. This systematic review aims to explore the immunological mechanisms of action underlying the abscopal effect and to investigate how hyperthermia (HT) can increase the chances of radiotherapy (RT) triggering systemic anti-tumor immune responses.</p><p><strong>Methods: </strong>This review is created in accordance with the PRISMA guidelines.</p><p><strong>Results and conclusion: </strong>HT and RT have both complementary and synergistic immunological effects. Both methods trigger danger signal release, promoting cytokine and chemokine secretion, which increases T-cell infiltration and facilitates cell death. Both treatments upregulate extracellular tumor HSP70, which could amplify DAMP recognition by macrophages and DCs, leading to stronger tumor antigen presentation and CTL-mediated immune responses. Additionally, the combined increase in cell adhesion molecules (VCAM-1, ICAM-1, E-selectin, L-selectin) could enhance leukocyte adhesion to tumors, improving lymphocyte trafficking and boosting systemic anti-tumor effects. Lastly, HT causes vasodilation and improves blood flow, which might exacerbate those distant effects. We suggest the combination of local radiotherapy with fever-range whole-body hyperthermia to optimally enhance the chances of triggering the abscopal effect mediated by the immune system.</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/PMC11545184/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615548","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}
Sarron Randall-Demllo, Ghanyah Al-Qadami, Anita E Raposo, Chenkai Ma, Ilka K Priebe, Maryam Hor, Rajvinder Singh, Kim Y C Fung
Despite improvements in participation in population-based screening programme, colorectal cancer remains a major cause of cancer-related mortality worldwide. Targeted interventions are desirable to reduce the health and economic burden of this disease. Two-dimensional monolayers of colorectal cancer cell lines represent the traditional in vitro models for disease and are often used for diverse purposes, including the delineation of molecular pathways associated with disease aetiology or the gauging of drug efficacy. The lack of complexity in such models, chiefly the limited epithelial cell diversity and differentiation, attenuated mucus production, lack of microbial interactions and mechanical stresses, has driven interest in the development of more holistic and physiologically relevant in vitro model systems. In particular, established ex vivo patient-derived explant and patient-derived tumour xenograft models have been supplemented by progress in organoid and microfluidic organ-on-a-chip cultures. Here, we discuss the applicability of advanced culturing technologies, such as organoid systems, as models for colorectal cancer and for testing chemotherapeutic drug sensitivity and efficacy. We highlight current challenges associated with organoid technologies and discuss their future for more accurate disease modelling and personalized medicine.
{"title":"Ex Vivo Intestinal Organoid Models: Current State-of-the-Art and Challenges in Disease Modelling and Therapeutic Testing for Colorectal Cancer.","authors":"Sarron Randall-Demllo, Ghanyah Al-Qadami, Anita E Raposo, Chenkai Ma, Ilka K Priebe, Maryam Hor, Rajvinder Singh, Kim Y C Fung","doi":"10.3390/cancers16213664","DOIUrl":"10.3390/cancers16213664","url":null,"abstract":"<p><p>Despite improvements in participation in population-based screening programme, colorectal cancer remains a major cause of cancer-related mortality worldwide. Targeted interventions are desirable to reduce the health and economic burden of this disease. Two-dimensional monolayers of colorectal cancer cell lines represent the traditional in vitro models for disease and are often used for diverse purposes, including the delineation of molecular pathways associated with disease aetiology or the gauging of drug efficacy. The lack of complexity in such models, chiefly the limited epithelial cell diversity and differentiation, attenuated mucus production, lack of microbial interactions and mechanical stresses, has driven interest in the development of more holistic and physiologically relevant in vitro model systems. In particular, established ex vivo patient-derived explant and patient-derived tumour xenograft models have been supplemented by progress in organoid and microfluidic organ-on-a-chip cultures. Here, we discuss the applicability of advanced culturing technologies, such as organoid systems, as models for colorectal cancer and for testing chemotherapeutic drug sensitivity and efficacy. We highlight current challenges associated with organoid technologies and discuss their future for more accurate disease modelling and personalized medicine.</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/PMC11544769/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615654","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}
Chetan V Vakkalagadda, Danielle B Dressler, Zequn Sun, Joseph Fuchs, Yingzhe Liu, Philip Silberman, Avanthi Ragam, Sheetal Kircher, Jyoti D Patel, Nisha A Mohindra
Purpose: Broad-based molecular testing with next-generation sequencing (NGS) is now the standard of care in advanced non-small cell lung cancer (NSCLC). Two approaches to molecular testing are (1) reflexive testing at pathologic NSCLC confirmation, often using an in-house molecular panel, and (2) send-out testing to private vendors, ordered by a clinician. This study explored the outcomes with reflex versus send-out testing.
Methods: A retrospective chart review was conducted of patients diagnosed with de novo stage IV NSCLC in 2019 and 2020 at three hospitals in the same system, one academic hospital (Northwestern Memorial Hospital, or NMH) utilizing reflex, in-house NGS, and two community-based hospitals (Central DuPage Hospital, or CDH, and Delnor, or D) sending out tissue samples for testing. The outcomes assessed were the time from biopsy to results, biopsy to treatment, the incidence of first-line targetable mutations and the use of first-line targeted therapies, and overall survival.
Results: In total, 191 patients met the inclusion criteria, 85 at NMH, 106 at CDH + D, and in total, 131 in 2019 and 60 in 2020. The time to results was significantly shorter with reflexive NGS when compared with send-out testing; the time to treatment was also shorter but not statistically significant. At CDH + D, the time to results was significantly shorter with a limited panel than with comprehensive testing, but the time to treatment was similar. NGS testing rates were 95% at NMH and 84.5% at CDH + D (p = 0.009), with 31.0% at NMH receiving 1L targeted therapies versus 20.8% at CDH + D (p = 0.08). In 2019, the median time from biopsy to treatment was 35 days at NMH and 38 days at CDH and Delnor; in 2020, time to treatment was 26 days and 37 days, respectively. Overall survival trended longer in 2020 relative to 2019 independent of site.
Conclusion: Reflexive NGS testing is associated with a shorter time to actionable results and higher rates of first-line targetable mutations than send-out testing. In practices with send-out testing, limited panels had slightly faster turnaround times but no difference in time to treatment. If resources allow, reflexive NGS should be considered in healthcare systems for patients with NSCLC.
{"title":"The Impact of Next-Generation Sequencing Workflows on Outcomes in Advanced Lung Cancer: A Retrospective Analysis at One Academic Health System.","authors":"Chetan V Vakkalagadda, Danielle B Dressler, Zequn Sun, Joseph Fuchs, Yingzhe Liu, Philip Silberman, Avanthi Ragam, Sheetal Kircher, Jyoti D Patel, Nisha A Mohindra","doi":"10.3390/cancers16213654","DOIUrl":"10.3390/cancers16213654","url":null,"abstract":"<p><strong>Purpose: </strong>Broad-based molecular testing with next-generation sequencing (NGS) is now the standard of care in advanced non-small cell lung cancer (NSCLC). Two approaches to molecular testing are (1) reflexive testing at pathologic NSCLC confirmation, often using an in-house molecular panel, and (2) send-out testing to private vendors, ordered by a clinician. This study explored the outcomes with reflex versus send-out testing.</p><p><strong>Methods: </strong>A retrospective chart review was conducted of patients diagnosed with de novo stage IV NSCLC in 2019 and 2020 at three hospitals in the same system, one academic hospital (Northwestern Memorial Hospital, or NMH) utilizing reflex, in-house NGS, and two community-based hospitals (Central DuPage Hospital, or CDH, and Delnor, or D) sending out tissue samples for testing. The outcomes assessed were the time from biopsy to results, biopsy to treatment, the incidence of first-line targetable mutations and the use of first-line targeted therapies, and overall survival.</p><p><strong>Results: </strong>In total, 191 patients met the inclusion criteria, 85 at NMH, 106 at CDH + D, and in total, 131 in 2019 and 60 in 2020. The time to results was significantly shorter with reflexive NGS when compared with send-out testing; the time to treatment was also shorter but not statistically significant. At CDH + D, the time to results was significantly shorter with a limited panel than with comprehensive testing, but the time to treatment was similar. NGS testing rates were 95% at NMH and 84.5% at CDH + D (<i>p</i> = 0.009), with 31.0% at NMH receiving 1L targeted therapies versus 20.8% at CDH + D (<i>p</i> = 0.08). In 2019, the median time from biopsy to treatment was 35 days at NMH and 38 days at CDH and Delnor; in 2020, time to treatment was 26 days and 37 days, respectively. Overall survival trended longer in 2020 relative to 2019 independent of site.</p><p><strong>Conclusion: </strong>Reflexive NGS testing is associated with a shorter time to actionable results and higher rates of first-line targetable mutations than send-out testing. In practices with send-out testing, limited panels had slightly faster turnaround times but no difference in time to treatment. If resources allow, reflexive NGS should be considered in healthcare systems for patients with NSCLC.</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/PMC11545180/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615750","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}
Lourdes Herrera-Quintana, Héctor Vázquez-Lorente, Rafael Cardoso Maciel Costa Silva, Jorge Olivares-Arancibia, Tomás Reyes-Amigo, Bruno Ricardo Barreto Pires, Julio Plaza-Diaz
The gut microbiome has emerged as a crucial player in modulating cancer therapies, including radiotherapy. In the case of breast cancer, the interplay between the microbiome and radiotherapy-derived metabolites may enhance therapeutic outcomes and minimize adverse effects. In this review, we explore the bidirectional relationship between the gut microbiome and breast cancer. We explain how gut microbiome composition influences cancer progression and treatment response, and how breast cancer and its treatments influence microbiome composition. A dual role for radiotherapy-derived metabolites is explored in this article, highlighting both their therapeutic benefits and potential hazards. By integrating genomics, metabolomics, and bioinformatics tools, we present a comprehensive overview of these interactions. The study provides real-world insight through case studies and clinical trials, while therapeutic innovations such as probiotics, and dietary interventions are examined for their potential to modulate the microbiome and enhance treatment effectiveness. Moreover, ethical considerations and patient perspectives are discussed, ensuring a comprehensive understanding of the subject. Towards revolutionizing treatment strategies and improving patient outcomes, the review concludes with future research directions. It also envisions integrating microbiome and metabolite research into personalized breast cancer therapy.
{"title":"The Role of the Microbiome and of Radiotherapy-Derived Metabolites in Breast Cancer.","authors":"Lourdes Herrera-Quintana, Héctor Vázquez-Lorente, Rafael Cardoso Maciel Costa Silva, Jorge Olivares-Arancibia, Tomás Reyes-Amigo, Bruno Ricardo Barreto Pires, Julio Plaza-Diaz","doi":"10.3390/cancers16213671","DOIUrl":"10.3390/cancers16213671","url":null,"abstract":"<p><p>The gut microbiome has emerged as a crucial player in modulating cancer therapies, including radiotherapy. In the case of breast cancer, the interplay between the microbiome and radiotherapy-derived metabolites may enhance therapeutic outcomes and minimize adverse effects. In this review, we explore the bidirectional relationship between the gut microbiome and breast cancer. We explain how gut microbiome composition influences cancer progression and treatment response, and how breast cancer and its treatments influence microbiome composition. A dual role for radiotherapy-derived metabolites is explored in this article, highlighting both their therapeutic benefits and potential hazards. By integrating genomics, metabolomics, and bioinformatics tools, we present a comprehensive overview of these interactions. The study provides real-world insight through case studies and clinical trials, while therapeutic innovations such as probiotics, and dietary interventions are examined for their potential to modulate the microbiome and enhance treatment effectiveness. Moreover, ethical considerations and patient perspectives are discussed, ensuring a comprehensive understanding of the subject. Towards revolutionizing treatment strategies and improving patient outcomes, the review concludes with future research directions. It also envisions integrating microbiome and metabolite research into personalized breast cancer therapy.</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/PMC11545256/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615756","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}
Background: The KRAS G12C mutation, prevalent in various malignancies, including non-small cell lung cancer (NSCLC), represents a unique therapeutic target. Adagrasib and sotorasib, two FDA-approved agents specifically targeting this mutation, have shown promise in clinical trials. This study aims to compare their efficacy in treating KRAS G12C-mutated NSCLC, drawing insights from pivotal clinical trials. Methods: We analyzed data from three key clinical trials: KRYSTAL-1, CodeBreak100, and CodeBreak200. Our methodology involved reconstructing individual patient data from published Kaplan-Meier curves using the IPDfromKM tool (Version 0.1.10). The primary endpoints were progression-free survival (PFS) and overall survival (OS), evaluated through hazard ratios (HRs) and the restricted mean survival time (RMST) method. Results: The HR for PFS favored adagrasib (HR: 0.90 [95% CI: 0.69, 1.19], p = 0.473), suggesting a non-significant trend toward better disease control compared to sotorasib. For OS, the HR was 0.99 [95% CI: 0.75, 1.33] (p = 0.969), indicating no significant difference between the two drugs. RMST analysis supported these findings, with adagrasib showing a consistently higher RMST in PFS at 6, 12, and 18 months. However, OS benefits converged over time, with adagrasib marginally surpassing sotorasib by the 18-month mark. Conclusions: This comprehensive analysis reveals that while adagrasib may offer a slight advantage in PFS, both drugs demonstrate comparable efficacy in OS for KRAS G12C-mutated NSCLC. The subtle differences observed, particularly in PFS, could inform clinical decision-making, emphasizing the need for personalized treatment strategies. Future research should focus on long-term effects and identifying patient subgroups that may benefit more from one drug over the other.
{"title":"Comparative Efficacy of Adagrasib and Sotorasib in KRAS G12C-Mutant NSCLC: Insights from Pivotal Trials.","authors":"Tzu-Rong Peng, Ta-Wei Wu, Tai-Yung Yi, An-Jan Wu","doi":"10.3390/cancers16213676","DOIUrl":"10.3390/cancers16213676","url":null,"abstract":"<p><p><b>Background</b>: The KRAS G12C mutation, prevalent in various malignancies, including non-small cell lung cancer (NSCLC), represents a unique therapeutic target. Adagrasib and sotorasib, two FDA-approved agents specifically targeting this mutation, have shown promise in clinical trials. This study aims to compare their efficacy in treating KRAS G12C-mutated NSCLC, drawing insights from pivotal clinical trials. <b>Methods</b>: We analyzed data from three key clinical trials: KRYSTAL-1, CodeBreak100, and CodeBreak200. Our methodology involved reconstructing individual patient data from published Kaplan-Meier curves using the IPDfromKM tool (Version 0.1.10). The primary endpoints were progression-free survival (PFS) and overall survival (OS), evaluated through hazard ratios (HRs) and the restricted mean survival time (RMST) method. <b>Results</b>: The HR for PFS favored adagrasib (HR: 0.90 [95% CI: 0.69, 1.19], <i>p</i> = 0.473), suggesting a non-significant trend toward better disease control compared to sotorasib. For OS, the HR was 0.99 [95% CI: 0.75, 1.33] (<i>p</i> = 0.969), indicating no significant difference between the two drugs. RMST analysis supported these findings, with adagrasib showing a consistently higher RMST in PFS at 6, 12, and 18 months. However, OS benefits converged over time, with adagrasib marginally surpassing sotorasib by the 18-month mark. <b>Conclusions</b>: This comprehensive analysis reveals that while adagrasib may offer a slight advantage in PFS, both drugs demonstrate comparable efficacy in OS for KRAS G12C-mutated NSCLC. The subtle differences observed, particularly in PFS, could inform clinical decision-making, emphasizing the need for personalized treatment strategies. Future research should focus on long-term effects and identifying patient subgroups that may benefit more from one drug over the other.</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/PMC11545475/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615550","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}
Abdulrahim Saleh Alrasheed, Abdulsalam Mohammed Aleid, Reema Ahmed Alharbi, Mostafa Habeeb Alhodibi, Abdulmonem Ali Alhussain, Awn Abdulmohsen Alessa, Sami Fadhel Almalki
Background: Proton beam radiation therapy (PBRT) is an advanced cancer treatment modality that utilizes the distinctive physical properties of protons to precisely deliver radiation to tumor targets while sparing healthy tissue. This cannot be obtained with photon radiation. In this systematic review and meta-analysis, we aimed to comprehensively assess the risk of brainstem toxicity in pediatric brain tumor patients undergoing PBRT. Methods: With adherence to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, a predetermined search strategy was used to identify eligible articles from PubMed, Web of Science, Scopus, and Cochrane Library through July 2024. Results: The current study included a total of 11 eligible articles. The pooled prevalence of patients who suffered from brainstem toxicity was 1.8% (95% CI: 1%, 2.6%). The pooled prevalences of patients with Grade 1 to Grade 5 brainstem toxicity were found to be 10.6% (95% CI: 8.8%, 30%), 1.5% (95% CI: 0.6%, 2.5%), 0.7% (95% CI: 0.3%, 1.1%), 0.4% (95% CI: 0.1%, 0.7%), and 0.4% (95% CI: 0.1%, 0.8%), respectively, with an overall pooled prevalence of 0.7% (95% CI: 0.4%, 1%). Conclusions: This study revealed a relatively low incidence of symptomatic brainstem toxicity and its related mortality in the pediatric population undergoing PBRT. However, further research is encouraged to study the broader effects of PBRT and to explore various factors that may influence the risk of brainstem toxicity in patients treated with PBRT.
{"title":"Brainstem Toxicity Following Proton Beam Radiation Therapy in Pediatric Brain Tumors: A Systematic Review and Meta-Analysis.","authors":"Abdulrahim Saleh Alrasheed, Abdulsalam Mohammed Aleid, Reema Ahmed Alharbi, Mostafa Habeeb Alhodibi, Abdulmonem Ali Alhussain, Awn Abdulmohsen Alessa, Sami Fadhel Almalki","doi":"10.3390/cancers16213655","DOIUrl":"10.3390/cancers16213655","url":null,"abstract":"<p><p><b>Background:</b> Proton beam radiation therapy (PBRT) is an advanced cancer treatment modality that utilizes the distinctive physical properties of protons to precisely deliver radiation to tumor targets while sparing healthy tissue. This cannot be obtained with photon radiation. In this systematic review and meta-analysis, we aimed to comprehensively assess the risk of brainstem toxicity in pediatric brain tumor patients undergoing PBRT. <b>Methods:</b> With adherence to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, a predetermined search strategy was used to identify eligible articles from PubMed, Web of Science, Scopus, and Cochrane Library through July 2024. <b>Results:</b> The current study included a total of 11 eligible articles. The pooled prevalence of patients who suffered from brainstem toxicity was 1.8% (95% CI: 1%, 2.6%). The pooled prevalences of patients with Grade 1 to Grade 5 brainstem toxicity were found to be 10.6% (95% CI: 8.8%, 30%), 1.5% (95% CI: 0.6%, 2.5%), 0.7% (95% CI: 0.3%, 1.1%), 0.4% (95% CI: 0.1%, 0.7%), and 0.4% (95% CI: 0.1%, 0.8%), respectively, with an overall pooled prevalence of 0.7% (95% CI: 0.4%, 1%). <b>Conclusions:</b> This study revealed a relatively low incidence of symptomatic brainstem toxicity and its related mortality in the pediatric population undergoing PBRT. However, further research is encouraged to study the broader effects of PBRT and to explore various factors that may influence the risk of brainstem toxicity in patients treated with PBRT.</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/PMC11545066/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615472","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}
Background: Breast cancer (BC) is the most common cancer among women in Palestine, where the need for supportive care frequently goes unmet. Therefore, this study aims to assess the supportive care services provided at the governmental hospitals in the southern area of the West Bank and to determine the factors associated with the unmet needs of these services. Methods: A cross-sectional study was conducted on 362 women with BC. Data were collected using a face-to-face questionnaire that included the Supportive Care Needs Survey (SCNS-SF34), patients' sociodemographic, economic, and clinical characteristics, as well as familial history of cancer and social support. Results: The study revealed that 61% of participants had unmet supportive care needs, with health system information, physical support, and psychological support being the most unmet needs. Factors contributing to unmet needs included age, marital status, familial support, and a family history of cancer. Chemotherapy and surgery increased the probability of physical care needs by fivefold, while hormone therapy reduced the probability of psychological needs (AOR = 0.36, p < 0.001) and patient care and support needs (AOR = 0.49, p = 0.01). Additionally, radiotherapy reduced sexual care needs by 58% and biological therapy by 60%. Conclusions: There is an urgent need for enhanced supportive care services for BC patients in the West Bank, especially regarding health system information, physical care, and psychological support. Addressing these needs through targeted interventions could significantly improve patients' quality of life.
背景:乳腺癌(BC)是巴勒斯坦妇女中最常见的癌症,其支持性护理需求经常得不到满足。因此,本研究旨在评估约旦河西岸南部地区政府医院提供的支持性护理服务,并确定未满足这些服务需求的相关因素。研究方法对 362 名患有 BC 的妇女进行了横断面研究。研究采用面对面问卷调查的方式收集数据,其中包括支持性护理需求调查(SCNS-SF34)、患者的社会人口、经济和临床特征以及家族癌症史和社会支持。结果显示研究显示,61%的参与者的支持性护理需求未得到满足,其中医疗系统信息、身体支持和心理支持是未满足需求最多的方面。导致需求未得到满足的因素包括年龄、婚姻状况、家庭支持和癌症家族史。化疗和手术将身体护理需求的概率提高了五倍,而激素治疗则降低了心理需求(AOR = 0.36,p < 0.001)和患者护理与支持需求(AOR = 0.49,p = 0.01)的概率。此外,放射治疗将性保健需求降低了 58%,生物治疗降低了 60%。结论约旦河西岸的 BC 患者急需加强支持性护理服务,尤其是在卫生系统信息、身体护理和心理支持方面。通过有针对性的干预措施来满足这些需求,可显著改善患者的生活质量。
{"title":"Factors Associated with Supportive Care Needs Among Palestinian Women with Breast Cancer in the West Bank: A Cross-Sectional Study.","authors":"Ibtisam Titi, Nuha El Sharif","doi":"10.3390/cancers16213663","DOIUrl":"10.3390/cancers16213663","url":null,"abstract":"<p><p><b>Background</b>: Breast cancer (BC) is the most common cancer among women in Palestine, where the need for supportive care frequently goes unmet. Therefore, this study aims to assess the supportive care services provided at the governmental hospitals in the southern area of the West Bank and to determine the factors associated with the unmet needs of these services. <b>Methods</b>: A cross-sectional study was conducted on 362 women with BC. Data were collected using a face-to-face questionnaire that included the Supportive Care Needs Survey (SCNS-SF34), patients' sociodemographic, economic, and clinical characteristics, as well as familial history of cancer and social support. <b>Results</b>: The study revealed that 61% of participants had unmet supportive care needs, with health system information, physical support, and psychological support being the most unmet needs. Factors contributing to unmet needs included age, marital status, familial support, and a family history of cancer. Chemotherapy and surgery increased the probability of physical care needs by fivefold, while hormone therapy reduced the probability of psychological needs (AOR = 0.36, <i>p</i> < 0.001) and patient care and support needs (AOR = 0.49, <i>p</i> = 0.01). Additionally, radiotherapy reduced sexual care needs by 58% and biological therapy by 60%. <b>Conclusions</b>: There is an urgent need for enhanced supportive care services for BC patients in the West Bank, especially regarding health system information, physical care, and psychological support. Addressing these needs through targeted interventions could significantly improve patients' quality of life.</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/PMC11545701/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615666","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}
Haizhen Du, Lijie Hou, Huan Yu, Fenghao Zhang, Ke Tong, Xiaowen Wu, Ziyi Zhang, Kaiping Liu, Xiangguang Miao, Wenhui Guo, Jun Guo, Yan Kong
Background: Mucosal melanoma (MM) is epidemiologically, biologically, and molecularly distinct from cutaneous melanoma. Current treatment strategies have failed to significantly improve the prognosis for MM patients. This study aims to identify therapeutic targets and develop combination strategies by investigating the mechanisms underlying the tumorigenesis and progression of MM.
Methods: We analyzed the copy number amplification of enhancer of zeste homolog 2 (EZH2) in 547 melanoma patients and investigated its correlation with clinical prognosis. Utilizing cell lines, organoids, and patient-derived xenograft models, we assessed the impact of EZH2 on cell proliferation and sensitivity to ferroptosis. Further, we explored the mechanisms of ferroptosis resistance associated with EZH2 by conducting RNA sequencing and chromatin immunoprecipitation sequencing.
Results: EZH2 copy number amplification was closely associated with malignant phenotype and poor prognosis in MM patients. EZH2 was essential for MM cell proliferation in vitro and in vivo. Moreover, genetic perturbation of EZH2 rendered MM cells sensitized to ferroptosis. Combination treatment of EZH2 inhibitor with ferroptosis inducer significantly inhibited the growth of MM. Mechanistically, EZH2 inhibited the expression of Krüpple-Like factor 14 (KLF14), which binds to the promoter of solute carrier family 7 member 11 (SLC7A11) to repress its transcription. Loss of EZH2 therefore reduced the expression of SLC7A11, leading to reduced intracellular SLC7A11-dependent glutathione synthesis to promote ferroptosis.
Conclusion: Our findings not only establish EZH2 as a biomarker for MM prognosis but also highlight the EZH2-KLF14-SLC7A11 axis as a potential target for MM treatment.
背景:粘膜黑色素瘤(MM)在流行病学、生物学和分子学上与皮肤黑色素瘤截然不同。目前的治疗策略未能明显改善粘膜黑色素瘤患者的预后。本研究旨在通过研究MM肿瘤发生和发展的内在机制,确定治疗靶点并制定联合治疗策略:方法:我们分析了547例黑色素瘤患者的泽斯特同源增强子2(EZH2)拷贝数扩增情况,并研究了其与临床预后的相关性。我们利用细胞系、器官组织和患者来源的异种移植模型,评估了EZH2对细胞增殖和铁中毒敏感性的影响。此外,我们还通过进行RNA测序和染色质免疫沉淀测序,探索了与EZH2相关的铁中毒抗性机制:结果:EZH2拷贝数扩增与MM患者的恶性表型和不良预后密切相关。EZH2对MM细胞的体外和体内增殖至关重要。此外,对EZH2进行遗传扰乱可使MM细胞对铁变态反应敏感。EZH2抑制剂与铁变态反应诱导剂联合治疗可显著抑制MM的生长。从机制上讲,EZH2抑制了Krüpple-Like因子14(KLF14)的表达,KLF14与溶质运载家族7成员11(SLC7A11)的启动子结合,抑制其转录。因此,EZH2的缺失会降低SLC7A11的表达,导致细胞内SLC7A11依赖的谷胱甘肽合成减少,从而促进铁变态反应:我们的研究结果不仅确定了 EZH2 是 MM 预后的生物标志物,还强调了 EZH2-KLF14-SLC7A11 轴是 MM 治疗的潜在靶点。
{"title":"Enhancer of Zeste Homolog 2 Protects Mucosal Melanoma from Ferroptosis via the KLF14-SLC7A11 Signaling Pathway.","authors":"Haizhen Du, Lijie Hou, Huan Yu, Fenghao Zhang, Ke Tong, Xiaowen Wu, Ziyi Zhang, Kaiping Liu, Xiangguang Miao, Wenhui Guo, Jun Guo, Yan Kong","doi":"10.3390/cancers16213660","DOIUrl":"10.3390/cancers16213660","url":null,"abstract":"<p><strong>Background: </strong>Mucosal melanoma (MM) is epidemiologically, biologically, and molecularly distinct from cutaneous melanoma. Current treatment strategies have failed to significantly improve the prognosis for MM patients. This study aims to identify therapeutic targets and develop combination strategies by investigating the mechanisms underlying the tumorigenesis and progression of MM.</p><p><strong>Methods: </strong>We analyzed the copy number amplification of enhancer of zeste homolog 2 (<i>EZH2</i>) in 547 melanoma patients and investigated its correlation with clinical prognosis. Utilizing cell lines, organoids, and patient-derived xenograft models, we assessed the impact of <i>EZH2</i> on cell proliferation and sensitivity to ferroptosis. Further, we explored the mechanisms of ferroptosis resistance associated with <i>EZH2</i> by conducting RNA sequencing and chromatin immunoprecipitation sequencing.</p><p><strong>Results: </strong><i>EZH2</i> copy number amplification was closely associated with malignant phenotype and poor prognosis in MM patients. <i>EZH2</i> was essential for MM cell proliferation in vitro and in vivo. Moreover, genetic perturbation of <i>EZH2</i> rendered MM cells sensitized to ferroptosis. Combination treatment of <i>EZH2</i> inhibitor with ferroptosis inducer significantly inhibited the growth of MM. Mechanistically, <i>EZH2</i> inhibited the expression of Krüpple-Like factor 14 (<i>KLF14</i>), which binds to the promoter of solute carrier family 7 member 11 (<i>SLC7A11</i>) to repress its transcription. Loss of <i>EZH2</i> therefore reduced the expression of <i>SLC7A11</i>, leading to reduced intracellular <i>SLC7A11</i>-dependent glutathione synthesis to promote ferroptosis.</p><p><strong>Conclusion: </strong>Our findings not only establish <i>EZH2</i> as a biomarker for MM prognosis but also highlight the <i>EZH2-KLF14-SLC7A11</i> axis as a potential target for MM treatment.</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/PMC11545276/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615593","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}
Muhammad Awais, Mais Al Taie, Caleb S O'Connor, Austin H Castelo, Belkacem Acidi, Hop S Tran Cao, Kristy K Brock
Background/objectives: In the field of surgical medicine, the planning and execution of liver resection procedures present formidable challenges, primarily attributable to the intricate and highly individualized nature of liver vascular anatomy. In the current surgical milieu, intraoperative ultrasonography (IOUS) has become indispensable; however, traditional 2D ultrasound imaging's interpretability is hindered by noise and speckle artifacts. Accurate identification of critical structures for preservation during hepatectomy requires advanced surgical skills.
Methods: An AI-based model that can help detect and recognize vessels including the inferior vena cava (IVC); the right (RHV), middle (MHV), and left (LVH) hepatic veins; the portal vein (PV) and its major first and second order branches the left portal vein (LPV), right portal vein (RPV), and right anterior (RAPV) and posterior (RPPV) portal veins, for real-time IOUS navigation can be of immense value in liver surgery. This research aims to advance the capabilities of IOUS-guided interventions by applying an innovative AI-based approach named the "2D-weigthed U-Net model" for the segmentation of multiple blood vessels in real-time IOUS video frames.
Results: Our proposed deep learning (DL) model achieved a mean Dice score of 0.92 for IVC, 0.90 for RHV, 0.89 for MHV, 0.86 for LHV, 0.95 for PV, 0.93 for LPV, 0.84 for RPV, 0.85 for RAPV, and 0.96 for RPPV.
Conclusion: In the future, this research will be extended for real-time multi-label segmentation of extended vasculature in the liver, followed by the translation of our model into the surgical suite.
{"title":"Enhancing Surgical Guidance: Deep Learning-Based Liver Vessel Segmentation in Real-Time Ultrasound Video Frames.","authors":"Muhammad Awais, Mais Al Taie, Caleb S O'Connor, Austin H Castelo, Belkacem Acidi, Hop S Tran Cao, Kristy K Brock","doi":"10.3390/cancers16213674","DOIUrl":"10.3390/cancers16213674","url":null,"abstract":"<p><strong>Background/objectives: </strong>In the field of surgical medicine, the planning and execution of liver resection procedures present formidable challenges, primarily attributable to the intricate and highly individualized nature of liver vascular anatomy. In the current surgical milieu, intraoperative ultrasonography (IOUS) has become indispensable; however, traditional 2D ultrasound imaging's interpretability is hindered by noise and speckle artifacts. Accurate identification of critical structures for preservation during hepatectomy requires advanced surgical skills.</p><p><strong>Methods: </strong>An AI-based model that can help detect and recognize vessels including the inferior vena cava (IVC); the right (RHV), middle (MHV), and left (LVH) hepatic veins; the portal vein (PV) and its major first and second order branches the left portal vein (LPV), right portal vein (RPV), and right anterior (RAPV) and posterior (RPPV) portal veins, for real-time IOUS navigation can be of immense value in liver surgery. This research aims to advance the capabilities of IOUS-guided interventions by applying an innovative AI-based approach named the \"2D-weigthed U-Net model\" for the segmentation of multiple blood vessels in real-time IOUS video frames.</p><p><strong>Results: </strong>Our proposed deep learning (DL) model achieved a mean Dice score of 0.92 for IVC, 0.90 for RHV, 0.89 for MHV, 0.86 for LHV, 0.95 for PV, 0.93 for LPV, 0.84 for RPV, 0.85 for RAPV, and 0.96 for RPPV.</p><p><strong>Conclusion: </strong>In the future, this research will be extended for real-time multi-label segmentation of extended vasculature in the liver, followed by the translation of our model into the surgical suite.</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/PMC11545685/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615595","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}
Marwan-Julien Sleiman, Annamaria Jelip, Nicolas Buchs, Christian Toso, Emilie Liot, Thibaud Koessler, Jeremy Meyer, Guillaume Meurette, Frederic Ris
Introduction: Pressurized intraperitoneal aerosol chemotherapy (PIPAC) consists of the administration of aerosolized chemotherapy into the abdominal cavity of patients suffering from peritoneal carcinomatosis. Our aim was to review the evidence supporting PIPAC in patients with peritoneal carcinomatosis from colorectal cancer.
Methods: A systematic review was performed in accordance with the 2020 PRISMA guideline. MEDLINE and CENTRAL were searched using combinations of terms including "Peritoneal carcinomatosis", "Peritoneal metastasis", "PIPAC", "Pressurized intraperitoneal aerosol chemotherapy" and "Colorectal cancer". Original studies, in English, including patients treated with PIPAC for colorectal peritoneal carcinomatosis, were considered eligible. Case reports, non-English or French language articles and secondary analyses were excluded.
Results: A total of 385 articles were screened and 374 articles were excluded, leaving 11 publications for inclusion in the qualitative analysis. The included studies totalized 949 patients who received PIPAC for peritoneal carcinomatosis due to colorectal cancer. The median peritoneal carcinomatosis index (PCI) ranged from 10 to 31. In all studies, the complete PIPAC protocol was achieved with an average of two to three 3 PIPAC sessions per patient. Oxaliplatin (OX) was used as a chemotherapeutic agent in all studies and could be associated with intravenous 5-FU and leucovorin. Most post-operative adverse events were recorded as mild to moderate with no intraoperative complications. Only four studies reported a decrease in the average PCI score for 50% of the patients. Median overall survival ranged from 8 to 37.8 months. Quality of life indicators were stable between PIPAC-OX cycles with a small but not statistically significant trend of improvement of most functional scales.
Conclusions: PIPAC for peritoneal carcinomatosis from colorectal origin is feasible, safe and tolerable. Its impact on survival outcomes or quality of life remains to be demonstrated by randomized trials.
{"title":"Pressurized Intraperitoneal Aerosol Chemotherapy for Peritoneal Carcinomatosis in Colorectal Cancer Patients: A Systematic Review of the Evidence.","authors":"Marwan-Julien Sleiman, Annamaria Jelip, Nicolas Buchs, Christian Toso, Emilie Liot, Thibaud Koessler, Jeremy Meyer, Guillaume Meurette, Frederic Ris","doi":"10.3390/cancers16213661","DOIUrl":"10.3390/cancers16213661","url":null,"abstract":"<p><strong>Introduction: </strong>Pressurized intraperitoneal aerosol chemotherapy (PIPAC) consists of the administration of aerosolized chemotherapy into the abdominal cavity of patients suffering from peritoneal carcinomatosis. Our aim was to review the evidence supporting PIPAC in patients with peritoneal carcinomatosis from colorectal cancer.</p><p><strong>Methods: </strong>A systematic review was performed in accordance with the 2020 PRISMA guideline. MEDLINE and CENTRAL were searched using combinations of terms including \"Peritoneal carcinomatosis\", \"Peritoneal metastasis\", \"PIPAC\", \"Pressurized intraperitoneal aerosol chemotherapy\" and \"Colorectal cancer\". Original studies, in English, including patients treated with PIPAC for colorectal peritoneal carcinomatosis, were considered eligible. Case reports, non-English or French language articles and secondary analyses were excluded.</p><p><strong>Results: </strong>A total of 385 articles were screened and 374 articles were excluded, leaving 11 publications for inclusion in the qualitative analysis. The included studies totalized 949 patients who received PIPAC for peritoneal carcinomatosis due to colorectal cancer. The median peritoneal carcinomatosis index (PCI) ranged from 10 to 31. In all studies, the complete PIPAC protocol was achieved with an average of two to three 3 PIPAC sessions per patient. Oxaliplatin (OX) was used as a chemotherapeutic agent in all studies and could be associated with intravenous 5-FU and leucovorin. Most post-operative adverse events were recorded as mild to moderate with no intraoperative complications. Only four studies reported a decrease in the average PCI score for 50% of the patients. Median overall survival ranged from 8 to 37.8 months. Quality of life indicators were stable between PIPAC-OX cycles with a small but not statistically significant trend of improvement of most functional scales.</p><p><strong>Conclusions: </strong>PIPAC for peritoneal carcinomatosis from colorectal origin is feasible, safe and tolerable. Its impact on survival outcomes or quality of life remains to be demonstrated by randomized trials.</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/PMC11544814/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615614","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}