Pub Date : 2025-10-24DOI: 10.1177/10849785251388809
Jun Fang, Ningjie Li, Hongbo Li, Mumo Wang, Li Wang
Malignant brain tumors remain a major therapeutic challenge due to poor intracellular delivery of therapeutics. Radiopharmaceuticals such as Technetium-99m (^99mTc) are valuable for imaging and therapy but suffer from limited tumor uptake caused by cellular and membrane barriers. Focused ultrasound (FUS) offers a noninvasive strategy to transiently enhance membrane permeability through sonoporation. Unlike prior studies largely focused on blood-brain barrier disruption, this work specifically investigates direct tumor cell sonoporation as an independent uptake mechanism. This study evaluates FUS-mediated enhancement of ^99mTc radiopharmaceutical uptake in brain tumor cells and determines optimal acoustic parameters balancing efficacy and safety. Human glioblastoma (U87-MG) and astrocytoma (A172) cells were cultured and exposed to FUS at intensities of 0.3, 0.5, and 0.7 W/cm2 for 30-120 s. Radiopharmaceutical uptake was quantified using γ-scintillation counting. Membrane integrity was assessed by live/dead fluorescence microscopy and lactate dehydrogenase release, while cell viability was evaluated via medical training therapy (MTT) assays. U87-MG cells exhibited up to a 3.1-fold increase at 0.7 W/cm2 for 120 s, with a 2.3-fold enhancement at the clinically relevant 0.5 W/cm2 for 60 s while maintaining >92% viability. A172 cells showed similar trends with slightly lower magnitudes. Safety assays confirmed reversible membrane permeabilization at ≤0.5 W/cm2. The temporal uptake kinetics aligned with established membrane pore resealing dynamics, supporting reversible sonoporation as the uptake mechanism. Importantly, while ^99mTc complexes are primarily diagnostic, enhanced intracellular delivery achieved by optimized FUS may also support future theranostic strategies, including radionuclide therapy. These findings underscore the translational potential of FUS in neuro-oncology, where tumor heterogeneity necessitates parameter optimization to maximize radiopharmaceutical delivery, improve imaging contrast, and overcome therapeutic resistance.
{"title":"<i>In Vitro</i> Assessment of Radiopharmaceutical Uptake in Brain Tumor Cells Using Focused Ultrasound Stimulation.","authors":"Jun Fang, Ningjie Li, Hongbo Li, Mumo Wang, Li Wang","doi":"10.1177/10849785251388809","DOIUrl":"https://doi.org/10.1177/10849785251388809","url":null,"abstract":"<p><p>Malignant brain tumors remain a major therapeutic challenge due to poor intracellular delivery of therapeutics. Radiopharmaceuticals such as Technetium-99m (^<sup>99m</sup>Tc) are valuable for imaging and therapy but suffer from limited tumor uptake caused by cellular and membrane barriers. Focused ultrasound (FUS) offers a noninvasive strategy to transiently enhance membrane permeability through sonoporation. Unlike prior studies largely focused on blood-brain barrier disruption, this work specifically investigates direct tumor cell sonoporation as an independent uptake mechanism. This study evaluates FUS-mediated enhancement of ^<sup>99m</sup>Tc radiopharmaceutical uptake in brain tumor cells and determines optimal acoustic parameters balancing efficacy and safety. Human glioblastoma (U87-MG) and astrocytoma (A172) cells were cultured and exposed to FUS at intensities of 0.3, 0.5, and 0.7 W/cm<sup>2</sup> for 30-120 s. Radiopharmaceutical uptake was quantified using γ-scintillation counting. Membrane integrity was assessed by live/dead fluorescence microscopy and lactate dehydrogenase release, while cell viability was evaluated via medical training therapy (MTT) assays. U87-MG cells exhibited up to a 3.1-fold increase at 0.7 W/cm<sup>2</sup> for 120 s, with a 2.3-fold enhancement at the clinically relevant 0.5 W/cm<sup>2</sup> for 60 s while maintaining >92% viability. A172 cells showed similar trends with slightly lower magnitudes. Safety assays confirmed reversible membrane permeabilization at ≤0.5 W/cm<sup>2</sup>. The temporal uptake kinetics aligned with established membrane pore resealing dynamics, supporting reversible sonoporation as the uptake mechanism. Importantly, while ^<sup>99m</sup>Tc complexes are primarily diagnostic, enhanced intracellular delivery achieved by optimized FUS may also support future theranostic strategies, including radionuclide therapy. These findings underscore the translational potential of FUS in neuro-oncology, where tumor heterogeneity necessitates parameter optimization to maximize radiopharmaceutical delivery, improve imaging contrast, and overcome therapeutic resistance.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.1,"publicationDate":"2025-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145356840","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-24DOI: 10.1177/10849785251389350
Omer Akay, Mert Guler, Husnu Ozan Sevik, Anil Demir, Sener Simsek, Mahmut Emin Cicek, Furkan Turkoglu, Ufuk Oguz Idiz, Cihad Tatar
Background: Response to neoadjuvant chemotherapy (NACT) in locally advanced gastric cancer varies. This study compares tumor response to NACT across anatomical locations, considering clinicopathological differences. Materials and Methods: This retrospective study included 212 patients with gastric adenocarcinoma who received NACT followed by surgery. Tumors were classified by location (antrum-pylorus, corpus-fundus, cardia). Treatment response was assessed using the Modified Ryan Scoring System (0 = complete, 1 = near complete, 2 = partial, 3 = minimal/none). Results: Tumor locations were antrum-pylorus (30.2%), corpus-fundus (28.3%), and cardia (41.5%). Localization showed no statistically significant differences in response (p = 0.337). However, cardia tumors were more frequent in Groups 3 (40.9%) and 4 (48.1%), which showed poorer pathological responses, whereas antrum (34.6%) and corpus (38.5%) tumors were more common in Group 1, representing patients with a pathological complete response. These findings suggest that cardia tumors may have a lower response to NACT, although definitive conclusions cannot be drawn. In multivariate analysis, only advanced T stage (T3-4) was independently associated with poor tumor regression grade response (odds ratio 14.3, 95% confidence interval 5.4-37.5, p < 0.001). Conclusions: Tumor response to NACT varied by anatomical location, although differences were not statistically significant. Cardia tumors showed a trend toward lower response rates. To the authors' knowledge, this is the first study evaluating gastric anatomical subgroups in this context. While not conclusive, the findings suggest that tumor location may influence treatment strategies, warranting validation in larger studies.
{"title":"The Impact of Gastric Anatomical Subdivisions on the Response to Neoadjuvant Chemotherapy in Gastric Cancer.","authors":"Omer Akay, Mert Guler, Husnu Ozan Sevik, Anil Demir, Sener Simsek, Mahmut Emin Cicek, Furkan Turkoglu, Ufuk Oguz Idiz, Cihad Tatar","doi":"10.1177/10849785251389350","DOIUrl":"https://doi.org/10.1177/10849785251389350","url":null,"abstract":"<p><p><b><i>Background:</i></b> Response to neoadjuvant chemotherapy (NACT) in locally advanced gastric cancer varies. This study compares tumor response to NACT across anatomical locations, considering clinicopathological differences. <b><i>Materials and Methods:</i></b> This retrospective study included 212 patients with gastric adenocarcinoma who received NACT followed by surgery. Tumors were classified by location (antrum-pylorus, corpus-fundus, cardia). Treatment response was assessed using the Modified Ryan Scoring System (0 = complete, 1 = near complete, 2 = partial, 3 = minimal/none). <b><i>Results:</i></b> Tumor locations were antrum-pylorus (30.2%), corpus-fundus (28.3%), and cardia (41.5%). Localization showed no statistically significant differences in response (<i>p</i> = 0.337). However, cardia tumors were more frequent in Groups 3 (40.9%) and 4 (48.1%), which showed poorer pathological responses, whereas antrum (34.6%) and corpus (38.5%) tumors were more common in Group 1, representing patients with a pathological complete response. These findings suggest that cardia tumors may have a lower response to NACT, although definitive conclusions cannot be drawn. In multivariate analysis, only advanced T stage (T3-4) was independently associated with poor tumor regression grade response (odds ratio 14.3, 95% confidence interval 5.4-37.5, <i>p</i> < 0.001). <b><i>Conclusions:</i></b> Tumor response to NACT varied by anatomical location, although differences were not statistically significant. Cardia tumors showed a trend toward lower response rates. To the authors' knowledge, this is the first study evaluating gastric anatomical subgroups in this context. While not conclusive, the findings suggest that tumor location may influence treatment strategies, warranting validation in larger studies.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.1,"publicationDate":"2025-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145369176","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-24DOI: 10.1177/10849785251388246
Yingying Ye
Ocular malignancies provide a unique therapeutic challenge because of their anatomical intricacy, limited accessibility, and vision-critical nature. Recent developments in radiopharmaceutical design have been paired with ultrasound-mediated medicine administration to create highly targeted, less invasive therapies for intraocular cancers. This research looks at the emerging topic of ultrasound-responsive radiopharmaceutical devices built specifically for ocular oncology. These methods enhance tumor selectivity, decrease off-target effects, and enable real-time imaging-guided therapy by utilizing targeted ultrasound to induce localized medication release or radiotherapeutic agent activation. Microbubble-assisted delivery, thermosensitive liposomes, and phase-transition nanodroplets carrying radionuclides have all been designed to optimize ocular pharmacokinetics and tissue penetration. Preclinical studies reveal promising results in increasing radiotherapeutic efficacy against retinoblastoma and uveal melanoma while sparing healthy ocular tissues.
{"title":"Advances in Ultrasound-Responsive Radiopharmaceutical Systems for the Management of Ocular Malignancies.","authors":"Yingying Ye","doi":"10.1177/10849785251388246","DOIUrl":"https://doi.org/10.1177/10849785251388246","url":null,"abstract":"<p><p>Ocular malignancies provide a unique therapeutic challenge because of their anatomical intricacy, limited accessibility, and vision-critical nature. Recent developments in radiopharmaceutical design have been paired with ultrasound-mediated medicine administration to create highly targeted, less invasive therapies for intraocular cancers. This research looks at the emerging topic of ultrasound-responsive radiopharmaceutical devices built specifically for ocular oncology. These methods enhance tumor selectivity, decrease off-target effects, and enable real-time imaging-guided therapy by utilizing targeted ultrasound to induce localized medication release or radiotherapeutic agent activation. Microbubble-assisted delivery, thermosensitive liposomes, and phase-transition nanodroplets carrying radionuclides have all been designed to optimize ocular pharmacokinetics and tissue penetration. Preclinical studies reveal promising results in increasing radiotherapeutic efficacy against retinoblastoma and uveal melanoma while sparing healthy ocular tissues.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.1,"publicationDate":"2025-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145356866","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Recent research has significantly altered the understanding of the immunogenic profile of certain processes of cancer cell death, leading to the recognition of a new subclass of apoptosis called "immunogenic apoptosis." This form of cell death, induced by specific chemotherapeutic agents, has been shown to elicit a "chemotherapy vaccine effect" in vivo, effectively stimulating an antitumor immune response. At the molecular level, "a collection of molecules" known as "damage-associated molecular patterns (DAMPs)" have been identified as key contributors to the immunogenicity of various cell death pathways. Intracellular molecules, such as heat-shock proteins, high-mobility group box 1 protein, and calreticulin, act as DAMPs when exposed or secreted in response to specific certain stressors, stimuli, and modes of cell death. These discoveries have fueled ongoing research focused on the identification of novel DAMPs, uncovering new mechanisms of their exposure or secretion and developing therapeutic agents capable of inducing immunogenic cell death (ICD). In addition, there is growing interest in addressing the current challenges and limitations within this emerging paradigm. The authors believe that this integrated strategy-combining DAMPs, ICD, and anticancer therapies-may hold the key to significantly reducing cancer-related mortality in the near future.
{"title":"Comprehensive Review on How Repurposed Drugs Modulate Antitumor Immunity: Harnessing Damage-Associated Molecular Patterns.","authors":"Rahaman Shaik, Maheshwari Sappidi, Deepika Vemula, Alekhya Munjala, Fatima Sarwar Syeda, Shireen Begum, Huda Khan, Kiranmai Mandava","doi":"10.1177/10849785251389355","DOIUrl":"https://doi.org/10.1177/10849785251389355","url":null,"abstract":"<p><p>Recent research has significantly altered the understanding of the immunogenic profile of certain processes of cancer cell death, leading to the recognition of a new subclass of apoptosis called \"immunogenic apoptosis.\" This form of cell death, induced by specific chemotherapeutic agents, has been shown to elicit a \"chemotherapy vaccine effect\" <i>in vivo</i>, effectively stimulating an antitumor immune response. At the molecular level, \"a collection of molecules\" known as \"damage-associated molecular patterns (DAMPs)\" have been identified as key contributors to the immunogenicity of various cell death pathways. Intracellular molecules, such as heat-shock proteins, high-mobility group box 1 protein, and calreticulin, act as DAMPs when exposed or secreted in response to specific certain stressors, stimuli, and modes of cell death. These discoveries have fueled ongoing research focused on the identification of novel DAMPs, uncovering new mechanisms of their exposure or secretion and developing therapeutic agents capable of inducing immunogenic cell death (ICD). In addition, there is growing interest in addressing the current challenges and limitations within this emerging paradigm. The authors believe that this integrated strategy-combining DAMPs, ICD, and anticancer therapies-may hold the key to significantly reducing cancer-related mortality in the near future.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.1,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145338214","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-08DOI: 10.1177/10849785251376447
Baogang Shi, Xiaobo Xu, Xiaofeng Xia, Quanyong Wang
To properly target tumors during preoperative chemoradiotherapy, differentiated thyroid carcinoma (DTC) must be careful. This method increases treatment success and decreases recurrence. Ultrasound coupled with SPECT/CT may provide novel localization and dose planning possibilities. Many systems solely use anatomical or functional imaging. This may result in insufficient dosage delivery and wasted radiation exposure to healthy tissues. These issues are addressed by Dual-Modality Imaging-Guided Adaptive Chemoradiotherapy Planning (DMI-ACP). This innovative approach combines real-time ultrasound imaging with 6 Å SPECT/CT imaging for precise tumor delineation and tailored dosimetry. This system enables clinicians to adjust chemoradiotherapy regimens by seamlessly integrating functional iodine absorption data with anatomical characteristics, thereby targeting therapy to cancerous areas. The outcomes of this method for patients with DTC were promising, including better lesion targeting, reduced radiation exposure to healthy tissues, and improved chemotherapeutic dose distribution. In clinical evaluations, the DMI-ACP framework demonstrated a sensitivity of 94% and a specificity of 89% in identifying malignant lesions compared with traditional imaging techniques. Furthermore, the integration of adaptive planning resulted in a 20% improvement in tumor control probability and a 15% reduction in exposure to surrounding healthy tissue, as assessed through dosimetric analysis. 2023075 Nanjing Drum Tower Hospital Group Suqian Hospital/The Affiliated Suqian Hospital of Xuzhou Medical University.
术前放化疗时,分化型甲状腺癌(DTC)必须谨慎选择合适的放化疗目标。这种方法提高了治疗成功率,减少了复发。超声结合SPECT/CT可能提供新的定位和剂量计划的可能性。许多系统仅使用解剖或功能成像。这可能导致剂量输送不足和对健康组织的辐射暴露浪费。这些问题通过双模成像引导的适应性放化疗计划(DMI-ACP)来解决。这种创新的方法结合了实时超声成像与6 Å SPECT/CT成像精确肿瘤划定和量身定制的剂量。该系统使临床医生能够通过无缝整合功能性碘吸收数据和解剖学特征来调整放化疗方案,从而靶向治疗癌区。该方法用于DTC患者的结果是有希望的,包括更好的病灶靶向,减少对健康组织的辐射暴露,改善化疗剂量分布。在临床评估中,与传统成像技术相比,DMI-ACP框架在识别恶性病变方面的敏感性为94%,特异性为89%。此外,通过剂量学分析评估,适应性规划的整合导致肿瘤控制概率提高20%,对周围健康组织的暴露减少15%。2023075南京鼓楼医院集团宿迁医院/徐州医科大学宿迁附属医院
{"title":"Ultrasound-Guided Optimization of Preoperative Chemoradiotherapy Using ¹³¹I SPECT/CT for Enhanced Targeting in Differentiated Thyroid Carcinoma.","authors":"Baogang Shi, Xiaobo Xu, Xiaofeng Xia, Quanyong Wang","doi":"10.1177/10849785251376447","DOIUrl":"https://doi.org/10.1177/10849785251376447","url":null,"abstract":"<p><p>To properly target tumors during preoperative chemoradiotherapy, differentiated thyroid carcinoma (DTC) must be careful. This method increases treatment success and decreases recurrence. Ultrasound coupled with SPECT/CT may provide novel localization and dose planning possibilities. Many systems solely use anatomical or functional imaging. This may result in insufficient dosage delivery and wasted radiation exposure to healthy tissues. These issues are addressed by Dual-Modality Imaging-Guided Adaptive Chemoradiotherapy Planning (DMI-ACP). This innovative approach combines real-time ultrasound imaging with 6 Å SPECT/CT imaging for precise tumor delineation and tailored dosimetry. This system enables clinicians to adjust chemoradiotherapy regimens by seamlessly integrating functional iodine absorption data with anatomical characteristics, thereby targeting therapy to cancerous areas. The outcomes of this method for patients with DTC were promising, including better lesion targeting, reduced radiation exposure to healthy tissues, and improved chemotherapeutic dose distribution. In clinical evaluations, the DMI-ACP framework demonstrated a sensitivity of 94% and a specificity of 89% in identifying malignant lesions compared with traditional imaging techniques. Furthermore, the integration of adaptive planning resulted in a 20% improvement in tumor control probability and a 15% reduction in exposure to surrounding healthy tissue, as assessed through dosimetric analysis. 2023075 Nanjing Drum Tower Hospital Group Suqian Hospital/The Affiliated Suqian Hospital of Xuzhou Medical University.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.1,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145253808","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Colorectal cancer (CRC) remains a significant factor contributing to the morbidity and mortality rates linked with cancer throughout the world, especially in its stages of progression. Increasingly attractive therapeutic options include immune modulation combined with preoperative chemotherapy and radiation therapy (CRT). Recent studies have revealed that the protein serine peptidase inhibitor Kazal type 4 (SPINK4), which is abundantly expressed in gastrointestinal tract tissues, plays a role in immune evasion and treatment resistance in cancers. This meta-analysis aims to assess the relationship between SPINK4 expression levels and the therapeutic effectiveness of radiolabeled immune modulators in patients with advanced CRC who are undergoing preoperative chemotherapy and radiation treatment. The degree of SPINK4 expression and a lower objective response to radiolabeled immune modulators showed a statistically significant link. Conversely, patients with low SPINK4 expression have more favorable treatment responses and ongoing clinical improvement following CRT. High SpINK4 expression can act as a negative prognostic biomarker for radiolabeled immune control in advanced CRC.
{"title":"SPINK4 Expression as a Predictive Biomarker for Radiolabeled Immune Modulator Therapy in Advanced Colorectal Cancer.","authors":"Haihua Long, Yongqi Shen, Shuting Li, Hongxiang Kong, Jianqin Liang","doi":"10.1177/10849785251379696","DOIUrl":"https://doi.org/10.1177/10849785251379696","url":null,"abstract":"<p><p>Colorectal cancer (CRC) remains a significant factor contributing to the morbidity and mortality rates linked with cancer throughout the world, especially in its stages of progression. Increasingly attractive therapeutic options include immune modulation combined with preoperative chemotherapy and radiation therapy (CRT). Recent studies have revealed that the protein serine peptidase inhibitor Kazal type 4 (SPINK4), which is abundantly expressed in gastrointestinal tract tissues, plays a role in immune evasion and treatment resistance in cancers. This meta-analysis aims to assess the relationship between SPINK4 expression levels and the therapeutic effectiveness of radiolabeled immune modulators in patients with advanced CRC who are undergoing preoperative chemotherapy and radiation treatment. The degree of SPINK4 expression and a lower objective response to radiolabeled immune modulators showed a statistically significant link. Conversely, patients with low SPINK4 expression have more favorable treatment responses and ongoing clinical improvement following CRT. High SpINK4 expression can act as a negative prognostic biomarker for radiolabeled immune control in advanced CRC.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.1,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145234322","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-06DOI: 10.1177/10849785251384813
Rahaman Shaik, Huda Khan, Mohammed Ziya Salomi, Fatima Uz Zehra, Srujan Kumar Vempati, Mohammed Riyaz, Shaik Azeeza
Chemotherapy, radiation, and targeted biological treatments are examples of cancer therapies that have a significant effect on the immune system. They frequently interfere with the manufacture of immunoglobulins (Igs), which results in immunodeficiency. The processes via which these medications affect B cell activity and antibody production are examined in this review, with an emphasis on cytokine regulation, bone marrow suppression, and therapy-induced lymphopenia. Reduced Ig levels can have clinical repercussions such as increased vulnerability to infections, decreased effectiveness of vaccinations, and compromised immune monitoring. This study also looks at new and existing methods to lessen these consequences, including immunomodulatory techniques, prophylactic antibiotics, and Ig replacement treatment. Optimizing patient outcomes, striking a balance between immunological protection and oncologic efficacy, and directing future research in supportive cancer care all depend on an understanding of how humoral immunity and cancer treatment interact.
{"title":"Effects of Cancer Therapies on Immunoglobulin Synthesis: A Review of Mechanisms, Clinical Implications, and Mitigation Strategies.","authors":"Rahaman Shaik, Huda Khan, Mohammed Ziya Salomi, Fatima Uz Zehra, Srujan Kumar Vempati, Mohammed Riyaz, Shaik Azeeza","doi":"10.1177/10849785251384813","DOIUrl":"https://doi.org/10.1177/10849785251384813","url":null,"abstract":"<p><p>Chemotherapy, radiation, and targeted biological treatments are examples of cancer therapies that have a significant effect on the immune system. They frequently interfere with the manufacture of immunoglobulins (Igs), which results in immunodeficiency. The processes via which these medications affect B cell activity and antibody production are examined in this review, with an emphasis on cytokine regulation, bone marrow suppression, and therapy-induced lymphopenia. Reduced Ig levels can have clinical repercussions such as increased vulnerability to infections, decreased effectiveness of vaccinations, and compromised immune monitoring. This study also looks at new and existing methods to lessen these consequences, including immunomodulatory techniques, prophylactic antibiotics, and Ig replacement treatment. Optimizing patient outcomes, striking a balance between immunological protection and oncologic efficacy, and directing future research in supportive cancer care all depend on an understanding of how humoral immunity and cancer treatment interact.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":""},"PeriodicalIF":2.1,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145240519","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-01Epub Date: 2025-06-16DOI: 10.1089/cbr.2025.0077
Yan Xu, Jinlong Dai, Biao Huang, Guoyuan Lu
Background: Chemotherapy sensitivity in renal carcinoma may be influenced by renal ischemia-reperfusion injury (RIRI). This study elucidates the underlying mechanism by investigating the regulatory role of MYDGF. Methods: The public dataset was downloaded, and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases were used to analyze functional and pathway enrichment of genes in the most significant modules. MitoTracker Green and MitoSOX were used to assess mitochondrial activity and superoxide production in oxygen-glucose deprivation/reoxygenation (OGD/R)-treated renal proximal tubular epithelial cells (RPTECs), with or without MYDGF treatment. Reactive oxygen species production and apoptosis were further analyzed through flow cytometry. A mouse model of RIRI was established and treated with MYDGF, followed by kidney evaluation after 24 h. Histological damage was assessed using hematoxylin-eosin and Masson staining in both RIRI mice and IR-induced patients with AKI. Immunohistochemistry and quantitative real-time polymerase chain reaction were performed to evaluate MYDGF, BCL2, and BAX expression levels in renal tissues. Results: A total of 557 differentially expressed genes were identified. GO and KEGG analyses revealed significant enrichment in oxidative phosphorylation and apoptosis pathways, both of which are relevant to chemosensitivity. MYDGF treatment significantly inhibited apoptosis, enhanced mitochondrial function, and reduced superoxide production in OGD/R-treated RPTECs. In vivo, MYDGF reduced tubular apoptosis and protected against kidney injury, as shown by TUNEL and Masson staining. Notably, MYDGF increased BCL2 and decreased BAX expression both in vitro and in vivo, suggesting an antiapoptotic shift. These changes may contribute not only to protection from RIRI but also to increased susceptibility of damaged renal cells to chemotherapy-induced apoptosis by maintaining mitochondrial integrity. Conclusions: Regulation of apoptotic signaling by MYDGF attenuates ischemia-reperfusion injury and improves chemotherapy outcomes in advanced renal carcinoma.
{"title":"<i>MYDGF</i> Regulates Apoptotic Signaling to Mitigate Renal Ischemia-Reperfusion Injury and Enhance Chemotherapy Sensitivity.","authors":"Yan Xu, Jinlong Dai, Biao Huang, Guoyuan Lu","doi":"10.1089/cbr.2025.0077","DOIUrl":"10.1089/cbr.2025.0077","url":null,"abstract":"<p><p><b><i>Background:</i></b> Chemotherapy sensitivity in renal carcinoma may be influenced by renal ischemia-reperfusion injury (RIRI). This study elucidates the underlying mechanism by investigating the regulatory role of <i>MYDGF</i>. <b><i>Methods:</i></b> The public dataset was downloaded, and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases were used to analyze functional and pathway enrichment of genes in the most significant modules. MitoTracker Green and MitoSOX were used to assess mitochondrial activity and superoxide production in oxygen-glucose deprivation/reoxygenation (OGD/R)-treated renal proximal tubular epithelial cells (RPTECs), with or without <i>MYDGF</i> treatment. Reactive oxygen species production and apoptosis were further analyzed through flow cytometry. A mouse model of RIRI was established and treated with <i>MYDGF</i>, followed by kidney evaluation after 24 h. Histological damage was assessed using hematoxylin-eosin and Masson staining in both RIRI mice and IR-induced patients with AKI. Immunohistochemistry and quantitative real-time polymerase chain reaction were performed to evaluate <i>MYDGF</i>, BCL2, and BAX expression levels in renal tissues. <b><i>Results:</i></b> A total of 557 differentially expressed genes were identified. GO and KEGG analyses revealed significant enrichment in oxidative phosphorylation and apoptosis pathways, both of which are relevant to chemosensitivity. <i>MYDGF</i> treatment significantly inhibited apoptosis, enhanced mitochondrial function, and reduced superoxide production in OGD/R-treated RPTECs. <i>In vivo</i>, <i>MYDGF</i> reduced tubular apoptosis and protected against kidney injury, as shown by TUNEL and Masson staining. Notably, <i>MYDGF</i> increased BCL2 and decreased BAX expression both <i>in vitro</i> and <i>in vivo</i>, suggesting an antiapoptotic shift. These changes may contribute not only to protection from RIRI but also to increased susceptibility of damaged renal cells to chemotherapy-induced apoptosis by maintaining mitochondrial integrity. <b><i>Conclusions:</i></b> Regulation of apoptotic signaling by <i>MYDGF</i> attenuates ischemia-reperfusion injury and improves chemotherapy outcomes in advanced renal carcinoma.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":"515-525"},"PeriodicalIF":2.1,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144310847","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-01Epub Date: 2025-06-02DOI: 10.1089/cbr.2025.0106
Weihong Tong, Zhengyong Zhu, Ruiyang Zhu, Zihe Wang, Jin Zhu
Objective: This study explored the role of monocarboxylate transporter 1 (MCT1) in nasopharyngeal carcinoma (NPC) metastasis and its regulation via DNA methyltransferase 3B (DNMT3B)-mediated methylation, to identify therapeutic targets for NPC. Methods:MCT1/DNMT3B expression was analyzed in NPC (n = 30) and normal tissues (n = 30) using quantitative polymerase chain reaction (qPCR) and immunohistochemistry. DNMT3B overexpression plasmids were transfected into NPC cells to assess MCT1 expression and promoter methylation via bisulfite sequencing PCR. Luciferase and chromatin immunoprecipitation (ChIP) assays identified DNMT3B-MCT1 promoter interactions. Migration/invasion assays and Western blot evaluated functional impacts of MCT1 silencing on metastasis-related pathways. Bioinformatic validation utilized GEO datasets. Results:MCT1 mRNA/protein levels were significantly elevated in NPC versus normal tissues (***p < 0.001), whereas DNMT3B was downregulated. DNMT3B overexpression reduced MCT1 expression (*p < 0.05) and increased MCT1 promoter methylation (**p < 0.01). Luciferase assays revealed that DNMT3B suppressed wild-type MCT1 promoter activity, dependent on an 80 bp CpG island (**p < 0.01). ChIP confirmed DNMT3B enrichment at hypermethylated MCT1 promoter regions (**p < 0.01). MCT1 silencing inhibited NPC cell migration/invasion (*p < 0.05) and downregulated p-AKT, p-mTOR, and p-NFκB (*p < 0.05). High MCT1 correlated with Epstein-Barr virus (EBV)-associated EBNA1BP2 (**p < 0.01), but not PD-L1 markers. DNMT3B inversely correlated with MCT1 (*p < 0.05) and was upregulated in advanced-stage NPC (Stage III + IV vs. I + II, ***p < 0.001), indicating stage-specific epigenetic dysregulation. Conclusion:MCT1 promotes NPC metastasis via NF-κB and PI3K/AKT/mTOR pathways, regulated by DNMT3B-driven promoter methylation. The MCT1-DNMT3B axis, linked to EBV-associated metabolic reprogramming, represents a prognostic biomarker and therapeutic target for advanced NPC.
{"title":"Mechanism of Monocarboxylate Transporter 1 and Its Methylation in Nasopharyngeal Carcinoma Pathogenesis.","authors":"Weihong Tong, Zhengyong Zhu, Ruiyang Zhu, Zihe Wang, Jin Zhu","doi":"10.1089/cbr.2025.0106","DOIUrl":"10.1089/cbr.2025.0106","url":null,"abstract":"<p><p><b><i>Objective:</i></b> This study explored the role of monocarboxylate transporter 1 (<i>MCT1</i>) in nasopharyngeal carcinoma (NPC) metastasis and its regulation via DNA methyltransferase 3B (<i>DNMT3B</i>)-mediated methylation, to identify therapeutic targets for NPC. <b><i>Methods:</i></b> <i>MCT1/DNMT3B</i> expression was analyzed in NPC (<i>n</i> = 30) and normal tissues (<i>n</i> = 30) using quantitative polymerase chain reaction (qPCR) and immunohistochemistry. <i>DNMT3B</i> overexpression plasmids were transfected into NPC cells to assess <i>MCT1</i> expression and promoter methylation via bisulfite sequencing PCR. Luciferase and chromatin immunoprecipitation (ChIP) assays identified <i>DNMT3B-MCT1</i> promoter interactions. Migration/invasion assays and Western blot evaluated functional impacts of <i>MCT1</i> silencing on metastasis-related pathways. Bioinformatic validation utilized GEO datasets. <b><i>Results:</i></b> <i>MCT1</i> mRNA/protein levels were significantly elevated in NPC versus normal tissues (***<i>p</i> < 0.001), whereas <i>DNMT3B</i> was downregulated. <i>DNMT3B</i> overexpression reduced <i>MCT1</i> expression (*<i>p</i> < 0.05) and increased <i>MCT1</i> promoter methylation (**<i>p</i> < 0.01). Luciferase assays revealed that <i>DNMT3B</i> suppressed wild-type <i>MCT1</i> promoter activity, dependent on an 80 bp CpG island (**<i>p</i> < 0.01). ChIP confirmed <i>DNMT3B</i> enrichment at hypermethylated <i>MCT1</i> promoter regions (**<i>p</i> < 0.01). <i>MCT1</i> silencing inhibited NPC cell migration/invasion (*<i>p</i> < 0.05) and downregulated p-AKT, p-mTOR, and p-NFκB (*<i>p</i> < 0.05). High <i>MCT1</i> correlated with Epstein-Barr virus (EBV)-associated EBNA1BP2 (**<i>p</i> < 0.01), but not PD-L1 markers. <i>DNMT3B</i> inversely correlated with <i>MCT1</i> (*<i>p</i> < 0.05) and was upregulated in advanced-stage NPC (Stage III + IV vs. I + II, ***<i>p</i> < 0.001), indicating stage-specific epigenetic dysregulation. <b><i>Conclusion:</i></b> <i>MCT1</i> promotes NPC metastasis via NF-κB and PI3K/AKT/mTOR pathways, regulated by <i>DNMT3B</i>-driven promoter methylation. The <i>MCT1-DNMT3B</i> axis, linked to EBV-associated metabolic reprogramming, represents a prognostic biomarker and therapeutic target for advanced NPC.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":"526-539"},"PeriodicalIF":2.1,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144200903","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-10-01Epub Date: 2025-08-19DOI: 10.1177/10849785251366774
Yiping Zheng, Yinghui Huang, Jianfeng Cai, Qiuhong Ji, Kaijun Liao, Jie Gao, Gengyun Sun
Objective: This study elucidated the role of the forkhead box E1 (FOXE1)-laminin γ2 (LAMC2) signaling axis in promoting brain metastasis (BM) of lung cancer and evaluated its potential as a therapeutic target to enhance the efficacy of preoperative chemoradiotherapy (CRT). Methods: Bioinformatics analysis of the GSE126548 dataset revealed a significant association between elevated FOXE1 expression and BM in lung cancer patients. Functional in vitro assays-including real-time polymerase chain reaction, Western blotting, migration, invasion, and endothelial permeability assays-were conducted in lung cancer cells and human umbilical vein endothelial cells exposed to tumor-conditioned media. In addition, in vivo xenograft and BM mouse models were established to assess the impact of FOXE1 on tumor growth, metastatic potential, and treatment responsiveness. Results:FOXE1 knockdown significantly inhibited lung cancer cell proliferation, migration, invasion, and epithelial-mesenchymal transition. Mechanistically, LAMC2 was identified as a downstream effector of FOXE1, with rescue experiments confirming that the FOXE1-LAMC2 axis plays a central role in driving tumor progression and brain metastatic potential. Notably, FOXE1 silencing enhanced sensitivity to CRT in preclinical models. Conclusions:FOXE1 promotes lung cancer progression and BM by upregulating LAMC2. Targeting the FOXE1-LAMC2 pathway may improve the efficacy of preoperative CRT and offers a promising strategy for therapeutic intervention in lung cancer patients at high risk of BM.
{"title":"Targeting <i>FOXE1</i>-Mediated LAMC2 Expression to Improve Preoperative Chemoradiotherapy Outcomes in Lung Cancer Patients at Risk of Brain Metastasis.","authors":"Yiping Zheng, Yinghui Huang, Jianfeng Cai, Qiuhong Ji, Kaijun Liao, Jie Gao, Gengyun Sun","doi":"10.1177/10849785251366774","DOIUrl":"10.1177/10849785251366774","url":null,"abstract":"<p><p><b><i>Objective:</i></b> This study elucidated the role of the forkhead box E1 (<i>FOXE1</i>)-laminin γ2 (LAMC2) signaling axis in promoting brain metastasis (BM) of lung cancer and evaluated its potential as a therapeutic target to enhance the efficacy of preoperative chemoradiotherapy (CRT). <b><i>Methods:</i></b> Bioinformatics analysis of the GSE126548 dataset revealed a significant association between elevated <i>FOXE1</i> expression and BM in lung cancer patients. Functional <i>in vitro</i> assays-including real-time polymerase chain reaction, Western blotting, migration, invasion, and endothelial permeability assays-were conducted in lung cancer cells and human umbilical vein endothelial cells exposed to tumor-conditioned media. In addition, <i>in vivo</i> xenograft and BM mouse models were established to assess the impact of <i>FOXE1</i> on tumor growth, metastatic potential, and treatment responsiveness. <b><i>Results:</i></b> <i>FOXE1</i> knockdown significantly inhibited lung cancer cell proliferation, migration, invasion, and epithelial-mesenchymal transition. Mechanistically, LAMC2 was identified as a downstream effector of <i>FOXE1</i>, with rescue experiments confirming that the <i>FOXE1</i>-LAMC2 axis plays a central role in driving tumor progression and brain metastatic potential. Notably, <i>FOXE1</i> silencing enhanced sensitivity to CRT in preclinical models. <b><i>Conclusions:</i></b> <i>FOXE1</i> promotes lung cancer progression and BM by upregulating LAMC2. Targeting the <i>FOXE1</i>-LAMC2 pathway may improve the efficacy of preoperative CRT and offers a promising strategy for therapeutic intervention in lung cancer patients at high risk of BM.</p>","PeriodicalId":55277,"journal":{"name":"Cancer Biotherapy and Radiopharmaceuticals","volume":" ","pages":"580-592"},"PeriodicalIF":2.1,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144884335","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}