Pub Date : 2026-01-01DOI: 10.21873/anticanres.17930
Jinsoo Kim, Qinghong Han, Shukuan Li, Byung Mo Kang, Kohei Mizuta, Yohei Asano, Yuta Miyashi, Michael Bouvet, Robert M Hoffman
Background/aim: Methionine addiction is a fundamental and general hallmark of cancer, known as the Hoffman effect. The aim of the present study was to target methionine addiction of cancer cells co-cultured with normal cells with various doses of recombinant methioninase (rMETase) to precisely eliminate cancer cells and to determine the timing of rescue of cancer cells by methionine supplementation.
Materials and methods: HCT116 human colon cancer cells were co-cultured with Hs-27 diploid human normal fibroblasts in 12-well tissue-culture plates containing Dulbecco's Modified Eagle's Medium (DMEM) with 10% fetal bovine serum. rMETase was added to the co-cultures at 0.25, 0.5, or 1.0 U/ml. In attempts to rescue the cancer cells, normal DMEM which contains 0.2 mM of methionine, replaced DMEM containing rMETase on days -2, -4 or -6. Co-cultures were observed for cell viability and proliferation using phase-contrast microscopy.
Results: The efficacy of rMETase to eliminate cancer cells co-cultured with normal cells was dose-dependent. rMETase at 0.25 U/ml allowed large numbers of cancer cells to remain in the co-cultures by day 12. rMETase at 0.5 U/ml and 1.0 U/ml largely eliminated the cancer cells from the co-cultures by day 12. rMETase at 1.0 U/ml was slightly toxic to the normal cells. However, at each dose of rMETase, even at 1.0 U/ml, DMEM could rescue the cancer cells even when added on day 6.
Conclusion: The ability of rMETase to eliminate cancer cells co-cultured with normal cells is dose-dependent. Even at high effective doses of rMETase, the cancer cells could be rescued by methionine supplementation up to at least day-6, indicating the need for the continuous presence of rMETase to selectively inhibit the cancer cells.
{"title":"Effect of Recombinant Methioninase Dose and Timing on the Selective and Precise Elimination of Cancer Cells from Co-Cultured Normal Cells and on Methionine-dependent Rescue of Cancer Cells.","authors":"Jinsoo Kim, Qinghong Han, Shukuan Li, Byung Mo Kang, Kohei Mizuta, Yohei Asano, Yuta Miyashi, Michael Bouvet, Robert M Hoffman","doi":"10.21873/anticanres.17930","DOIUrl":"https://doi.org/10.21873/anticanres.17930","url":null,"abstract":"<p><strong>Background/aim: </strong>Methionine addiction is a fundamental and general hallmark of cancer, known as the Hoffman effect. The aim of the present study was to target methionine addiction of cancer cells co-cultured with normal cells with various doses of recombinant <i>methioninase</i> (rMETase) to precisely eliminate cancer cells and to determine the timing of rescue of cancer cells by methionine supplementation.</p><p><strong>Materials and methods: </strong>HCT116 human colon cancer cells were co-cultured with Hs-27 diploid human normal fibroblasts in 12-well tissue-culture plates containing Dulbecco's Modified Eagle's Medium (DMEM) with 10% fetal bovine serum. rMETase was added to the co-cultures at 0.25, 0.5, or 1.0 U/ml. In attempts to rescue the cancer cells, normal DMEM which contains 0.2 mM of methionine, replaced DMEM containing rMETase on days -2, -4 or -6. Co-cultures were observed for cell viability and proliferation using phase-contrast microscopy.</p><p><strong>Results: </strong>The efficacy of rMETase to eliminate cancer cells co-cultured with normal cells was dose-dependent. rMETase at 0.25 U/ml allowed large numbers of cancer cells to remain in the co-cultures by day 12. rMETase at 0.5 U/ml and 1.0 U/ml largely eliminated the cancer cells from the co-cultures by day 12. rMETase at 1.0 U/ml was slightly toxic to the normal cells. However, at each dose of rMETase, even at 1.0 U/ml, DMEM could rescue the cancer cells even when added on day 6.</p><p><strong>Conclusion: </strong>The ability of rMETase to eliminate cancer cells co-cultured with normal cells is dose-dependent. Even at high effective doses of rMETase, the cancer cells could be rescued by methionine supplementation up to at least day-6, indicating the need for the continuous presence of rMETase to selectively inhibit the cancer cells.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"46 1","pages":"143-151"},"PeriodicalIF":1.7,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145861825","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 : 2026-01-01DOI: 10.21873/anticanres.17928
Bo-Hyung Kim, Jiyoon Lee, Donghwan Lee, Kwoneel Kim, Jung-Won Kim, Boram Ha, Hyun Joo Jang, Mee Soo Chang, Eun Shin
Background/aim: Response to neoadjuvant chemoradiotherapy (nCRT) in rectal cancer varies. Recent studies have highlighted the role of the tumor immune microenvironment in influencing tumor behavior. Herein, we aimed to assess immune-related gene expression in rectal cancer following nCRT and to investigate their potential as predictive and prognostic biomarkers.
Materials and methods: Expression profiling of 730 immune-related genes was conducted in 48 post-nCRT rectal cancer using the NanoString nCounter platform and the PanCancer Immune Profiling panel. Differentially expressed genes were compared between good and poor responders, and gene set enrichment analysis was conducted. The prognostic significance of these genes was analyzed. A genetic model was generated to predict nCRT responses.
Results: We identified 24 immune-associated genes that were differentially expressed between good and poor responders, among which S100A8, SPINK5, ANXA1, FOXJ1, and CLEC7A showed high expression levels in good responders (Log2 fold change >1, p<0.05). Pathway analysis revealed that these genes were mainly involved in biological process associated with natural killer cell-mediated cytotoxicity. S100A8 and SPINK5 expression levels were associated with relapse-free survival (p=0.001 and 0.036, respectively), and these findings were validated in a publicly available dataset (S100A8; p=0.015, and SPINK5; p=0.024). The accuracy of the predictive model comprising TLR4, CCND3, TCF7, CREB5, TNFRSF10B, DPP4, PBK, DUSP4, and MUC1 was 85.7%.
Conclusion: Immune-related gene expression patterns are associated with response to nCRT in rectal cancer. High expression levels of S100A8, SPINK5, ANXA1, FOXJ1, and CLEC7A were indicative of favorable treatment response, and S100A8 and SPINK5 were associated with prognosis. A machine learning-based model composed of immune-related genes showed strong predictive potential. Our results support the use of immune gene signatures to guide personalized therapy in rectal cancer.
{"title":"Immune Microenvironment Signatures Predict Response and Survival in Rectal Cancer Patients After Neoadjuvant Chemoradiation.","authors":"Bo-Hyung Kim, Jiyoon Lee, Donghwan Lee, Kwoneel Kim, Jung-Won Kim, Boram Ha, Hyun Joo Jang, Mee Soo Chang, Eun Shin","doi":"10.21873/anticanres.17928","DOIUrl":"https://doi.org/10.21873/anticanres.17928","url":null,"abstract":"<p><strong>Background/aim: </strong>Response to neoadjuvant chemoradiotherapy (nCRT) in rectal cancer varies. Recent studies have highlighted the role of the tumor immune microenvironment in influencing tumor behavior. Herein, we aimed to assess immune-related gene expression in rectal cancer following nCRT and to investigate their potential as predictive and prognostic biomarkers.</p><p><strong>Materials and methods: </strong>Expression profiling of 730 immune-related genes was conducted in 48 post-nCRT rectal cancer using the NanoString nCounter platform and the PanCancer Immune Profiling panel. Differentially expressed genes were compared between good and poor responders, and gene set enrichment analysis was conducted. The prognostic significance of these genes was analyzed. A genetic model was generated to predict nCRT responses.</p><p><strong>Results: </strong>We identified 24 immune-associated genes that were differentially expressed between good and poor responders, among which <i>S100A8, SPINK5, ANXA1, FOXJ1</i>, and <i>CLEC7A</i> showed high expression levels in good responders (Log2 fold change >1, <i>p</i><0.05). Pathway analysis revealed that these genes were mainly involved in biological process associated with natural killer cell-mediated cytotoxicity. <i>S100A8</i> and <i>SPINK5</i> expression levels were associated with relapse-free survival (<i>p</i>=0.001 and 0.036, respectively), and these findings were validated in a publicly available dataset (<i>S100A8</i>; <i>p</i>=0.015, and <i>SPINK5</i>; <i>p</i>=0.024). The accuracy of the predictive model comprising <i>TLR4, CCND3, TCF7, CREB5, TNFRSF10B, DPP4, PBK, DUSP4</i>, and <i>MUC1</i> was 85.7%.</p><p><strong>Conclusion: </strong>Immune-related gene expression patterns are associated with response to nCRT in rectal cancer. High expression levels of <i>S100A8, SPINK5, ANXA1, FOXJ1</i>, and <i>CLEC7A</i> were indicative of favorable treatment response, and <i>S100A8</i> and <i>SPINK5</i> were associated with prognosis. A machine learning-based model composed of immune-related genes showed strong predictive potential. Our results support the use of immune gene signatures to guide personalized therapy in rectal cancer.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"46 1","pages":"123-134"},"PeriodicalIF":1.7,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145861828","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 : 2026-01-01DOI: 10.21873/anticanres.17935
Hong-Beum Kim, Yong Sub Na, Seong-Hun Kim, Hee-Jeong Lee, Sang-Gon Park
Background/aim: Small-cell lung cancer (SCLC) is an aggressive malignancy with limited therapeutic progress. Adenylate kinase 2 (AK2), a mitochondrial isoenzyme involved in nucleotide metabolism and apoptosis regulation, has been implicated in tumorigenesis, but its role in SCLC remains undefined. This study aimed to investigate the oncogenic function of AK2 in SCLC and its regulatory mechanism through the PI3K/AKT/mTOR signaling pathway.
Materials and methods: AK2 expression was analyzed using the GSE60052 dataset and validated using western blotting in SCLC cell lines. Functional assays, including MTT, proliferation, colony formation, and phospho-kinase array profiling, were performed following siRNA-mediated AK2 knockdown in HCC-33 and H417 cells. Western blotting was used to confirm pathway alterations. In vivo tumorigenicity was assessed in xenograft models.
Results: AK2 was markedly up-regulated in SCLC tissues and cell lines. Silencing AK2 significantly inhibited proliferation and clonogenic growth of SCLC cells and reduced tumorigenicity in vivo. Phospho-kinase analysis and western blot validation demonstrated that AK2 knockdown altered phosphorylation of key components in the PI3K/AKT/mTOR pathway, including AKT and mTOR.
Conclusion: AK2 acts as an oncogenic driver in SCLC, promoting tumor progression through PI3K/AKT/mTOR signaling. Targeting AK2 may represent a novel therapeutic strategy and predictive biomarker in SCLC management.
{"title":"Adenylate Kinase 2 Promotes Tumor Progression in Small-cell Lung Cancer <i>via</i> PI3K/AKT/mTOR Signaling.","authors":"Hong-Beum Kim, Yong Sub Na, Seong-Hun Kim, Hee-Jeong Lee, Sang-Gon Park","doi":"10.21873/anticanres.17935","DOIUrl":"https://doi.org/10.21873/anticanres.17935","url":null,"abstract":"<p><strong>Background/aim: </strong>Small-cell lung cancer (SCLC) is an aggressive malignancy with limited therapeutic progress. Adenylate kinase 2 (AK2), a mitochondrial isoenzyme involved in nucleotide metabolism and apoptosis regulation, has been implicated in tumorigenesis, but its role in SCLC remains undefined. This study aimed to investigate the oncogenic function of AK2 in SCLC and its regulatory mechanism through the PI3K/AKT/mTOR signaling pathway.</p><p><strong>Materials and methods: </strong>AK2 expression was analyzed using the GSE60052 dataset and validated using western blotting in SCLC cell lines. Functional assays, including MTT, proliferation, colony formation, and phospho-kinase array profiling, were performed following siRNA-mediated AK2 knockdown in HCC-33 and H417 cells. Western blotting was used to confirm pathway alterations. <i>In vivo</i> tumorigenicity was assessed in xenograft models.</p><p><strong>Results: </strong>AK2 was markedly up-regulated in SCLC tissues and cell lines. Silencing AK2 significantly inhibited proliferation and clonogenic growth of SCLC cells and reduced tumorigenicity <i>in vivo</i>. Phospho-kinase analysis and western blot validation demonstrated that AK2 knockdown altered phosphorylation of key components in the PI3K/AKT/mTOR pathway, including AKT and mTOR.</p><p><strong>Conclusion: </strong>AK2 acts as an oncogenic driver in SCLC, promoting tumor progression through PI3K/AKT/mTOR signaling. Targeting AK2 may represent a novel therapeutic strategy and predictive biomarker in SCLC management.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"46 1","pages":"205-212"},"PeriodicalIF":1.7,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145861838","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}
Background/aim: Regorafenib, a multikinase inhibitor widely used to treat metastatic colorectal cancer (mCRC), is associated with elevations in pancreatic enzyme levels. However, clinical predictors of this adverse event have yet to be adequately defined. This study aimed to identify risk factors for regorafenib-induced enzyme elevation in routine clinical practice.
Patients and methods: We retrospectively evaluated 47 patients with mCRC who received regorafenib between May 2013 and October 2024. Pancreatic enzyme elevation was defined as a Common Terminology Criteria for Adverse Events (CTCAE) grade ≥1 increase in serum lipase or amylase levels within 28 days of treatment initiation. Clinical data were extracted from medical records, and risk factors were assessed using univariate and multivariate logistic regression. Sensitivity analyses were performed to confirm the robustness of the findings.
Results: Lipase elevation occurred in 48.9% of patients, whereas amylase elevation was observed in 8.5% of patients. Severe enzyme elevation (CTCAE grade ≥3) was detected in 17.0% and exclusively involved lipase. The median onset time was 7 days, with no cases of acute pancreatitis reported. Multivariate analysis identified prior anti-vascular endothelial growth factor (VEGF) therapy lasting ≥300 days as an independent risk factor (adjusted odds ratio 5.99, 95% confidence interval 1.49-31.41, p=0.01). Sensitivity analyses supported this association.
Conclusion: The elevation of pancreatic enzymes, predominantly lipase, is a frequent early adverse event associated with regorafenib treatment. A history of prolonged anti-VEGF therapy may predispose patients to toxicity, highlighting the need for close monitoring during the initial treatment phase. These findings provide novel insights that may help to optimize the safe clinical use of regorafenib.
{"title":"Evaluation of Regorafenib-induced Pancreatic Enzyme Elevation in Metastatic Colorectal Cancer Treatment.","authors":"Ko-Ichi Ohtaki, Riu Takeuchi, Kuninori Iwayama, Masayuki Chuma, Yoshikazu Tasaki, Yoshitaka Saito","doi":"10.21873/anticanres.17950","DOIUrl":"https://doi.org/10.21873/anticanres.17950","url":null,"abstract":"<p><strong>Background/aim: </strong>Regorafenib, a multikinase inhibitor widely used to treat metastatic colorectal cancer (mCRC), is associated with elevations in pancreatic enzyme levels. However, clinical predictors of this adverse event have yet to be adequately defined. This study aimed to identify risk factors for regorafenib-induced enzyme elevation in routine clinical practice.</p><p><strong>Patients and methods: </strong>We retrospectively evaluated 47 patients with mCRC who received regorafenib between May 2013 and October 2024. Pancreatic enzyme elevation was defined as a Common Terminology Criteria for Adverse Events (CTCAE) grade ≥1 increase in serum lipase or amylase levels within 28 days of treatment initiation. Clinical data were extracted from medical records, and risk factors were assessed using univariate and multivariate logistic regression. Sensitivity analyses were performed to confirm the robustness of the findings.</p><p><strong>Results: </strong>Lipase elevation occurred in 48.9% of patients, whereas amylase elevation was observed in 8.5% of patients. Severe enzyme elevation (CTCAE grade ≥3) was detected in 17.0% and exclusively involved lipase. The median onset time was 7 days, with no cases of acute pancreatitis reported. Multivariate analysis identified prior anti-vascular endothelial growth factor (VEGF) therapy lasting ≥300 days as an independent risk factor (adjusted odds ratio 5.99, 95% confidence interval 1.49-31.41, <i>p</i>=0.01). Sensitivity analyses supported this association.</p><p><strong>Conclusion: </strong>The elevation of pancreatic enzymes, predominantly lipase, is a frequent early adverse event associated with regorafenib treatment. A history of prolonged anti-VEGF therapy may predispose patients to toxicity, highlighting the need for close monitoring during the initial treatment phase. These findings provide novel insights that may help to optimize the safe clinical use of regorafenib.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"46 1","pages":"359-369"},"PeriodicalIF":1.7,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145861863","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 : 2026-01-01DOI: 10.21873/anticanres.17929
Yuta Miyashi, Kohei Mizuta, Yohei Asano, Byung Mo Kang, Jin Soo Kim, Qinghong Han, Shukuan Li, Michael Bouvet, Yasunori Tome, Kotaro Nishida, Robert M Hoffman
Background/aim: Recently, there have been numerous publications on the induction of ferroptosis by cysteine restriction in cancer cells. The present report aimed to determine whether cysteine restriction (CR) is a cancer-specific vulnerability in comparison with methionine restriction (MR), which is a known cancer-specific vulnerability.
Materials and methods: Human cancer cell lines (HCT116 colon cancer, 143B osteosarcoma or HT1080 fibrosarcoma) and normal human fibroblasts (Hs27) were cultured in Dulbecco's modified Eagle's medium (DMEM) with dialyzed fetal bovine serum from which methionine or cysteine or both or neither had been depleted. Cancer and normal cells were co-cultured in 12-well plates under the above conditions. HCT116 cells expressing green fluorescent protein, and 143B and HT1080 cells expressing red fluorescent protein, were visualized by fluorescence microscopy. Normal fibroblasts and cancer cells were visualized by phase-contrast microscopy as well.
Results: In co-culture, of either 143B, HCT116 or HT1080 with Hs27 human fibrosarcoma, CR was toxic to Hs27 normal fibroblasts as well as to all three cancer cell lines. In contrast, MR was toxic only to the cancer cells but not normal fibroblasts. Dual CR and MR was toxic to normal and cancer cells.
Conclusion: For all three cancer cell lines, HCT116 colon cancer, HT1080 fibrosarcoma and 143B osteosarcoma, both MR and CR were highly inhibitory in the co-cultures with Hs27 normal fibroblasts. In all cases MR had only a slight effect on normal fibroblasts, but CR was highly toxic to normal fibroblasts. Thus, MR is a cancer-specific vulnerability in contrast to CR which is toxic to both normal and cancer cells and is not a cancer-specificity vulnerability. Therefore, attempting to induce ferroptosis of cancer cells by CR does not appear to have potential as an effective cancer therapy.
{"title":"Methionine Restriction, Not Cysteine Restriction, Is a Cancer-specific Vulnerability.","authors":"Yuta Miyashi, Kohei Mizuta, Yohei Asano, Byung Mo Kang, Jin Soo Kim, Qinghong Han, Shukuan Li, Michael Bouvet, Yasunori Tome, Kotaro Nishida, Robert M Hoffman","doi":"10.21873/anticanres.17929","DOIUrl":"https://doi.org/10.21873/anticanres.17929","url":null,"abstract":"<p><strong>Background/aim: </strong>Recently, there have been numerous publications on the induction of ferroptosis by cysteine restriction in cancer cells. The present report aimed to determine whether cysteine restriction (CR) is a cancer-specific vulnerability in comparison with methionine restriction (MR), which is a known cancer-specific vulnerability.</p><p><strong>Materials and methods: </strong>Human cancer cell lines (HCT116 colon cancer, 143B osteosarcoma or HT1080 fibrosarcoma) and normal human fibroblasts (Hs27) were cultured in Dulbecco's modified Eagle's medium (DMEM) with dialyzed fetal bovine serum from which methionine or cysteine or both or neither had been depleted. Cancer and normal cells were co-cultured in 12-well plates under the above conditions. HCT116 cells expressing green fluorescent protein, and 143B and HT1080 cells expressing red fluorescent protein, were visualized by fluorescence microscopy. Normal fibroblasts and cancer cells were visualized by phase-contrast microscopy as well.</p><p><strong>Results: </strong>In co-culture, of either 143B, HCT116 or HT1080 with Hs27 human fibrosarcoma, CR was toxic to Hs27 normal fibroblasts as well as to all three cancer cell lines. In contrast, MR was toxic only to the cancer cells but not normal fibroblasts. Dual CR and MR was toxic to normal and cancer cells.</p><p><strong>Conclusion: </strong>For all three cancer cell lines, HCT116 colon cancer, HT1080 fibrosarcoma and 143B osteosarcoma, both MR and CR were highly inhibitory in the co-cultures with Hs27 normal fibroblasts. In all cases MR had only a slight effect on normal fibroblasts, but CR was highly toxic to normal fibroblasts. Thus, MR is a cancer-specific vulnerability in contrast to CR which is toxic to both normal and cancer cells and is not a cancer-specificity vulnerability. Therefore, attempting to induce ferroptosis of cancer cells by CR does not appear to have potential as an effective cancer therapy.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"46 1","pages":"135-141"},"PeriodicalIF":1.7,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145861936","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 : 2026-01-01DOI: 10.21873/anticanres.17946
Wataru Goto, Shinichiro Kashiwagi, Mariko Nishikawa, Chika Watanabe, Koji Takada, Yukie Tauchi, Kana Ogisawa, Masatsune Shibutani, Haruhito Kinoshita, Tamami Morisaki
Background/aim: Endocrine therapy combined with cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) is standard first-line treatment for hormone receptor (HR)-positive metastatic breast cancer (MBC). Although radiological complete response (rCR) is rarely achieved, the optimal management of such patients remains unclear. In human epidermal growth factor receptor 2 (HER2)-positive MBC, discontinuation of anti-HER2 therapy after rCR has been explored but treatment de-escalation in HR-positive MBC has not been sufficiently investigated.
Patients and methods: We retrospectively analyzed 178 patients with HR-positive MBC treated with CDK4/6i-based endocrine therapy at our Institution between December 2017 and October 2024. Clinicopathological factors, including immune-related markers (absolute lymphocyte count and neutrophil-to-lymphocyte ratio), were evaluated for associations with rCR. Bone metastases were considered rCR-equivalent only if radiological stability was maintained for ≥6 months. Patients who discontinued CDK4/6i after achieving rCR were further assessed.
Results: Nineteen patients (10.7%) achieved rCR (13 with measurable non-bone metastases and six with bone metastases). The median time to rCR was 11.9 months. rCR occurred more frequently in patients treated with abemaciclib (p=0.008) and those with smaller primary tumors (≤2 cm, p=0.040). Earlier-line treatment also showed a trend towards achieving higher rCR rates (p=0.104). Among the 19 patients with rCR, five discontinued CDK4/6i while maintaining endocrine therapy. Fatigue and diarrhea improved after discontinuation. Two patients experienced disease progression after 14.5 and 20.5 months of CDK4/6i withdrawal, respectively.
Conclusion: rCR following CDK4/6i therapy was an independent prognostic factor in HR-positive MBC. Patients with smaller primary tumors and early administration of abemaciclib were more likely to achieve rCR. Selected patients achieving rCR may be candidates for discontinuation of CDK4/6i as part of individualized treatment de-escalation strategies.
背景/目的:内分泌治疗联合细胞周期蛋白依赖性激酶4/6抑制剂(CDK4/6i)是激素受体(HR)阳性转移性乳腺癌(MBC)的标准一线治疗方法。虽然很少达到放射完全缓解(rCR),但这类患者的最佳管理仍不清楚。在人表皮生长因子受体2 (HER2)阳性的MBC中,已经探索了rCR后停止抗HER2治疗,但对hr阳性MBC的治疗降级尚未进行充分的研究。患者和方法:我们回顾性分析了2017年12月至2024年10月在我院接受cdk4 /6i内分泌治疗的178例hr阳性MBC患者。临床病理因素,包括免疫相关标志物(绝对淋巴细胞计数和中性粒细胞与淋巴细胞比率),评估与rCR的关系。只有放射学稳定性维持≥6个月时,骨转移才被认为是rcr等效的。在达到rCR后停用CDK4/6i的患者进行进一步评估。结果:19例患者(10.7%)达到rCR(可测量的非骨转移13例,骨转移6例)。达到rCR的中位时间为11.9个月。rCR在接受阿贝马昔利布治疗的患者(p=0.008)和原发肿瘤较小(≤2 cm, p=0.040)的患者中发生率更高。早期治疗也显示出更高rCR率的趋势(p=0.104)。在19例rCR患者中,5例在维持内分泌治疗的同时停止了CDK4/6i治疗。停药后疲劳和腹泻有所改善。两名患者分别在CDK4/6i停药14.5和20.5个月后出现疾病进展。结论:CDK4/6i治疗后的rCR是hr阳性MBC的独立预后因素。原发肿瘤较小和早期给予阿贝马昔利布的患者更有可能达到rCR。选定的达到rCR的患者可以作为个体化治疗降级策略的一部分,停用CDK4/6i。
{"title":"Radiological Complete Response as a Prognostic Marker in HR-positive Metastatic Breast Cancer Treated With CDK4/6 Inhibitors: Implications for Treatment De-escalation.","authors":"Wataru Goto, Shinichiro Kashiwagi, Mariko Nishikawa, Chika Watanabe, Koji Takada, Yukie Tauchi, Kana Ogisawa, Masatsune Shibutani, Haruhito Kinoshita, Tamami Morisaki","doi":"10.21873/anticanres.17946","DOIUrl":"https://doi.org/10.21873/anticanres.17946","url":null,"abstract":"<p><strong>Background/aim: </strong>Endocrine therapy combined with cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) is standard first-line treatment for hormone receptor (HR)-positive metastatic breast cancer (MBC). Although radiological complete response (rCR) is rarely achieved, the optimal management of such patients remains unclear. In human epidermal growth factor receptor 2 (HER2)-positive MBC, discontinuation of anti-HER2 therapy after rCR has been explored but treatment de-escalation in HR-positive MBC has not been sufficiently investigated.</p><p><strong>Patients and methods: </strong>We retrospectively analyzed 178 patients with HR-positive MBC treated with CDK4/6i-based endocrine therapy at our Institution between December 2017 and October 2024. Clinicopathological factors, including immune-related markers (absolute lymphocyte count and neutrophil-to-lymphocyte ratio), were evaluated for associations with rCR. Bone metastases were considered rCR-equivalent only if radiological stability was maintained for ≥6 months. Patients who discontinued CDK4/6i after achieving rCR were further assessed.</p><p><strong>Results: </strong>Nineteen patients (10.7%) achieved rCR (13 with measurable non-bone metastases and six with bone metastases). The median time to rCR was 11.9 months. rCR occurred more frequently in patients treated with abemaciclib (<i>p</i>=0.008) and those with smaller primary tumors (≤2 cm, <i>p</i>=0.040). Earlier-line treatment also showed a trend towards achieving higher rCR rates (<i>p</i>=0.104). Among the 19 patients with rCR, five discontinued CDK4/6i while maintaining endocrine therapy. Fatigue and diarrhea improved after discontinuation. Two patients experienced disease progression after 14.5 and 20.5 months of CDK4/6i withdrawal, respectively.</p><p><strong>Conclusion: </strong>rCR following CDK4/6i therapy was an independent prognostic factor in HR-positive MBC. Patients with smaller primary tumors and early administration of abemaciclib were more likely to achieve rCR. Selected patients achieving rCR may be candidates for discontinuation of CDK4/6i as part of individualized treatment de-escalation strategies.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"46 1","pages":"319-325"},"PeriodicalIF":1.7,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145861905","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}
Background/aim: Evidence regarding the benefits of nutritional therapy after robotic pancreatectomy is limited. This randomized controlled trial aimed to investigate the effects of a polymeric formula (PF) on preventing body weight loss (BWL) following robotic pancreatectomy.
Patients and methods: This single-center, open-label, randomized trial was conducted to assign 46 patients undergoing robotic pancreatectomy in a 1:1 ratio to either the PF (ISOCAL Clear) or control group. The primary endpoint was the percentage of BWL on postoperative days 14 and 28. The secondary endpoints were postoperative outcomes.
Results: Of the 52 eligible patients between December 2023 and November 2024, 46 were analyzed using intention-to-treat principles: 23 in the ISOCAL group and 23 in the control group. The %BWL was significantly lower in the ISOCAL group compared with that in the control group on postoperative days 14 (4.8±3.5% vs. 6.6±3.2%, p=0.02) and 28 (6.4±3.0% vs. 8.4±3.5%, p=0.047). Postoperative outcomes, including major complications (p=0.55) and hospital stay (p=0.83), did not differ significantly between the groups.
Conclusion: This study demonstrates the safety and feasibility of administering PF to patients undergoing robotic pancreatectomy. The results showed the beneficial effects of PF on mitigating BWL without compromising short-term outcomes.
{"title":"Impact of Polymeric Formula on Outcomes in Robotic Pancreatectomy: A Randomized Controlled Trial.","authors":"Kosei Takagi, Tomokazu Fuji, Kazuya Yasui, Motohiko Yamada, Takeyoshi Nishiyama, Yasuo Nagai, Masashi Hashimoto, Toshiharu Mitsuhashi, Toshiyoshi Fujiwara","doi":"10.21873/anticanres.17954","DOIUrl":"10.21873/anticanres.17954","url":null,"abstract":"<p><strong>Background/aim: </strong>Evidence regarding the benefits of nutritional therapy after robotic pancreatectomy is limited. This randomized controlled trial aimed to investigate the effects of a polymeric formula (PF) on preventing body weight loss (BWL) following robotic pancreatectomy.</p><p><strong>Patients and methods: </strong>This single-center, open-label, randomized trial was conducted to assign 46 patients undergoing robotic pancreatectomy in a 1:1 ratio to either the PF (ISOCAL Clear) or control group. The primary endpoint was the percentage of BWL on postoperative days 14 and 28. The secondary endpoints were postoperative outcomes.</p><p><strong>Results: </strong>Of the 52 eligible patients between December 2023 and November 2024, 46 were analyzed using intention-to-treat principles: 23 in the ISOCAL group and 23 in the control group. The %BWL was significantly lower in the ISOCAL group compared with that in the control group on postoperative days 14 (4.8±3.5% <i>vs.</i> 6.6±3.2%, <i>p</i>=0.02) and 28 (6.4±3.0% <i>vs.</i> 8.4±3.5%, <i>p</i>=0.047). Postoperative outcomes, including major complications (<i>p</i>=0.55) and hospital stay (<i>p</i>=0.83), did not differ significantly between the groups.</p><p><strong>Conclusion: </strong>This study demonstrates the safety and feasibility of administering PF to patients undergoing robotic pancreatectomy. The results showed the beneficial effects of PF on mitigating BWL without compromising short-term outcomes.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"46 1","pages":"407-414"},"PeriodicalIF":1.7,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145861861","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}
Background/aim: Partial or radical nephrectomy are the gold standard treatments for localized renal cell carcinoma; however, these treatments can induce kidney disease. This study aimed to investigate the functional outcomes of chronic kidney disease due to medical causes and end-stage renal disease in patients with localized renal cell carcinoma.
Patients and methods: This retrospective study enrolled 243 patients diagnosed with localized renal cell carcinoma at our Institution. Chronic kidney disease was classified as either surgically induced or attributable to medical causes, and outcomes were compared. Furthermore, a machine-learning model was used to identify important factors related to end-stage renal disease.
Results: Of the 243 patients, 5 died of progressive disease, 18 died of other causes, and 8 developed end-stage renal disease. Patients with chronic kidney disease due to medical causes had a higher risk of mortality (p=0.049), and none of the patients with surgically-induced chronic kidney disease developed end-stage renal disease (p<0.0001). Preoperative renal function and proteinuria were key factors related to end-stage renal disease after surgery. Patients with chronic kidney disease due to medical causes exhibited a 0.1% higher risk of end-stage renal disease than those with surgically-induced chronic kidney disease. Patients with proteinuria exhibited a 0.4% higher risk of end-stage renal disease than those without proteinuria.
Conclusion: Chronic kidney disease due to medical causes should be considered in surgical decision-making as it is a significant risk factor for mortality and end-stage renal disease. Chronic kidney disease due to medical causes and proteinuria before and after surgery must be treated with care to improve patient survival and avoid the onset of end-stage renal disease.
{"title":"Impact of Chronic Kidney Disease on End-stage Renal Disease After Treatment for Localized Renal Cell Carcinoma.","authors":"Shunta Hori, Tomonori Nakahama, Mitsuru Tomizawa, Kuniaki Inoue, Kenta Onishi, Yosuke Morizawa, Daisuke Gotoh, Yasushi Nakai, Makito Miyake, Tatsuo Yoneda, Nobumichi Tanaka, Kiyohide Fujimoto","doi":"10.21873/anticanres.17964","DOIUrl":"https://doi.org/10.21873/anticanres.17964","url":null,"abstract":"<p><strong>Background/aim: </strong>Partial or radical nephrectomy are the gold standard treatments for localized renal cell carcinoma; however, these treatments can induce kidney disease. This study aimed to investigate the functional outcomes of chronic kidney disease due to medical causes and end-stage renal disease in patients with localized renal cell carcinoma.</p><p><strong>Patients and methods: </strong>This retrospective study enrolled 243 patients diagnosed with localized renal cell carcinoma at our Institution. Chronic kidney disease was classified as either surgically induced or attributable to medical causes, and outcomes were compared. Furthermore, a machine-learning model was used to identify important factors related to end-stage renal disease.</p><p><strong>Results: </strong>Of the 243 patients, 5 died of progressive disease, 18 died of other causes, and 8 developed end-stage renal disease. Patients with chronic kidney disease due to medical causes had a higher risk of mortality (<i>p</i>=0.049), and none of the patients with surgically-induced chronic kidney disease developed end-stage renal disease (<i>p</i><0.0001). Preoperative renal function and proteinuria were key factors related to end-stage renal disease after surgery. Patients with chronic kidney disease due to medical causes exhibited a 0.1% higher risk of end-stage renal disease than those with surgically-induced chronic kidney disease. Patients with proteinuria exhibited a 0.4% higher risk of end-stage renal disease than those without proteinuria.</p><p><strong>Conclusion: </strong>Chronic kidney disease due to medical causes should be considered in surgical decision-making as it is a significant risk factor for mortality and end-stage renal disease. Chronic kidney disease due to medical causes and proteinuria before and after surgery must be treated with care to improve patient survival and avoid the onset of end-stage renal disease.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"46 1","pages":"503-514"},"PeriodicalIF":1.7,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145861870","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 : 2026-01-01DOI: 10.21873/anticanres.17955
Priscilla Nardi, Eugenio Varcasia, Valerio Rinaldi, Rocco Pasqua, Enrico Coletta, Paolina Saullo, Roberto Caronna, Giovanni Casella, Lidia Castagneto Gissey, Giampaolo Prezioso, Vito D'Andrea, Marco Angrisani, Roberto Luca Meniconi, Giulio Illuminati, Giuseppe Maria Ettorre
Background/aim: Intraductal papillary mucinous neoplasms (IPMN) are the most common cystic pancreatic lesions, with increasing incidence due to advances in imaging. Their management is complex due to their malignant potential and association with other neoplasms, weighed against morbidity and mortality of pancreatic resection. This study aimed to evaluate survival and surgical outcomes in patients with IPMN exhibiting high-risk features for malignant transformation.
Patients and methods: This retrospective study reviewed the medical records of 22 patients with adenocarcinoma arising in degenerated IPMN, diagnosed between 2008 and 2024. Inclusion criteria were clinical and radiological signs of pancreatic neoplasm consistent with degenerated IPMN (2023 Kyoto guidelines) and histopathological confirmation. Endoscopic ultrasound (EUS) was not performed in most cases due to the study's timeframe and strong Magnetic Resonance Imaging (MRI) indication of malignancy. Whipple procedures or total pancreatectomy were performed, excluding patients with advanced arterial infiltration or metastases. Demographic, clinical, surgical, and histopathological data, including tumor markers, tumor size, postoperative complications (pancreatic fistula, hemorrhage, delayed gastric emptying) and 30-day mortality were analyzed. Overall survival (OS) and disease- free survival (DFS) were estimated using the Kaplan-Meier method.
Results: The cohort comprised 68.2% men with a median age of 70.5 years. Common comorbidities included hypertension, chronic obstructive pulmonary disease (COPD) and diabetes. Most patients were symptomatic presenting with jaundice, pain, or weight loss. Preoperative findings included elevated bilirubin and CA 19-9 levels. Based on Kyoto guidelines, most patients exhibited high-risk stigmata. Surgical procedures primarily involved Whipple procedures, with a median operative time of 360 minutes. Postoperative complications occurred in 45.5% of patients. Median hospital stay was 19 days. Median follow-up was 23 months. Overall survival was 86.4% at 12 months, 72.7% at 24 months, and 68.2% at 60 months. Disease-free survival was 86.4% at 12 months, 81.8% at 24 months, and 72.7% at 60 months. Recurrence occurred in 45.5% of patients, primarily in the lungs, liver, and residual pancreas.
Conclusion: Despite the limitations of the small sample size and retrospective design, this study supports the 2023 Kyoto guidelines, demonstrating that surgical management of invasive IPMN can achieve substantially longer survival similar to classic pancreatic adenocarcinoma. Multidisciplinary evaluation is crucial for identifying signs of invasion and malignant degeneration, guiding surgical intervention.
{"title":"Intraductal Papillary Mucinous Neoplasms (IPMN): An Overall Review and a Retrospective Analysis of 22 Patients.","authors":"Priscilla Nardi, Eugenio Varcasia, Valerio Rinaldi, Rocco Pasqua, Enrico Coletta, Paolina Saullo, Roberto Caronna, Giovanni Casella, Lidia Castagneto Gissey, Giampaolo Prezioso, Vito D'Andrea, Marco Angrisani, Roberto Luca Meniconi, Giulio Illuminati, Giuseppe Maria Ettorre","doi":"10.21873/anticanres.17955","DOIUrl":"https://doi.org/10.21873/anticanres.17955","url":null,"abstract":"<p><strong>Background/aim: </strong>Intraductal papillary mucinous neoplasms (IPMN) are the most common cystic pancreatic lesions, with increasing incidence due to advances in imaging. Their management is complex due to their malignant potential and association with other neoplasms, weighed against morbidity and mortality of pancreatic resection. This study aimed to evaluate survival and surgical outcomes in patients with IPMN exhibiting high-risk features for malignant transformation.</p><p><strong>Patients and methods: </strong>This retrospective study reviewed the medical records of 22 patients with adenocarcinoma arising in degenerated IPMN, diagnosed between 2008 and 2024. Inclusion criteria were clinical and radiological signs of pancreatic neoplasm consistent with degenerated IPMN (2023 Kyoto guidelines) and histopathological confirmation. Endoscopic ultrasound (EUS) was not performed in most cases due to the study's timeframe and strong Magnetic Resonance Imaging (MRI) indication of malignancy. Whipple procedures or total pancreatectomy were performed, excluding patients with advanced arterial infiltration or metastases. Demographic, clinical, surgical, and histopathological data, including tumor markers, tumor size, postoperative complications (pancreatic fistula, hemorrhage, delayed gastric emptying) and 30-day mortality were analyzed. Overall survival (OS) and disease- free survival (DFS) were estimated using the Kaplan-Meier method.</p><p><strong>Results: </strong>The cohort comprised 68.2% men with a median age of 70.5 years. Common comorbidities included hypertension, chronic obstructive pulmonary disease (COPD) and diabetes. Most patients were symptomatic presenting with jaundice, pain, or weight loss. Preoperative findings included elevated bilirubin and CA 19-9 levels. Based on Kyoto guidelines, most patients exhibited high-risk stigmata. Surgical procedures primarily involved Whipple procedures, with a median operative time of 360 minutes. Postoperative complications occurred in 45.5% of patients. Median hospital stay was 19 days. Median follow-up was 23 months. Overall survival was 86.4% at 12 months, 72.7% at 24 months, and 68.2% at 60 months. Disease-free survival was 86.4% at 12 months, 81.8% at 24 months, and 72.7% at 60 months. Recurrence occurred in 45.5% of patients, primarily in the lungs, liver, and residual pancreas.</p><p><strong>Conclusion: </strong>Despite the limitations of the small sample size and retrospective design, this study supports the 2023 Kyoto guidelines, demonstrating that surgical management of invasive IPMN can achieve substantially longer survival similar to classic pancreatic adenocarcinoma. Multidisciplinary evaluation is crucial for identifying signs of invasion and malignant degeneration, guiding surgical intervention.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":"46 1","pages":"415-428"},"PeriodicalIF":1.7,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145861823","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}