Pub Date : 2025-01-01DOI: 10.1016/j.ctarc.2025.101024
Shyamaladevi Babu, Madhan Krishnan
{"title":"RUVBL1: A promising prognostic indicator for HPV-associated oral squamous cell carcinoma","authors":"Shyamaladevi Babu, Madhan Krishnan","doi":"10.1016/j.ctarc.2025.101024","DOIUrl":"10.1016/j.ctarc.2025.101024","url":null,"abstract":"","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"45 ","pages":"Article 101024"},"PeriodicalIF":2.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145408245","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1016/j.ctarc.2025.101030
P K Jha , Bipin Chaurasia
Surgical removal of meningioma sometimes become challenging due to its nature of slow growing and being huge at the time of diagnosis. Another problems exists when it becomes adhere to critical structures posing threat to post -op neurological deficits after removal. The art of debulking completely without neurological deficit comes well when a surgeon has good planning, intraoperative techniques, monitoring and good bleeding controls etc. We delve into different steps here to minimize the subsequent risks.
{"title":"The art of debulking meningioma","authors":"P K Jha , Bipin Chaurasia","doi":"10.1016/j.ctarc.2025.101030","DOIUrl":"10.1016/j.ctarc.2025.101030","url":null,"abstract":"<div><div>Surgical removal of meningioma sometimes become challenging due to its nature of slow growing and being huge at the time of diagnosis. Another problems exists when it becomes adhere to critical structures posing threat to post -op neurological deficits after removal. The art of debulking completely without neurological deficit comes well when a surgeon has good planning, intraoperative techniques, monitoring and good bleeding controls etc. We delve into different steps here to minimize the subsequent risks.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"45 ","pages":"Article 101030"},"PeriodicalIF":2.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145476652","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1016/j.ctarc.2023.100784
Meagan S. Whisenant , Jessica Treviño Jones , Anneliese O. Gonzalez , Therese Bartholomew Bevers , Kelly Brassil , Darcy A. Ponce , Sharvari Kamat , Emily Solis , Ann Maliackal , Hannah Warlick , Amie Walters , Chloe Denham , Loretta A. Williams
Background
For the estimated 10 million women in the United States who meet the high-risk criteria for breast cancer, evidence-based interventions may reduce the risk of breast cancer by 50–65 %. Even with substantial evidence supporting preventive medication for risk reduction, there is significant lack of uptake and adherence. The purpose of this study was to characterize the experience of women at high risk for breast cancer and define the content domain for a patient-reported outcomes (PRO) measure of symptom burden from breast cancer risk and risk reducing medication.
Methods
Thirty women at high risk for breast cancer and receiving risk reducing medication participated in single qualitative interviews. Content analysis was used to identify the symptom burden. An expert panel review rated the relevance of symptoms identified in the qualitative interviews to establish the items for inclusion in a PRO symptom burden measure.
Results
Participants had a mean age of 54.6 years; 43.3 % self-identified as Hispanic and 20.0 % self-identified as Black. Content analysis found 20 symptoms related to both risk and preventive treatment, with 8 symptoms reported by ≥ 20 % of women. All women described distress related to their risk and preventive care. Treatment-related symptoms varied based on history of risk-reducing surgery and type of endocrine therapy. Women described symptom-related interference with relationships, work, enjoyment of life, and adherence to risk reducing medication.
Conclusions
Women with a known high risk of breast cancer and receiving preventive care experience a unique symptom burden including multiple symptoms and functional impact related to symptoms.
{"title":"The symptom burden of women with a known risk of breast cancer receiving risk reducing medication","authors":"Meagan S. Whisenant , Jessica Treviño Jones , Anneliese O. Gonzalez , Therese Bartholomew Bevers , Kelly Brassil , Darcy A. Ponce , Sharvari Kamat , Emily Solis , Ann Maliackal , Hannah Warlick , Amie Walters , Chloe Denham , Loretta A. Williams","doi":"10.1016/j.ctarc.2023.100784","DOIUrl":"10.1016/j.ctarc.2023.100784","url":null,"abstract":"<div><h3>Background</h3><div>For the estimated 10 million women in the United States who meet the high-risk criteria for breast cancer, evidence-based interventions may reduce the risk of breast cancer by 50–65 %. Even with substantial evidence supporting preventive medication for risk reduction, there is significant lack of uptake and adherence. The purpose of this study was to characterize the experience of women at high risk for breast cancer and define the content domain for a patient-reported outcomes (PRO) measure of symptom burden from breast cancer risk and risk reducing medication.</div></div><div><h3>Methods</h3><div>Thirty women at high risk for breast cancer and receiving risk reducing medication participated in single qualitative interviews. Content analysis was used to identify the symptom burden. An expert panel review rated the relevance of symptoms identified in the qualitative interviews to establish the items for inclusion in a PRO symptom burden measure.</div></div><div><h3>Results</h3><div>Participants had a mean age of 54.6 years; 43.3 % self-identified as Hispanic and 20.0 % self-identified as Black. Content analysis found 20 symptoms related to both risk and preventive treatment, with 8 symptoms reported by ≥ 20 % of women. All women described distress related to their risk and preventive care. Treatment-related symptoms varied based on history of risk-reducing surgery and type of endocrine therapy. Women described symptom-related interference with relationships, work, enjoyment of life, and adherence to risk reducing medication.</div></div><div><h3>Conclusions</h3><div>Women with a known high risk of breast cancer and receiving preventive care experience a unique symptom burden including multiple symptoms and functional impact related to symptoms.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"43 ","pages":"Article 100784"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139022859","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1016/j.ctarc.2025.100903
Caroline B. van den Berg , Gatske M. Nieuwenhuyzen-de Boer , Ingrid A. Boere , Ruben G. Boers , Joachim B. Boers , Wilfred F.J. van-IJcken , Maurice P.H.M. Jansen , Tugce S. Kirmizitas , Joost H. Gribnau , Heleen J. van Beekhuizen
The aim of this study was to identify differentially methylated regions in cell-free DNA (cfDNA) between healthy persons and patients with advanced stage ovarian cancer (ASOC) and to identify differences in cfDNA methylation before and after cytoreductive surgery. Plasma-derived cfDNA was analyzed by a high-throughput genome wide DNA methylation sequencing technique: MeD-seq. A training set of therapy naïve cfDNA samples of patients with ASOC (≥FIGO stage IIIB, n=10) was compared with cfDNA of healthy controls (n=10) to define a ASOC specific cfDNA methylation signature. A cumulative hypermethylation score was constructed and a validation set of pre- and postoperative samples of 39 patients were compared using this score. MeD-seq results of tumor tissue samples were correlated with cfDNA results. Patients with ASOC showed a clear distinct cfDNA methylation signature from healthy controls (p<0.0001). This cfDNA-methylation signature resulted in preoperative hypermethylation scores (135; interquartile range 110–163) that were significantly higher than postoperative hypermethylation scores (91; interquartile range 76–101) (p<0.001). The cfDNA methylation signature at baseline differed from tumor tissue and was more closely related to DNA methylation of immune-related cells (T-lymphocytes, neutrophil granulocytes, monocytes, and B-lymphocytes) than to ASOC tissue.
MeD-seq provides a promising method for genome wide methylation profiling of cfDNA. Patients with ASOC could clearly be distinguished from healthy controls and differed pre- and postoperatively.
{"title":"Genome-wide cell-free DNA methylation profiling in advanced stage ovarian cancer. Are we looking at the tumor or the patient's immune response to the tumor?","authors":"Caroline B. van den Berg , Gatske M. Nieuwenhuyzen-de Boer , Ingrid A. Boere , Ruben G. Boers , Joachim B. Boers , Wilfred F.J. van-IJcken , Maurice P.H.M. Jansen , Tugce S. Kirmizitas , Joost H. Gribnau , Heleen J. van Beekhuizen","doi":"10.1016/j.ctarc.2025.100903","DOIUrl":"10.1016/j.ctarc.2025.100903","url":null,"abstract":"<div><div>The aim of this study was to identify differentially methylated regions in cell-free DNA (cfDNA) between healthy persons and patients with advanced stage ovarian cancer (ASOC) and to identify differences in cfDNA methylation before and after cytoreductive surgery. Plasma-derived cfDNA was analyzed by a high-throughput genome wide DNA methylation sequencing technique: MeD-seq. A training set of therapy naïve cfDNA samples of patients with ASOC (≥FIGO stage IIIB, n=10) was compared with cfDNA of healthy controls (n=10) to define a ASOC specific cfDNA methylation signature. A cumulative hypermethylation score was constructed and a validation set of pre- and postoperative samples of 39 patients were compared using this score. MeD-seq results of tumor tissue samples were correlated with cfDNA results. Patients with ASOC showed a clear distinct cfDNA methylation signature from healthy controls (p<0.0001). This cfDNA-methylation signature resulted in preoperative hypermethylation scores (135; interquartile range 110–163) that were significantly higher than postoperative hypermethylation scores (91; interquartile range 76–101) (p<0.001). The cfDNA methylation signature at baseline differed from tumor tissue and was more closely related to DNA methylation of immune-related cells (T-lymphocytes, neutrophil granulocytes, monocytes, and B-lymphocytes) than to ASOC tissue.</div><div>MeD-seq provides a promising method for genome wide methylation profiling of cfDNA. Patients with ASOC could clearly be distinguished from healthy controls and differed pre- and postoperatively.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"43 ","pages":"Article 100903"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143714721","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
While chimeric antigen receptor T-cell (CAR T) therapy has shown promise in treating B-cell non-Hodgkin lymphoma, its efficacy is variable in patients with poor initial responses. This study investigates the efficacy and safety of zanubrutinib combined with CAR T therapy targeting CD19 in refractory/relapsed diffuse large B cell lymphoma (DLBCL).
Methods
We conducted a retrospective study of 17 patients with R/R DLBCL who received zanubrutinib combined with anti-CD19 CAR T-cell therapy. We assessed overall survival (OS) and progression-free survival (PFS) and conducted subgroup analyses. We also monitored CAR T-cell expansion by quantifying vector copy numbers (VCN). Safety and tolerability were assessed by documenting adverse events, including cytokine release syndrome and neurotoxicity.
Results
The median follow-up was 30 months (range, 4–36). The overall response rate(ORR) was 88.2 %, with 70.5 % achieving complete remission. At 24 months, the estimated PFS was 59 % (95 %CI, 40–88 %), and the OS was 71 % (95 %CI, 52–90 %). At 36 months, the estimated OS was 65 % (95 %CI, 46–92 %). Patients with non-elevated lactate dehydrogenase(LDH) levels showed a higher PFS probability (100 %) compared to those with elevated LDH (42 %, 95 %CI: 21 %-81 %) (log-rank, p = 0.071). The area under the receiver-operating characteristic (ROC) curve for peak CAR T-cell expansion was 0.773, suggesting an optimal cutoff at day 13 for enhancing survival (sensitivity 66.7 %, specificity 90.9 %; p = 0.07). Early peak expansion (before day 13) correlated with better PFS and OS (log-rank, p = 0.0018 and p = 0.0053, respectively). The total VCN AUC was 0.636, with a cutoff of 12,690 significantly predicting survival (sensitivity 83.3 %, specificity 72.7 %; p = 0.366). Kaplan-Meier analysis indicated statistically significant differences in OS for patients with VCN below 12,690 (log-rank, p = 0.017) in comparison to those exceeding 12,690, though trends in PFS did not reach statistical significance (log-rank, p = 0.12). Safety profiles indicated manageable cytokine release syndrome, with no severe neurotoxicity reported.
Conclusions
Zanubrutinib combined with CAR T-cell therapy offers an effective treatment option for patients with R/R DLBCL, enhancing response rates and survival. Detailed analysis of CAR T-cell expansion provides insight into the dynamics of cellular responses, underscoring the potential for tailored therapeutic approaches based on biological markers.
{"title":"Efficacy and safety of zanubrutinib combined with chimeric antigen receptor T-cell therapy targeting CD19 in refractory or relapsed diffuse large B cell lymphoma: A retrospective analysis","authors":"Jinhuan Xu , Xiaoying Zhang , Yun Li , Fankai Meng , Yicheng Zhang , Miao Zheng","doi":"10.1016/j.ctarc.2025.100902","DOIUrl":"10.1016/j.ctarc.2025.100902","url":null,"abstract":"<div><h3>Background</h3><div>While chimeric antigen receptor T-cell (CAR T) therapy has shown promise in treating B-cell non-Hodgkin lymphoma, its efficacy is variable in patients with poor initial responses. This study investigates the efficacy and safety of zanubrutinib combined with CAR T therapy targeting CD19 in refractory/relapsed diffuse large B cell lymphoma (DLBCL).</div></div><div><h3>Methods</h3><div>We conducted a retrospective study of 17 patients with R/R DLBCL who received zanubrutinib combined with anti-CD19 CAR T-cell therapy. We assessed overall survival (OS) and progression-free survival (PFS) and conducted subgroup analyses. We also monitored CAR T-cell expansion by quantifying vector copy numbers (VCN). Safety and tolerability were assessed by documenting adverse events, including cytokine release syndrome and neurotoxicity.</div></div><div><h3>Results</h3><div>The median follow-up was 30 months (range, 4–36). The overall response rate(ORR) was 88.2 %, with 70.5 % achieving complete remission. At 24 months, the estimated PFS was 59 % (95 %CI, 40–88 %), and the OS was 71 % (95 %CI, 52–90 %). At 36 months, the estimated OS was 65 % (95 %CI, 46–92 %). Patients with non-elevated lactate dehydrogenase(LDH) levels showed a higher PFS probability (100 %) compared to those with elevated LDH (42 %, 95 %CI: 21 %-81 %) (log-rank, <em>p</em> = 0.071). The area under the receiver-operating characteristic (ROC) curve for peak CAR T-cell expansion was 0.773, suggesting an optimal cutoff at day 13 for enhancing survival (sensitivity 66.7 %, specificity 90.9 %; <em>p</em> = 0.07). Early peak expansion (before day 13) correlated with better PFS and OS (log-rank, <em>p</em> = 0.0018 and <em>p</em> = 0.0053, respectively). The total VCN AUC was 0.636, with a cutoff of 12,690 significantly predicting survival (sensitivity 83.3 %, specificity 72.7 %; <em>p</em> = 0.366). Kaplan-Meier analysis indicated statistically significant differences in OS for patients with VCN below 12,690 (log-rank, <em>p</em> = 0.017) in comparison to those exceeding 12,690, though trends in PFS did not reach statistical significance (log-rank, <em>p</em> = 0.12). Safety profiles indicated manageable cytokine release syndrome, with no severe neurotoxicity reported.</div></div><div><h3>Conclusions</h3><div>Zanubrutinib combined with CAR T-cell therapy offers an effective treatment option for patients with R/R DLBCL, enhancing response rates and survival. Detailed analysis of CAR T-cell expansion provides insight into the dynamics of cellular responses, underscoring the potential for tailored therapeutic approaches based on biological markers.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"43 ","pages":"Article 100902"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143725012","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1016/j.ctarc.2025.100970
Zoran Andric , Fedor Moiseenko , Tamta Makharadze , Alona Oleksiienko , Eduardo Yañez Ruiz , SungHyun Kim , KeumYoung Ahn , TaeHong Park , Hana Ju , Eric Hyungseok Baek , Soonbum Kwon , IlSung Chang , SinHye Kim , HyunAh Kim , EunKyung Lee , Claire Verschraegen
Purpose
Data from the CT-P16 3.1 study demonstrated equivalent efficacy between CT-P16, a bevacizumab biosimilar, and European-approved reference bevacizumab (EU-bevacizumab) for the primary endpoint of objective response rate (ORR) during induction treatment in patients with metastatic or recurrent non-squamous non-small cell lung cancer (NSCLC). We now present long-term findings.
Methods
In this randomized, double-blind, multicenter phase 3 study, patients with metastatic or recurrent non-squamous NSCLC received CT-P16 or EU-bevacizumab (15 mg/kg every 3 weeks; ≤6 cycles) together with paclitaxel (200 mg/m2) and carboplatin (area under the curve 6.0) for 4–6 cycles during the induction period. Patients with controlled disease entered the maintenance period, continuing with CT-P16 or EU-bevacizumab as monotherapy until disease progression/intolerable toxicity. They were then followed up every 9 weeks until death or end of study. Outcomes were evaluated ≤3 years after the last patient enrollment.
Results
Of 689 patients receiving CT-P16 (N = 342) or EU-bevacizumab (N = 347), 499 (72.4 %) completed the induction period (CT-P16, n = 258 [75.4 %]; EU-bevacizumab, n = 241 [69.5 %]), 466 (67.6 %) initiated the maintenance period (CT-P16, n = 239 [69.9 %]; EU-bevacizumab, n = 227 [65.4 %]), and 389 (56.5 %) entered follow-up (CT-P16, n = 190 [55.6 %]; EU-bevacizumab, n = 199 [57.3 %]). Whole study ORRs were similar for CT-P16 (45.61 % [95 % confidence interval 40.34–50.89]) and EU-bevacizumab (46.11 % [95 % confidence interval 40.86–51.35]). Response duration, time to progression, progression-free survival, and overall survival were similar. No new safety signals were detected.
Conclusions
Long-term results of the CT-P16 3.1 study confirm equivalent efficacy of CT-P16 and EU-bevacizumab in patients with metastatic or recurrent non-squamous NSCLC.
Trial registration number
NCT03676192.
Micro abstract
Equivalent efficacy between CT-P16, a bevacizumab biosimilar, and European-approved reference bevacizumab (EU-bevacizumab) was demonstrated in a phase 3 study of patients with metastatic or recurrent non-squamous NSCLC. Long-term findings in 689 patients confirmed equivalent efficacy of CT-P16 and EU-bevacizumab; objective response rates were 45.6 % and 46.1 %, respectively. Response duration, time to progression, progression-free survival, and overall survival were also similar.
{"title":"Long-term results of a randomized controlled trial of biosimilar CT-P16 and reference bevacizumab in patients with metastatic or recurrent non-small cell lung cancer","authors":"Zoran Andric , Fedor Moiseenko , Tamta Makharadze , Alona Oleksiienko , Eduardo Yañez Ruiz , SungHyun Kim , KeumYoung Ahn , TaeHong Park , Hana Ju , Eric Hyungseok Baek , Soonbum Kwon , IlSung Chang , SinHye Kim , HyunAh Kim , EunKyung Lee , Claire Verschraegen","doi":"10.1016/j.ctarc.2025.100970","DOIUrl":"10.1016/j.ctarc.2025.100970","url":null,"abstract":"<div><h3>Purpose</h3><div>Data from the CT-P16 3.1 study demonstrated equivalent efficacy between CT-P16, a bevacizumab biosimilar, and European-approved reference bevacizumab (EU-bevacizumab) for the primary endpoint of objective response rate (ORR) during induction treatment in patients with metastatic or recurrent non-squamous non-small cell lung cancer (NSCLC). We now present long-term findings.</div></div><div><h3>Methods</h3><div>In this randomized, double-blind, multicenter phase 3 study, patients with metastatic or recurrent non-squamous NSCLC received CT-P16 or EU-bevacizumab (15 mg/kg every 3 weeks; ≤6 cycles) together with paclitaxel (200 mg/m<sup>2</sup>) and carboplatin (area under the curve 6.0) for 4–6 cycles during the induction period. Patients with controlled disease entered the maintenance period, continuing with CT-P16 or EU-bevacizumab as monotherapy until disease progression/intolerable toxicity. They were then followed up every 9 weeks until death or end of study. Outcomes were evaluated ≤3 years after the last patient enrollment.</div></div><div><h3>Results</h3><div>Of 689 patients receiving CT-P16 (<em>N</em> = 342) or EU-bevacizumab (<em>N</em> = 347), 499 (72.4 %) completed the induction period (CT-P16, <em>n</em> = 258 [75.4 %]; EU-bevacizumab, <em>n</em> = 241 [69.5 %]), 466 (67.6 %) initiated the maintenance period (CT-P16, <em>n</em> = 239 [69.9 %]; EU-bevacizumab, <em>n</em> = 227 [65.4 %]), and 389 (56.5 %) entered follow-up (CT-P16, <em>n</em> = 190 [55.6 %]; EU-bevacizumab, <em>n</em> = 199 [57.3 %]). Whole study ORRs were similar for CT-P16 (45.61 % [95 % confidence interval 40.34–50.89]) and EU-bevacizumab (46.11 % [95 % confidence interval 40.86–51.35]). Response duration, time to progression, progression-free survival, and overall survival were similar. No new safety signals were detected.</div></div><div><h3>Conclusions</h3><div>Long-term results of the CT-P16 3.1 study confirm equivalent efficacy of CT-P16 and EU-bevacizumab in patients with metastatic or recurrent non-squamous NSCLC.</div></div><div><h3>Trial registration number</h3><div>NCT03676192.</div></div><div><h3>Micro abstract</h3><div>Equivalent efficacy between CT-P16, a bevacizumab biosimilar, and European-approved reference bevacizumab (EU-bevacizumab) was demonstrated in a phase 3 study of patients with metastatic or recurrent non-squamous NSCLC. Long-term findings in 689 patients confirmed equivalent efficacy of CT-P16 and EU-bevacizumab; objective response rates were 45.6 % and 46.1 %, respectively. Response duration, time to progression, progression-free survival, and overall survival were also similar.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"44 ","pages":"Article 100970"},"PeriodicalIF":2.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144827334","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Curcumin (Cur), a yellow polyphenolic compound derived from the rhizome of turmeric, exhibits potent chemopreventive and chemotherapeutic properties. However, its clinical application as an anticancer agent is hindered by poor water solubility and low bioavailability. To overcome these limitations, the present study introduces a novel nanoformulation in which curcumin is effectively encapsulated within 142 nm spherical dendrosomes, which serve as a non-toxic nanocarrier.
Methods
The morphological changes in OVCAR3 ovarian cancer cells were assessed using both inverted light microscopy and fluorescence microscopy. Cell cycle distribution was analyzed by flow cytometry using the MultiCycler software. Cells were treated with dendrosomal curcumin (DNC), oxaliplatin (Oxa), or a combination of both for 48 hours. Protein expression levels were analyzed by western blotting using enhanced chemiluminescence (ECL) detection.
Results
Dendrosomal curcumin improves curcumin’s bioavailability and therapeutic potential in cancer treatment. Treatment with DNC and Oxa individually resulted in significant induction of apoptosis in OVCAR3 cells. The combination therapy further amplified this effect compared to either agent alone. Molecular analyses revealed upregulation of pro-apoptotic Bax and downregulation of anti-apoptotic Bcl-2 at both mRNA and protein levels, supporting the activation of apoptotic pathways.
Conclusion
Our findings demonstrate that both DNC and Oxa induce apoptosis in OVCAR3 ovarian cancer cells through modulation of Bax and Bcl-2 expression. The enhanced efficacy observed in combination therapy highlights the therapeutic potential of DNC, in conjunction with conventional chemotherapeutics, as a promising strategy for ovarian cancer treatment.
{"title":"Dendrosomal Curcumin Nanoformulation induces apoptosis via Bax/Bcl-2 in human ovarian cancer OVCAR3 cells","authors":"Marziyeh Alizadeh Zarei , Hossein Nikzad , Zahra Shabani , Hamed Haddad Kashani , Elahe Seyed Hosseini","doi":"10.1016/j.ctarc.2025.100981","DOIUrl":"10.1016/j.ctarc.2025.100981","url":null,"abstract":"<div><h3>Introduction</h3><div>Curcumin (Cur), a yellow polyphenolic compound derived from the rhizome of turmeric, exhibits potent chemopreventive and chemotherapeutic properties. However, its clinical application as an anticancer agent is hindered by poor water solubility and low bioavailability. To overcome these limitations, the present study introduces a novel nanoformulation in which curcumin is effectively encapsulated within 142 nm spherical dendrosomes, which serve as a non-toxic nanocarrier.</div></div><div><h3>Methods</h3><div>The morphological changes in OVCAR3 ovarian cancer cells were assessed using both inverted light microscopy and fluorescence microscopy. Cell cycle distribution was analyzed by flow cytometry using the MultiCycler software. Cells were treated with dendrosomal curcumin (DNC), oxaliplatin (Oxa), or a combination of both for 48 hours. Protein expression levels were analyzed by western blotting using enhanced chemiluminescence (ECL) detection.</div></div><div><h3>Results</h3><div>Dendrosomal curcumin improves curcumin’s bioavailability and therapeutic potential in cancer treatment. Treatment with DNC and Oxa individually resulted in significant induction of apoptosis in OVCAR3 cells. The combination therapy further amplified this effect compared to either agent alone. Molecular analyses revealed upregulation of pro-apoptotic Bax and downregulation of anti-apoptotic Bcl-2 at both mRNA and protein levels, supporting the activation of apoptotic pathways.</div></div><div><h3>Conclusion</h3><div>Our findings demonstrate that both DNC and Oxa induce apoptosis in OVCAR3 ovarian cancer cells through modulation of Bax and Bcl-2 expression. The enhanced efficacy observed in combination therapy highlights the therapeutic potential of DNC, in conjunction with conventional chemotherapeutics, as a promising strategy for ovarian cancer treatment.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"44 ","pages":"Article 100981"},"PeriodicalIF":2.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144887077","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Through comprehensive analysis of cisplatin-resistant HeLa cervical cancer cells, we reveal that curcumin exerts potent anti-cancer effects by modulating autophagic processes, and further identify Endoplasmic reticulum oxidoreductase 1-alpha (ERO1α) as a key regulator mediating this biological response.
Methods
Cell proliferation was assessed using Cell Counting Kit-8 (CCK-8). The immunoblotting analysis measured the Phosphoinositide 3-Kinase/ Protein Kinase B (PI3K/AKT) pathway-related proteins, Microtubule-associated protein 1A/1B-light chain 3 (LC3II/I) ratio, and Beclin-1 expression. The cell migration ability was evaluated through a scratch assay.
Results
Curcumin significantly inhibits the proliferation and migration of cisplatin-resistant HeLa cells. It promotes autophagy in these cells, as shown by the upregulation of autophagy-related proteins Beclin-1 and LC3II/I. Curcumin also downregulates ERO1α expression, subsequently modulating the PI3K/AKT pathway. Interestingly, the overexpression of ERO1α or activation of PI3K negated the autophagy-promoting effects of curcumin, indicated by the absence of changes in autophagy-related proteins and phosphorylation levels of PI3K/AKT.
Conclusions
Our study provides compelling evidence that curcumin inhibits the proliferation and migration of cisplatin-resistant HeLa cells by promoting autophagy through the downregulation of ERO1α and inhibition of the PI3K/AKT pathway.
{"title":"ERO1α regulates curcumin-induced autophagy via PI3K/AKT pathway","authors":"Dandan Huang , Bolorchimeg Baldandorj , Erdenezaya Odkhuu , Egshiglen Amartuvshin , Damdindorj Boldbaatar , Hairong Guo , Adilsaikhan Mendjargal","doi":"10.1016/j.ctarc.2025.100957","DOIUrl":"10.1016/j.ctarc.2025.100957","url":null,"abstract":"<div><h3>Objective</h3><div>Through comprehensive analysis of cisplatin-resistant HeLa cervical cancer cells, we reveal that curcumin exerts potent anti-cancer effects by modulating autophagic processes, and further identify Endoplasmic reticulum oxidoreductase 1-alpha (ERO1α) as a key regulator mediating this biological response.</div></div><div><h3>Methods</h3><div>Cell proliferation was assessed using Cell Counting Kit-8 (CCK-8). The immunoblotting analysis measured the Phosphoinositide 3-Kinase/ Protein Kinase B (PI3K/AKT) pathway-related proteins, Microtubule-associated protein 1A/1B-light chain 3 (LC3II/I) ratio, and Beclin-1 expression. The cell migration ability was evaluated through a scratch assay.</div></div><div><h3>Results</h3><div>Curcumin significantly inhibits the proliferation and migration of cisplatin-resistant HeLa cells. It promotes autophagy in these cells, as shown by the upregulation of autophagy-related proteins Beclin-1 and LC3II/I. Curcumin also downregulates ERO1α expression, subsequently modulating the PI3K/AKT pathway. Interestingly, the overexpression of ERO1α or activation of PI3K negated the autophagy-promoting effects of curcumin, indicated by the absence of changes in autophagy-related proteins and phosphorylation levels of PI3K/AKT.</div></div><div><h3>Conclusions</h3><div>Our study provides compelling evidence that curcumin inhibits the proliferation and migration of cisplatin-resistant HeLa cells by promoting autophagy through the downregulation of ERO1α and inhibition of the PI3K/AKT pathway.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"44 ","pages":"Article 100957"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144470227","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1016/j.ctarc.2025.100976
Ariana Orlić, Issam Alsamara, Martina Paric
Background
Fertility preservation (FP) is an important aspect of cancer care for male patients of reproductive age; however, there is a lack of research on male experiences of FP. This scoping review aims to identify barriers and facilitators to FP among male cancer patients, based on both patient and healthcare provider/researcher perspectives.
Method
We conducted a scoping review following the Joanna Briggs Institute methodology and reported results according to the PRISMA-ScR checklist. We searched MEDLINE/PubMed, CINAHL, APA PsycInfo, and Web of Science, with supplemented hand-searching of the ESHRE website and reference lists. We included primary studies published in English (2001–2023) reporting on FP in male cancer patients. Findings were mapped to the Theoretical Domains Framework (TDF).
Results
A total of 56 studies met the inclusion criteria. Eleven TDF domains were applicable, with one additional domain, “Patient Characteristics”, added inductively. Among patients, the most frequently coded domains were “Social Influences”, “Goals, Beliefs about Capabilities”, and “Emotion”. For providers/researchers, key domains included “Environmental Context and Resources”, “Patient Characteristics” and “Knowledge”.
Conclusion
A combination of social, psychological, and contextual factors shapes barriers and facilitators to FP in male cancer patients. Addressing these barriers could improve male cancer patients’ quality of life and well-being during treatment and beyond. Recommendations include formalised FP care pathways with an educated, multidisciplinary team, which should be supported by concrete legal and ethical guidelines integrated in policy frameworks concerning cancer care. Applying the TDF can support the development of targeted, evidence-informed strategies to improve FP integration in cancer care.
生育能力保存(fertility preservation, FP)是育龄男性患者癌症护理的一个重要方面;然而,对男性生育经验的研究却很缺乏。本综述旨在根据患者和医疗保健提供者/研究人员的观点,确定男性癌症患者实施计划生育的障碍和促进因素。方法我们按照Joanna Briggs研究所的方法进行了范围审查,并根据PRISMA-ScR检查表报告结果。我们检索了MEDLINE/PubMed、CINAHL、APA PsycInfo和Web of Science,并辅以手工检索了ESHRE网站和参考文献列表。我们纳入了2001-2023年发表的关于男性癌症患者生育激素的主要英文研究。研究结果被映射到理论领域框架(TDF)。结果56项研究符合纳入标准。11个TDF域是适用的,还有一个额外的域,“患者特征”,是电感添加的。在患者中,最常见的编码域是“社会影响”、“目标、关于能力的信念”和“情感”。对于提供者/研究人员,关键领域包括“环境背景和资源”,“患者特征”和“知识”。结论社会、心理和环境因素共同构成了男性癌症患者计划生育的障碍和促进因素。解决这些障碍可以改善男性癌症患者在治疗期间和之后的生活质量和幸福感。建议包括由受过教育的多学科团队提供正规的计划生育护理途径,并应得到纳入癌症护理政策框架的具体法律和伦理准则的支持。应用TDF可以支持制定有针对性的循证战略,以改善计划生育在癌症治疗中的整合。
{"title":"Improving fertility preservation for male cancer patients: A scoping review of barriers and facilitators","authors":"Ariana Orlić, Issam Alsamara, Martina Paric","doi":"10.1016/j.ctarc.2025.100976","DOIUrl":"10.1016/j.ctarc.2025.100976","url":null,"abstract":"<div><h3>Background</h3><div>Fertility preservation (FP) is an important aspect of cancer care for male patients of reproductive age; however, there is a lack of research on male experiences of FP. This scoping review aims to identify barriers and facilitators to FP among male cancer patients, based on both patient and healthcare provider/researcher perspectives.</div></div><div><h3>Method</h3><div>We conducted a scoping review following the Joanna Briggs Institute methodology and reported results according to the PRISMA-ScR checklist. We searched MEDLINE/PubMed, CINAHL, APA PsycInfo, and Web of Science, with supplemented hand-searching of the ESHRE website and reference lists. We included primary studies published in English (2001–2023) reporting on FP in male cancer patients. Findings were mapped to the Theoretical Domains Framework (TDF).</div></div><div><h3>Results</h3><div>A total of 56 studies met the inclusion criteria. Eleven TDF domains were applicable, with one additional domain, “Patient Characteristics”, added inductively. Among patients, the most frequently coded domains were “Social Influences”, “Goals, Beliefs about Capabilities”, and “Emotion”. For providers/researchers, key domains included “Environmental Context and Resources”, “Patient Characteristics” and “Knowledge”.</div></div><div><h3>Conclusion</h3><div>A combination of social, psychological, and contextual factors shapes barriers and facilitators to FP in male cancer patients. Addressing these barriers could improve male cancer patients’ quality of life and well-being during treatment and beyond. Recommendations include formalised FP care pathways with an educated, multidisciplinary team, which should be supported by concrete legal and ethical guidelines integrated in policy frameworks concerning cancer care. Applying the TDF can support the development of targeted, evidence-informed strategies to improve FP integration in cancer care.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"44 ","pages":"Article 100976"},"PeriodicalIF":2.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144887121","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1016/j.ctarc.2025.100995
Shuang Wang , Tao Li
NSD2 is a histone lysine N-methyltransferase that plays a crucial role in complex cancer biology. Despite mounting evidence highlighting the critical role of NSD2 in tumor progression and immunity, a comprehensive pan-cancer analysis of NSD2 has yet to be performed to determine whether NSD2 can be used as a viable biomarker, for cancer screening, prognostic prediction, and for the accurate design of therapeutic regimens for a variety of human malignancies. malignancies. This study was based on TCGA, TIMER2.0, cBioPortal, TCGA plot R software package and comprehensively investigated the expression of NSD2 in 33 different tumors, clinical phenotype, survival analysis, immune infiltration, genetic alterations, correlation with TMB and MSI, enrichment analysis and prognostic significance. The results showed that NSD2 was significantly overexpressed in most tumors and significantly correlated with low OS in different tumor types. Our findings suggest its potential oncogenic and epigenetic role. Amplification was the most common type of NSD2 variant. Immune infiltration analysis revealed a strong correlation between NSD2 and different immune cells. NSD2 was significantly associated with TMB and MSI in several cancers. Furthermore, enrichment analysis revealed associations between NSD2 and cell cycle and DNA repair pathways as well as various cellular functions. These findings suggest that NSD2 may serve as a potential prognostic biomarker and target for immunotherapy of various cancers.
{"title":"Comprehensive pan-cancer analysis identified NSD2 as a potential prognostic and diagnostic biomarker","authors":"Shuang Wang , Tao Li","doi":"10.1016/j.ctarc.2025.100995","DOIUrl":"10.1016/j.ctarc.2025.100995","url":null,"abstract":"<div><div>NSD2 is a histone lysine N-methyltransferase that plays a crucial role in complex cancer biology. Despite mounting evidence highlighting the critical role of NSD2 in tumor progression and immunity, a comprehensive pan-cancer analysis of NSD2 has yet to be performed to determine whether NSD2 can be used as a viable biomarker, for cancer screening, prognostic prediction, and for the accurate design of therapeutic regimens for a variety of human malignancies. malignancies. This study was based on TCGA, TIMER2.0, cBioPortal, TCGA plot R software package and comprehensively investigated the expression of NSD2 in 33 different tumors, clinical phenotype, survival analysis, immune infiltration, genetic alterations, correlation with TMB and MSI, enrichment analysis and prognostic significance. The results showed that NSD2 was significantly overexpressed in most tumors and significantly correlated with low OS in different tumor types. Our findings suggest its potential oncogenic and epigenetic role. Amplification was the most common type of NSD2 variant. Immune infiltration analysis revealed a strong correlation between NSD2 and different immune cells. NSD2 was significantly associated with TMB and MSI in several cancers. Furthermore, enrichment analysis revealed associations between NSD2 and cell cycle and DNA repair pathways as well as various cellular functions. These findings suggest that NSD2 may serve as a potential prognostic biomarker and target for immunotherapy of various cancers.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"45 ","pages":"Article 100995"},"PeriodicalIF":2.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145046077","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}