1
|
Artusa F, Lamatsch S, Phan MD, Özdirik B, Berger H, Egerer M, Knorr‐Klocke J, Fischer J, Veelken R, van Bömmel F, Berg T, Kappert K, Tauber R, Puengel T, Engelmann C, Demir M, Tacke F, Mohr R. Soluble Urokinase Plasminogen Activator Receptor Predicts Survival and Hepatic Decompensation in Advanced Hepatocellular Carcinoma. Liver Int 2025; 45:e70121. [PMID: 40317602 PMCID: PMC12046945 DOI: 10.1111/liv.70121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 04/01/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND AND AIMS The introduction of immune checkpoint inhibitor (ICI) based therapies has significantly improved the prognosis of patients with unresectable hepatocellular carcinoma (HCC). However, the variable treatment response and the uncertain benefit in patients with advanced liver cirrhosis emphasise the urgent need for prognostic and predictive biomarkers guiding patient selection. The soluble urokinase plasminogen activator receptor (suPAR) is strongly associated with inflammation, liver cirrhosis and various types of cancer. In this study, we investigated suPAR as a potential novel biomarker in patients with unresectable HCC. METHODS This multicenter retrospective study, conducted at three German tertiary care centers, included 90 patients with unresectable HCC and suPAR measurements prior to and during atezolizumab/bevacizumab therapy. Patients with liver cirrhosis without HCC (n = 235) and non-cirrhotic patients with other gastrointestinal tumours (n = 155) were selected as control cohorts. RESULTS Median suPAR levels were significantly higher in patients with liver cirrhosis compared to non-cirrhotic cancer patients. A strong association with parameters of liver function, but not with HCC characteristics, was observed. In patients with HCC receiving atezolizumab/bevacizumab, suPAR was the most accurate independent predictor of hepatic decompensation and overall survival (OS). In addition, suPAR was able to stratify the risk of hepatic decompensation within the different Child-Pugh classes. CONCLUSIONS SuPAR represents a promising novel biomarker in patients with HCC treated with ICI-based therapies and bears the potential to guide the selection of antitumoral systemic therapies in patients with advanced liver cirrhosis.
Collapse
Affiliation(s)
- Fabian Artusa
- Department of Hepatology and GastroenterologyCharité ‐ Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | - Sven Lamatsch
- Department of Hepatology and GastroenterologyCharité ‐ Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | - Minh Duc Phan
- Department of Hepatology and GastroenterologyCharité ‐ Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | - Burcin Özdirik
- Department of Hepatology and GastroenterologyCharité ‐ Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | - Hilmar Berger
- Department of Hepatology and GastroenterologyCharité ‐ Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | - Mara Egerer
- Department of Hepatology and GastroenterologyCharité ‐ Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | - Jana Knorr‐Klocke
- Department of Hepatology and GastroenterologyCharité ‐ Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | - Janett Fischer
- Division of Hepatology, Department of Medicine IILeipzig University Medical CenterLeipzigGermany
| | - Rhea Veelken
- Division of Hepatology, Department of Medicine IILeipzig University Medical CenterLeipzigGermany
| | - Florian van Bömmel
- Division of Hepatology, Department of Medicine IILeipzig University Medical CenterLeipzigGermany
| | - Thomas Berg
- Division of Hepatology, Department of Medicine IILeipzig University Medical CenterLeipzigGermany
| | - Kai Kappert
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and PathobiochemistryCharité ‐ Universitätsmedizin BerlinBerlinGermany
- Labor Berlin – Charité Vivantes GmbHBerlinGermany
| | - Rudolf Tauber
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and PathobiochemistryCharité ‐ Universitätsmedizin BerlinBerlinGermany
- Labor Berlin – Charité Vivantes GmbHBerlinGermany
| | - Tobias Puengel
- Department of Hepatology and GastroenterologyCharité ‐ Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | - Cornelius Engelmann
- Department of Hepatology and GastroenterologyCharité ‐ Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
- Institute for Liver and Digestive HealthUniversity College LondonLondonUK
| | - Münevver Demir
- Department of Hepatology and GastroenterologyCharité ‐ Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | - Frank Tacke
- Department of Hepatology and GastroenterologyCharité ‐ Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | - Raphael Mohr
- Department of Hepatology and GastroenterologyCharité ‐ Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| |
Collapse
|
2
|
Sólymos P, Rédei M, Turan C, Szabó B, Ádám A, Molnár Z, Duray G, Hegyi P, Horváthy DB. Holmium-166 Radioembolization Is a Safe and Effective Locoregional Treatment for Primary and Secondary Liver Tumors: A Systematic Review and Meta-Analysis. Cancers (Basel) 2025; 17:1841. [PMID: 40507322 PMCID: PMC12153601 DOI: 10.3390/cancers17111841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Revised: 05/21/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES This systematic review and meta-analysis evaluated the effectiveness and the safety of transarterial radioembolization using Holmium-166 microspheres (Ho-166-TARE) for the treatment of primary and secondary liver tumors. The aim of the study was to offer a detailed analysis of clinical outcomes and the potential benefits of this innovative therapy. METHODS The study was conducted according to the PRISMA 2020 guidelines. The systematic search was performed in five databases in November 2023 and updated in June 2024. All 16 eligible studies were original research that evaluated Ho-166-TARE. The endpoints analyzed were disease control rate (DCR), overall survival (OS), progression-free survival (PFS), clinical and laboratory adverse events, healthy-liver- and tumor-liver-absorbed doses. The risk of bias was assessed using the MINORS checklist. RESULTS The pooled overall disease control rate (DCR) was 72% (95% CI, 46-89%); by mRECIST, it was 93% (95% CI, 71-99%); and by RECIST 1.1, it was 54% (95% CI, 22-83%) at 3-month follow-up. Overall survival (OS) at 3, 6, 12, and 30 months was 98%, 89%, 74%, and 39%, respectively. Severe clinical adverse events were minimal, although some patients showed elevated GGT levels and lymphocytopenia. Tumor-absorbed doses were nearly three times higher than those in healthy liver tissue. CONCLUSIONS These findings suggest that Ho-166-TARE is a safe and effective locoregional treatment option for liver tumors, especially in cases where systemic therapy alone is insufficient or surgical resection is not feasible. Further studies are needed to investigate tumor-specific response, optimize dosimetry strategies, and establish standardized protocols for long-term outcome assessment.
Collapse
Affiliation(s)
- Petra Sólymos
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (P.S.); (M.R.); (C.T.); (B.S.); (A.Á.); (Z.M.); (G.D.); (P.H.)
- Department of Radiology, Medical Imaging Centre, Semmelweis University, 1082 Budapest, Hungary
| | - Mátyás Rédei
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (P.S.); (M.R.); (C.T.); (B.S.); (A.Á.); (Z.M.); (G.D.); (P.H.)
- Department of Radiology, Medical Imaging Centre, Semmelweis University, 1082 Budapest, Hungary
| | - Caner Turan
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (P.S.); (M.R.); (C.T.); (B.S.); (A.Á.); (Z.M.); (G.D.); (P.H.)
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, 1082 Budapest, Hungary
| | - Bence Szabó
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (P.S.); (M.R.); (C.T.); (B.S.); (A.Á.); (Z.M.); (G.D.); (P.H.)
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Alexandra Ádám
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (P.S.); (M.R.); (C.T.); (B.S.); (A.Á.); (Z.M.); (G.D.); (P.H.)
| | - Zsolt Molnár
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (P.S.); (M.R.); (C.T.); (B.S.); (A.Á.); (Z.M.); (G.D.); (P.H.)
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, 1082 Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Poznan University of Medical Sciences, 61701 Poznan, Poland
| | - Gábor Duray
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (P.S.); (M.R.); (C.T.); (B.S.); (A.Á.); (Z.M.); (G.D.); (P.H.)
- Department of Cardiology, Central Hospital of Northern Pest—Military Hospital, 1134 Budapest, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (P.S.); (M.R.); (C.T.); (B.S.); (A.Á.); (Z.M.); (G.D.); (P.H.)
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, 1083 Budapest, Hungary
| | - Dénes B. Horváthy
- Centre for Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (P.S.); (M.R.); (C.T.); (B.S.); (A.Á.); (Z.M.); (G.D.); (P.H.)
- Department of Interventional Radiology, Heart and Vascular Centre, Semmelweis University, 1122 Budapest, Hungary
| |
Collapse
|
3
|
Tomonari T, Shimose S, Saeki I, Tani J, Honma Y, Ito T, Takeuchi M, Naito T, Takeuchi Y, Sasaki R, Sasaki K, Hatanaka T, Kakizaki S, Kanayama Y, Naganuma A, Tanabe N, Tanaka H, Kawano Y, Sato Y, Nishina S, Miyaaki H, Otsuka M, Kawashima H, Harada M, Kobara H, Takami T, Kawaguchi T, Takayama T, Hepatology InVestigator Experts in Japan (HIVE‐J) Study Group. A novel approach to evaluate the therapeutic efficacy of durvalumab and tremelimumab combination therapy in hepatocellular carcinoma. Hepatol Res 2025. [PMID: 40433908 DOI: 10.1111/hepr.14212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 04/18/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025]
Abstract
AIM This study aimed to evaluate the efficacy of durvalumab + tremelimumab (Dur + Tre) in real-world clinical practice, characterize "stable disease (SD)," and identify SD responders (SD-R) who benefit from Dur + Tre treatment. METHODS This multicenter observational study included 212 patients with unresectable hepatocellular carcinoma (u-HCC) treated with Dur + Tre between March 2023 and November 2024. The patients were categorized into 95 first-line and 117 later-line cases, respectively. Sequential cutoff points for depth of response (DOR) and progression-free survival (PFS) were tested to identify subgroups with survival outcomes comparable to those of responders. RESULTS Disease control rate (DCR) and PFS were significantly better in the first-line setting for both response evaluation criteria in solid tumors (RECIST) and modified RECIST (mRECIST) criteria. Patients who achieved PFS of ≥84 days or RECIST DOR of ≤-10% were classified as SD-R, as they had long-term survival outcomes similar to those with PR or CR. Furthermore, the CR + PR + SD-R group had significantly better survival outcomes than the other groups (p < 0.01), and multivariate analysis confirmed that SD-R was an independent prognostic factor with the strongest impact on survival outcomes (hazard ratio = 0.11). CONCLUSIONS In real-world clinical practice, Dur + Tre is highly effective as a first-line treatment for u-HCC. Additionally, patients with SD who met the SD-R criteria (PFS ≥84 days or RECIST DOR ≤-10%) showed survival outcomes comparable to those of patients with PR or CR. These findings may help identify patients who are most likely to benefit from treatment and improve their prognoses.
Collapse
Affiliation(s)
- Tetsu Tomonari
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima, Japan
- Tomonari Gastroenterology and Hepatology Clinic, Tokushima, Japan
| | - Shigeo Shimose
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Issei Saeki
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Joji Tani
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yuichi Honma
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Hospital, Nagoya, Japan
| | - Mamiko Takeuchi
- Department of Gastroenterology, Anjo Kosei Hospital, Anjo, Japan
| | - Takehito Naito
- Department of Gastroenterology, Toyohashi Municipal Hospital, Toyohashi, Japan
| | - Yasuto Takeuchi
- Department of Gastroenterology, Okayama University Hospital, Okayama, Japan
| | - Ryu Sasaki
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kyo Sasaki
- Department of Gastroenterology and Hepatology, Kawasaki Medical School, Kurashiki, Japan
| | - Takeshi Hatanaka
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Satoru Kakizaki
- Department of Clinical Research, NHO Takasaki General Medical Center, Takasaki, Japan
| | - Yuki Kanayama
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Atsushi Naganuma
- Department of Clinical Research, NHO Takasaki General Medical Center, Takasaki, Japan
| | - Norikazu Tanabe
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hironori Tanaka
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima, Japan
| | - Yutaka Kawano
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima, Japan
| | - Yasushi Sato
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima, Japan
| | - Sohji Nishina
- Department of Gastroenterology and Hepatology, Kawasaki Medical School, Kurashiki, Japan
| | - Hisamitsu Miyaaki
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology, Okayama University Hospital, Okayama, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Hospital, Nagoya, Japan
| | - Masaru Harada
- Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hideki Kobara
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Taro Takami
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima, Japan
| | | |
Collapse
|
4
|
Liao J, Ye H, Wang J, Liu F, Zhu H, Xie G, Pan J. Comparison of immune-related adverse events and analysis of risk factors in older and younger rectal cancer patients receiving immunotherapy. Am J Cancer Res 2025; 15:2153-2169. [PMID: 40520854 PMCID: PMC12163441 DOI: 10.62347/qlfx5040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 05/09/2025] [Indexed: 06/18/2025] Open
Abstract
BACKGROUND Immunotherapy has transformed rectal cancer treatment but poses risks of immune-related adverse events (irAEs), particularly in elderly patients who exhibit immunosenescence and inflammaging. This study compares the incidence and severity of irAEs in elderly and young rectal cancer patients receiving immunotherapy and identifies predictive biomarkers for these events. METHODS We retrospectively analyzed 405 rectal cancer patients treated with immunotherapy from January 2015 to December 2023. Patients were categorized into younger (< 60 years) and older (≥ 60 years) groups. Incidence and severity of irAEs were assessed using the Common Terminology Criteria for Adverse Events (CTCAE) standards. Blood samples were analyzed for hematological and immunological markers. RESULTS The older group displayed a significantly higher incidence of irAEs at 48.65% compared to 32.11% in the younger group (P = 0.003). Severity varied, with 69.72% of younger patients experiencing irAEs of grade ≤ 2 versus 51.69% in the older group (P = 0.001). Notably, higher absolute lymphocyte count (ALC), interleukin-6 (IL-6), and C-reactive protein (CRP) levels were associated with increased irAEs (P = 0.002, P = 0.001, P = 0.007, respectively). The multivariate analysis identified ALC, IL-6, CRP, B and T Lymphocyte Attenuator, Human Granulocyte-macrophage Colony Stimulating Factor, Programmed Death-1 and Programmed Death-Ligand 1 as significant predictors of irAEs, with ALC showing an odds ratio (OR) of 9.700 (P = 0.001) and IL-6 an OR of 58.961 (P < 0.001). Furthermore, the platelet-to-lymphocyte ratio (PLR) inversely correlated with irAEs (P = 0.013). CONCLUSION Older rectal cancer patients receiving immunotherapy were at increased risk for both greater incidence and severity of irAEs. Specific biomarkers, such as ALC and IL-6, were associated with a heightened risk of these events.
Collapse
Affiliation(s)
- Jie Liao
- Department of General Surgery, The Second People’s Hospital of YibinYibin 644000, Sichuan, China
| | - Haiwen Ye
- Department of General Surgery, The Second People’s Hospital of YibinYibin 644000, Sichuan, China
| | - Jian Wang
- Department of General Surgery, The Second People’s Hospital of YibinYibin 644000, Sichuan, China
| | - Fei Liu
- Department of General Surgery, The Second People’s Hospital of YibinYibin 644000, Sichuan, China
| | - Hongjia Zhu
- Cancer Center, The Second People’s Hospital of YibinYibin 644000, Sichuan, China
| | - Guowei Xie
- Department of General Surgery, The Second People’s Hospital of YibinYibin 644000, Sichuan, China
| | - Junjiang Pan
- Department of General Surgery, The Second People’s Hospital of YibinYibin 644000, Sichuan, China
| |
Collapse
|
5
|
Vogl TJ, Stein LV, Rödel C, Bielfeldt J, Adwan H. Efficacy and safety of intra-arterial chemoperfusion as palliative treatment of symptomatic primary brain malignancies. J Neuroradiol 2025; 52:101351. [PMID: 40379097 DOI: 10.1016/j.neurad.2025.101351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
BACKGROUND To retrospectively analyze the safety and efficacy of intra-arterial chemoperfusion (IAC) for symptomatic patients with recurrent or post-therapeutic progressive primary malignant brain tumors in regard to safety, survival and therapy response as a palliative treatment. METHODS Thirty-nine patients (24 men, 15 women; mean age: 52) who were treated by IAC in 181 sessions (mean: 4.6 sessions/patient) were enrolled and evaluated in this study. Out of total 39 patients with primary malignant brain tumor, 27 (69 %) were diagnosed with glioblastoma. The overall survival (OS) was calculated using the Kaplan-Meier method. Therapy response was determined according to RECIST 1.1. RESULTS All IAC treatments were performed without major adverse events or treatment-related deaths. The median OS time was 10.3 months (95 %CI: 0.6 - 19.9). The median PFS time was 5.9 months (95 %CI: 3.6 - 8.3). The rate of partial response was 10.8 % (4/37) and stable disease was achieved in 56.8 % (21/37) of patients. Progressive disease was observed in 32.4 % of cases (12/37). There were no cases of complete response. In two cases RECIST could not be analyzed due to loss of radiological follow-up. CONCLUSION This study could show that IAC may serve as a safe palliative strategy for symptomatic patients with recurrent or post-therapeutic progressive primary malignant brain tumors.
Collapse
Affiliation(s)
- Thomas J Vogl
- Clinic for Radiology and Nuclear Medicine, University Hospital Frankfurt, Goethe University, Theodor-Stern-Kai 7 60590, Frankfurt Am Main, Germany
| | - Leon Vincent Stein
- Clinic for Radiology and Nuclear Medicine, University Hospital Frankfurt, Goethe University, Theodor-Stern-Kai 7 60590, Frankfurt Am Main, Germany
| | - Claus Rödel
- Department of Radiotherapy and Oncology, University Hospital Frankfurt, Goethe University, Theodor-Stern-Kai 7 60590, Frankfurt Am Main, Germany
| | - John Bielfeldt
- Clinic for Radiology and Nuclear Medicine, University Hospital Frankfurt, Goethe University, Theodor-Stern-Kai 7 60590, Frankfurt Am Main, Germany
| | - Hamzah Adwan
- Clinic for Radiology and Nuclear Medicine, University Hospital Frankfurt, Goethe University, Theodor-Stern-Kai 7 60590, Frankfurt Am Main, Germany.
| |
Collapse
|
6
|
Li G, Wang J, Fang Q, Dai L, Du W. Oncologic outcomes following neoadjuvant immunochemotherapy in locally advanced oral squamous cell carcinoma. Front Immunol 2025; 16:1571285. [PMID: 40406105 PMCID: PMC12095181 DOI: 10.3389/fimmu.2025.1571285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/07/2025] [Indexed: 05/26/2025] Open
Abstract
Background To assess the oncologic outcomes in patients with oral squamous cell carcinoma (SCC) who underwent treatment with radiotherapy (RT) or chemoradiation therapy (CRT) following neoadjuvant immunochemotherapy and surgery. Methods Data from patients who underwent neoadjuvant immunochemotherapy, surgery, and adjuvant therapy were collected prospectively and analyzed retrospectively. The primary outcomes assessed were 3-year overall survival and locoregional control. Secondary endpoints included the objective response rate (ORR), rates of pathologic complete response (pCR) and major pathologic response (MPR), as well as safety. Results A total of 137 patients were included in the analysis. Neoadjuvant therapy yielded an ORR of 81.7%, with pCR and MPR achieved in 47 and 73 patients, respectively. Grade III and IV adverse events were rare, comprising only 1.6% of all events. The addition of adjuvant chemotherapy to RT did not show a significant reduction in the risk of locoregional recurrence. However, with regards to overall survival, the hazard ratios were 0.85 (95% CI: 0.73-0.96) for the MPR group and 0.66 (95% CI: 0.37-0.89) for the pCR group, both significantly higher than that in patients with incomplete pathologic response. The addition of adjuvant chemotherapy to RT was associated with a 5% reduction in the risk of mortality (95% CI: 1%-14%), the protective effect of CRT was the most obvious in patients with MPR. Conclusion Neoadjuvant immunochemotherapy demonstrated high safety and efficacy in oral SCC. CRT was superior to RT in terms of overall survival especially in patients with MPR when administered following neoadjuvant immunochemotherapy and surgery.
Collapse
Affiliation(s)
| | | | | | | | - Wei Du
- Department of Head Neck and Thyroid, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
7
|
Ball D, Nault JC, Vithayathil M, Allaire M, Ganne-Carrié N, Campani C, Marra F, Sharma R. Survival in patients receiving reduced dose intensity of bevacizumab for unresectable hepatocellular carcinoma. NPJ Precis Oncol 2025; 9:129. [PMID: 40328959 PMCID: PMC12056213 DOI: 10.1038/s41698-025-00908-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
IMBrave 150 established atezolizumab and bevacizumab as the new standard for advanced hepatocellular carcinoma (HCC) treatment. However, the trial reported significant adverse events leading to bevacizumab dose interruptions or discontinuations. This retrospective, real-world analysis evaluated the effect of reduced bevacizumab dose intensity on clinical outcomes in 354 patients receiving first-line combination immunotherapy for advanced HCC. To minimize immortal time bias, only those on therapy for over 3 months were included. Of 219 patients included in the landmark analysis, 52 received a reduced dose intensity of bevacizumab. The median relative dose intensity (RDTI) of bevacizumab was 75% (range 9.1-96.9%). There was no significant difference in progression-free survival (11.2 vs. 14.8 months, p = 0.5) or overall survival (20.4 vs. 26.8 months, p = 0.1) between those receiving 100% vs. reduced RDTI. Exploratory analysis showed that even doses under 75% had no survival impact. Treatment-related grade 3/4 adverse events occurred more frequently with RDTI (30.7% vs. 15.5%). Reduced bevacizumab doses do not impact survival.
Collapse
Affiliation(s)
- Dimity Ball
- Department of Medical Oncology, Imperial College NHS Healthcare Trust, Hammersmith Hospital, Du Cane Road, W12 0HS, London, UK
| | - Jean-Charles Nault
- Centre de recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, team « Functional Genomics of Solid Tumors », Equipe labellisée Ligue Nationale Contre le Cancer, Labex OncoImmunology, F-75006, Paris, France
- 3. Liver unit, Avicenne Hospital, APHP, Bobigny, France, University Sorbonne Paris Nord, Bobigny, France
| | - Mathew Vithayathil
- Division of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, W12 0NN, London, UK
| | - Manon Allaire
- Service d'Hépatolo-gastroentérologie, Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix, AP-HP, Sorbonne Université, Paris, France
| | - Nathalie Ganne-Carrié
- Centre de recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, team « Functional Genomics of Solid Tumors », Equipe labellisée Ligue Nationale Contre le Cancer, Labex OncoImmunology, F-75006, Paris, France
- 3. Liver unit, Avicenne Hospital, APHP, Bobigny, France, University Sorbonne Paris Nord, Bobigny, France
| | - Claudia Campani
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, Florence, Italy
| | - Fabio Marra
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, Florence, Italy
| | - Rohini Sharma
- Division of Surgery and Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, W12 0NN, London, UK.
| |
Collapse
|
8
|
Galvani L, Zappi A, Pusceddu S, Gelsomino F, La Salvia A, Oldani S, Panzuto F, Andrini E, Lamberti G, Campana D. Capecitabine and temozolomide or temozolomide alone in patients with atypical carcinoids. Endocrine 2025; 88:660-667. [PMID: 39853630 PMCID: PMC12069480 DOI: 10.1007/s12020-025-04171-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/14/2025] [Indexed: 01/26/2025]
Abstract
BACKGROUND Lung neuroendocrine neoplasms (NENs) represent about 20% of all lung cancers. Few therapeutic options are available for atypical carcinoids (ACs). Single-agent temozolomide (TEM) is active in lung NENs, but whether the addition of capecitabine (CAPTEM) is associated with improved outcomes, is unknown. We sought to investigate the TEM-based therapies (TEM or CAPTEM) in patients with advanced AC. MATERIAL AND METHODS This was a retrospective analysis of prospectively collected data from patients with AC of the lung referred to our institution from January 2003 to January 2023 who have received chemotherapy with either TEM or CAPTEM as any line treatment. Primary endpoint was progression free survival (PFS), secondary endpoints included overall response rate (ORR) and overall survival (OS). RESULTS In this study we included 31 patients with advanced AC. Median Ki-67 was 14.4% (3-30). CAPTEM in 17 patients (54.8%), while TEM was administered in 14 patients (45.2%). Overall, ORR was 39% (N = 12/31, all partial responses), while median PFS and OS were 57.4 months (95%CI: 43.2-71.7) and 24.4 months (95% confidence interval [95%CI]: 14.7-34.1). Median PFS was 33.9 months (15.6-52.1) in the CAPTEM group, while it was 15.5 (7.3-23.8) in the TEM group (p = 0.047). When adjusting for potential confounding factors, treatment with TEM vs CAPTEM retained its independent association with an increased risk of progression (HR: 4.01 [95%CI: 1.18-13.68]; p = 0.027). CONCLUSIONS Treatment with CAPTEM is associated with longer PFS than TEM alone in patients with AC. Prospective studies with larger sample size are needed to validate this finding.
Collapse
Affiliation(s)
- Linda Galvani
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - Università di Bologna, Bologna, Italy
| | - Arianna Zappi
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - Università di Bologna, Bologna, Italy
| | - Sara Pusceddu
- Medical Oncology and Hematology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Fabio Gelsomino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Anna La Salvia
- Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- National Center for Drug Research and Evaluation, National Institute of Health (ISS), Rome, Italy
| | - Simone Oldani
- Medical Oncology and Hematology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Francesco Panzuto
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
- Digestive Disease Unit, Sant'Andrea University Hospital, ENETS Center of Excellence, Rome, Italy
| | - Elisa Andrini
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - Università di Bologna, Bologna, Italy
| | - Giuseppe Lamberti
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - Università di Bologna, Bologna, Italy.
| | - Davide Campana
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - Università di Bologna, Bologna, Italy
- Medical Oncology Department, IRCCS Azienda Ospedaliero-Universitaria Sant'Orsola-Malpighi di Bologna, Bologna, Italy
| |
Collapse
|
9
|
Toyoda H, Hiraoka A, Ochi H, Tsuji K, Tajiri K, Tani J, Tada T, Okubo T, Atsukawa M, Hirooka M, Itobayashi E, Hatanaka T, Kariyama K, Ishikawa T, Kuroda H, Takaguchi K, Kosaka H, Kawada K, Kakizaki S, Yada Y, Ogawa C, Nishimura T, Yasuda S, Deguchi A, Morishita A, Itokawa N, Arai T, Tsutsui A, Naganuma A, Enomoto H, Kaibori M, Nouso K, Hiasa Y, Kumada T, Akita T, Tanaka J, Johnson PJ. Prognostic Significance of the BALAD Serological Model in Systemic Therapies for Hepatocellular Carcinoma: A Personalized Approach to the Prediction of Survival Benefit. JCO Clin Cancer Inform 2025; 9:e2400175. [PMID: 40354590 DOI: 10.1200/cci-24-00175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 01/27/2025] [Accepted: 03/05/2025] [Indexed: 05/14/2025] Open
Abstract
PURPOSE The BALAD model, a scoring system for staging hepatocellular carcinoma (HCC), is based on five serum markers: bilirubin, albumin, lens culinaris agglutinin-reactive alpha-fetoprotein [AFP], AFP, and des-gamma-carboxy prothrombin. It has shown good ability to predict survival in patients with HCC irrespective of stage and treatment, a high BALAD value being associated with a poor prognosis. However, its prognostic significance is unclear in patients with advanced unresectable HCC (uHCC) who undergo systemic therapies. We assessed the prognostic ability of BALAD in this subpopulation. METHODS In a multicenter cohort of 1,510 patients with advanced uHCC treated with first-line systemic therapies, the baseline BALAD score was calculated on the basis of pretreatment serum levels. Overall survival (OS), progression-free survival (PFS), overall response rate (ORR), and disease control rate (DCR) were calculated and related to the BALAD score. RESULTS In all, 502 patients were treated with sorafenib, 435 with lenvatinib, and 573 with atezolizumab plus bevacizumab. Irrespective of treatment regimen, OS, PFS, ORR, and DCR were all independently negatively correlated with the BALAD score. The beneficial effects of specific systemic therapy regimens differed according to the BALAD score. CONCLUSION The BALAD score had good prognostic ability for predicting OS and PFS in patients with advanced uHCC who underwent systemic therapies and was associated with treatment response. Application of the BALAD score offers increased precision in the prediction of outcome both for individual patients and for specific subgroups of patients with HCC.
Collapse
Affiliation(s)
- Hidenori Toyoda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Atsushi Hiraoka
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama, Japan
| | - Hironori Ochi
- Hepato-biliary Center, Japanese Red Cross Matsuyama Hospital, Matsuyama, Japan
| | - Kunihiko Tsuji
- Center of Gastroenterology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Kazuto Tajiri
- Department of Gastroenterology, Toyama University Hospital, Toyama, Japan
| | - Joji Tani
- Department of Gastroenterology and Hepatology, Kagawa University, Kagawa, Japan
| | - Toshifumi Tada
- Department of Internal Medicine, Japanese Red Cross Himeji Hospital, Himeji, Japan
| | - Tomomi Okubo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School Chiba Hokuso Hospital, Inzai, Japan
| | - Masanori Atsukawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Masashi Hirooka
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Japan
| | - Ei Itobayashi
- Department of Gastroenterology, Asahi General Hospital, Asahi, Japan
| | - Takeshi Hatanaka
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Kazuya Kariyama
- Department of Hepatology, Okayama City Hospital, Okayama, Japan
| | - Toru Ishikawa
- Department of Hepatology, Saiseikai Niigata Hospital, Niigata, Japan
| | - Hidekatsu Kuroda
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, School of Medicine, Morioka, Japan
| | - Koichi Takaguchi
- Department of Hepatology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Hisashi Kosaka
- Department of Surgery, Kansai Medical University, Hirakata, Japan
| | - Kazuhito Kawada
- Hepatology Division, Department of Internal Medicine II, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Satoru Kakizaki
- Department of Gastroenterology, National Hospital Organization Takasaki General Medical Center, Takasaki, Japan
| | - Yutaka Yada
- Department of Gastroenterology, Hanwa Memorial Hospital, Osaka, Japan
| | - Chikara Ogawa
- Department of Gastroenterology, Japanese Red Cross Takamatsu Hospital, Takamatsu, Japan
| | - Takashi Nishimura
- Department of Gastroenterology and Hepatology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Satoshi Yasuda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Akihiro Deguchi
- Department of Gastroenterology and Hepatology, Kagawa University, Kagawa, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Hepatology, Kagawa University, Kagawa, Japan
| | - Norio Itokawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Taeang Arai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Akemi Tsutsui
- Department of Hepatology, Kagawa Prefectural Central Hospital, Takamatsu, Japan
| | - Atsushi Naganuma
- Department of Gastroenterology, National Hospital Organization Takasaki General Medical Center, Takasaki, Japan
| | - Hirayuki Enomoto
- Department of Gastroenterology and Hepatology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Masaki Kaibori
- Department of Surgery, Kansai Medical University, Hirakata, Japan
| | - Kazuhiro Nouso
- Department of Hepatology, Okayama City Hospital, Okayama, Japan
| | - Yoichi Hiasa
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Japan
| | - Takashi Kumada
- Department of Nursing, Gifu Kyoritsu University, Ogaki, Japan
| | - Tomoyuki Akita
- Department of Epidemiology, Infectious Disease Control, and Prevention, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| | - Junko Tanaka
- Department of Epidemiology, Infectious Disease Control, and Prevention, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| | - Philip J Johnson
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
10
|
Zeng Z, Yi Z, Xu B. The biological and technical challenges facing utilizing circulating tumor DNA in non-metastatic breast cancer patients. Cancer Lett 2025; 616:217574. [PMID: 39983895 DOI: 10.1016/j.canlet.2025.217574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Breast cancer is one of the most prevalent cancers and has emerged as a major global challenge. Circulating tumor DNA (ctDNA), a liquid biopsy method, overcomes the accessibility limitations of tissue-based testing and is widely used for monitoring minimal residual disease and molecular relapse, predicting prognosis, evaluating the response of neoadjuvant therapy, and optimizing treatment decisions in non-metastatic breast cancer. However, the application of ctDNA still faces many challenges. Here, we survey the clinical applications of ctDNA in non-metastatic breast cancer and discuss the significant biological and technical challenges of utilizing ctDNA. Importantly, we investigate potential avenues for addressing the challenges. In addition, emerging technologies, including fragmentomics detection, methylation sequencing, and long-read sequencing, have clinical potential and could be a future direction. Proper utilization of machine learning facilitates the identification of meaningful patterns from complex fragment and methylation profiles of ctDNA. There is still a lack of clinical trials focused on the subsets of ctDNA (e.g., circulating mitochondrial DNA), ctDNA-inferred drug-resistant clonal evolution, tumor heterogeneity, and ctDNA-guided clinical decision-making in non-metastatic breast cancer. Due to regional differences in the number of registered clinical trials, it is essential to enhance communication and foster global collaboration to advance the field.
Collapse
Affiliation(s)
- Zihang Zeng
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China
| | - Zongbi Yi
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, 430071, Wuhan, China.
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| |
Collapse
|
11
|
Huang X, Ji M, Shang X, Zhang H, Zhang X, Zhou J, Yin T. Smart on-demand drug release strategies for cancer combination therapy. J Control Release 2025; 383:113782. [PMID: 40294796 DOI: 10.1016/j.jconrel.2025.113782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/06/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
In cancer therapy, enhancing therapeutic indices and patient compliance has been a central focus in recent drug delivery technology development. However, achieving a delicate balance between improving anti-tumor efficacy and minimizing toxicity to normal tissues remains a significant challenge. With the advent of smart on-demand drug release strategies, new opportunities have emerged. These strategies represent a promising approach to drug delivery, enabling precise control over the release of therapeutic agents in a programmed and spatiotemporal manner. Recent studies have focused on designing delivery systems capable of releasing multiple therapeutic agents sequentially, while achieving spatial resolution in vivo. Smart on-demand drug release strategies have demonstrated considerable potential in tumor combination therapy for achieving precision drug delivery and controlled release by responding to specific physiological signals or external physical stimuli in the tumor microenvironment. These strategies not only improve tumor targeting and reduce toxicity to healthy tissues but also enable sequential release in combination therapy, allowing multiple drugs to be released in a specific spatiotemporal order to enhance synergistic treatment effects. In this paper, we systematically reviewed the current research progress of smart on-demand drug release drug delivery strategies in anti-tumor combination therapy. We highlighted representative integrated drug delivery systems and discussed the challenges associated with their clinical application. Additionally, potential future research directions are proposed to further advance this promising field.
Collapse
Affiliation(s)
- Xiaolin Huang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Mengfei Ji
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xinyu Shang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Hengchuan Zhang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xin Zhang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jianping Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| | - Tingjie Yin
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| |
Collapse
|
12
|
Okada Y, Sato Y, Shinomiya R, Miyake T, Takahashi T, Yokoyama R, Mitsui Y, Tomonari T, Okamoto K, Sogabe M, Miyamoto H, Kawano Y, Takayama T. Conditions for effective use of liposomal irinotecan with fluorouracil and leucovorin in unresectable pancreatic cancer after FOLFIRINOX treatment. Int J Clin Oncol 2025:10.1007/s10147-024-02677-y. [PMID: 40266453 DOI: 10.1007/s10147-024-02677-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/01/2024] [Indexed: 04/24/2025]
Abstract
BACKGROUND Liposomal irinotecan + fluorouracil/leucovorin (nal-IRI + 5FU/LV) is commonly used as a second- or later-line treatment for pancreatic ductal adenocarcinoma (PDAC) and offers survival benefits. However, its efficacy and safety in patients previously treated with FOLFIRINOX, which includes irinotecan, remain unclear. We evaluated the efficacy and safety of nal-IRI + 5FU/LV in patients with unresectable PDAC who received previous FOLFIRINOX therapy and those who did not. METHODS This retrospective observational study included 42 patients with PDAC who were treated with nal-IRI + 5FU/LV (October 2020-November 2023). Patients were grouped based on prior FOLFIRINOX treatment. RESULTS The progression-free survival (PFS) in patients who previously received modified FOLFIRINOX (mFFX) therapy was shorter than that in patients who did not (2.5 vs. 3.5 months, P = 0.07). When patients with greater than- and less than the cut-off value of irinotecan-free interval (IFI) were classified into the long and short IFI groups, respectively, PFS was significantly longer in the long-IFI group than that in the short IFI group (4.0 vs. 2.1 months, P = 0.01). Moreover, the C-reactive protein/albumin ratio (CAR) was also a significant predictor of PFS (P = 0.03). Furthermore, both factors were found to be independent factors influencing PFS in the univariate Cox regression analysis (P = 0.02 and P = 0.04). CONCLUSION Nal-IRI + 5FU/LV therapy may be a safe and effective option as a second- or later-line treatment, particularly for patients who have not previously received mFFX therapy. For patients who received prior mFFX exposure, a longer IFI and lower CAR may indicate greater potential benefit, thus aiding in more personalized treatment approaches.
Collapse
Affiliation(s)
- Yasuyuki Okada
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Yasushi Sato
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan.
- Department of Community Medicine for Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Ryo Shinomiya
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Takanori Miyake
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Taku Takahashi
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Reiko Yokoyama
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Yasuhiro Mitsui
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Tetsu Tomonari
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Koichi Okamoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Masahiro Sogabe
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Hiroshi Miyamoto
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Yutaka Kawano
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
13
|
Ceylan F, Aktürk Esen S, Ünal OÜ, Aslan F, Onur İD, Ateş Ö, Demirciler E, Ünek İT, Gülmez A, Özen Engin E, Taş S, Gököz Doğu G, Şimşek M, Türk HM, İnal A, Şahin G, Yüksel HÇ, Tenekeci AK, Hızal M, Şendur MAN, Uncu D. Long-Term Outcomes of Patients Undergoing Conversion Surgery After Induction Chemotherapy: Turkish Oncology Group Study. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:776. [PMID: 40428735 PMCID: PMC12113550 DOI: 10.3390/medicina61050776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/13/2025] [Accepted: 04/20/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: Conversion surgery for liver metastatic colorectal cancer (mCRC) has been associated with prolonged survival. This study aimed to evaluate the efficacy and safety of integrating biological therapies with fluorouracil-based induction chemotherapy in patients with isolated liver mCRC who subsequently underwent curative resection of both the primary tumor and liver metastases. Materials and Methods: This multicenter, retrospective study, conducted by the Turkish Oncology Group (TOG), included 116 patients from 11 tertiary centers who underwent conversion surgery following induction chemotherapy between 2009 and 2024. Results: The median age was 57 years, with 62% male patients. The median follow-up period was 55.3 months. The median progression-free survival (PFS) and overall survival (OS) were 21.1 and 53.7 months, respectively. No significant differences in PFS or OS were observed based on biological therapy use or tumor localization. Among patients with RAS/RAF wild-type tumors, PFS and OS were comparable between those receiving Anti-EGFR and Anti-VEGF therapy. In RAS/RAF mutant tumors, the addition of Anti-VEGF therapy did not confer a survival benefit. Factors associated with shorter PFS included advanced tumor stage (ypT3-T4), lymph node metastasis, and multiple metastases, while shorter OS was linked to advanced tumor stage and lack of objective response. Conclusions: Surgical resection plays a pivotal role in improving survival outcomes in patients with potentially resectable liver mCRC. Optimizing induction chemotherapy regimens may enhance conversion rates and prolong long-term survival. Further studies are needed to refine treatment selection based on tumor localization, mutation status, and molecular biomarkers.
Collapse
Affiliation(s)
- Furkan Ceylan
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara 06800, Turkey; (S.A.E.); (M.H.); (M.A.N.Ş.); (D.U.)
| | - Selin Aktürk Esen
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara 06800, Turkey; (S.A.E.); (M.H.); (M.A.N.Ş.); (D.U.)
| | - Olçun Ümit Ünal
- Department of Medical Oncology, UHS Izmir Faculty of Medicine, Izmir 35020, Turkey;
| | - Ferit Aslan
- Department of Medical Oncology, Ankara Batıkent Medical Park Hospital, Ankara 06680, Turkey;
| | - İlknur Deliktaş Onur
- Department of Medical Oncology, A.Y. Ankara Oncology Training and Research Hospital, Ankara 06200, Turkey; (İ.D.O.); (Ö.A.)
| | - Öztürk Ateş
- Department of Medical Oncology, A.Y. Ankara Oncology Training and Research Hospital, Ankara 06200, Turkey; (İ.D.O.); (Ö.A.)
| | - Erkut Demirciler
- Department of Medical Oncology, Dokuz Eylül University, Izmir 35390, Turkey; (E.D.); (İ.T.Ü.)
| | - İlkay Tuğba Ünek
- Department of Medical Oncology, Dokuz Eylül University, Izmir 35390, Turkey; (E.D.); (İ.T.Ü.)
| | - Ahmet Gülmez
- Department of Medical Oncology, Adana City Hospital, Adana 01230, Turkey;
| | - Esra Özen Engin
- Department of Medical Oncology, Sakarya Training and Research Hospital, Sakarya 54100, Turkey;
| | - Semra Taş
- Department of Medical Oncology, Pamukkale University, Denizli 20070, Turkey; (S.T.); (G.G.D.)
| | - Gamze Gököz Doğu
- Department of Medical Oncology, Pamukkale University, Denizli 20070, Turkey; (S.T.); (G.G.D.)
| | - Melih Şimşek
- Department of Medical Oncology, Bezmialem University, Istanbul 34093, Turkey; (M.Ş.); (H.M.T.)
| | - Hacı Mehmet Türk
- Department of Medical Oncology, Bezmialem University, Istanbul 34093, Turkey; (M.Ş.); (H.M.T.)
| | - Ali İnal
- Department of Medical Oncology, Mersin City Hospital, Mersin 33330, Turkey;
| | - Gökhan Şahin
- Department of Medical Oncology, Ege University, Izmir 35100, Turkey; (G.Ş.); (H.Ç.Y.)
| | - Haydar Çağatay Yüksel
- Department of Medical Oncology, Ege University, Izmir 35100, Turkey; (G.Ş.); (H.Ç.Y.)
| | | | - Mutlu Hızal
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara 06800, Turkey; (S.A.E.); (M.H.); (M.A.N.Ş.); (D.U.)
| | - Mehmet Ali Nahit Şendur
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara 06800, Turkey; (S.A.E.); (M.H.); (M.A.N.Ş.); (D.U.)
- Department of Medical Oncology, Ankara Yıldırım Beyazıt University, Çankaya 06800, Turkey
| | - Doğan Uncu
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara 06800, Turkey; (S.A.E.); (M.H.); (M.A.N.Ş.); (D.U.)
| |
Collapse
|
14
|
Bindal P, Trottier CA, Elavalakanar P, Dodge LE, Kim S, Logan E, Ma S, Liegel J, Arnason J, Alonso CD. Early versus late infectious complications following chimeric antigen receptor-modified T-cell therapy. Leuk Lymphoma 2025; 66:702-712. [PMID: 39704413 DOI: 10.1080/10428194.2024.2439513] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/28/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024]
Abstract
Despite increasing utilization of CAR T-cell therapy, data are lacking regarding long term follow up and risk of infectious complications after the early period following CAR T-cell infusion. In this study, we sought to compare epidemiology and risk factors for early (≤ 3 months) and late (3 months to 1 year) infections. Data were retrospectively collected at six time points: pre-CAR T, day of infusion, and at 3, 6, 9, and 12 months post CAR-T infusion for all consecutive adult patients treated at our institution. In this cohort, the cumulative incidence of any infection was 73.2% in the first year. Bridging therapy, CRS, neurotoxicity and steroid use were identified as contributing risk factors for early bacterial infections. After 3 months, community acquired respiratory infections were common. We characterize bacterial, viral and fungal pathogens based on time elapsed after CAR T-cell infusion.
Collapse
Affiliation(s)
- P Bindal
- Division of Oncology, Department of Medicine, University of Massachusetts, Worcester, Massachusetts, USA
| | - C A Trottier
- Division of Geographic Medicine and Infectious Diseases, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - P Elavalakanar
- Division of Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - L E Dodge
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - S Kim
- Division of Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - E Logan
- Division of Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - S Ma
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - J Liegel
- Division of Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - J Arnason
- Division of Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - C D Alonso
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Infectious Diseases, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Huang M, Zhu X, Xu W, Zhu J, Xun X, Su B, Chen H. TTC7A-ALK, a novel ALK fusion variant identified in a patient with metastatic lung adenocarcinoma, exhibits excellent response to crizotinib. Transl Oncol 2025; 54:102345. [PMID: 40054123 PMCID: PMC11930134 DOI: 10.1016/j.tranon.2025.102345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/20/2025] [Accepted: 03/02/2025] [Indexed: 03/18/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. ALK gene rearrangement has been identified in 3 % to 5 % of the patients with NSCLC. Thanks to the advancements in second-generation sequencing technology, an increasing number of novel fusion partners have been identified. In our research, we discovered a rare ALK fusion, TTC7A-ALK, in a patient with advanced lung adenocarcinoma using targeted next-generation sequencing (NGS). After being diagnosed with advanced lung adenocarcinoma with TTC7A-ALK fusion, the patient received crizotinib treatment and achieved a progression-free survival of 29 months. Additonanlly, we conducted further functional analyses on this fusion protein to assess its oncogenic potential. Similar to EML4-ALK, the TTC7A-ALK fusion protein can promote the growth of Ba/F3 cells under IL-3-independent conditions in vitro. In vivo studies demonstrate that the TTC7A-ALK fusion protein could enhance the tumorigenesis of NIH3T3 cells in nude mice, which can be suppressed by crizotinib. Mechanistic studies indicated that the ectopic expression of TTC7A-ALK in 293T cells led to the hyperactivation of downstream MAPK and PI3K/Akt pathways, which can be inhibited by crizotinib. Furthermore, upon tumor progression, the patient transitioned to alectinib, which provided rapid symptom relief and controlled the majority of lesions. Conclusionly, we identified and validated TTC7A-ALK as a oncogenic fusion in NSCLC. The patient demonstrated a significant clinical benefit from sequential treatment with crizotinib and alectinib, highlighting TTC7A-ALK as a novel therapeutic target for ALK inhibitors. These findings extend the spectrum of actionable ALK fusions and promote the inclusion of rare fusion detection in clinical diagnostic processes and treatment strategies.
Collapse
Affiliation(s)
- Meijin Huang
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China
| | - Xiangqing Zhu
- Department of Basic Medical Laboratory, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China
| | - Wenmang Xu
- Department of Pathology, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China
| | - Jun Zhu
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China
| | - Xin Xun
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China
| | - Bin Su
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China
| | - Hong Chen
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, PLA, Yunnan, China.
| |
Collapse
|
16
|
Woźnicki P, Laqua C, Fiku I, Hekalo A, Truhn D, Engelhardt S, Kather J, Foersch S, D'Antonoli TA, Pinto Dos Santos D, Baeßler B, Laqua FC. Automatic structuring of radiology reports with on-premise open-source large language models. Eur Radiol 2025; 35:2018-2029. [PMID: 39390261 PMCID: PMC11913902 DOI: 10.1007/s00330-024-11074-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/26/2024] [Accepted: 08/21/2024] [Indexed: 10/12/2024]
Abstract
OBJECTIVES Structured reporting enhances comparability, readability, and content detail. Large language models (LLMs) could convert free text into structured data without disrupting radiologists' reporting workflow. This study evaluated an on-premise, privacy-preserving LLM for automatically structuring free-text radiology reports. MATERIALS AND METHODS We developed an approach to controlling the LLM output, ensuring the validity and completeness of structured reports produced by a locally hosted Llama-2-70B-chat model. A dataset with de-identified narrative chest radiograph (CXR) reports was compiled retrospectively. It included 202 English reports from a publicly available MIMIC-CXR dataset and 197 German reports from our university hospital. Senior radiologist prepared a detailed, fully structured reporting template with 48 question-answer pairs. All reports were independently structured by the LLM and two human readers. Bayesian inference (Markov chain Monte Carlo sampling) was used to estimate the distributions of Matthews correlation coefficient (MCC), with [-0.05, 0.05] as the region of practical equivalence (ROPE). RESULTS The LLM generated valid structured reports in all cases, achieving an average MCC of 0.75 (94% HDI: 0.70-0.80) and F1 score of 0.70 (0.70-0.80) for English, and 0.66 (0.62-0.70) and 0.68 (0.64-0.72) for German reports, respectively. The MCC differences between LLM and humans were within ROPE for both languages: 0.01 (-0.05 to 0.07), 0.01 (-0.05 to 0.07) for English, and -0.01 (-0.07 to 0.05), 0.00 (-0.06 to 0.06) for German, indicating approximately comparable performance. CONCLUSION Locally hosted, open-source LLMs can automatically structure free-text radiology reports with approximately human accuracy. However, the understanding of semantics varied across languages and imaging findings. KEY POINTS Question Why has structured reporting not been widely adopted in radiology despite clear benefits and how can we improve this? Findings A locally hosted large language model successfully structured narrative reports, showing variation between languages and findings. Critical relevance Structured reporting provides many benefits, but its integration into the clinical routine is limited. Automating the extraction of structured information from radiology reports enables the capture of structured data while allowing the radiologist to maintain their reporting workflow.
Collapse
Affiliation(s)
- Piotr Woźnicki
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, Würzburg, Germany.
| | - Caroline Laqua
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, Würzburg, Germany
| | - Ina Fiku
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, Würzburg, Germany
| | - Amar Hekalo
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, Würzburg, Germany
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital Aachen, Aachen, Germany
| | - Sandy Engelhardt
- Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Jakob Kather
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center Mainz, Mainz, Germany
| | - Tugba Akinci D'Antonoli
- Institute of Radiology and Nuclear Medicine, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Daniel Pinto Dos Santos
- Department of Diagnostic and Interventional Radiology, University of Cologne, Cologne, Germany
- Department of Radiology, University Hospital of Frankfurt, Frankfurt, Germany
| | - Bettina Baeßler
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, Würzburg, Germany
| | - Fabian Christopher Laqua
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
17
|
Zhao J, Li Y, Li R, Yao X, Dong X, Su L, Li Y. Nomogram based on computed tomography radiomics features and clinicopathological factors to predict the prognosis of patients with non-small cell lung cancer receiving immune checkpoint inhibitor rechallenge. Transl Lung Cancer Res 2025; 14:842-856. [PMID: 40248725 PMCID: PMC12000940 DOI: 10.21037/tlcr-24-876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/13/2025] [Indexed: 04/19/2025]
Abstract
Background Whether patients with advanced non-small cell lung cancer (NSCLC) who experience progressive disease (PD) after the initial immunotherapy treatment benefit from subsequent immunotherapy remains unclear. In this study, we aimed to identify predictive factors and develop a nomogram to predict successful immunotherapy rechallenge for such patients with NSCLC to guide clinical treatment and improve prognosis. Methods Between January 2019 and December 2022, 352 patients with advanced NSCLC who received immunotherapy rechallenge after experiencing PD were divided into the training (n=246) and validation (n=106) cohorts. Clinicopathological factors and radiomics features were included in the univariate and multivariate analyses, with significant predictive factors being used to develop the nomogram. Results Univariate and multivariate analyses showed that time from the initial immunotherapy to PD occurrence (duration), clinical N stage, liver metastasis, treatment after PD following the first immunotherapy (post-PD treatment), and radiomics features were independent predictive factors for progression-free survival (PFS). In addition, age, duration, clinical N stage, post-PD treatment, and radiomics were independent predictive factors for overall survival (OS). Accordingly, these predictive factors were used to develop a nomogram. The area under the curves (AUCs) of the nomogram for predicting 6-, 12-, and 18-month PFS and 12-, 18-, and 24-month OS were 0.731, 0.809, 0.878, 0.742, 0.782, and 0.868, respectively, in the training cohorts, whereas the corresponding values in the validation cohort were 0.672, 0.774, 0.826, 0.833, 0.705, and 0.762. This indicated good discrimination. Conclusions We developed and validated a predictive nomogram based on clinicopathological factors and radiomics features for the prognosis of patients with advanced NSCLC who received immunotherapy rechallenge following PD after the first immunotherapy. The nomogram showed strong predictive utility and can be a suitable tool for such patients with advanced NSCLC.
Collapse
Affiliation(s)
- Junfeng Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ying Li
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ruyue Li
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Xiujing Yao
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Xue Dong
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Lin Su
- Department of Respiratory Medicine, Jinan Fourth People’s Hospital, Jinan, China
| | - Yintao Li
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
18
|
Jian L, Sheng C, Liu H, Li H, Hu P, Ai Z, Yu X, Liu H. Early prediction of progression-free survival of patients with locally advanced nasopharyngeal carcinoma using multi-parametric MRI radiomics. BMC Cancer 2025; 25:519. [PMID: 40119284 PMCID: PMC11929181 DOI: 10.1186/s12885-025-13899-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 03/10/2025] [Indexed: 03/24/2025] Open
Abstract
PURPOSE Prognostic prediction plays a pivotal role in guiding personalized treatment for patients with locoregionally advanced nasopharyngeal carcinoma (LANPC). However, few studies have investigated the incremental value of functional MRI to the conventional MRI-based radiomic models. Here, we aimed to develop a radiomic model including functional MRI to predict the prognosis of LANPC patients. METHODS One hundred and twenty-six patients (training dataset, n = 88; validation dataset, n = 38) with LANPC were retrospectively included. Radiomic features were extracted from T1-weighted imaging (T1WI), T2-weighted imaging (T2WI), contrast-enhanced T1WI (cT1WI), and diffusion-weighted imaging (DWI). Pearson correlation analysis and recursive feature elimination or Relief were used for identifying features associated with progression-free survival (PFS). Five machine learning algorithms with cross-validation were compared to develop the optimal single-layer and fusion radiomic models. Clinical and combined models were developed via multivariate Cox regression model. RESULTS The clinical model based on TNM stage achieved a C-index of 0.544 in the validation dataset. The fusion radiomic model, incorporating DWI-, T1WI-, and cT1WI-derived imaging features, yielded the highest C-index of 0.788, outperforming DWI-based (C-index = 0.739), T1WI-based (C-index = 0.734), cT1WI-based (C-index = 0.722), and T1WI plus cT1WI-based models (C-index = 0.747) in predicting PFS. The fusion radiomic model yielded the C-index of 0.786 and 0.690 in predicting distant metastasis-free survival and overall survival, respectively. However, the addition of TNM stage to the fusion radiomic model could not improve the predictive power. CONCLUSION The fusion radiomic model demonstrates favorable performance in predicting survival outcomes in LANPC patients, surpassing TNM staging alone. Integration of DWI-derived features into conventional MRI radiomic models could enhance predictive accuracy.
Collapse
Affiliation(s)
- Lian Jian
- Hunan Cancer Hospital, Central South University, No.283, TongzipoRoad, Changsha, 410013, China
| | - Cai Sheng
- the Affiliated Changsha Central Hospital, University of South China, No.283, TongzipoRoad, Changsha, 410004, China
| | - Huaping Liu
- Hunan Cancer Hospital, Central South University, No.283, TongzipoRoad, Changsha, 410013, China
| | - Handong Li
- Hunan Cancer Hospital, Central South University, No.283, TongzipoRoad, Changsha, 410013, China
| | - Pingsheng Hu
- Hunan Cancer Hospital, Central South University, No.283, TongzipoRoad, Changsha, 410013, China
| | - Zhaodong Ai
- Hunan Cancer Hospital, Central South University, No.283, TongzipoRoad, Changsha, 410013, China
| | - Xiaoping Yu
- Hunan Cancer Hospital, Central South University, No.283, TongzipoRoad, Changsha, 410013, China.
| | - Huai Liu
- Hunan Cancer Hospital, Central South University, No.283, TongzipoRoad, Changsha, 410013, China
| |
Collapse
|
19
|
Li X, Fan F, Zhang T. Efficacy and influencing factors of immunotherapy crossover combined with targeted therapy in advanced esophageal cancer patients following first-line chemotherapy combined with immunotherapy failure. Am J Cancer Res 2025; 15:1321-1334. [PMID: 40226448 PMCID: PMC11982723 DOI: 10.62347/gboq6704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/28/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Advanced esophageal cancer presents significant treatment challenges, especially after immunochemotherapy failure. This study evaluates the efficacy of further treatment with combination chemotherapy versus combination immunotherapy crossover in terms of tumor regression, quality of life, and identifies factors influencing treatment outcomes. METHODS In a retrospective case-control study, clinical data from 293 patients with advanced esophageal cancer treated at Shanxi Province Cancer Hospital between February 2021 and February 2023 were analyzed. Patients excluded from radical resection due to failure of first-line immunotherapy were divided into two groups: 95 received combination chemotherapy with Irinotecan and Tigio (S-1, Tegafur/Gimeracil/Oteracil Potassium), and 198 underwent Anlotinib targeted therapy combined with immunotherapy crossover. Treatment efficacy was assessed using tumor regression grading (TRG), and quality of life was evaluated using EORTC QLQ-C30 and QLQ-OES18 scales. Potential factors affecting treatment efficacy were examined using multivariate logistic regression analysis. RESULTS Baseline characteristics, including age, gender, body mass index (BMI), and history of smoking and alcohol consumption, were comparable between the two groups. TRG showed no significant differences in distribution, with objective response rates of 40% in the Irinotecan/S-1 group and 44.44% in the combined immunotherapy crossover group (P = 0.472). However, quality of life measures indicated superior outcomes from immunotherapy crossover in physical (P = 0.024), emotional (P = 0.002), and general health scores (P = 0.003). Factors negatively impacting treatment success included male gender, smoking, alcohol consumption history, and certain tumor locations. Elevated CEA levels positively correlated with treatment efficacy. Logistic regression analysis identified male gender (OR, 2.109; P = 0.021), smoking (OR, 2.575; P = 0.003), alcohol consumption (OR, 1.995; P = 0.043), and CEA levels (OR, 0.742; P = 0.017) as significant predictors of treatment efficacy. CONCLUSION Immunotherapy combined with targeted therapy and chemotherapy alone showed comparable efficacy in tumor regression. However, immunotherapy combined with targeted therapy improved certain aspects of quality of life. Factors such as gender, lifestyle habits, and CEA levels can significantly influence treatment outcomes.
Collapse
Affiliation(s)
- Xiuxiu Li
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive CancerTianjin 300060, China
- Department of Gastroenterology, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuan 030001, Shanxi, China
| | - Fan Fan
- Department of Gastroenterology, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuan 030001, Shanxi, China
| | - Ti Zhang
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive CancerTianjin 300060, China
- Department of Hepatobiliary Surgery, Fudan University Shanghai Cancer CenterShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| |
Collapse
|
20
|
Yang M, Zheng C, Miao Y, Yin C, Tang L, Zhang C, Yu P, Han Q, Ma Y, Li S, Jiang G, Li W, Xia P. BTLA promoter hypomethylation correlates with enhanced immune cell infiltration, favorable prognosis, and immunotherapy response in melanoma. J Immunother Cancer 2025; 13:e009841. [PMID: 40081944 PMCID: PMC11907004 DOI: 10.1136/jitc-2024-009841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB)-based immunotherapy has significantly improved survival in advanced melanoma. However, many patients exhibit resistance to these therapies. This study examines the impact of BTLA promoter methylation on its expression, immune cell infiltration, and clinical outcomes, evaluating its potential as a prognostic and predictive biomarker for immunotherapy response. METHODS We analyzed methylation and gene expression data from public datasets (The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO)) and an in-house cohort of melanoma patients treated with ICB therapy at the First Affiliated Hospital of Zhengzhou University. We developed a quantitative methylation-specific PCR (qMSP) assay to measure methylation levels of the cg24157392 and cg03995631 CpG sites, and a targeted bisulfite sequencing assay was used to validate the accuracy of qMSP. We measured BTLA protein expression using multiplex immunofluorescence and immunohistochemical staining methods. Pearson correlation, survival analysis, and immune cell infiltration estimation were conducted to explore the associations between BTLA promoter methylation, mRNA and protein expression, clinical outcomes, and immune characteristics. RESULTS Hypomethylation at CpG sites cg24157392 and cg03995631 in the BTLA promoter were significantly associated with higher BTLA mRNA and protein expression. In the TCGA dataset, low methylation at these sites predicted longer overall survival and was validated in an independent cohort of 50 stage III/IV melanoma patients, with an area under the curve of 0.94 for predicting 5-year survival. Furthermore, BTLA promoter hypomethylation correlated with higher infiltration of immune cells, such as CD8+T cells, CD4+T cells, B cells, and macrophages. Additionally, low methylation at cg24157392 and cg03995631, as quantified by the qMSP assay, was significantly associated with better progression-free survival in patients treated with immune checkpoint inhibitors. These findings were further validated using GEO datasets. CONCLUSIONS BTLA promoter hypomethylation serves as a significant biomarker for favorable prognosis and enhanced response to ICB therapy in melanoma. The developed qMSP assays for cg24157392 and cg03995631 accurately quantified methylation levels and demonstrated their potential for clinical application in patient stratification and personalized immunotherapy.
Collapse
Affiliation(s)
- Minglei Yang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chenxi Zheng
- The Department of Dermatology, Henan Second Provincial People's Hospital, Zhengzhou, China
| | - Yu Miao
- Henan Academy of Sciences, Zhengzhou, Henan, China
| | - Cuicui Yin
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Longfei Tang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chongli Zhang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pu Yu
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Hennan, China
| | - Qingfang Han
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yihui Ma
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shenglei Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guozhong Jiang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wencai Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peiyi Xia
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
21
|
Ahn HM, Park SY, Choi Y, Kim J, Lee Y. Molecular subtype changes after acquiring resistance to tarlatamab in small cell lung cancer. Drug Resist Updat 2025; 79:101198. [PMID: 39742708 DOI: 10.1016/j.drup.2024.101198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/08/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
Tarlatamab, a novel bispecific T-cell engager, has demonstrated unprecedented efficacy in patients with small cell lung cancer. However, there is no known mechanism of resistance to tarlatamab. This study suggests that a transcriptional expression shift might be associated with acquired resistance to tarlatamab.
Collapse
Affiliation(s)
- Hyung-Min Ahn
- Center for Lung Cancer, National Cancer Center Hospital, Goyang, South Korea
| | - Seog-Yun Park
- Department of Pathology, National Cancer Center Hospital, Goyang, South Korea
| | - Yura Choi
- Targeted Therapy Branch, Research Institute, National Cancer Center, Goyang, South Korea
| | - Jaemin Kim
- Targeted Therapy Branch, Research Institute, National Cancer Center, Goyang, South Korea
| | - Youngjoo Lee
- Center for Lung Cancer, National Cancer Center Hospital, Goyang, South Korea; Targeted Therapy Branch, Research Institute, National Cancer Center, Goyang, South Korea.
| |
Collapse
|
22
|
Guigal-Stephan N, Lockhart B, Moser T, Heitzer E. A perspective review on the systematic implementation of ctDNA in phase I clinical trial drug development. J Exp Clin Cancer Res 2025; 44:79. [PMID: 40022112 PMCID: PMC11871688 DOI: 10.1186/s13046-025-03328-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/13/2025] [Indexed: 03/03/2025] Open
Abstract
Circulating tumour DNA (ctDNA) represents an increasingly important biomarker for the screening, diagnosis and management of patients in clinical practice in advanced/metastatic disease across multiple cancer types. In this context, ctDNA-based comprehensive genomic profiling is now available for patient management decisions, and several ctDNA-based companion diagnostic assays have been approved by regulatory agencies. However, although the assessment of ctDNA levels in Phase II-III drug development is now gathering momentum, it remains somewhat surprisingly limited in the early Phase I phases in light of the potential opportunities provided by such analysis. In this perspective review, we investigate the potential and hurdles of applying ctDNA testing for the inclusion and monitoring of patients in phase 1 clinical trials. This will enable more informed decisions regarding patient inclusion, dose optimization, and proof-of-mechanism of drug biological activity and molecular response, thereby supporting the evolving oncology drug development paradigm. Furthermore, we will highlight the use of cost-efficient, agnostic genome-wide techniques (such as low-pass whole genome sequencing and fragmentomics) and methylation-based methods to facilitate a more systematic integration of ctDNA in early clinical trial settings.
Collapse
Affiliation(s)
- Nolwen Guigal-Stephan
- Translational Medicine, Institut de Recherches Servier, 22 route 128, Gif-sur-Yvette, Saclay, 91190, France.
| | - Brian Lockhart
- Translational Medicine, Institut de Recherches Servier, 22 route 128, Gif-sur-Yvette, Saclay, 91190, France
| | - Tina Moser
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, Graz, 8010, Austria
| | - Ellen Heitzer
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, Graz, 8010, Austria.
| |
Collapse
|
23
|
Liu K, Huang Z, Zhao L, Zhao H. Significant Response to Palbociclib Plus Lenvatinib as Second-line Treatment for CDKN2A/2B Deletion Intrahepatic Cholangiocarcinoma: A Case Report. J Clin Transl Hepatol 2025; 13:169-172. [PMID: 39917465 PMCID: PMC11797823 DOI: 10.14218/jcth.2024.00404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/03/2024] [Accepted: 12/17/2024] [Indexed: 02/09/2025] Open
Abstract
Cyclin-dependent kinase inhibitor 2A/2B (CDKN2A/2B) deletions are frequently identified in patients with biliary tract cancer; however, standard treatment options for this genetic alteration are lacking. Here, we present the case of a 64-year-old woman diagnosed with intrahepatic cholangiocarcinoma and hilar lymph node metastasis who underwent radical surgery. Postoperative pathology confirmed moderately differentiated adenocarcinoma. The tumor recurred during the second cycle of adjuvant chemotherapy following surgery, and the metastatic sites included the cranial region, right lung, and right adrenal gland. Genetic analysis revealed a CDKN2A/2B deletion, indicating palbociclib sensitivity. Subsequently, the patient received palbociclib plus lenvatinib as systemic therapy, along with stereotactic radiotherapy for the intracranial lesion. Notably, the right pulmonary metastasis significantly regressed after 12 months of treatment, with the complete disappearance of the intracranial tumor. However, the disease progressed at 32.2 months, with significant enlargement of the right adrenal gland metastasis and new metastasis in the right lung. The progression-free survival and overall survival were 32.2 months and 34.4 months, respectively. In conclusion, our case demonstrates that palbociclib plus lenvatinib is a promising chemotherapy-free second-line treatment for intrahepatic cholangiocarcinoma with a CDKN2A/2B deletion.
Collapse
Affiliation(s)
- Kai Liu
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziyue Huang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lijin Zhao
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
24
|
Ma HY, Tai QW, Song H. PD-1 inhibitor combined with chemotherapy or lenvatinib in advanced gallbladder cancer: a retrospective comparative study. BMC Gastroenterol 2025; 25:111. [PMID: 39994570 PMCID: PMC11849360 DOI: 10.1186/s12876-025-03688-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Gallbladder cancer (GBC) is a refractory primary cancer. Some GBC patients are prone to recurrence even after surgical resection. In such cases, chemotherapy is the most common non-surgical treatment. The emergence of programmed cell death protein 1 (PD-1) inhibitors and targeted therapy have provided an additional option for those suffering from advanced tumors. METHODS This was a retrospective study involving patients with advanced GBC treated at the Shanghai Eastern Hepatobiliary Surgery Hospital between June 2019 and June 2022. The patients who received a PD-1 inhibitor (tislelizumab) with chemotherapy or with lenvatinib were retrospectively analyzed. The Response Evaluation Criteria in Solid Tumors (RECIST 1.1) was used as the efficacy evaluation standard. The overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), and tumor marker CA199 were evaluated. RESULTS This study involved 61 patients with advanced GBC. Of these, 32 patients received tislelizumab and GS (gemcitabine and TS-1) chemotherapy, whereas 29 patients received tislelizumab and lenvatinib. For the Tislelizumab plus GS chemotherapy group, the median OS and PFS were 19.64 ± 11.81 (95% CI: 16.47-25.20) and 15.44 ± 13.42 (95% CI: 12.08-22.25) months, respectively. For the lenvatinib group, the OS and PFS were 13.06 ± 9.41 (95% CI: 9.72-16.63) and 10.34 ± 10.03 (95% CI: 6.56-14.13) months, respectively. The ORR and DCR were 59.38% and 81.3%, respectively, for the Tislelizumab plus GS chemotherapy group, which were significantly longer than those for the Tislelizumab plus Lenvatinib group. Treatment-related adverse events were similar between the groups. CONCLUSION Tislelizumab combined with GS chemotherapy provides a safe and more efficient treatment option for advanced GBC patients.
Collapse
Affiliation(s)
- Hong-Yan Ma
- Pharmaceutical Department, Longhua District Central Hospital, ShenZhen, China
| | - Qin-Wen Tai
- Department of Hepatobiliary Surgery, Shenzhen HospitalSouthern Medical University, ShenZhen, China
| | - Hao Song
- Department of Hepatobiliary Surgery, Shenzhen HospitalSouthern Medical University, ShenZhen, China.
| |
Collapse
|
25
|
Pietrantonio F, Morano F, Niger M, Ghelardi F, Chiodoni C, Palazzo M, Nichetti F, Manca P, Cristarella E, Doldi V, Zaffaroni N, Sabella G, Brambilla N, Benincasa E, Giacovelli G, Vitalini C, Girolami F, Rovati LC. The Prostaglandin EP4 Antagonist Vorbipiprant Combined with PD-1 Blockade for Refractory Microsatellite-Stable Metastatic Colorectal Cancer: A Phase Ib/IIa Trial. Clin Cancer Res 2025; 31:649-658. [PMID: 39620921 PMCID: PMC11831105 DOI: 10.1158/1078-0432.ccr-24-2611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/04/2024] [Accepted: 11/26/2024] [Indexed: 02/18/2025]
Abstract
PURPOSE Novel combinations are required to overcome resistance to immune checkpoint inhibitors in proficient mismatch repair (pMMR) or microsatellite-stable (MSS) metastatic colorectal cancer (mCRC). We aimed to determine whether vorbipiprant, a prostaglandin E2 receptor EP4 subtype antagonist, can convert immune-resistant mCRC into a tumor responsive to anti-PD-1 inhibition. PATIENTS AND METHODS This phase Ib/IIa prospective, open-label, single-arm trial followed a 3 + 3 dose-escalation and dose-optimization design. A total of 28 patients with chemorefractory pMMR/MSS mCRC were given dose-escalated oral vorbipiprant (30, 90, or 180 mg twice daily), along with biweekly intravenous balstilimab (3 mg/kg), an anti-PD-1 antibody. The primary endpoints included safety and the disease control rate (DCR). Secondary endpoints were the overall response rate, duration of response, progression-free survival, and overall survival. RESULTS No dose-limiting toxicities were observed. Of the 28 patients, seven (25%) experienced serious adverse events, but only one was attributed to vorbipiprant and one to balstilimab. The trial achieved a DCR of 50% observed across the entire cohort. In the subgroup of patients with liver metastases (n = 12), the DCR was 25%. The overall response rate was 11%, with three patients showing a partial response (median duration of response, 7.4 months). The median progression-free survival was 2.6 months, and the median overall survival was 14.2 months. Translational exploratory analyses suggested that vorbipiprant may boost response to anti-PD-1 in patients with immunogenic tumors. CONCLUSIONS The combination of vorbipiprant and a PD-1 inhibitor (balstilimab) yielded sufficient activity in refractory pMMR/MSS mCRC, which is worthy of confirmation in future clinical trials in biomarker-enriched populations.
Collapse
Affiliation(s)
- Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Niger
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo Ghelardi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claudia Chiodoni
- Molecular Immunology Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Palazzo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federico Nichetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Computational Oncology, Molecular Diagnostics Program, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Paolo Manca
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eleonora Cristarella
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Valentina Doldi
- Molecular Pharmacology Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Experimental Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanna Sabella
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Nadia Brambilla
- Department of Clinical Research, Rottapharm Biotech, Monza, Italy
| | - Elena Benincasa
- Department of Clinical Research, Rottapharm Biotech, Monza, Italy
| | | | | | | | - Lucio C. Rovati
- Department of Clinical Research, Rottapharm Biotech, Monza, Italy
- School of Medicine, University of Milano–Bicocca, Milan, Italy
| |
Collapse
|
26
|
Chen YQ, Tan JX, Gao LL, Yang JX, Huang J, Yang JJ, Zhao Q. Exploring YAP1-related TIME in SCLC: implications for survival and treatment response to immuno-chemotherapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:8. [PMID: 40051494 PMCID: PMC11883233 DOI: 10.20517/cdr.2024.177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/30/2024] [Accepted: 01/25/2025] [Indexed: 03/09/2025]
Abstract
Aim: Small-cell lung cancer (SCLC) is usually diagnosed as an advanced stage with a poor outcome. SCLC has limited response to immunotherapy due to the absence or lack of immune cell infiltration, so studying its tumor immune microenvironment (TIME) is essential. Methods: The study involved patients with extensive-stage small-cell lung cancer (ES-SCLC) diagnosed at the Guangdong Lung Cancer Institute between January 2018 and April 2022 who had received the atezolizumab/carboplatin/etoposide (ECT) treatment. We used multi-immunohistochemistry (mIHC) to assess the prognostic value of YAP1 and TIME in SCLC, with results confirmed using public data. Results: 15 patients with sufficient baseline biopsy samples were included in this study. The total population of YAP1-positive cells is inversely related to progression-free survival (PFS) and shows a potential negative correlation with overall survival (OS). CD56-positive cells are the primary components of TIME in SCLC tumor parenchyma and stroma. The total population and cell density of YAP1-positive cells are significantly positively correlated with CD4-positive cells. Furthermore, in the tumor parenchyma, both the proportion and the cell density of YAP1-positive cells are positively correlated with that of FOXP3-positive cells. The total population of CD56-positive cells showed a negative correlation trend with YAP1-positive cells but without significant difference. Conclusion: YAP1 has shown prognostic value in SCLC patients receiving ECT regimen treatment. The high expression level of YAP1 seems related to the inhibitory TIME. However, some prospective studies with larger populations are warranted.
Collapse
Affiliation(s)
- Yu-Qing Chen
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Authors contributed equally
| | - Jia-Xiong Tan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Authors contributed equally
| | - Ling-Ling Gao
- Beihang University, Beijing 100191, China
- Authors contributed equally
| | - Jia-Xing Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Jie Huang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Qiang Zhao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
27
|
Lei Z, Chai H, Liu X, Jiang Y. Key prognostic factors in transarterial chemoembolization combined with sorafenib treatment for hepatocellular carcinoma with portal vein tumor thrombosis. Am J Cancer Res 2025; 15:517-532. [PMID: 40084372 PMCID: PMC11897632 DOI: 10.62347/sxmj5155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a prevalent malignancy worldwide, with portal vein tumor thrombosis (PVTT) worsening its prognosis and complicating management. The combination of transarterial chemoembolization (TACE) and the targeted agent sorafenib has been proposed to improve treatment outcomes. This study investigates the prognostic factors influencing the effectiveness of this combined treatment in HCC patients with PVTT. METHODS A retrospective cohort study was conducted on 299 patients diagnosed with HCC and PVTT who underwent TACE and sorafenib treatment between January 2018 and December 2022. Patients were categorized into good-prognosis (n = 197) and poor-prognosis (n = 102) groups based on Response Evaluation Criteria in Solid Tumors (RECIST) assessed four weeks post-treatment. Prognostic factors were analyzed using univariate and multivariate analyses to identify significant determinants affecting therapeutic outcomes. RESULTS Key prognostic factors included tumor number, differentiation, size, PVTT extent, Child-Pugh class, ECOG performance status, hospitalization duration, and AFP levels. Patients with a single tumor had better outcomes (OR 0.358, P = 0.002), whereas poor differentiation (OR 4.561, P = 0.005) and larger tumor size (OR 0.347, P < 0.001) were associated with worse prognosis. A higher Child-Pugh class (OR 0.563, P = 0.035) and better ECOG performance (OR 2.710, P = 0.025) improved prognosis, while prolonged hospitalization and elevated AFP levels were linked to poorer outcomes. ASA classification and HCC morphology did not significantly impact prognosis. CONCLUSION The prognosis of HCC with PVTT treated with TACE and sorafenib is significantly influenced by tumor characteristics, liver function, and overall patient health. Identifying these factors can aid in refining personalized treatment strategies to improve survival outcomes.
Collapse
Affiliation(s)
- Zilun Lei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical UniversityChongqing 400016, China
| | - Hao Chai
- The Cancer Hospital Affiliated to Chongqing UniversityChongqing 400030, China
| | - Xiaoya Liu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical UniversityChongqing 400016, China
| | - Yingsong Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical UniversityChongqing 400016, China
| |
Collapse
|
28
|
Kim J, Hong SS, Kim SH, Hwang HK, Hong N, Rhee Y, Kang CM. Genotype-based prognosis prediction for MEN1-Related pancreatic neuroendocrine tumors in Korean patients a single-center retrospective study. Pancreatology 2025; 25:134-141. [PMID: 39638700 DOI: 10.1016/j.pan.2024.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/25/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Pancreatic neuroendocrine tumors (PNETs) are the leading cause of death related to multiple endocrine neoplasia type 1 (MEN1). Previous studies have linked certain mutations in the MEN1 gene and loss of interactions with MENIN's functional partners to the mortality or aggressiveness of PNETs. This study aimed to evaluate the genotype-phenotype correlations of MEN1-related PNETs in Korean patients and to summarize the treatment outcomes comprehensively. METHODS We retrospectively analyzed 72 patients diagnosed with MEN1 at a tertiary care center in Korea between January 2003 and September 2022. MEN1 mutations were analyzed using direct or next-generation sequencing. RESULTS Among 40 families with MEN1, 10 had exon 2 mutations, which were the most frequently observed. Of these, 50 (69.4 %) were diagnosed with PNETs; 20 underwent pancreatic resection. Patients with truncating mutations showed a significant difference in age-related penetrance of PNET (p = 0.029). No distinct genotype was associated with malignant transformation (lymph node or distant metastasis) in MEN1-related PNETs. In the subgroup Cox model, mutations in exons 3 or 10 showed significant differences in tumor progression in the observation group (adjusted hazard ratio: 8.164,(95 % CI: 1.648-40.436), p = 0.010, HR: 8.300, (95 % CI: 1.808-38.113), p = 0.007). CONCLUSION PNETs in Korean patients with MEN1 exhibit a stable prognosis. An individualized follow-up strategy may be necessary, particularly for young patients with truncating mutation in the MEN1 gene. In addition, those with mutations in exons 3 or 10 may require more active surveillance to decrease the risk of progression.
Collapse
Affiliation(s)
- Juwan Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung Soo Hong
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, South Korea; Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Hyun Kim
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, South Korea; Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Ho Kyoung Hwang
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, South Korea; Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Namki Hong
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Yumie Rhee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea.
| | - Chang Moo Kang
- Pancreatobiliary Cancer Center, Yonsei Cancer Center, Severance Hospital, Seoul, South Korea; Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
29
|
Qu C, Zeng P, Hu W, Yang D, Wang H, Yuan H, Cao J, Xiu D. Multiparametric quantitative diffusion weighted magnetic resonance imaging can effectively predict the response to neoadjuvant therapy in borderline resectable pancreatic ductal adenocarcinoma. Eur J Radiol 2025; 183:111893. [PMID: 39753006 DOI: 10.1016/j.ejrad.2024.111893] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/08/2024] [Accepted: 12/16/2024] [Indexed: 02/08/2025]
Abstract
PURPOSE To investigate whether multiparametric quantitative diffusion weighted magnetic resonance imaging (DWI) can effectively predict the neoadjuvant therapy (NAT) response in borderline resectable pancreatic ductal adenocarcinoma (BRPC). METHODS The clinicopathological data, including tumor size, location, and CA19-9 values, as well as DWI parameters(ADC, D, and f values) from 72 patients with BRPC, were analyzed. The differences and changes in these factors before and after NAT were compared to identify those most accurately reflect the response to NAT. ROC analysis was used to evaluate the diagnostic efficacy, and Kaplan-Meier survival analysis explored the relationship between DWI parameters and prognosis. Subgroup survival analysis to further identify populations potentially benefiting from NAT based on multiparametric DWI. RESULTS After-NAT, the response group showed significantly higher ADC and D values and lower f values compared to the non-response group. The ΔADC (OR: 12.24, P = 0.013) emerged as the most important independent factor for tumor response, demonstrating the highest diagnostic accuracy for NAT response with an AUC of 0.936. Kaplan-Meier showed the high ADC value group, high D value group and low f value group were associated with better prognosis after NAT; and the ΔADC ≥ 0 group, ΔD ≥ 0 group, and Δf < 0 group was significantly associated with better prognosis. In addition, subgroup analysis suggested two groups of patients might potentially benefit from NAT. CONCLUSIONS Multiparametric quantitative DWI may offer valuable insights into the efficacy and prognosis of NAT in BRPC. These findings have the potential to support the evaluation and decision-making process for patients undergoing NAT.
Collapse
Affiliation(s)
- Chao Qu
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Shandong Province, China; Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Piaoe Zeng
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Weiyu Hu
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Shandong Province, China
| | - Dongxia Yang
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Shandong Province, China
| | - Hangyan Wang
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Huishu Yuan
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Jingyu Cao
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Shandong Province, China.
| | - Dianrong Xiu
- Department of General Surgery, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
30
|
Li J, Wang XT, Wang Y, Chen K, Li GG, Long YF, Chen MF, Peng C, Liu Y, Cheng W. Multimodal treatment combining neoadjuvant therapy, laparoscopic subtotal distal pancreatectomy and adjuvant therapy for pancreatic neck-body cancer: Case series. World J Gastrointest Surg 2025; 17:97897. [PMID: 39872794 PMCID: PMC11757209 DOI: 10.4240/wjgs.v17.i1.97897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 12/27/2024] Open
Abstract
BACKGROUND Pancreatic cancer involving the pancreas neck and body often invades the retroperitoneal vessels, making its radical resection challenging. Multimodal treatment strategies, including neoadjuvant therapy, surgery, and postoperative adjuvant therapy, are contributing to a paradigm shift in the treatment of pancreatic cancer. This strategy is also promising in the treatment of pancreatic neck-body cancer. AIM To evaluate the feasibility and effectiveness of a multimodal strategy for the treatment of borderline/locally advanced pancreatic neck-body cancer. METHODS From January 2019 to December 2021, we reviewed the demographic characteristics, neoadjuvant and adjuvant treatment data, intraoperative and postoperative variables, and follow-up outcomes of patients who underwent multimodal treatment for pancreatic neck-body cancer in a prospectively collected database of our hospital. This investigation was reported in line with the Preferred Reporting of Case Series in Surgery criteria. RESULTS A total of 11 patients with pancreatic neck-body cancer were included in this study, of whom 6 patients were borderline resectable and 5 were locally advanced. Through multidisciplinary team discussion, all patients received neoadjuvant therapy, of whom 8 (73%) patients achieved a partial response and 3 patients maintained stable disease. After multidisciplinary team reassessment, all patients underwent laparoscopic subtotal distal pancreatectomy and portal vein reconstruction and achieved R0 resection. Postoperatively, two patients (18%) developed ascites, and two patients (18%) developed pancreatic fistulae. The median length of stay of the patients was 11 days (range: 10-15 days). All patients received postoperative adjuvant therapy. During the follow-up, three patients experienced tumor recurrence, with a median disease-free survival time of 13.3 months and a median overall survival time of 20.5 months. CONCLUSION A multimodal treatment strategy combining neoadjuvant therapy, laparoscopic subtotal distal pancreatectomy, and adjuvant therapy is safe and feasible in patients with pancreatic neck-body cancer.
Collapse
Affiliation(s)
- Jia Li
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China
| | - Xi-Tao Wang
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China
| | - Yi Wang
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China
| | - Kang Chen
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China
| | - Guo-Guang Li
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China
| | - Yan-Fei Long
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China
| | - Mei-Fu Chen
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China
| | - Chuang Peng
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China
| | - Yi Liu
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China
| | - Wei Cheng
- Department of Hepatobiliary Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China
| |
Collapse
|
31
|
Arita Y, Kwee TC, Akin O, Shigeta K, Paudyal R, Roest C, Ueda R, Lema-Dopico A, Nalavenkata S, Ruby L, Nissan N, Edo H, Yoshida S, Shukla-Dave A, Schwartz LH. Multiparametric MRI and artificial intelligence in predicting and monitoring treatment response in bladder cancer. Insights Imaging 2025; 16:7. [PMID: 39747744 PMCID: PMC11695553 DOI: 10.1186/s13244-024-01884-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Bladder cancer is the 10th most common and 13th most deadly cancer worldwide, with urothelial carcinomas being the most common type. Distinguishing between non-muscle-invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC) is essential due to significant differences in management and prognosis. MRI may play an important diagnostic role in this setting. The Vesical Imaging Reporting and Data System (VI-RADS), a multiparametric MRI (mpMRI)-based consensus reporting platform, allows for standardized preoperative muscle invasion assessment in BCa with proven diagnostic accuracy. However, post-treatment assessment using VI-RADS is challenging because of anatomical changes, especially in the interpretation of the muscle layer. MRI techniques that provide tumor tissue physiological information, including diffusion-weighted (DW)- and dynamic contrast-enhanced (DCE)-MRI, combined with derived quantitative imaging biomarkers (QIBs), may potentially overcome the limitations of BCa evaluation when predominantly focusing on anatomic changes at MRI, particularly in the therapy response setting. Delta-radiomics, which encompasses the assessment of changes (Δ) in image features extracted from mpMRI data, has the potential to monitor treatment response. In comparison to the current Response Evaluation Criteria in Solid Tumors (RECIST), QIBs and mpMRI-based radiomics, in combination with artificial intelligence (AI)-based image analysis, may potentially allow for earlier identification of therapy-induced tumor changes. This review provides an update on the potential of QIBs and mpMRI-based radiomics and discusses the future applications of AI in BCa management, particularly in assessing treatment response. CRITICAL RELEVANCE STATEMENT: Incorporating mpMRI-based quantitative imaging biomarkers, radiomics, and artificial intelligence into bladder cancer management has the potential to enhance treatment response assessment and prognosis prediction. KEY POINTS: Quantitative imaging biomarkers (QIBs) from mpMRI and radiomics can outperform RECIST for bladder cancer treatments. AI improves mpMRI segmentation and enhances radiomics feature extraction effectively. Predictive models integrate imaging biomarkers and clinical data using AI tools. Multicenter studies with strict criteria validate radiomics and QIBs clinically. Consistent mpMRI and AI applications need reliable validation in clinical practice.
Collapse
Affiliation(s)
- Yuki Arita
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Thomas C Kwee
- Department of Radiology, Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands
| | - Oguz Akin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Keisuke Shigeta
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Urology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Ramesh Paudyal
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christian Roest
- Department of Radiology, Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, The Netherlands
| | - Ryo Ueda
- Office of Radiation Technology, Keio University Hospital, Shinjuku-ku, Tokyo, Japan
| | - Alfonso Lema-Dopico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sunny Nalavenkata
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lisa Ruby
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Noam Nissan
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hiromi Edo
- Department of Radiology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Soichiro Yoshida
- Department of Urology, Institute of Science Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Amita Shukla-Dave
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lawrence H Schwartz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
32
|
Cui Y, Gao Y, Zhou Y, Ma X, Wang Y, Zhou T, Wen J, Chen S, Lu L, Tong A, Li Y. A novel strategy for predicting the efficacy of temozolomide treatment for metastatic pheochromocytomas/paragangliomas. J Endocrinol Invest 2024; 47:3039-3048. [PMID: 38837102 DOI: 10.1007/s40618-024-02398-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 05/20/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND There are few studies on the efficacy of temozolomide (TMZ) in the treatment of Metastatic pheochromocytoma / paraganglioma (MPP) patients. And it remains unclear which MPP patients may benefit from TMZ treatment. METHODS This was a prospective study. MPP patients were enrolled. Patients were treated with TMZ until disease progression or intolerable toxicities. The primary endpoints were disease control rate (DCR) and objective response rate (ORR). Secondary endpoints included biochemical response rate progression-free survival (PFS) and safety. We compared the difference between effective and ineffective groups, to explore which patients are more suitable for TMZ treatment. RESULTS 62 patients with MPP were enrolled and tumor response were evaluated in 54 patients. The DCR was 83% (35/42), and the ORR was 24% (10/41) among the progressive patients. PFS was 25.2 ± 3.1 months. The most common adverse event was nausea (41/55). We found that 92.9% (13/14) of patients with MGMT methylation greater than 7% respond to treatment. For the patients with MGMT methylation less than 7%, Ki-67 index could be used to guide the use of TMZ in these patients. Among the patients with Ki-67 index less than 5%, 66% (8/12) patients showed respond to treatment, and only 33% (4/12) patients with Ki-67 index more than 5% showed respond to TMZ. CONCLUSIONS This study indicated that TMZ is a potential choice for the treatment of MPP with the high ability on disease control and well tolerability. We recommended to MGMT methylation analysis test and Ki-67 index to guide TMZ application.
Collapse
Affiliation(s)
- Y Cui
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, 100730, Beijing, People's Republic of China
| | - Y Gao
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, 100730, Beijing, People's Republic of China
| | - Y Zhou
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, 100730, Beijing, People's Republic of China
| | - X Ma
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, 100730, Beijing, People's Republic of China
| | - Y Wang
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, 100730, Beijing, People's Republic of China
| | - T Zhou
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, 100730, Beijing, People's Republic of China
| | - J Wen
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, 100730, Beijing, People's Republic of China
| | - S Chen
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, 100730, Beijing, People's Republic of China
| | - L Lu
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, 100730, Beijing, People's Republic of China
| | - A Tong
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, 100730, Beijing, People's Republic of China.
| | - Y Li
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health Commission of the People's Republic of China, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, No.1 Shuaifuyuan, 100730, Beijing, People's Republic of China
| |
Collapse
|
33
|
Wyatt AW, Litiere S, Bidard FC, Cabel L, Dyrskjøt L, Karlovich CA, Pantel K, Petrie J, Philip R, Andrews HS, Vellanki PJ, Tolmeijer SH, Villalobos Alberu X, Alfano C, Bogaerts J, Calvo E, Chen AP, Toledo RA, de Vries EGE, Seymour L, Laurie SA, Garralda E. Plasma ctDNA as a Treatment Response Biomarker in Metastatic Cancers: Evaluation by the RECIST Working Group. Clin Cancer Res 2024; 30:5034-5041. [PMID: 39269996 DOI: 10.1158/1078-0432.ccr-24-1883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/08/2024] [Accepted: 09/13/2024] [Indexed: 09/15/2024]
Abstract
Early indicators of metastatic cancer response to therapy are important for evaluating new drugs and stopping ineffective treatment. The RECIST guidelines based on repeat cancer imaging are widely adopted in clinical trials, are used to identify active regimens that may change practice, and contribute to regulatory approvals. However, these criteria do not provide insight before 6 to 12 weeks of treatment and typically require that patients have measurable disease. Recent data suggest that measuring on-treatment changes in the amount or proportion of ctDNA in peripheral blood plasma may accurately identify responding and nonresponding cancers at earlier time points. Over the past year, the RECIST working group has evaluated current evidence for plasma ctDNA kinetics as a treatment response biomarker in metastatic cancers and early endpoint in clinical trials to identify areas of focus for future research and validation. Here, we outline the requirement for large standardized trial datasets, greater scrutiny of optimal ctDNA collection time points and assay thresholds, and consideration of regulatory body guidelines and patient opinions. In particular, clinically meaningful changes in plasma ctDNA abundance are likely to differ by cancer type and therapy class and must be assessed before ctDNA can be considered a potential pan-cancer response evaluation biomarker. Despite the need for additional data, minimally invasive on-treatment ctDNA measurements hold promise to build upon existing response assessments such as RECIST and offer opportunities for developing novel early endpoints for modern clinical trials.
Collapse
Affiliation(s)
- Alexander W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Genome Sciences Centre and Clinical Cancer Genomics Program, BC Cancer, Vancouver, British Columbia, Canada
| | - Saskia Litiere
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Francois-Clément Bidard
- Department of Medical Oncology, Institut Curie, Université Versailles Saint-Quentin, Université Paris-Saclay, Saint-Cloud, France
| | - Luc Cabel
- Department of Medical Oncology, Institut Curie, Université Versailles Saint-Quentin, Université Paris-Saclay, Saint-Cloud, France
| | - Lars Dyrskjøt
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Chris A Karlovich
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Klaus Pantel
- Department of Tumor Biology, Center for Experimental Medicine, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joan Petrie
- Canadian Cancer Trials Group, Kingston, Ontario, Canada
| | - Reena Philip
- Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, Maryland
| | | | - Paz J Vellanki
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Sofie H Tolmeijer
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Christian Alfano
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Jan Bogaerts
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Emiliano Calvo
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Alice P Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland
| | | | - Elisabeth G E de Vries
- University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Lesley Seymour
- Canadian Cancer Trials Group, Queen's University, Kingston, Ontario, Canada
| | - Scott A Laurie
- Division of Medical Oncology, The Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada
| | - Elena Garralda
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
34
|
Li Y, Zhao J, Li R, Yao X, Dong X, Zhang R, Li Y. Treatment options for tumor progression after initial immunotherapy in advanced non-small cell lung cancer: A real-world study. Neoplasia 2024; 57:101043. [PMID: 39226660 PMCID: PMC11403516 DOI: 10.1016/j.neo.2024.101043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/14/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024]
Abstract
OBJECTIVE Whether to continue administering immunotherapy to patients with advanced non-small cell lung cancer (NSCLC) who have experienced tumor progression remains controversial after immunotherapy. The aims were to explore survival outcomes after further immunotherapy post-progression and to determine the optimal combination therapy in such cases. METHODS Overall, 507 patients with NSCLC who underwent immunotherapy and experienced tumor progression were retrospectively divided into Immuno-combination and No-immuno groups according to whether additional combination therapy involving immunotherapy was administered post-progression. Progression-free survival (PFS) and overall survival (OS) were evaluated. Subgroup analyses were performed according to the different treatment regimens for patients in the Immuno-combination group. RESULTS After propensity score matching, there were 150 patients in the No-immuno group and 300 patients in the Immuno combination group. Superior PFS was observed in the Immuno-combination group compared with those in the No-immuno group (6-month PFS: 25.3 % vs. 60.6 %; 12-month PFS: 6.7 % vs. 24.4 %; P < 0.001). Similar intergroup differences were observed for OS (12-month OS: 22.3 % vs. 69.4 %; 18-month OS: 6.4 % vs. 40.4 %; P < 0.001). Superior PFS outcomes were observed in the Immuno+Antiangiogenic group compared with the Immuno+Chemo group (6-month PFS: 51.3 % vs. 71.5 %; 12-month PFS: 23.1 % vs. 25.7 %; P = 0.017). Similar differences in OS were observed between those same subgroups (12-month OS: 62.1 % vs. 77.9 %; 18-month OS: 33.3 % vs. 48.7 %; P = 0.006). CONCLUSION Patients with NSCLC experiencing tumor progression post-immunotherapy can still benefit from further treatment, with immunotherapy combined with antiangiogenic therapy the most efficacious option.
Collapse
Affiliation(s)
- Ying Li
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, China
| | - Junfeng Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, China
| | - Ruyue Li
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shan Dong, 261000, China
| | - Xiujing Yao
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shan Dong, 261000, China
| | - Xue Dong
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, China
| | - Ruidan Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, China
| | - Yintao Li
- Department of Respiratory Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, China.
| |
Collapse
|
35
|
Egerer M, Schuch K, Schöler D, Artusa F, Püngel T, Holtman TM, Loosen SH, Demir M, Wree A, Luedde T, Tacke F, Roderburg C, Mohr R. Extracellular Vesicles May Predict Response to Atezolizumab Plus Bevacizumab in Patients with Advanced Hepatocellular Carcinoma. Cancers (Basel) 2024; 16:3651. [PMID: 39518089 PMCID: PMC11545167 DOI: 10.3390/cancers16213651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND AND AIMS Treatment with atezolizumab and bevacizumab has been approved as one of the standards of care for patients with advanced hepatocellular carcinoma (HCC). The median overall survival (OS) upon available treatments still remains below 2 years, urgently suggesting better stratification tools to identify ideal candidates for this treatment and potentially allowing personalized approaches. In this study, we evaluated the potential role of extracellular vesicles (EVs) as a novel biomarker in patients receiving atezolizumab and bevacizumab for HCC. METHODS We characterized EVs in 212 longitudinal serum samples from an observational cohort of 53 individuals with advanced HCC, who started therapy with atezolizumab plus bevacizumab at our center between January 2020 and March 2022. RESULTS In our cohort, the overall efficacy of atezolizumab and bevacizumab was comparable to previously published phase III data. We detected significantly smaller EVs in treatment responders, while enlarged EVs were associated with significantly decreased efficacy of atezolizumab and bevacizumab in terms of OS. A decrease in vesicle size during immunotherapy was related to a longer progression-free survival (PFS). A univariate Cox regression analysis including various clinicopathological parameters (e.g., tumor stage, markers of inflammation, organ dysfunction, or tumor markers) revealed vesicle size as an independent prognostic marker in HCC patients receiving atezolizumab and bevacizumab. Moreover, higher vesicle concentrations and lower zeta potentials were identified as a positive prognostic factor throughout treatment. CONCLUSIONS Distinct EV characteristics such as vesicle size, concentration, and zeta potential represent promising novel biomarkers in patients with advanced HCC receiving atezolizumab and bevacizumab, potentially helping to identify optimal candidates for checkpoint inhibitor-based treatments.
Collapse
Affiliation(s)
- Mara Egerer
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.E.); (F.A.); (T.P.); (T.M.H.); (M.D.); (A.W.); (F.T.)
| | - Kathrin Schuch
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (K.S.); (D.S.); (S.H.L.); (T.L.); (C.R.)
| | - David Schöler
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (K.S.); (D.S.); (S.H.L.); (T.L.); (C.R.)
| | - Fabian Artusa
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.E.); (F.A.); (T.P.); (T.M.H.); (M.D.); (A.W.); (F.T.)
| | - Tobias Püngel
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.E.); (F.A.); (T.P.); (T.M.H.); (M.D.); (A.W.); (F.T.)
- Berlin Institute of Health, 10178 Berlin, Germany
| | - Theresa Maria Holtman
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.E.); (F.A.); (T.P.); (T.M.H.); (M.D.); (A.W.); (F.T.)
| | - Sven H. Loosen
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (K.S.); (D.S.); (S.H.L.); (T.L.); (C.R.)
| | - Münevver Demir
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.E.); (F.A.); (T.P.); (T.M.H.); (M.D.); (A.W.); (F.T.)
| | - Alexander Wree
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.E.); (F.A.); (T.P.); (T.M.H.); (M.D.); (A.W.); (F.T.)
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (K.S.); (D.S.); (S.H.L.); (T.L.); (C.R.)
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.E.); (F.A.); (T.P.); (T.M.H.); (M.D.); (A.W.); (F.T.)
| | - Christoph Roderburg
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (K.S.); (D.S.); (S.H.L.); (T.L.); (C.R.)
| | - Raphael Mohr
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité–Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.E.); (F.A.); (T.P.); (T.M.H.); (M.D.); (A.W.); (F.T.)
| |
Collapse
|
36
|
Xu Z, Jiang G, Dai J. Tumor therapeutics in the era of "RECIST": past, current insights, and future prospects. Oncol Rev 2024; 18:1435922. [PMID: 39493769 PMCID: PMC11527623 DOI: 10.3389/or.2024.1435922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/30/2024] [Indexed: 11/05/2024] Open
Abstract
In recent years, advancements in medical treatment and imaging technologies have revolutionized the assessment of tumor response. However, the Response Evaluation Criteria in Solid Tumors (RECIST) has long been established as the gold standard for evaluating tumor treatment. As treatment modalities evolve, the need for continuous refinement and adaptation of RECIST becomes increasingly apparent. This review explores the historical evolution, current applications, limitations, and future directions of RECIST. It discusses the challenges of distinguishing true progression from pseudo-progression in ICIs (immune checkpoint inhibitors), the integration of advanced imaging tools, and the necessity for RECIST criteria tailored to specific therapies like neoadjuvant treatments. The review highlights the ongoing efforts to enhance RECIST's accuracy and reliability in clinical decision-making and the potential for developing new standards to better evaluate treatment efficacy in the rapidly evolving landscape of oncology.
Collapse
Affiliation(s)
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Dai
- *Correspondence: Gening Jiang, ; Jie Dai,
| |
Collapse
|
37
|
Tan Y, Liu K, Zhu C, Wang S, Wang Y, Xue J, Ning C, Zhang N, Chao J, Zhang L, Long J, Yang X, Zeng D, Zhao L, Zhao H. Efficacy and safety of lenvatinib combined with anti-PD-1 antibodies plus GEMOX chemotherapy as non-first-line systemic therapy in advanced gallbladder cancer. Cancer Immunol Immunother 2024; 73:240. [PMID: 39358463 PMCID: PMC11447189 DOI: 10.1007/s00262-024-03831-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Lenvatinib, programmed cell death 1 (PD-1) antibodies, and gemcitabine and oxaliplatin (GEMOX) chemotherapy have shown significant antitumor activity as first-line therapy against biliary tract cancer. This study evaluated their efficacy and safety as non-first-line therapy in advanced gallbladder cancer (GBC). METHODS Patients with advanced GBC who received lenvatinib combined with anti-PD-1 antibodies and GEMOX chemotherapy as a non-first-line therapy were retrospectively analyzed. The primary endpoints were overall survival (OS) and progression-free survival (PFS), and the secondary endpoints were objective response rate (ORR) and safety. RESULTS A total of 36 patients with advanced GBC were included in this study. The median follow-up time was 11.53 (95% confidence interval (CI): 2.2-20.9) months, and the ORR was 36.1%. The median OS and PFS were 15.1 (95% CI: 3.2-26.9) and 6.1 (95% CI: 4.9-7.2) months, respectively. The disease control rate (DCR) and clinical benefit rate (CBR) were 75% and 61.1%, respectively. Subgroup analysis demonstrated that patients with programmed cell death-ligand 1 (PD-L1) expression had significantly longer PFS and OS than those without PD-L1 expression. Additionally, patients with a neutrophil-lymphocyte ratio (NLR) < 5.57 had a longer OS than those with an NLR ≥ 5.57. All patients experienced adverse events (AEs), with 61.1% experiencing grade 3 or 4 AEs, including myelosuppression (13.9%) and fatigue (13.3%), alanine transaminase or aspartate transaminase levels (8.3%), and diarrhea (8.3%). No grade 5 AEs were reported. CONCLUSION Anti-PD-1 antibodies combined with lenvatinib and GEMOX chemotherapy are effective and well-tolerated as a non-first-line therapy in advanced GBC. PD-L1 expression and baseline NLR may potentially predict treatment efficacy.
Collapse
Affiliation(s)
- Yang Tan
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563000, Guizhou, China
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Kai Liu
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563000, Guizhou, China
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Chengpei Zhu
- Department of General Surgery Center, Beijing YouAn Hospital, Clinical Center for Liver Cancer, Capital Medical University, 8 Xitoutiao, Youanmenwai, Fengtai, Beijing, 100069, China
| | - Shanshan Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Yunchao Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Jingnan Xue
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563000, Guizhou, China
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Cong Ning
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Nan Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Jiashuo Chao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Longhao Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Junyu Long
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China
| | - Xiaobo Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China.
| | - Daobing Zeng
- Department of General Surgery Center, Beijing YouAn Hospital, Clinical Center for Liver Cancer, Capital Medical University, 8 Xitoutiao, Youanmenwai, Fengtai, Beijing, 100069, China.
| | - Lijin Zhao
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563000, Guizhou, China.
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College (CAMS & PUMC), #1 Shuaifuyuan, Wangfujing, Beijing, 100730, China.
| |
Collapse
|
38
|
Kale P, Datta S, Kalbande P, Aher P, Singh A, Datta NR. Is it time to move from the Unidimensional RECIST 1.1 Response Assessment Criteria to a Volumetric Evaluation in the Present Era of Image-based Oncology? An Evaluation in Locally Advanced Head Neck Cancers Undergoing Treatment. Clin Oncol (R Coll Radiol) 2024; 36:615-623. [PMID: 39112341 DOI: 10.1016/j.clon.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/12/2024] [Accepted: 07/17/2024] [Indexed: 09/15/2024]
Abstract
AIMS Tumour response assessments, as per Response Evaluation Criteria in Solid Tumours (RECIST 1.1), are based on the sum of diameters (SODs) of the primary tumour (longest diameter) and/or short axis diameter of lymph nodes. This study evaluates the response categorisation as per RECIST 1.1 vs Computed tomography (CT) based volumetric assessment of RECIST (proposed as vRECIST) in locally advanced head and neck cancers (LAHNCs) undergoing treatment. MATERIAL AND METHODS The pre-treatment SODs and CT estimated tumour volumes were recorded in 45 LAHNCs treated with radiotherapy (RT), chemoradiotherapy (CTRT) or thermochemoradiotherapy (HTCTRT). Tumour responses were assessed independently as per RECIST 1.1 and vRECIST by two radiation oncologists and grouped into complete response (CR), partial response (PR), stable disease (SD) or progressive disease (PD). These response groups were evaluated for the likely congruence of the two approaches, as categorised independently by these two observers. RESULTS All patients in stages III (n = 7), IVA (n = 16) and IVB (n = 22) were inoperable and had received either RT alone (n = 1), CTRT (n = 12) or HTCTRT (n = 32). Based on SODs criteria of RECIST 1.1, of the 45 patients, 5 and 40 were grouped as PR and SD by the first observer, while this changed to 34 and 10, respectively and 1 PD, with vRECIST (p < 0.001). Similarly, for the second observer, the 4 PR and 41 SD grouped using RECIST 1.1 were recategorised to 34 PR, 10 SD, and 1 PD by vRECIST (p < 0.001). Thus, a mismatch of 66.6% and 68.8%, respectively, was evident by observers first and second in categorising SD based on SODs of RECIST 1.1 vs PR on vRECIST. CONCLUSIONS Treatment responses in LAHNCs assessed using SODs resulted in significant uncertainties and failed to reflect actual volumetric changes in tumours during treatment. It is perhaps time to consider replacing the SODs of RECIST 1.1 with vRECIST for unequivocal tumour response categorisation in the present era of image-based oncology practice.
Collapse
Affiliation(s)
- P Kale
- Department of Radiotherapy, Mahatma Gandhi Institute of Medical Sciences, Sevagram, Maharashtra, India; Department of Radiotherapy, Government Medical College, Nagpur, India.
| | - S Datta
- Department of Radiotherapy, Mahatma Gandhi Institute of Medical Sciences, Sevagram, Maharashtra, India; Independent Researcher in Data Science, London, UK.
| | - P Kalbande
- Department of Radiotherapy, Mahatma Gandhi Institute of Medical Sciences, Sevagram, Maharashtra, India; Department of Radiotherapy, University Hospital, Leicester, UK.
| | - P Aher
- Department of Radiotherapy, Mahatma Gandhi Institute of Medical Sciences, Sevagram, Maharashtra, India.
| | - A Singh
- Department of Radiotherapy, Mahatma Gandhi Institute of Medical Sciences, Sevagram, Maharashtra, India.
| | - N R Datta
- Department of Radiotherapy, Mahatma Gandhi Institute of Medical Sciences, Sevagram, Maharashtra, India.
| |
Collapse
|
39
|
Tomonari T, Tani J, Sato Y, Tanaka H, Morishita A, Okamoto K, Kawano Y, Sogabe M, Miyamoto H, Takayama T. Initial treatment efficacy and safety of durvalumab plus tremelimumab combination therapy in unresectable hepatocellular carcinoma in clinical practice. JGH Open 2024; 8:e70033. [PMID: 39371045 PMCID: PMC11450737 DOI: 10.1002/jgh3.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 10/08/2024]
Abstract
Background and Aims We aimed to evaluate the efficacy and safety of durvalumab plus tremelimumab (Dur + Tre) combination therapy in patients with unresectable hepatocellular carcinoma (uHCC) in clinical practice. Methods We retrospectively evaluated 37 patients with uHCC from our institutions between April 2023 and January 2024. Patients were divided into first- and later-line groups for analysis of antitumor efficacy, adverse events (AEs), and transition rate to second-line treatment according to the Response Evaluation Criteria in Solid Tumors (RECIST). Results The disease control rate (DCR) for the first-line group was 80.9%, which was significantly higher than that for the later-line group (50%). The incidence of immune-related AEs (irAEs) was 24.3%, with grade 3 or higher irAEs including increased transaminase (8.1%), diarrhea (8.1%), and adrenal insufficiency (2.7%). The rates of drug withdrawal and discontinuation owing to AEs were 23.8% and 19%, respectively, in the first-line treatment and 31.2% and 12.5%, respectively, in the later-line treatment, with no significant difference. Analysis of changes in liver reserve using the albumin-bilirubin (ALBI) score showed no obvious loss of liver reserve for up to 12 weeks. The transition rate from first- to second-line therapy after progressive disease (PD) was as high as 94.7%. Conclusion The efficacy and safety of Dur + Tre in clinical practice were comparable to those reported in a recent phase III trial. The first-line Dur + Tre therapy had a higher DCR than that of the later lines, and the transition rate to second-line therapy was considerably high, suggesting that Dur + Tre therapy would be more beneficial in first-line treatment.
Collapse
Affiliation(s)
- Tetsu Tomonari
- Department of Gastroenterology and Oncology, Institute of Biomedical SciencesTokushima University Graduate School of MedicineTokushimaJapan
| | - Joji Tani
- Department of Gastroenterology and NeurologyKagawa University Graduate School of MedicineKagawaJapan
| | - Yasushi Sato
- Department of Community Medicine for Gastroenterology and Oncology, Institute of Biomedical SciencesTokushima University Graduate School of MedicineTokushimaJapan
| | - Hironori Tanaka
- Department of Gastroenterology and Oncology, Institute of Biomedical SciencesTokushima University Graduate School of MedicineTokushimaJapan
| | - Akihiro Morishita
- Department of Gastroenterology and NeurologyKagawa University Graduate School of MedicineKagawaJapan
| | - Koichi Okamoto
- Department of Gastroenterology and Oncology, Institute of Biomedical SciencesTokushima University Graduate School of MedicineTokushimaJapan
| | - Yutaka Kawano
- Department of Gastroenterology and Oncology, Institute of Biomedical SciencesTokushima University Graduate School of MedicineTokushimaJapan
| | - Masahiro Sogabe
- Department of Gastroenterology and Oncology, Institute of Biomedical SciencesTokushima University Graduate School of MedicineTokushimaJapan
| | - Hiroshi Miyamoto
- Department of Gastroenterology and Oncology, Institute of Biomedical SciencesTokushima University Graduate School of MedicineTokushimaJapan
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Institute of Biomedical SciencesTokushima University Graduate School of MedicineTokushimaJapan
| |
Collapse
|
40
|
Liu T, Chen X, Mo S, Zhou T, Ma W, Chen G, Chen X, Shi M, Yang Y, Huang Y, Zhao H, Fang W, Yang Y, Li J, Zhang L, Zhao Y. Molecular subtypes and prognostic factors of lung large cell neuroendocrine carcinoma. Transl Lung Cancer Res 2024; 13:2222-2235. [PMID: 39430332 PMCID: PMC11484736 DOI: 10.21037/tlcr-24-292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/26/2024] [Indexed: 10/22/2024]
Abstract
Background Lung large cell neuroendocrine carcinoma (LCNEC) is an aggressive disease with poor prognosis and short-term survival, which lacks effective prognostic indicators. The study aims to investigate the molecular subtypes and prognostic markers of lung LCNEC. Methods Patients diagnosed with lung LCNEC at Sun Yat-sen University Cancer Center (SYSUCC) between November 2007 and January 2021 were screened. Baseline clinical data were collected and routine blood indexes including lymphocyte-to-monocyte ratio (LMR), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR) and systemic immune-inflammation index (SII) were calculated. Immunohistochemistry (IHC) of ASCL1, NEUROD1, POU2F3, YAP1 were done to perform molecular subtyping, while CD56, Syn, CgA, CD3, CD8, CD20, CD68, and CD163 were also stained on tissue samples. Then prognostic factors of lung LCNEC were explored. Results One hundred and fifty-one lung LCNEC patients were identified, 103 of whom had complete clinical information, available routine blood and biochemical indexes were eventually included in the present study. Tumor tissue specimens were available from 64 patients. Positive expression rates of ASCL1, NEUROD1, and YAP1 were 82.8%, 50.0%, and 28.1%, respectively. No POU2F3+ cases were detected. Forty (62.5%) patients co-expressed with two or three markers. High LMR (>3.3) was an independent predictor of favorable prognosis of disease-free survival (DFS) [hazard ratio (HR), 0.391; 95% confidence interval (CI): 0.161-0.948; P=0.04] and overall survival (OS) (HR, 0.201; 95% CI: 0.071-0.574; P=0.003). Notably, high LMR was correlated with higher intra-tumoral CD3+ (P=0.004), CD8+ (P=0.01), and CD68+ (P<0.001) immune cell infiltration compared to low LMR in lung LCNEC. Conclusions Our study validated molecular subtypes by IHC in lung LCNEC, and co-expression was found among different subtypes, with no prognostic effect. High blood LMR level was associated with a favorable prognosis in lung LCNEC, which might partly reflect a hot tumor tissue immune microenvironment. Our findings may benefit clinical practice, and further studies are warranted.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xueyuan Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Silang Mo
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ting Zhou
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenjuan Ma
- Department of Intensive Care Unit, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Gang Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiang Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mengting Shi
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuwen Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hongyun Zhao
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenfeng Fang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yunpeng Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing Li
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuanyuan Zhao
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
41
|
Rimbaș M, Dumitru AC, Tripodi G, Larghi A. EUS-Guided Radiofrequency Ablation Therapy for Pancreatic Neoplasia. Diagnostics (Basel) 2024; 14:2111. [PMID: 39410514 PMCID: PMC11476430 DOI: 10.3390/diagnostics14192111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
Radiofrequency ablation (RFA) under endoscopic ultrasound (EUS) guidance has been developed and utilized over the last decade to provide the loco-regional treatment of solid and cystic pancreatic neoplastic lesions. The advantage of this approach relies on the close proximity of the EUS transducer to the target pancreatic lesion, which, coupled with the development of specifically designed RFA ablation devices, has made the procedure minimally invasive, with a clear reduction in adverse events as compared to the high morbidity of the surgical approach. EUS-RFA has been applied so far to pancreatic functional and non-functional neuroendocrine neoplasms, pancreatic ductal adenocarcinoma or metastases to the pancreas, and pancreatic neoplastic cysts. Excluding neuroendocrine tumors, for other indications, most of these procedures have been performed in patients who refused surgery or were at high surgical risk. More studies evaluating EUS-RFA in selected patients, not at surgical risk, are gradually becoming available and will pave the road to extend the indications for this therapeutic approach, also in association with other oncological therapies. The present manuscript will critically review the available evidence in the field of the EUS-guided RFA of solid and cystic pancreatic neoplasms.
Collapse
Affiliation(s)
- Mihai Rimbaș
- Gastroenterology Department, Colentina Clinical Hospital, Carol Davila University of Medicine, 020125 Bucharest, Romania; (M.R.); (A.-C.D.)
| | - Andra-Cristiana Dumitru
- Gastroenterology Department, Colentina Clinical Hospital, Carol Davila University of Medicine, 020125 Bucharest, Romania; (M.R.); (A.-C.D.)
| | - Giulia Tripodi
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- CERTT, Center for Endoscopic Research Therapeutics and Training, Catholic University, 00168 Rome, Italy
| | - Alberto Larghi
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- CERTT, Center for Endoscopic Research Therapeutics and Training, Catholic University, 00168 Rome, Italy
| |
Collapse
|
42
|
Shi Y, Wu Z, Liu S, Zuo D, Niu Y, Qiu Y, Qiao L, He W, Qiu J, Yuan Y, Wang G, Li B. Targeting PRMT3 impairs methylation and oligomerization of HSP60 to boost anti-tumor immunity by activating cGAS/STING signaling. Nat Commun 2024; 15:7930. [PMID: 39256398 PMCID: PMC11387718 DOI: 10.1038/s41467-024-52170-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 08/28/2024] [Indexed: 09/12/2024] Open
Abstract
Immune checkpoint blockade (ICB) has emerged as a promising therapeutic option for hepatocellular carcinoma (HCC), but resistance to ICB occurs and patient responses vary. Here, we uncover protein arginine methyltransferase 3 (PRMT3) as a driver for immunotherapy resistance in HCC. We show that PRMT3 expression is induced by ICB-activated T cells via an interferon-gamma (IFNγ)-STAT1 signaling pathway, and higher PRMT3 expression levels correlate with reduced numbers of tumor-infiltrating CD8+ T cells and poorer response to ICB. Genetic depletion or pharmacological inhibition of PRMT3 elicits an influx of T cells into tumors and reduces tumor size in HCC mouse models. Mechanistically, PRMT3 methylates HSP60 at R446 to induce HSP60 oligomerization and maintain mitochondrial homeostasis. Targeting PRMT3-dependent HSP60 methylation disrupts mitochondrial integrity and increases mitochondrial DNA (mtDNA) leakage, which results in cGAS/STING-mediated anti-tumor immunity. Lastly, blocking PRMT3 functions synergize with PD-1 blockade in HCC mouse models. Our study thus identifies PRMT3 as a potential biomarker and therapeutic target to overcome immunotherapy resistance in HCC.
Collapse
Affiliation(s)
- Yunxing Shi
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Colorectal Surgery, Guangdong Institute of Gastroenterology, and Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zongfeng Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shaoru Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Dinglan Zuo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yi Niu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yuxiong Qiu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Liang Qiao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wei He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jiliang Qiu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yunfei Yuan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Guocan Wang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Binkui Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
43
|
Lian J, Wang R, Wang X, Pang X, Xu B, Tang S, Shao J, Lu H. Irinotecan plus raltitrexed as second‑line chemotherapy for metastatic colorectal cancer: A retrospective study. Oncol Lett 2024; 28:409. [PMID: 38988448 PMCID: PMC11234808 DOI: 10.3892/ol.2024.14542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/28/2023] [Indexed: 07/12/2024] Open
Abstract
The present study evaluated the efficiency, prognostic factors for and the safety of irinotecan combined with raltitrexed (TOMIRI) in patients with metastatic colorectal cancer (CRC). Outcome data of patients who received TOMIRI as first-, second- and third- or later-line treatment regimens were assessed to compare the efficacy of this regimen. Progression-free survival (PFS), overall survival (OS), objective response rate (ORR) and disease control rate (DCR) were evaluated for each group. Kaplan-Meier curves and univariate and multivariate analyses were performed to evaluate efficacy. From January 2017 to December 2019, TOMIRI was administered as a first-line treatment in 23 patients, second-line treatment in 164 patients and third- or later-line treatment in 18 patients. Irinotecan and 5-fluorouracil (FOLFIRI) was administered to another 50 patients, who served as the control group. The median PFS was 9, 7 and 6 months and the median OS was 37, 21 and 17 months for first-, second- and third- or later-line treatments, respectively. The ORRs of the included patients were 21.7, 13.4 and 11.1%, respectively, and the DCRs were 91.3, 81.7 and 66.7%, respectively. Compared with FOLFIRI, TOMIRI as a second-line chemotherapy treatment was associated with longer survival of the patients with CRC. Further analysis demonstrated that pathologic tumor-node-metastasis category, carcinoembryonic antigen, carbohydrate antigen 19-9, treatment cycles, targeted therapy, treatment of local metastases and first-line PFS were prognostic factors for second-line treatment. Among these, the number of treatment cycles was of vital importance. Hepatic dysfunction was the most commonly reported grade 1-2 (55.1%) and grade 3-4 (7.3%) adverse event. Neutropenia (12.2%), thrombocytopenia (10.2%), anemia (27.3%), proteinuria (38.1%) and hematuria (21.0%) were also common grade 1-2 adverse events. In conclusion, TOMIRI may be recommended as an effective and safe second-line treatment for metastatic CRC in the clinic.
Collapse
Affiliation(s)
- Jie Lian
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150040, P.R. China
| | - Ren Wang
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150040, P.R. China
| | - Xin Wang
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150040, P.R. China
| | - Xiangyi Pang
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150040, P.R. China
| | - Benjie Xu
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150040, P.R. China
| | - Shuli Tang
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150040, P.R. China
| | - Jiayue Shao
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150040, P.R. China
| | - Haibo Lu
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
44
|
Zhao D, Wang A, Li Y, Cai X, Zhao J, Zhang T, Zhao Y, Dong Y, Zhou F, Li Y, Wang J. Establishing the homologous recombination score threshold in metastatic prostate cancer patients to predict the efficacy of PARP inhibitors. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:280-287. [PMID: 39281716 PMCID: PMC11401495 DOI: 10.1016/j.jncc.2024.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 09/18/2024] Open
Abstract
Background The homologous recombination deficiency (HRD) score serves as a promising biomarker to identify patients who are eligible for treatment with PARP inhibitors (PARPi). Previous studies have suggested a 3-biomarker Genomic Instability Score (GIS) threshold of ≥ 42 as a valid biomarker to predict response to PARPi in patients with ovarian cancer and breast cancer. However, the GIS threshold for prostate cancer (PCa) is still lacking. Here, we conducted an exploratory analysis to investigate an appropriate HRD score threshold and to evaluate its ability to predict response to PARPi in PCa patients. Methods A total of 181 patients with metastatic castration-resistant PCa were included in this study. Tumor tissue specimens were collected for targeted next-generation sequencing for homologous recombination repair (HRR) genes and copy number variation (CNV) analysis. The HRD score was calculated based on over 50,000 single-nucleotide polymorphisms (SNP) distributed across the human genome, incorporating three SNP-based assays: loss of heterozygosity, telomeric allelic imbalance, and large-scale state transition. The HRD score threshold was set at the last 5th percentile of the HRD scores in our cohort of known HRR-deficient tumors. The relationship between the HRD score and the efficacy in 16 patients of our cohort who received PARPi treatment were retrospectively analyzed. Results Genomic testing was succeeded in 162 patients. In our cohort, 61 patients (37.7%) had HRR mutations (HRRm). BRCA mutations occurred in 15 patients (9.3%). The median HRD score was 4 (ranged from 0 to 57) in the total cohort, which is much lower than that in breast and ovarian cancers. Patients who harbored HRRm and BRCA or TP53 mutations had higher HRD scores. CNV occured more frequently in patients with HRRm. The last 5th percentile of HRD scores was 43 in the HRR-mutant cohort and consequently HRD high was defined as HRD scores ≥ 43. In the 16 patients who received PARPi in our cohort, 4 patients with a high HRD score achieved an objective response rate (ORR) of 100% while 12 patients with a low HRD score achieved an ORR of 8.3%. Progression-free survival (PFS) in HRD high patients was longer compared to HRD low patients, regardless of HRRm. Conclusions A HRD score threshold of 43 was established and preliminarily validated to predict the efficacy of PARPi in this study. Future studies are needed to further verify this threshold.
Collapse
Affiliation(s)
- Diwei Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Anqi Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuanwei Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xinyang Cai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Junliang Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tianyou Zhang
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Zhao
- Precision Scientific (Beijing) Co. Ltd., Beijing, China
| | - Yu Dong
- Precision Scientific (Beijing) Co. Ltd., Beijing, China
| | - Fangjian Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yonghong Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jun Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
45
|
Promsorn P, Yamaguchi T, Kosaka H, Aoi K, Yoshida K, Matsushima H, Matsui K, Shimoda S, Kaibori M, Naganuma M. Efficacy of lenvatinib and transarterial chemoembolization combination therapy in patients with hepatocellular carcinoma administered an insufficient dose of early lenvatinib. Mol Clin Oncol 2024; 21:63. [PMID: 39071976 PMCID: PMC11273243 DOI: 10.3892/mco.2024.2761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/12/2024] [Indexed: 07/30/2024] Open
Abstract
Recently, the relationship between the relative dose intensity (RDI) and efficacy was demonstrated for lenvatinib therapy in patients with advanced hepatocellular carcinoma (HCC), with a higher RDI of lenvatinib monotherapy indicating a higher efficacy. However, not every patient can tolerate a high RDI during the course of treatment; therefore, add-on combination therapy may be necessary for patients requiring a low RDI. The addition of transarterial chemoembolization (TACE) to lenvatinib therapy improves clinical outcomes. Therefore, the aim of the present study was to compare the clinical outcomes of lenvatinib plus TACE (the LEN-TACE group) with those of lenvatinib alone (the LEN group) in patients with unresectable HCC with a high- or low-RDI. A total of 66 patients with advanced HCC were enrolled in the present retrospective study. Eligible patients were those who initiated lenvatinib monotherapy between April 2018 and September 2020. Of these patients, 29 had an 8-week RDI of ≥60%, 6 of which received LEN-TACE. A further 37 patients had an 8-week RDI of <60%, 7 of which received LEN-TACE. In the high-RDI group, both the radiological evaluations and the overall survival (OS) time were improved in those in the low-RDI group. In addition, the median OS of patients treated with LEN-TACE was longer compared with that of patients treated with lenvatinib alone in the low-RDI group (P=0.0467). Therefore, the results of the present study revealed that early TACE should be considered instead of continuing lenvatinib only treatment in patients receiving an insufficient dose of lenvatinib, such as those with an 8-week RDI of <60%.
Collapse
Affiliation(s)
- Panuwat Promsorn
- Department of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1101, Japan
| | - Takashi Yamaguchi
- Department of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1101, Japan
| | - Hisashi Kosaka
- Department of Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Kazunori Aoi
- Department of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1101, Japan
| | - Katsunori Yoshida
- Department of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1101, Japan
| | - Hideyuki Matsushima
- Department of Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Kosuke Matsui
- Department of Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Shinji Shimoda
- Department of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1101, Japan
| | - Masaki Kaibori
- Department of Surgery, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Makoto Naganuma
- Department of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka 573-1101, Japan
| |
Collapse
|
46
|
Chen Y, Zhang T, Zhang R, Cao X. Pyrotinib and Trastuzumab Plus Chemotherapy Serve as an Acceptable Neoadjuvant Regimen Exhibiting Good Efficacy and Tolerance in HER2-Positive Breast Cancer Patients. Cancer Biother Radiopharm 2024; 39:435-440. [PMID: 38527247 DOI: 10.1089/cbr.2023.0175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024] Open
Abstract
Objective: Pyrotinib, a new irreversible dual pan-human epidermal growth factor receptor 2 (HER2) receptor tyrosine kinase inhibitor blocking EGFR and HER2, has achieved a promising efficacy for advanced HER2-positive (HER2+) breast cancer. This study intended to further investigate the efficacy and safety of neoadjuvant pyrotinib and trastuzumab plus chemotherapy for HER2+ breast cancer treatment. Methods: Thirty-eight HER2+ breast cancer patients who received neoadjuvant pyrotinib and trastuzumab plus chemotherapy (docetaxel and carboplatin) were retrospectively reviewed. Clinical response by Response Evaluation Criteria in Solid Tumors (RECIST), pathological complete response (pCR), and adverse events data was retrieved. Results: According to the RECIST, the complete response rate was 0.0%, 10.5%, and 15.8% after second-cycle, fourth-cycle, and sixth-cycle therapy, respectively; whereas the objective response rate was 76.3%, 92.1%, and 100.0%, accordingly. The total pCR (tpCR) rate was 52.6%, the pCR rate of the breast was also 52.6%, and the pCR rate of lymph nodes was 86.8%. The tpCR rate was lower in patients with HER2 immunohistochemistry (IHC)++ and amplification by fluorescent in situ hybridization (FISH) than in those with HER2 IHC+++ (14.3% vs. 61.3%, p = 0.024), which was also lower in patients with Ki-67 expression ≥30% than in those with Ki-67 expression <30% (40.0% vs. 76.9%, p = 0.031). The common adverse events included diarrhea (84.2%), anemia (73.7%), nausea and vomiting (63.2%), fatigue (50.0%), hypomagnesemia (44.7%), leukopenia (42.1%), thrombocytopenia (39.5%), elevated transaminase (36.8%), and pruritus (31.6%). Conclusions: Pyrotinib and trastuzumab plus chemotherapy serve as an acceptable neoadjuvant regimen exhibiting good efficacy and tolerance in HER2+ breast cancer patients, while further large-scale validation is warranted.
Collapse
Affiliation(s)
- Yibo Chen
- Department of General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tianyi Zhang
- School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Rui Zhang
- Department of General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, China
| | - Xuchen Cao
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
47
|
Gupta PK, Orlovskiy S, Arias-Mendoza F, Nelson DS, Nath K. 1H and 31P Magnetic Resonance Spectroscopic Metabolomic Imaging: Assessing Mitogen-Activated Protein Kinase Inhibition in Melanoma. Cells 2024; 13:1220. [PMID: 39056801 PMCID: PMC11274771 DOI: 10.3390/cells13141220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
The MAPK signaling pathway with BRAF mutations has been shown to drive the pathogenesis of 40-60% of melanomas. Inhibitors of this pathway's BRAF and MEK components are currently used to treat these malignancies. However, responses to these treatments are not always successful. Therefore, identifying noninvasive biomarkers to predict treatment responses is essential for personalized medicine in melanoma. Using noninvasive 1H magnetic resonance spectroscopy (1H MRS), we previously showed that BRAF inhibition reduces lactate and alanine tumor levels in the early stages of effective therapy and could be considered as metabolic imaging biomarkers for drug response. The present work demonstrates that these metabolic changes observed by 1H MRS and those assessed by 31P MRS are also found in preclinical human melanoma models treated with MEK inhibitors. Apart from 1H and 31P MRS, additional supporting in vitro biochemical analyses are described. Our results indicate significant early metabolic correlations with response levels to MEK inhibition in the melanoma models and are consistent with our previous study of BRAF inhibition. Given these results, our study supports the potential clinical utility of noninvasive MRS to objectively image metabolic biomarkers for the early prediction of melanoma's response to MEK inhibition.
Collapse
Affiliation(s)
- Pradeep Kumar Gupta
- Molecular Imaging Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (F.A.-M.); (D.S.N.)
| | - Stepan Orlovskiy
- Molecular Imaging Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (F.A.-M.); (D.S.N.)
| | - Fernando Arias-Mendoza
- Molecular Imaging Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (F.A.-M.); (D.S.N.)
- Advanced Imaging Research, Inc., Cleveland, OH 44114, USA
| | - David S. Nelson
- Molecular Imaging Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (F.A.-M.); (D.S.N.)
| | - Kavindra Nath
- Molecular Imaging Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; (S.O.); (F.A.-M.); (D.S.N.)
| |
Collapse
|
48
|
Chon YE, Kim DY, Kim MN, Kim BK, Kim SU, Park JY, Ahn SH, Ha Y, Lee JH, Lee KS, Kang B, Kim JS, Chon HJ, Kim DY. Sorafenib vs. Lenvatinib in advanced hepatocellular carcinoma after atezolizumab/bevacizumab failure: A real-world study. Clin Mol Hepatol 2024; 30:345-359. [PMID: 38468561 PMCID: PMC11261222 DOI: 10.3350/cmh.2023.0553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND/AIMS Atezolizumab plus bevacizumab (ATE+BEV) therapy has become the recommended first-line therapy for patients with unresectable hepatocellular carcinoma (HCC) because of favorable treatment responses. However, there is a lack of data on sequential regimens after ATE+BEV treatment failure. We aimed to investigate the clinical outcomes of patients with advanced HCC who received subsequent systemic therapy for disease progression after ATE+BEV. METHODS This multicenter, retrospective study included patients who started second-line systemic treatment with sorafenib or lenvatinib after HCC progressed on ATE+BEV between August 2019 and December 2022. Treatment response was assessed using the Response Evaluation Criteria in Solid Tumors (version 1.1.). Clinical features of the two groups were balanced through propensity score (PS) matching. RESULTS This study enrolled 126 patients, 40 (31.7%) in the lenvatinib group, and 86 (68.3%) in the sorafenib group. The median age was 63 years, and males were predominant (88.1%). In PS-matched cohorts (36 patients in each group), the objective response rate was similar between the lenvatinib- and sorafenib-treated groups (5.6% vs. 8.3%; P=0.643), but the disease control rate was superior in the lenvatinib group (66.7% vs. 22.2%; P<0.001). Despite the superior progression- free survival (PFS) in the lenvatinib group (3.5 vs. 1.8 months, P=0.001), the overall survival (OS, 10.3 vs. 7.5 months, P=0.353) did not differ between the two PS-matched treatment groups. CONCLUSION In second-line therapy for unresectable HCC after ATE+BEV failure, lenvatinib showed better PFS and comparable OS to sorafenib in a real-world setting. Future studies with larger sample sizes and longer follow-ups are needed to optimize second-line treatment.
Collapse
Affiliation(s)
- Young Eun Chon
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Dong Yun Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Mi Na Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Beom Kyung Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Up Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jun Yong Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Hoon Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Yeonjung Ha
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Joo Ho Lee
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Kwan Sik Lee
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Beodeul Kang
- Department of Medical Oncology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Jung Sun Kim
- Department of Medical Oncology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Hong Jae Chon
- Department of Medical Oncology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Do Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
49
|
Pastò B, Buzzatti G, Schettino C, Malapelle U, Bergamini A, De Angelis C, Musacchio L, Dieci MV, Kuhn E, Lambertini M, Passarelli A, Toss A, Farolfi A, Roncato R, Capoluongo E, Vida R, Pignata S, Callari M, Baldassarre G, Bartoletti M, Gerratana L, Puglisi F. Unlocking the potential of Molecular Tumor Boards: from cutting-edge data interpretation to innovative clinical pathways. Crit Rev Oncol Hematol 2024; 199:104379. [PMID: 38718940 DOI: 10.1016/j.critrevonc.2024.104379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/02/2024] [Accepted: 05/01/2024] [Indexed: 05/22/2024] Open
Abstract
The emerging era of precision medicine is characterized by an increasing availability of targeted anticancer therapies and by the parallel development of techniques to obtain more refined molecular data, whose interpretation may not always be straightforward. Molecular tumor boards gather various professional figures, in order to leverage the analysis of molecular data and provide prognostic and predictive insights for clinicians. In addition to healthcare development, they could also become a tool to promote knowledge and research spreading. A growing body of evidence on the application of molecular tumor boards to clinical practice is forming and positive signals are emerging, although a certain degree of heterogeneity exists. This work analyzes molecular tumor boards' potential workflows, figures involved, data sources, sample matrices and eligible patients, as well as available evidence and learning examples. The emerging concept of multi-institutional, disease-specific molecular tumor boards is also considered by presenting two ongoing nationwide experiences.
Collapse
Affiliation(s)
- Brenno Pastò
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Giulia Buzzatti
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova 16132, Italy
| | - Clorinda Schettino
- Clinical Trials Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Napoli 80131, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Napoli 80131, Italy
| | - Alice Bergamini
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milano 20132, Italy; Unit of Obstetrics and Gynaecology, IRCCS San Raffaele Scientific Institute, Milano 20132, Italy
| | - Carmine De Angelis
- Oncology Unit - Department of Clinical Medicine and Surgery, University of Naples Federico II, Napoli 80131, Italy
| | - Lucia Musacchio
- Department of Women and Child Health, Division of Gynaecologic Oncology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Roma 00168, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova 35122, Italy; Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova 35128, Italy
| | - Elisabetta Kuhn
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milano 20122, Italy; Pathology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Matteo Lambertini
- Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova 16132, Italy; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova 16132, Italy
| | - Anna Passarelli
- Department of Urology and Gynaecology, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Napoli 80131, Italy
| | - Angela Toss
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena 41124, Italy; Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena 41124, Italy
| | - Alberto Farolfi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola 47014, Italy
| | - Rossana Roncato
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
| | - Ettore Capoluongo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Napoli 80131, Italy; Clinical Pathology Unit, Azienda Ospedaliera San Giovanni Addolorata, Roma 00184, Italy
| | - Riccardo Vida
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Sandro Pignata
- Department of Urology and Gynaecology, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Napoli 80131, Italy
| | | | - Gustavo Baldassarre
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano 33081, Italy
| | - Michele Bartoletti
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Lorenzo Gerratana
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy.
| | - Fabio Puglisi
- Department of Medicine (DMED), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| |
Collapse
|
50
|
Sakurai Y, Iwasaki F, Hirose A, Matsumoto N, Miyagawa N, Keino D, Yokosuka T, Hamanoue S, Yanagimachi M, Shiomi M, Goto S, Tanaka M, Tanaka Y, Nozawa K, Goto H. Metronomic Chemotherapy for Pediatric Refractory Solid Tumors: A Retrospective Single-center Study. J Pediatr Hematol Oncol 2024; 46:233-240. [PMID: 38691057 DOI: 10.1097/mph.0000000000002870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/28/2024] [Indexed: 05/03/2024]
Abstract
Metronomic chemotherapy (MC) is based on chronic administration of chemotherapeutic agents at minimally toxic doses without prolonged drug-free breaks, that inhibits tumor angiogenesis and induces tumor dormancy. This study aimed to determine the efficacy of MC for pediatric refractory solid tumors. We retrospectively analyzed the data of pediatric patients with relapsed/refractory solid tumors who received treatment, including low-dose continuous administration of anticancer drugs, at our institute. Of the 18 patients, the disease statuses at the initiation of MC were complete remission (n=2), partial remission/stable disease (n=5), and progressive disease (n=11). The overall survival rate was 61% at 12 months and 34% at 24 months, and the progression-free survival rate was 21% at 12 and 24 months. Although only 5 of the 18 patients showed certain tumor regression or maintained remission, tumors that stabilized, maintained remission/stable disease, and showed certain advantages in terms of overall survival rate, even if limited to progressive disease. Approximately half of the patients demonstrated temporal tumor stabilization and improved survival time. Overall, previous reports and the present study support the conclusion that MC has the potential to play an important role in pediatric cancer treatment during the advanced stage.
Collapse
Affiliation(s)
- Yukari Sakurai
- Division of Hematology/Oncology, Kanagawa Children's Medical Center
| | - Fuminori Iwasaki
- Division of Hematology/Oncology, Kanagawa Children's Medical Center
| | - Ayana Hirose
- Division of Hematology/Oncology, Kanagawa Children's Medical Center
| | - Naoya Matsumoto
- Division of Hematology/Oncology, Kanagawa Children's Medical Center
| | - Naoyuki Miyagawa
- Division of Hematology/Oncology, Kanagawa Children's Medical Center
| | - Dai Keino
- Division of Hematology/Oncology, Kanagawa Children's Medical Center
| | - Tomoko Yokosuka
- Division of Hematology/Oncology, Kanagawa Children's Medical Center
| | - Satoshi Hamanoue
- Division of Hematology/Oncology, Kanagawa Children's Medical Center
| | | | - Masae Shiomi
- Division of Hematology/Oncology, Kanagawa Children's Medical Center
| | - Shoko Goto
- Division of Hematology/Oncology, Kanagawa Children's Medical Center
| | | | | | - Kumiko Nozawa
- Radiology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hiroaki Goto
- Division of Hematology/Oncology, Kanagawa Children's Medical Center
| |
Collapse
|