Pub Date : 2026-01-01DOI: 10.1016/j.ctarc.2025.101060
Nouf Khalifa ALaqeel
Immunotherapy is currently one of the most promising fields for novel cancer treatments and discoveries. Scientists now understand how intricately our immune system interacts with cancer. The way we think about and treat cancer is evolving as a result of novel medicines that are made possible by our growing understanding of how the immune system functions. Treatment is now more tumor-specific and less toxic thanks to new cancer targeted medicines that employ therapeutic antibodies or tiny chemicals. Using ligand-targeted treatment to make targeting more precise and deliver bigger dosages of anti-cancer medication to tumor tissue is one of the most promising approaches to overcoming such obstacles. Optical biosensors are commonly used in clinical diagnostics, offering the advantage of continuous monitoring of biochemical reactions. The development of targeted medication delivery systems for a variety of illnesses, including cancer cells, is a potential area of nanomedicine. Researchers are concentrating on cancer cells because of their high rates of proliferation and resistance to conventional treatment techniques. .Lipid-based drug delivery employs nanoparticles to encapsulate and distribute medications to particular cells or tissues The present review is focused on personalized cancer vaccines are a promising immunotherapy targeting patient specific tumor neoantigens, and nanoparticles.
{"title":"Advances in personalized cancer vaccine to nanotechnology innovations","authors":"Nouf Khalifa ALaqeel","doi":"10.1016/j.ctarc.2025.101060","DOIUrl":"10.1016/j.ctarc.2025.101060","url":null,"abstract":"<div><div>Immunotherapy is currently one of the most promising fields for novel cancer treatments and discoveries. Scientists now understand how intricately our immune system interacts with cancer. The way we think about and treat cancer is evolving as a result of novel medicines that are made possible by our growing understanding of how the immune system functions. Treatment is now more tumor-specific and less toxic thanks to new cancer targeted medicines that employ therapeutic antibodies or tiny chemicals. Using ligand-targeted treatment to make targeting more precise and deliver bigger dosages of anti-cancer medication to tumor tissue is one of the most promising approaches to overcoming such obstacles. Optical biosensors are commonly used in clinical diagnostics, offering the advantage of continuous monitoring of biochemical reactions. The development of targeted medication delivery systems for a variety of illnesses, including cancer cells, is a potential area of nanomedicine. Researchers are concentrating on cancer cells because of their high rates of proliferation and resistance to conventional treatment techniques. .Lipid-based drug delivery employs nanoparticles to encapsulate and distribute medications to particular cells or tissues The present review is focused on personalized cancer vaccines are a promising immunotherapy targeting patient specific tumor neoantigens, and nanoparticles.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"46 ","pages":"Article 101060"},"PeriodicalIF":2.4,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145910630","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 : 2026-01-01DOI: 10.1016/j.ctarc.2025.101078
Ludovic Doucet , Camille Moreau-Bachelard , Laurent Mathiot , Olivier Kerdraon , Marie Robert , Julie Quintin , François Bocquet , Morgan Zenatri , Mario Campone , Jean-Sébastien Frenel
Background
Adjuvant trastuzumab emtansine (T-DM1) is the standard treatment for HER2-positive early breast cancer (EBC) patients who do not achieve a pathologic complete response (pCR) after neoadjuvant chemotherapy (NAC). Given the heterogeneity within this population, we analyzed long-term outcomes in non-pCR HER2-positive EBC patients who received adjuvant trastuzumab prior to the availability of the KATHERINE trial results
Methods
This single-center cohort study included all HER2-positive EBC patients treated with neoadjuvant chemotherapy at our hospital between 2006 and 2017. Kaplan–Meier and multivariable Cox regression models were employed to assess long-term invasive disease-free survival (iDFS) and overall survival (OS).
Results
This study included 103 patients, of which 67.0 % had a residual tumor size of ypT1, and 57.3 % had no residual tumor in the lymph nodes. The median follow-up was 9.3 years. At 3, 5, and 7 years, the iDFS rates were 78.3 %, 72.2 %, and 69.7 %, and the overall survival rates were 90.2 %, 83.3 %, and 79.8 %, respectively. Multivariate analysis identified ypT<2, ypN0, and estrogen receptor positivity as independent predictors of improved iDFS. Among patients with ypT<2, ypN0, and estrogen receptor-positive disease (34 out of 72 with estrogen receptor positivity), iDFS rates were notably high: 97 % (95 % CI: 91.3–100) at 3 years, and 90.9 % (95 % CI: 81.6–100) at both 5 and 7 years.
Conclusion
Patients without a pathologic complete response after neoadjuvant chemotherapy plus trastuzumab represent a heterogeneous group. Those with ypT<2, ypN0, and ER-positive residual disease may still achieve excellent iDFS and OS despite absence of adjuvant T-DM1.
{"title":"Favourable long-term outcomes in selected HER2-Positive early breast cancer patients without pathological complete response after neoadjuvant chemotherapy and trastuzumab: A pre–T-DM1 era cohort study","authors":"Ludovic Doucet , Camille Moreau-Bachelard , Laurent Mathiot , Olivier Kerdraon , Marie Robert , Julie Quintin , François Bocquet , Morgan Zenatri , Mario Campone , Jean-Sébastien Frenel","doi":"10.1016/j.ctarc.2025.101078","DOIUrl":"10.1016/j.ctarc.2025.101078","url":null,"abstract":"<div><h3>Background</h3><div>Adjuvant trastuzumab emtansine (T-DM1) is the standard treatment for HER2-positive early breast cancer (EBC) patients who do not achieve a pathologic complete response (pCR) after neoadjuvant chemotherapy (NAC). Given the heterogeneity within this population, we analyzed long-term outcomes in non-pCR HER2-positive EBC patients who received adjuvant trastuzumab prior to the availability of the KATHERINE trial results</div></div><div><h3>Methods</h3><div>This single-center cohort study included all HER2-positive EBC patients treated with neoadjuvant chemotherapy at our hospital between 2006 and 2017. Kaplan–Meier and multivariable Cox regression models were employed to assess long-term invasive disease-free survival (iDFS) and overall survival (OS).</div></div><div><h3>Results</h3><div>This study included 103 patients, of which 67.0 % had a residual tumor size of ypT1, and 57.3 % had no residual tumor in the lymph nodes. The median follow-up was 9.3 years. At 3, 5, and 7 years, the iDFS rates were 78.3 %, 72.2 %, and 69.7 %, and the overall survival rates were 90.2 %, 83.3 %, and 79.8 %, respectively. Multivariate analysis identified ypT<2, ypN0, and estrogen receptor positivity as independent predictors of improved iDFS. Among patients with ypT<2, ypN0, and estrogen receptor-positive disease (34 out of 72 with estrogen receptor positivity), iDFS rates were notably high: 97 % (95 % CI: 91.3–100) at 3 years, and 90.9 % (95 % CI: 81.6–100) at both 5 and 7 years.</div></div><div><h3>Conclusion</h3><div>Patients without a pathologic complete response after neoadjuvant chemotherapy plus trastuzumab represent a heterogeneous group. Those with ypT<2, ypN0, and ER-positive residual disease may still achieve excellent iDFS and OS despite absence of adjuvant T-DM1.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"46 ","pages":"Article 101078"},"PeriodicalIF":2.4,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145910767","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 : 2026-01-01DOI: 10.1016/j.ctarc.2026.101095
Filip Ambrozkiewicz , Esraa Ali , Martina Bradova , Petr Slavík , Petr Martinek , Martin Svaton , Akseli Hemminki , Kari Hemminki
Limited DNA sequencing data on lung cancer (LC) patients are available from Eastern Europe where male smoking is commonest in Europe. As sequence analysis is the prerequisite for targeted therapy we provide such data from Czechia (CZ). Additionally, we present novel sequencing data for adenocarcinoma subtypes. Panel sequencing data on 1218 adenocarcinoma patients were available from a single histology laboratory for 2016 to 2024. The highest variant frequencies were observed for KRAS (51.6%), EGFR (18.8%) and TP53 (16.3%). Commonest types of variants were codon 12 mutations for KRAS, exon 21 L858R for EGFR and V600E BRAF. EGFR variants were more common in females (25.3% vs 11.5%). KRAS mutations were frequent in males (56.9% vs 46.9%), as were PIK3CA mutations (3.8% vs 1.9%). KRAS mutations were frequent in patients diagnosed below 70 years, whereas EGFR, PIK3CA and MET alterations were frequent in patients above age 70 years. As stage distinctions, high prevalence of KRAS mutations was found in early stage tumors (II and IIIA) and of TP53 mutations in stages IIIB and IV. As to adenocarcinoma subtypes, lepidic type showed a high frequency of EGFR variants. We could show differential distribution of gene variants by sex, diagnostic age, stage and subtype of adenocarcinoma. The data are in line with current literature that targeted treatment is being applied for selected LC patients in CZ. It can be expected that LC patients benefit from therapeutic and other clinical improvements, which however do not reduce the primary importance of anti-smoking campaigns.
来自东欧的肺癌(LC)患者的DNA测序数据有限,东欧男性吸烟是欧洲最常见的。由于序列分析是靶向治疗的先决条件,我们从捷克(CZ)提供了这些数据。此外,我们提出了新的腺癌亚型测序数据。2016年至2024年,1218名腺癌患者的小组测序数据来自单一组织学实验室。KRAS(51.6%)、EGFR(18.8%)和TP53(16.3%)的变异频率最高。最常见的变异类型是KRAS密码子12突变,EGFR外显子21 L858R突变和V600E BRAF突变。EGFR变异在女性中更为常见(25.3% vs 11.5%)。KRAS突变在男性中较为常见(56.9%对46.9%),PIK3CA突变在男性中较为常见(3.8%对1.9%)。KRAS突变常见于70岁以下的患者,而EGFR、PIK3CA和MET突变常见于70岁以上的患者。作为分期区分,KRAS突变在早期肿瘤(II期和IIIA期)中高发,TP53突变在IIIB期和IV期高发。在腺癌亚型中,lepidic型EGFR变异频率较高。我们可以显示基因变异在性别、诊断年龄、腺癌分期和亚型上的差异分布。数据与目前文献一致,CZ选择的LC患者正在应用靶向治疗。可以预期,LC患者受益于治疗和其他临床改善,然而,这并没有降低反吸烟运动的首要重要性。
{"title":"Mutational landscape of lung adenocarcinoma in Czechia","authors":"Filip Ambrozkiewicz , Esraa Ali , Martina Bradova , Petr Slavík , Petr Martinek , Martin Svaton , Akseli Hemminki , Kari Hemminki","doi":"10.1016/j.ctarc.2026.101095","DOIUrl":"10.1016/j.ctarc.2026.101095","url":null,"abstract":"<div><div>Limited DNA sequencing data on lung cancer (LC) patients are available from Eastern Europe where male smoking is commonest in Europe. As sequence analysis is the prerequisite for targeted therapy we provide such data from Czechia (CZ). Additionally, we present novel sequencing data for adenocarcinoma subtypes. Panel sequencing data on 1218 adenocarcinoma patients were available from a single histology laboratory for 2016 to 2024. The highest variant frequencies were observed for <em>KRAS</em> (51.6%), <em>EGFR</em> (18.8%) and <em>TP53</em> (16.3%). Commonest types of variants were codon 12 mutations for <em>KRAS</em>, exon 21 L858R for <em>EGFR</em> and V600E <em>BRAF. EGFR</em> variants were more common in females (25.3% vs 11.5%). <em>KRAS</em> mutations were frequent in males (56.9% vs 46.9%), as were <em>PIK3CA</em> mutations (3.8% vs 1.9%). <em>KRAS</em> mutations were frequent in patients diagnosed below 70 years, whereas <em>EGFR, PIK3CA</em> and <em>MET</em> alterations were frequent in patients above age 70 years. As stage distinctions, high prevalence of <em>KRAS</em> mutations was found in early stage tumors (II and IIIA) and of <em>TP53</em> mutations in stages IIIB and IV. As to adenocarcinoma subtypes, lepidic type showed a high frequency of <em>EGFR</em> variants. We could show differential distribution of gene variants by sex, diagnostic age, stage and subtype of adenocarcinoma. The data are in line with current literature that targeted treatment is being applied for selected LC patients in CZ. It can be expected that LC patients benefit from therapeutic and other clinical improvements, which however do not reduce the primary importance of anti-smoking campaigns.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"46 ","pages":"Article 101095"},"PeriodicalIF":2.4,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145965438","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 : 2026-01-01DOI: 10.1016/j.ctarc.2026.101108
Wen Zhang , Zhou Long , Xiaoli He , Xuemei Wu , Jian Wen , Dongfan Ye , Jia Chen , Guansong Wang
Introduction
Liver metastasis (LM) confers a poor prognosis in patients with epidermal growth factor receptor (EGFR)–mutant non–small cell lung cancer (NSCLC). Although EGFR tyrosine kinase inhibitors (TKIs) are the standard first-line treatment, the optimal approach for this high-risk subgroup remains uncertain. This study compared the efficacy and safety of EGFR-TKI monotherapy versus EGFR-TKI combined with chemotherapy in EGFR-mutant NSCLC patients with LM.
Patients and Methods
We retrospectively analyzed 98 patients with stage IV EGFR-mutant NSCLC and LM treated between 2020 and 2025. Fifty-nine patients received EGFR-TKI monotherapy, and 39 received EGFR-TKI plus platinum-based chemotherapy. Propensity score matching (PSM) was performed to balance baseline characteristics. Progression-free survival (PFS) and overall survival (OS) were estimated using the Kaplan–Meier method and compared by Cox regression. Safety profiles were analyzed according to CTCAE v5.0.
Results
After PSM, baseline characteristics were well balanced. The combination group achieved a higher objective response rate (59.0% vs 35.6%; P = .038), a higher disease control rate (100% vs 86.4%; P = .020), and a longer median PFS (14.0 vs 10.4 months; HR, 0.47; 95% CI, 0.26–0.86; P = .012). Median OS was 30.0 months with combination therapy versus 24.0 months with monotherapy (HR, 0.78; 95% CI, 0.39–1.55; P = .474). Severe (grade ≥3) adverse events were infrequent and comparable between groups, with no treatment-related deaths.
Conclusion
First-line EGFR-TKI plus chemotherapy significantly improved PFS and disease control without added toxicity, suggesting a promising therapeutic strategy for EGFR-mutant NSCLC with LM.
表皮生长因子受体(EGFR)突变的非小细胞肺癌(NSCLC)患者发生肝转移(LM)预后较差。虽然EGFR酪氨酸激酶抑制剂(TKIs)是标准的一线治疗方法,但对于这一高危亚群的最佳方法仍不确定。本研究比较了EGFR-TKI单药治疗与EGFR-TKI联合化疗治疗EGFR-TKI突变NSCLC LM患者的疗效和安全性。患者和方法我们回顾性分析了2020年至2025年间治疗的98例IV期egfr突变型NSCLC和LM患者。59例患者接受EGFR-TKI单药治疗,39例患者接受EGFR-TKI联合铂类化疗。采用倾向评分匹配(PSM)来平衡基线特征。采用Kaplan-Meier法估计无进展生存期(PFS)和总生存期(OS),并采用Cox回归进行比较。根据CTCAE v5.0对安全性概况进行分析。结果经PSM后,基线特征平衡良好。联合治疗组的客观有效率更高(59.0% vs 35.6%, P = 0.038),疾病控制率更高(100% vs 86.4%, P = 0.020),中位PFS更长(14.0 vs 10.4个月;HR, 0.47; 95% CI, 0.26-0.86; P = 0.012)。联合治疗的中位OS为30.0个月,而单药治疗的中位OS为24.0个月(HR, 0.78; 95% CI, 0.39-1.55; P = 0.474)。严重(≥3级)不良事件很少发生,组间具有可比性,无治疗相关死亡。结论一线EGFR-TKI联合化疗可显著改善PFS和疾病控制,且不增加毒性,提示egfr -突变型NSCLC合并LM的治疗策略很有前景。
{"title":"Efficacy and safety of first-line EGFR-TKI combined with chemotherapy versus EGFR-TKI monotherapy in patients with EGFR-mutant non-small cell lung cancer and liver metastases: A propensity score–matched analysis","authors":"Wen Zhang , Zhou Long , Xiaoli He , Xuemei Wu , Jian Wen , Dongfan Ye , Jia Chen , Guansong Wang","doi":"10.1016/j.ctarc.2026.101108","DOIUrl":"10.1016/j.ctarc.2026.101108","url":null,"abstract":"<div><h3>Introduction</h3><div>Liver metastasis (LM) confers a poor prognosis in patients with epidermal growth factor receptor (EGFR)–mutant non–small cell lung cancer (NSCLC). Although EGFR tyrosine kinase inhibitors (TKIs) are the standard first-line treatment, the optimal approach for this high-risk subgroup remains uncertain. This study compared the efficacy and safety of EGFR-TKI monotherapy versus EGFR-TKI combined with chemotherapy in EGFR-mutant NSCLC patients with LM.</div></div><div><h3>Patients and Methods</h3><div>We retrospectively analyzed 98 patients with stage IV EGFR-mutant NSCLC and LM treated between 2020 and 2025. Fifty-nine patients received EGFR-TKI monotherapy, and 39 received EGFR-TKI plus platinum-based chemotherapy. Propensity score matching (PSM) was performed to balance baseline characteristics. Progression-free survival (PFS) and overall survival (OS) were estimated using the Kaplan–Meier method and compared by Cox regression. Safety profiles were analyzed according to CTCAE v5.0.</div></div><div><h3>Results</h3><div>After PSM, baseline characteristics were well balanced. The combination group achieved a higher objective response rate (59.0% vs 35.6%; <em>P</em> = .038), a higher disease control rate (100% vs 86.4%; <em>P</em> = .020), and a longer median PFS (14.0 vs 10.4 months; HR, 0.47; 95% CI, 0.26–0.86; <em>P</em> = .012). Median OS was 30.0 months with combination therapy versus 24.0 months with monotherapy (HR, 0.78; 95% CI, 0.39–1.55; <em>P</em> = .474). Severe (grade ≥3) adverse events were infrequent and comparable between groups, with no treatment-related deaths.</div></div><div><h3>Conclusion</h3><div>First-line EGFR-TKI plus chemotherapy significantly improved PFS and disease control without added toxicity, suggesting a promising therapeutic strategy for EGFR-mutant NSCLC with LM.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"46 ","pages":"Article 101108"},"PeriodicalIF":2.4,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145973007","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 : 2026-01-01DOI: 10.1016/j.ctarc.2025.101067
Omid Yazdanpanah , Aditya Mahadevan , David J. Benjamin , Elham Vosoughi , Ali Raad , Madina Popal , Piyanuch Kongtim , Nataliya Mar , Arash Rezazadeh Kalebasty
Introduction
Treatment of metastatic renal cell carcinoma (mRCC) has expanded with development of combination therapies containing tyrosine kinase inhibitors (TKIs) with immune checkpoint inhibitors (ICI) agents or dual ICI agents. However, patient preferences may not necessarily be incorporated into management decisions. We aimed to understand patient preferences in the management of mRCC and compare it with oncologists’ perspectives.
Methods
A single-arm, prospective study surveyed patients with mRCC utilizing a questionnaire containing descriptions of 5 treatment options in simple language. Patients rated their preference for each option on a scale from 1 (least preferred) to 10 (most preferred). The same questionnaire plus basic de-identified patients' characteristics were provided to 2 academic oncologists and 1 community oncologist.
Results
A total of 54 patients were surveyed. The most preferred patient treatment option was TKI with mean score of 7.9 while the least popular was early phase clinical trials (CTs) with a mean score of 5.9 (p < 0.01). Patient’s employment status, speaking language, and denovo metastatic disease were the variables found to be associated with likelihood of picking a particular treatment option. Among oncologists, the most selected treatment options were enrollment in early and late phase CTs with mean scores of 7.61 and 7.52 respectively and single-agent TKI was least preferred with a mean score of 5.69 (p < 0.01). Age and performance status influenced oncologist therapy choices based on the multivariable analysis.
Conclusions
This study unveils differences between patients and oncologists’ treatment preferences for mRCC and underscores the importance of individualized discussion with each patient to evaluate his or her therapeutic objectives.
{"title":"Understanding Patient and Oncologist Preferences in the Management of Metastatic Renal Cell Carcinoma","authors":"Omid Yazdanpanah , Aditya Mahadevan , David J. Benjamin , Elham Vosoughi , Ali Raad , Madina Popal , Piyanuch Kongtim , Nataliya Mar , Arash Rezazadeh Kalebasty","doi":"10.1016/j.ctarc.2025.101067","DOIUrl":"10.1016/j.ctarc.2025.101067","url":null,"abstract":"<div><h3>Introduction</h3><div>Treatment of metastatic renal cell carcinoma (mRCC) has expanded with development of combination therapies containing tyrosine kinase inhibitors (TKIs) with immune checkpoint inhibitors (ICI) agents or dual ICI agents. However, patient preferences may not necessarily be incorporated into management decisions. We aimed to understand patient preferences in the management of mRCC and compare it with oncologists’ perspectives.</div></div><div><h3>Methods</h3><div>A single-arm, prospective study surveyed patients with mRCC utilizing a questionnaire containing descriptions of 5 treatment options in simple language. Patients rated their preference for each option on a scale from 1 (least preferred) to 10 (most preferred). The same questionnaire plus basic de-identified patients' characteristics were provided to 2 academic oncologists and 1 community oncologist.</div></div><div><h3>Results</h3><div>A total of 54 patients were surveyed. The most preferred patient treatment option was TKI with mean score of 7.9 while the least popular was early phase clinical trials (CTs) with a mean score of 5.9 (<em>p</em> < 0.01). Patient’s employment status, speaking language, and denovo metastatic disease were the variables found to be associated with likelihood of picking a particular treatment option. Among oncologists, the most selected treatment options were enrollment in early and late phase CTs with mean scores of 7.61 and 7.52 respectively and single-agent TKI was least preferred with a mean score of 5.69 (<em>p</em> < 0.01). Age and performance status influenced oncologist therapy choices based on the multivariable analysis.</div></div><div><h3>Conclusions</h3><div>This study unveils differences between patients and oncologists’ treatment preferences for mRCC and underscores the importance of individualized discussion with each patient to evaluate his or her therapeutic objectives.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"46 ","pages":"Article 101067"},"PeriodicalIF":2.4,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145909942","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 : 2026-01-01DOI: 10.1016/j.ctarc.2026.101099
Anke Richters , Mira D. Franken , Richard P. Meijer , Antoine G. van der Heijden , ProBCI Study Group, Franchette W.P.J. van den Berkmortel , Katja K.H. Aben
Objective
To describe and compare clinicopathological characteristics, treatment patterns and overall survival outcomes of contemporary patients diagnosed with synchronous (smBC) or metachronous metastatic bladder cancer (mmBC) in the Netherlands.
Methods
All patients newly diagnosed with bladder cancer in the Netherlands in 2020–2023 were identified through the Netherlands Cancer Registry. The smBC group consisted of patients with mBC at initial diagnosis in 2020 – 2023. The mmBC group included all patients who developed metastatic disease <3 years after being diagnosed with muscle-invasive bladder cancer (MIBC) between January 2020 – June 2022 and was stratified into early (≤6 months) and late mmBC (>6 months). Vital status was available until January 2025.
Results
The cohort included 2375 patients (1584 smBC, 371 early mmBC and 420 late mmBC). Lung and liver metastases were equally common across subcohorts; visceral disease was more common among mmBC patients. Less than half of patients received systemic therapy in all subcohorts (carboplatin most common for smBC and late mmBC patients; immunotherapy for early mmBC patients). Median OS regardless of treatment was 4.6, 3.9 and 7.4 months for smBC, early mmBC and late mmBC patients, respectively. Among those who received systemic treatment, median OS was 11.8, 8.5 and 12.6 months. Median OS differences were smaller when further restricting to systemic treatment-naïve patients.
Conclusions
Patient populations with smBC and early and late mmBC showed moderate differences in patient or disease characteristics that may be of clinical relevance. Many patients were unable to receive systemic treatment, with only small differences between groups, resulting from prior treatment, emphasizing the unmet need among both smBC and mmBC patients. Patients with mBC have a poor prognosis, irrespective of time to metastasis with limited OS differences.
{"title":"Treatments and survival outcomes of patients with synchronous and metachronous metastatic bladder cancer: A population-based nationwide cohort study","authors":"Anke Richters , Mira D. Franken , Richard P. Meijer , Antoine G. van der Heijden , ProBCI Study Group, Franchette W.P.J. van den Berkmortel , Katja K.H. Aben","doi":"10.1016/j.ctarc.2026.101099","DOIUrl":"10.1016/j.ctarc.2026.101099","url":null,"abstract":"<div><h3>Objective</h3><div>To describe and compare clinicopathological characteristics, treatment patterns and overall survival outcomes of contemporary patients diagnosed with synchronous (smBC) or metachronous metastatic bladder cancer (mmBC) in the Netherlands.</div></div><div><h3>Methods</h3><div>All patients newly diagnosed with bladder cancer in the Netherlands in 2020–2023 were identified through the Netherlands Cancer Registry. The smBC group consisted of patients with mBC at initial diagnosis in 2020 – 2023. The mmBC group included all patients who developed metastatic disease <3 years after being diagnosed with muscle-invasive bladder cancer (MIBC) between January 2020 – June 2022 and was stratified into early (≤6 months) and late mmBC (>6 months). Vital status was available until January 2025.</div></div><div><h3>Results</h3><div>The cohort included 2375 patients (1584 smBC, 371 early mmBC and 420 late mmBC). Lung and liver metastases were equally common across subcohorts; visceral disease was more common among mmBC patients. Less than half of patients received systemic therapy in all subcohorts (carboplatin most common for smBC and late mmBC patients; immunotherapy for early mmBC patients). Median OS regardless of treatment was 4.6, 3.9 and 7.4 months for smBC, early mmBC and late mmBC patients, respectively. Among those who received systemic treatment, median OS was 11.8, 8.5 and 12.6 months. Median OS differences were smaller when further restricting to systemic treatment-naïve patients.</div></div><div><h3>Conclusions</h3><div>Patient populations with smBC and early and late mmBC showed moderate differences in patient or disease characteristics that may be of clinical relevance. Many patients were unable to receive systemic treatment, with only small differences between groups, resulting from prior treatment, emphasizing the unmet need among both smBC and mmBC patients. Patients with mBC have a poor prognosis, irrespective of time to metastasis with limited OS differences.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"46 ","pages":"Article 101099"},"PeriodicalIF":2.4,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145972900","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}
Lung cancer is a leading cause of cancer-related mortality. While cisplatin is a cornerstone of chemotherapy, metformin (Glucophage) has shown anti-cancer potential. This study investigated the effects of cisplatin and metformin (Glucophage) on non-small cell lung cancer (NSCLC) cells, focusing on their combined impact on apoptosis-related genes and lncRNAs.
Methods
The A549 (NSCLC) and MRC5 (normal fibroblast) cell lines were treated with cisplatin, metformin (Glucophage), or a combination thereof. Cell viability was assessed by MTT assay, and apoptosis was quantified by flow cytometry. Expression of Bax, Bcl-2, Caspase 3, PVT1, and MEG3 was analyzed by qRT-PCR.
Results
Both cisplatin and metformin (Glucophage) individually reduced A549 cell viability in a dose-dependent manner. Combination Index (CI) analysis revealed an additive interaction (CI = 0.935) between the two agents. This combination was associated with an upregulation of pro-apoptotic Bax and lncRNA MEG3, and a downregulation of anti-apoptotic Bcl-2 and oncogenic lncRNA PVT1.
Conclusion
The combination of cisplatin and metformin (Glucophage) exerts an additive cytotoxic effect and induces apoptosis in NSCLC cells in vitro. This effect is associated with a favorable modulation of key apoptosis-regulating genes and lncRNAs. These findings provide a strong rationale for further mechanistic and preclinical investigation of this combination therapy for lung cancer.
{"title":"Pro-apoptotic effects of metformin and cisplatin in non-small cell Lung Cancer: Modulation of apoptosis-related genes and LncRNAs","authors":"Nazanin Sadeghi , Fatemeh Soleimanifar , Elmira Attar , Hamed Haddad Kashani , Romina Ghayoumi , Seyed-Alireza Etesami","doi":"10.1016/j.ctarc.2025.101069","DOIUrl":"10.1016/j.ctarc.2025.101069","url":null,"abstract":"<div><h3>Background</h3><div>Lung cancer is a leading cause of cancer-related mortality. While cisplatin is a cornerstone of chemotherapy, metformin (Glucophage) has shown anti-cancer potential. This study investigated the effects of cisplatin and metformin (Glucophage) on non-small cell lung cancer (NSCLC) cells, focusing on their combined impact on apoptosis-related genes and lncRNAs.</div></div><div><h3>Methods</h3><div>The A549 (NSCLC) and MRC5 (normal fibroblast) cell lines were treated with cisplatin, metformin (Glucophage), or a combination thereof. Cell viability was assessed by MTT assay, and apoptosis was quantified by flow cytometry. Expression of <em>Bax, Bcl-2, Caspase 3, PVT1</em>, and <em>MEG3</em> was analyzed by qRT-PCR.</div></div><div><h3>Results</h3><div>Both cisplatin and metformin (Glucophage) individually reduced A549 cell viability in a dose-dependent manner. Combination Index (CI) analysis revealed an additive interaction (CI = 0.935) between the two agents. This combination was associated with an upregulation of pro-apoptotic <em>Bax</em> and lncRNA <em>MEG3</em>, and a downregulation of anti-apoptotic <em>Bcl-2</em> and oncogenic lncRNA <em>PVT1</em>.</div></div><div><h3>Conclusion</h3><div>The combination of cisplatin and metformin (Glucophage) exerts an additive cytotoxic effect and induces apoptosis in NSCLC cells <em>in vitro</em>. This effect is associated with a favorable modulation of key apoptosis-regulating genes and lncRNAs. These findings provide a strong rationale for further mechanistic and preclinical investigation of this combination therapy for lung cancer.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"46 ","pages":"Article 101069"},"PeriodicalIF":2.4,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145909962","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 : 2026-01-01DOI: 10.1016/j.ctarc.2025.101081
Jessica Vadaketh , Jake Konigsberg , Omar Elghawy , Kevin Dinh , Linda Zhu , Julia Youngman , Michaela Dungan , Marya Pulaski , Jessica M. Long , Kole H. Buckley , Bryson W. Katona
Background
Upper gastrointestinal surveillance in Lynch syndrome (LS) remains an ongoing debate; some guidelines recommend upper endoscopy with non-targeted biopsies of the gastric antrum/body to detect Helicobacter pylori (HP) and/or gastric intestinal metaplasia (GIM). However, whether non-targeted gastric biopsies should be repeated on successive upper endoscopies remains uncertain. Therefore, we aimed to determine the yield of repeat non-targeted gastric antrum/body biopsies and assess the HP seropositivity of a LS cohort.
Methods
Clinical and pathology data were collected retrospectively from LS carriers who underwent upper endoscopy with non-targeted gastric biopsies. Plasma samples from a LS biobank were tested for HP IgG positivity.
Results
Amongst 683 LS carriers, there were 291 (43 %) with 1+, 145 (21 %) with 2+, and 45 (7 %) with 3+ upper endoscopies with non-targeted gastric antrum and body biopsies performed. The overall prevalence of GIM on those endoscopies was 8 % and the rate of HP was 3 %. Of individuals without GIM detected on the initial upper endoscopy, 4 % had GIM identified on their second, and 2 % had GIM identified on a third or greater upper endoscopy after having two prior upper endoscopies without GIM identified. There was no additional HP identified on subsequent endoscopies. Plasma HP IgG positivity amongst 257 LS carriers was 14 %.
Conclusions
Amongst a LS cohort undergoing serial upper endoscopies, repeat gastric antrum/body biopsies yielded new cases of GIM, providing support for consideration of non-targeted gastric biopsies on all upper endoscopies performed in LS. Additionally, although endoscopic HP detection rates are low, HP exposure in LS is more common.
{"title":"Yield of repeat gastric biopsies and Helicobacter pylori serological assessment in Lynch syndrome","authors":"Jessica Vadaketh , Jake Konigsberg , Omar Elghawy , Kevin Dinh , Linda Zhu , Julia Youngman , Michaela Dungan , Marya Pulaski , Jessica M. Long , Kole H. Buckley , Bryson W. Katona","doi":"10.1016/j.ctarc.2025.101081","DOIUrl":"10.1016/j.ctarc.2025.101081","url":null,"abstract":"<div><h3>Background</h3><div>Upper gastrointestinal surveillance in Lynch syndrome (LS) remains an ongoing debate; some guidelines recommend upper endoscopy with non-targeted biopsies of the gastric antrum/body to detect <em>Helicobacter pylori</em> (<em>HP</em>) and/or gastric intestinal metaplasia (GIM). However, whether non-targeted gastric biopsies should be repeated on successive upper endoscopies remains uncertain. Therefore, we aimed to determine the yield of repeat non-targeted gastric antrum/body biopsies and assess the <em>HP</em> seropositivity of a LS cohort.</div></div><div><h3>Methods</h3><div>Clinical and pathology data were collected retrospectively from LS carriers who underwent upper endoscopy with non-targeted gastric biopsies. Plasma samples from a LS biobank were tested for <em>HP</em> IgG positivity.</div></div><div><h3>Results</h3><div>Amongst 683 LS carriers, there were 291 (43 %) with 1+, 145 (21 %) with 2+, and 45 (7 %) with 3+ upper endoscopies with non-targeted gastric antrum and body biopsies performed. The overall prevalence of GIM on those endoscopies was 8 % and the rate of <em>HP</em> was 3 %. Of individuals without GIM detected on the initial upper endoscopy, 4 % had GIM identified on their second, and 2 % had GIM identified on a third or greater upper endoscopy after having two prior upper endoscopies without GIM identified. There was no additional <em>HP</em> identified on subsequent endoscopies. Plasma <em>HP</em> IgG positivity amongst 257 LS carriers was 14 %.</div></div><div><h3>Conclusions</h3><div>Amongst a LS cohort undergoing serial upper endoscopies, repeat gastric antrum/body biopsies yielded new cases of GIM, providing support for consideration of non-targeted gastric biopsies on all upper endoscopies performed in LS. Additionally, although endoscopic <em>HP</em> detection rates are low, <em>HP</em> exposure in LS is more common.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"46 ","pages":"Article 101081"},"PeriodicalIF":2.4,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145909937","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 : 2026-01-01DOI: 10.1016/j.ctarc.2026.101100
Yuanqing Yang, Xiaobing Li, Yongqiang Yin
Bone metastasis is one of the most common complications in patients with advanced breast cancer, severely impairing quality of life and significantly increasing the risk of mortality. This process is driven by complex interactions between tumor cells and the bone microenvironment, where non-coding RNAs (ncRNAs) act as key regulatory molecules playing a central role. This article systematically reviews the expression changes, molecular mechanisms, and biological functions of microRNAs, long non-coding RNAs, and circular RNAs in breast cancer bone metastasis. Studies have demonstrated that these ncRNAs precisely regulate tumor cell colonization and proliferation in bone, osteoclast-mediated bone resorption, and osteoblast-mediated bone formation through multiple mechanisms — including competitive endogenous RNA networks, signaling pathway modulation, exosome-mediated intercellular communication, and even the encoding of functional short peptides — thereby disrupting bone homeostasis. Simultaneously, ncRNAs can also regulate the occurrence of bone metastasis by modulating the immune microenvironment in breast cancer. Furthermore, ncRNAs exhibit significant clinical translational potential in the early diagnosis, prognostic evaluation, and targeted therapy of breast cancer bone metastasis. By synthesizing the ncRNA regulatory networks, this review aims to provide a theoretical basis and research directions for further exploring the molecular mechanisms of breast cancer bone metastasis and developing novel therapeutic strategies.
{"title":"Non-Coding RNAs in breast cancer bone metastasis: Regulators and novel therapeutic targets","authors":"Yuanqing Yang, Xiaobing Li, Yongqiang Yin","doi":"10.1016/j.ctarc.2026.101100","DOIUrl":"10.1016/j.ctarc.2026.101100","url":null,"abstract":"<div><div>Bone metastasis is one of the most common complications in patients with advanced breast cancer, severely impairing quality of life and significantly increasing the risk of mortality. This process is driven by complex interactions between tumor cells and the bone microenvironment, where non-coding RNAs (ncRNAs) act as key regulatory molecules playing a central role. This article systematically reviews the expression changes, molecular mechanisms, and biological functions of microRNAs, long non-coding RNAs, and circular RNAs in breast cancer bone metastasis. Studies have demonstrated that these ncRNAs precisely regulate tumor cell colonization and proliferation in bone, osteoclast-mediated bone resorption, and osteoblast-mediated bone formation through multiple mechanisms — including competitive endogenous RNA networks, signaling pathway modulation, exosome-mediated intercellular communication, and even the encoding of functional short peptides — thereby disrupting bone homeostasis. Simultaneously, ncRNAs can also regulate the occurrence of bone metastasis by modulating the immune microenvironment in breast cancer. Furthermore, ncRNAs exhibit significant clinical translational potential in the early diagnosis, prognostic evaluation, and targeted therapy of breast cancer bone metastasis. By synthesizing the ncRNA regulatory networks, this review aims to provide a theoretical basis and research directions for further exploring the molecular mechanisms of breast cancer bone metastasis and developing novel therapeutic strategies.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"46 ","pages":"Article 101100"},"PeriodicalIF":2.4,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145973008","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}
This study investigateted the malignant transformation of endometriosis (EMS) into ovarian clear cell carcinoma (OCCC) using spatial transcriptomics and single-cell RNA sequencing (scRNA-seq) integration. Tissues with coexisting EMS and OCCC regions were analyzed to elucidate their molecular connections. Spatial transcriptomic profiling revealed shared molecular features between OCCC and EMS, including overexpression of genes related to tissue development and apoptosis. Integration with scRNA-seq data revealed that severe uterine EMS exhibited greater transcriptomic similarities to OCCC than to non-EAOC ovarian cancer subtypes, such as high-grade serous ovarian cancer (HGSOC). Further analysis classified the OCCC-specific genes into EMS epithelial cell-specific and tumor microenvironment (TME)-related categories, highlighting their roles in pathways associated with tumor progression, invasion, and metabolic reprogramming. These findings support the transcriptomic progression pathway from EMS to OCCC, and advance our understanding of the etiology of OCCC, suggesting strategies for targeted therapy and improved diagnostics.
{"title":"Transcriptomic landscape of coexisting ovarian clear cell carcinoma and endometriosis: Insights into malignant transformation","authors":"Mami Shibata , Yuji Kamei , Keisuke Kawamoto , Reiji Muto , Yutaka Ueda , Makoto Hamasaki , Koichi Ohshima , Fusanori Yotsumoto","doi":"10.1016/j.ctarc.2026.101098","DOIUrl":"10.1016/j.ctarc.2026.101098","url":null,"abstract":"<div><div>This study investigateted the malignant transformation of endometriosis (EMS) into ovarian clear cell carcinoma (OCCC) using spatial transcriptomics and single-cell RNA sequencing (scRNA-seq) integration. Tissues with coexisting EMS and OCCC regions were analyzed to elucidate their molecular connections. Spatial transcriptomic profiling revealed shared molecular features between OCCC and EMS, including overexpression of genes related to tissue development and apoptosis. Integration with scRNA-seq data revealed that severe uterine EMS exhibited greater transcriptomic similarities to OCCC than to non-EAOC ovarian cancer subtypes, such as high-grade serous ovarian cancer (HGSOC). Further analysis classified the OCCC-specific genes into EMS epithelial cell-specific and tumor microenvironment (TME)-related categories, highlighting their roles in pathways associated with tumor progression, invasion, and metabolic reprogramming. These findings support the transcriptomic progression pathway from EMS to OCCC, and advance our understanding of the etiology of OCCC, suggesting strategies for targeted therapy and improved diagnostics.</div></div>","PeriodicalId":9507,"journal":{"name":"Cancer treatment and research communications","volume":"46 ","pages":"Article 101098"},"PeriodicalIF":2.4,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145965466","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}