51
|
Popp D, Siedlecki M, Friedrich L, Haerting M, Scheurig-Muenkler C, Schwarz F, Kroencke T, Bette S, Decker JA. Potential of photon-counting detector CT technology for contrast medium reduction in portal venous phase thoracoabdominal CT. Eur Radiol 2025:10.1007/s00330-025-11409-3. [PMID: 39939421 DOI: 10.1007/s00330-025-11409-3] [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: 10/01/2024] [Revised: 12/17/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025]
Abstract
OBJECTIVES To compare image quality and iodine attenuation intra-individually in portal venous phase photon-counting detector CT (PCD-CT) scans using protocols with different contrast medium (CM) volume. MATERIALS AND METHODS A prospectively acquired patient cohort between 04/2021 and 11/2023 was retrospectively screened if patients had the following combination of portal venous phase thoracoabdominal CT scans: (a) PCD-CT with 120 mL CM volume (PCD-CT120 mL), (b) PCD-CT with 100 mL CM volume (PCD-CT100 mL), and (c) prior energy-integrating detector CT (EID-CT) with 120 mL CM volume. On PCD-CT, virtual monoenergetic image (VMI) reconstructions at 70 keV were applied for both groups as well as additional VMI at 60 keV for PCD‑CT100 mL. Quantitative analyses including signal-to-noise (SNR) and contrast-to-noise ratios (CNR) and qualitative analyses were performed using a mixed linear effects model. RESULTS The final study cohort comprised 49 patients (mean age 67 [31-86] years, 12 female). Comparison to EID-CT was available in 33 patients. In standard 70 keV VMI reconstructions, PCD-CT100 mL was non-inferior to PCD-CT120 mL as well as to EID-CT120 mL for CNR in abdominal organs (all p > 0.050). The mixed linear effects model revealed significant differences between contrast volume groups for both contrast enhancement and image quality ratings. PCD-CT100 mL/70 keV demonstrated the smallest deviation from optimal contrast enhancement (-0.306, p < 0.001). CONCLUSION In portal venous phase thoracoabdominal PCD-CT, a nearly 17% reduction in CM was achievable while maintaining subjective and objective image quality compared to prior higher CM volume PCD-CT scans within the same patients and still surpassing image quality of previous exams on an EID-CT system. KEY POINTS Question How do image quality and iodine attenuation intra-individually compare in portal venous phase photon-counting detector CT (PCD-CT) scans using protocols with different contrast medium volume. Findings PCD-CT scans exhibit superior quantitative and qualitative image quality compared to energy-integrating detector-CT acquisitions and are not negatively affected by contrast volume reductions up to 17%. Clinical relevance This study provides further evidence that PCD-CT enables a considerable reduction in iodine dose for portal venous phase acquisition, benefiting both patients and healthcare system costs.
Collapse
Affiliation(s)
- Daniel Popp
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Faculty of Medicine, University of Augsburg, Augsburg, Germany
- Department of Radiology and Nuclear medicine, Kantonspital St. Gallen, St. Gallen, Switzerland
| | - Martin Siedlecki
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Lena Friedrich
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Mark Haerting
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Christian Scheurig-Muenkler
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Florian Schwarz
- Institute for Radiology, DONAUISAR Hospital Deggendorf-Dingolfing-Landau, Deggendorf, Germany
- Medical Faculty, Ludwig Maximilian University Munich, Munich, Germany
| | - Thomas Kroencke
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Faculty of Medicine, University of Augsburg, Augsburg, Germany.
- Centre for Advanced Analytics and Predictive Sciences (CAAPS), University of Augsburg, Augsburg, Germany.
| | - Stefanie Bette
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Josua A Decker
- Department of Diagnostic and Interventional Radiology, University Hospital Augsburg, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| |
Collapse
|
52
|
Medici B, Caffari E, Maculan Y, Benatti S, Piacentini F, Dominici M, Gelsomino F. Everolimus in the Treatment of Neuroendocrine Tumors: Lights and Shadows. Biomedicines 2025; 13:455. [PMID: 40002868 PMCID: PMC11853220 DOI: 10.3390/biomedicines13020455] [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: 01/22/2025] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Neuroendocrine tumors (NETs) comprise a heterogeneous group of neoplasms that originate from neuroendocrine cells, characterized by their ability to secrete hormones and peptides. Once considered rare, the incidence of NETs has steadily increased due to improved diagnostic modalities. The therapeutic landscape is multifaceted, ranging from surgery for localized disease to pharmacological interventions for advanced cases. However, the absence of robust predictive biomarkers precludes patient stratification and optimization of therapy. Everolimus, an oral mTOR inhibitor, has emerged as a key therapeutic agent due to its dual role in inhibiting cell proliferation and angiogenesis. Nevertheless, challenges such as resistance mechanisms, toxicity and optimal treatment sequencing remain unresolved. This article provides a comprehensive review of the role of everolimus in the management of NETs, focusing in particular on unresolved issues, from the absence of predictive biomarkers to the unavailability of defined guidelines for determining the correct therapeutic sequence.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fabio Gelsomino
- Department of Oncology and Hematology, Division of Oncology, University Hospital of Modena, 41124 Modena, Italy; (B.M.); (E.C.); (Y.M.); (S.B.); (F.P.); (M.D.)
| |
Collapse
|
53
|
Liu R, Ji Z, Wang X, Zhu L, Xin J, Ma L, Zhang J, Ge S, Zhang L, Yang Y, Ning T, Bai M, Duan J, Wang F, Sun Y, Li H, Deng T, Ba Y, Hao J. Regorafenib plus sintilimab as a salvage treatment for microsatellite stable metastatic colorectal cancer: a single-arm, open-label, phase II clinical trial. Nat Commun 2025; 16:1481. [PMID: 39929851 PMCID: PMC11811139 DOI: 10.1038/s41467-025-56748-3] [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: 06/30/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have limited efficacy in microsatellite stable (MSS) metastatic colorectal cancer (mCRC), and combination therapy needs to be further explored. In this single-arm, open-label, phase II trial (NCT04745130), we evaluate the efficacy and safety of the combination therapy of antiangiogenesis (regorafenib) and ICI (sintilimab) in patients with MSS mCRC. The primary endpoint is overall survival (OS). Secondary endpoints include progression free survival (PFS), objective response rate (ORR), disease control rate (DCR), duration of response (DoR) and safety. The median OS and PFS are 14.1 months (95% CI: 10.5-17.7) and 4.1 months (95% CI: 3.4-4.8), respectively. The ORR is 21.4%, DCR is 63.1%, and DoR is 13.0 months (95% CI: 2.5-23.5). Patients with RAS/RAF wild-type exhibit significantly longer median OS (23.3 months, 95% CI: 10.0-36.6) compared to those with mutations (12.1 months, 95% CI: 8.4-15.8). The combination therapy is well tolerated and has limited toxicity. Biomarker analysis, including transcriptome sequencing and multiplex immunohistochemistry staining are performed. The efficacy of this combination treatment is tied to specific gene expressions governing tumor metabolism. Moreover, the effectiveness of immunotherapy depends on the abundance of immune cells, as well as the distance between immune cells and tumor cells.
Collapse
Affiliation(s)
- Rui Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China.
| | - Zhi Ji
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Xia Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Lila Zhu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Jiaqi Xin
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Lijun Ma
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Jiayu Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Shaohua Ge
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Le Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Yuchong Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Tao Ning
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Ming Bai
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Jingjing Duan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Feixue Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Yansha Sun
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Hongli Li
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China.
| | - Ting Deng
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China.
| | - Yi Ba
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China.
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.
| | - Jihui Hao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin, China.
| |
Collapse
|
54
|
Massironi S, Albertelli M, Hasballa I, Paravani P, Ferone D, Faggiano A, Danese S. "Cold" Somatostatin Analogs in Neuroendocrine Neoplasms: Decoding Mechanisms, Overcoming Resistance, and Shaping the Future of Therapy. Cells 2025; 14:245. [PMID: 39996718 PMCID: PMC11854070 DOI: 10.3390/cells14040245] [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: 01/12/2025] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Neuroendocrine neoplasms (NENs) represent a heterogeneous group of tumors that pose significant therapeutic challenges due to their potential for progression, metastasis, and hormonal syndromes. Somatostatin analogs (SSAs) have emerged as a cornerstone in NEN treatment, offering both antisecretory and antiproliferative effects by targeting somatostatin receptors (SSTRs). Despite their proven efficacy, intrinsic and acquired resistance mechanisms, including receptor downregulation, tumor heterogeneity, and microenvironmental influences, limit their long-term effectiveness. Recent advances, including high-dose SSA regimens and novel formulations, have aimed to optimize their therapeutic utility and address these limitations. Body of the review. This review explores the cellular and molecular mechanisms underlying the antitumor effects of SSAs, including receptor-mediated signaling pathways, cell cycle arrest, apoptosis induction, and antiangiogenesis. The role of SSAs in combination therapies with mTOR inhibitors and peptide receptor radionuclide therapy (PRRT) is analyzed, emphasizing their synergistic potential. Key clinical trials, such as RADIANT-2, EVERLAR, and NETTER-1, support the efficacy of these approaches, demonstrating improved outcomes when SSAs are combined with targeted agents or radiolabeled therapies. Emerging strategies include high-dose SSA regimens, particularly in progressive cases with low Ki67 indices. Finally, novel formulations, including oral octreotide, paltusotine, and subcutaneous depot formulations like CAM2029, offer improved pharmacokinetics, bioavailability, and patient adherence. Ongoing clinical trials, including SORENTO, further evaluate their efficacy and safety profiles. CONCLUSIONS This paper provides a comprehensive analysis of the cellular and molecular mechanisms of SSAs. SSAs remain integral to the management of NENs, providing effective tumor stabilization and symptom control. However, resistance mechanisms and tumor heterogeneity necessitate innovative approaches, including high-dose regimens, combination strategies, and next-generation formulations. Future research should focus on refining these strategies to optimize patient outcomes, enhance long-term efficacy, and expand the therapeutic landscape for NENs.
Collapse
Affiliation(s)
- Sara Massironi
- Faculty of Medicine and Surgery, Vita e Salute San Raffaele University, Via Olgettina, 20132 Milan, Italy;
- Gastroenterology Unit, Istituti Ospedalieri Bergamaschi, 24046 Bergamo, Italy
| | - Manuela Albertelli
- Endocrinology, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, 16132 Genova, Italy (I.H.); (D.F.)
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Iderina Hasballa
- Endocrinology, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, 16132 Genova, Italy (I.H.); (D.F.)
| | - Piero Paravani
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sant’Andrea Hospital, Sapienza University, 00189 Rome, Italy; (P.P.); (A.F.)
| | - Diego Ferone
- Endocrinology, Department of Internal Medicine and Medical Specialties (DiMI), University of Genova, 16132 Genova, Italy (I.H.); (D.F.)
| | - Antongiulio Faggiano
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sant’Andrea Hospital, Sapienza University, 00189 Rome, Italy; (P.P.); (A.F.)
| | - Silvio Danese
- Faculty of Medicine and Surgery, Vita e Salute San Raffaele University, Via Olgettina, 20132 Milan, Italy;
- Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy
| |
Collapse
|
55
|
Finn RS, Iyer R, Kalman RS, Parikh ND, Cabrera R, Babajanyan S, Kaseb AO. Tolerability and Effectiveness of Regorafenib Treatment in Patients with Unresectable Hepatocellular Carcinoma: Real-World Data from the United States. J Hepatocell Carcinoma 2025; 12:231-246. [PMID: 39935697 PMCID: PMC11812560 DOI: 10.2147/jhc.s459983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
Introduction While several systemic therapies are available for unresectable hepatocellular carcinoma (uHCC), there is a lack of granular real-world evidence to support the efficacy and safety of these therapies. The REFINE study evaluated safety and effectiveness of regorafenib in a global population under real-world practice conditions. This sub-analysis describes the safety and effectiveness of regorafenib among the United States (US) subset of patients in the REFINE study relative to patients in the non-US subset. Materials and Methods REFINE was an international, prospective, multicenter observational study. Eligible patients were those with uHCC for whom a decision to treat with regorafenib had already been made. The primary study endpoint was the frequency of documented treatment-emergent adverse events (TEAEs). Additional endpoints included overall survival and progression-free survival. Groups were compared descriptively. Results Of 1005 patients, 65 were from the US and 940 were from other countries. 91% of patients in the US subset (n=59) and 92% in the non-US subset (n=862) experienced ≥1 TEAE. Common adverse events (AEs) included gastrointestinal disorders, fatigue, and hand-foot skin reaction. Median overall survival for patients in the US subset was 11.4 months (interquartile range [IQR]: 4.7-25.4) and 13.2 months (IQR: 5.8-26.3) in the non-US subset. Median progression-free survival was 3.4 months (IQR: 2.4-6.1) for patients in the US subset and 3.9 months (IQR: 2.2-8.5) in the non-US subset. Conclusion Regorafenib was associated with similar safety and effectiveness outcomes for patients in the US and non-US subsets of the REFINE study. Differences in the incidence of certain AEs may be due to differences in treatment management between study sites or baseline disease status. These findings are consistent with the phase 3 RESORCE trial and corroborate the safety and effectiveness of regorafenib as a subsequent-line treatment in US patients with uHCC.
Collapse
Affiliation(s)
- Richard S Finn
- Department of Medicine, University of California, Los Angeles, CA, USA
| | - Renuka Iyer
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Richard S Kalman
- Department of Medicine, Einstein Medical Center, Philadelphia, PA, USA
| | - Neehar D Parikh
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, USA
| | - Roniel Cabrera
- Department of Medicine, University of Florida Health, Gainesville, FL, USA
| | | | - Ahmed O Kaseb
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
56
|
Ding Y, Feng Y, Ye Y, Shen J, Guo C, He X, Zhu L, Wang L. High and low dose radiotherapy combined with ICIs for MSS colorectal cancer patients with liver metastases: a phase I study (HaRyPOT). Front Oncol 2025; 15:1503517. [PMID: 39980556 PMCID: PMC11839429 DOI: 10.3389/fonc.2025.1503517] [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: 09/29/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Microsatellite stable (MSS) colorectal cancer with liver metastases (CLRM) responds poorly to immunotherapy, and various approaches to break immune tolerance have been tried. Radiotherapy in combination with immune checkpoint inhibitors is one of promising therapies, and the choice of radiotherapy and immunotherapy modalities is also a hot issue. Methods Here, we report on a Phase I trial treating nine patients with MSS CLRM using a combination of high and low dose radiotherapy and immune checkpoint inhibitors (ICIs). Results The primary endpoint of the trial was the safety and tolerability of this combination treatment modality. Secondary endpoints included the objective response rate (ORR), progression-free survival (PFS) and overall survival (OS). The study results showed that at three dose levels-single doses of 6Gy (n=3), 8Gy (n=3), and 10Gy (n=3)-the most common treatment-related adverse events (TRAEs) were nausea, vomiting, fatigue, and abnormal liver function. At the first condition assessment, four patients were observed to have stable disease (SD) and one patient achieved partial response (PR). In exploratory endpoint analyses, tissue biopsies and paired hematologic samples from patients showed M2 macrophage reduction. Plasma cytokines IL-10, IL-17, and INF-α increased after treatment with both drugs. Discussion In summary, this is the first clinical trial demonstrating the safety and immunogenic activity of combined high and low dose radiotherapy with ICIs in MSS colorectal cancer liver metastases (CRLMs). The combination therapy stimulated the immune response and altered the tumour microenvironment, warranting further exploration in the future.
Collapse
Affiliation(s)
- Yuxuan Ding
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yong Feng
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yangfan Ye
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiayi Shen
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Chang Guo
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xia He
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Liangjun Zhu
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Lijun Wang
- Department of Radiotherapy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| |
Collapse
|
57
|
Liu C, Chen J, Liu Y. Rechallenge therapy versus tyrosine kinase inhibitor (TKI) for advanced metastatic colorectal cancer: a retrospective study. Sci Rep 2025; 15:4237. [PMID: 39905022 PMCID: PMC11794864 DOI: 10.1038/s41598-025-86575-x] [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: 08/14/2024] [Accepted: 01/13/2025] [Indexed: 02/06/2025] Open
Abstract
Currently, the standard third-line treatment options for advanced metastatic colorectal cancer (mCRC) include regorafenib, fruquintinib, and trifluridine/tipiracil (TAS-102), but these drugs have limited clinical efficacy. This study aims to evaluate the efficacy and survival outcomes of tyrosine kinase inhibitor (TKI) versus chemotherapy rechallenge in third-line treatment for advanced mCRC patients. From January 2019 to December 2022, 107 patients met the inclusion criteria. After 1:1 matching, there were 34 patients in both the TKI and rechallenge groups. Cox regression analysis was performed to identify independent predictive factors. The result indicated that Eastern Cooperative Oncology Group (ECOG) score of 0-1 (hazard ratio [HR] = 0.03, 95% confidence interval [CI]: 0.01-0.17; P < 0.001) and chemotherapy rechallenge (HR = 0.43, 95% CI: 0.23-0.79; P = 0.007) were independent protective factors for overall survival (OS). Patient-Generated Subjective Global Assessment (PG-SGA) score of 0-3 (HR = 0.43, 95% CI: 0.23-0.80; P = 0.008), progression-free survival (PFS) of frontline treatment (HR = 0.66, 95% CI: 0.43-1.00, P = 0.047) and chemotherapy rechallenge (HR = 0.42, 95% CI: 0.24-0.73; P = 0.002) were independent protective factors for PFS. Kaplan-Meier survival analysis showed that the median OS in the rechallenge group was 13.2 months (95% CI: 9.6-19.3), compared with 7.7 months (95% CI: 6.5-14.5) in the TKI group, with a statistically significant difference (HR = 0.47, 95% CI: 0.26-0.86; P = 0.013). The median PFS for the rechallenge group was 5.0 months (95% CI: 4.3-6.7), compared with 3.4 months (95% CI: 2.7-4.8) in the TKI group, with a statistically significant difference too (HR = 0.54, 95% CI: 0.32-0.91; P = 0.019). Furthermore, subgroup analysis showed that in Group A, the rechallenge group also had superior median OS and PFS compared with the TKI group. In conclusion, while TKIs are the current standard for third-line treatment in mCRC patients, chemotherapy rechallenge is a more effective option for patients who have achieved disease control in the first two lines of treatment.
Collapse
Affiliation(s)
- Chenming Liu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jintao Chen
- Fujian Changle District Hospital, Fuzhou, Fujian, China
| | - Yuxing Liu
- Department of General Surgery, Affiliated Taikang Xianlin Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China.
| |
Collapse
|
58
|
Jain A, Stebbing J. The Relationship Between Response Rate and Survival Benefits in Randomized Immunotherapy Studies. Cancers (Basel) 2025; 17:495. [PMID: 39941863 PMCID: PMC11815975 DOI: 10.3390/cancers17030495] [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: 12/25/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Understanding the relationship between the Objective Response Rate (ORR) and survival outcomes, notably Progression-Free Survival (PFS) and Overall Survival (OS), is relevant for assessing the efficacy of regimens in oncology. We evaluate the relationship between ORR, PFS and OS in immuno-oncology (IO) trials. Data from 68 clinical trials submitted to the FDA were evaluated, examining immunotherapy regimens, notably immune checkpoint inhibitors such as anti-programmed death (ligand)-1 [anti-PD-(L)1], cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) inhibitors and combination therapies [e.g., IO + IO, anti-PD-L1 + chemotherapy, anti-PD-L1 + CTLA-4, anti-PD-L1 + TKI (tyrosine kinase inhibitors)]. Studies were included based on their reporting of ORR, PFS, and OS. Of the 68 clinical trials reviewed, 55 were included in the analysis. The correlation between ORR and PFS was moderate across most immunotherapy regimens, indicating that ORR can serve as a useful predictor of short-term disease control. However, the correlation between ORR and OS was weaker, especially in trials including combination therapies, indicating that ORR alone may not reliably predict long-term survival outcomes. ORR predicts PFS better in first-line treatment but declines in later lines and remains a weak OS predictor overall. Differing degrees of correlation between ORR and survival metrics, particularly across treatment lines and combinations, are observed. While ORR can serve as a surrogate marker for PFS in IO trials, its utility in predicting OS is restricted and the interpretation of the relationship between ORR and PFS or OS is a key limitation. Rather, a decline in PFS with increasing ORR may reflect trial differences rather than a direct relationship. Future analyses should adopt better methodologies to capture these dynamics and focus on improving surrogate endpoints for immunotherapy to improve clinical trial design and patient outcomes.
Collapse
Affiliation(s)
- Aditi Jain
- Edinburgh Medical School, Biomedical Sciences, The University of Edinburgh, Edinburgh EH8 9YL, UK
| | - Justin Stebbing
- School of Life Sciences, Anglia Ruskin University, Cambridge CB1 1PT, UK
| |
Collapse
|
59
|
Piercey O, Chantrill L, Hsu H, Ma B, Price T, Tan IB, Teng H, Tie J, Desai J. Expert consensus on the optimal management of BRAF V600E-mutant metastatic colorectal cancer in the Asia-Pacific region. Asia Pac J Clin Oncol 2025; 21:31-45. [PMID: 39456063 PMCID: PMC11733838 DOI: 10.1111/ajco.14132] [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: 05/15/2024] [Revised: 09/14/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024]
Abstract
The burden of colorectal cancer (CRC) is high in the Asia-Pacific region, and several countries in this region have among the highest and/or fastest growing rates of CRC in the world. A significant proportion of patients will present with or develop metastatic CRC (mCRC), and BRAFV600E-mutant mCRC represents a particularly aggressive phenotype that is less responsive to standard chemotherapies. In light of recent therapeutic advances, an Asia-Pacific expert consensus panel was convened to develop evidence-based recommendations for the diagnosis, treatment, and management of patients with BRAFV600E-mutant mCRC. The expert panel comprised nine medical oncologists from Australia, Hong Kong, Singapore, and Taiwan (the authors), who met to review current literature and develop eight consensus statements that describe the optimal management of BRAFV600E-mutant mCRC in the Asia-Pacific region. As agreed by the expert panel, the consensus statements recommend molecular testing at diagnosis to guide individualized treatment decisions, propose optimal treatment pathways according to microsatellite stability status, advocate for more frequent monitoring of BRAFV600E-mutant mCRC, and discuss local treatment strategies for oligometastatic disease. Together, these expert consensus statements are intended to optimize treatment and improve outcomes for patients with BRAFV600E-mutant mCRC in the Asia-Pacific region.
Collapse
Affiliation(s)
| | - Lorraine Chantrill
- Illawarra Shoalhaven Local Health DistrictIllawarraNew South WalesAustralia
- Faculty of Science, Medicine and HealthUniversity of WollongongWollongongNew South WalesAustralia
| | - Hung‐Chih Hsu
- Division of Hematology OncologyChang Gung Memorial HospitalNew TaipeiTaiwan
- College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Brigette Ma
- State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer, Department of Clinical Oncology, Hong Kong Cancer InstituteThe Chinese University of Hong KongHong Kong SARChina
| | - Timothy Price
- The Queen Elizabeth HospitalAdelaideSouth AustraliaAustralia
| | - Iain Beehuat Tan
- Division of Medical OncologyNational Cancer Centre SingaporeSingaporeSingapore
| | - Hao‐Wei Teng
- Department of OncologyTaipei Veterans General HospitalTaipeiTaiwan
| | - Jeanne Tie
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Jayesh Desai
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
60
|
Vienot A, Vernerey D, Bouard A, Klajer E, Kim S, Tournigand C, Louvet C, André T, Rousseau B, Wespiser M, Spehner L, Wang YA, Weispfenning A, Dochy E, Borg C. Stanniocalcin 1 in Patients with Refractory Colorectal Cancer Treated with Regorafenib: A Post Hoc Biomarker Analysis of the TEXCAN and CORRECT Trials. CANCER RESEARCH COMMUNICATIONS 2025; 5:287-294. [PMID: 39807836 PMCID: PMC11811826 DOI: 10.1158/2767-9764.crc-24-0246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 09/27/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
SIGNIFICANCE STC1 is a protein secreted by intratumor endothelial cells in which plasma concentrations increase in patients with chemorefractory mCRC. Based on analyses of patients with refractory mCRC in the TEXCAN and CORRECT trials, we found that STC1 plasma levels had a prognostic role for OS, with high levels associated with poor outcome. A predictive role for baseline STC1 levels was pointed out for regorafenib efficacy.
Collapse
Affiliation(s)
- Angélique Vienot
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- University of Franche Comté, EFS, INSERM, UMR RIGHT, Besançon, France
| | - Dewi Vernerey
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- University of Franche Comté, EFS, INSERM, UMR RIGHT, Besançon, France
| | - Adeline Bouard
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- University of Franche Comté, EFS, INSERM, UMR RIGHT, Besançon, France
| | - Elodie Klajer
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Stefano Kim
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | | | | | - Thierry André
- Sorbonne Université and Hôpital Saint-Antoine, Paris, France
| | | | - Mylène Wespiser
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Laurie Spehner
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- University of Franche Comté, EFS, INSERM, UMR RIGHT, Besançon, France
| | - Ying A. Wang
- Bayer HealthCare Pharmaceuticals, Cambridge, Massachusetts
| | | | | | - Christophe Borg
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- University of Franche Comté, EFS, INSERM, UMR RIGHT, Besançon, France
| |
Collapse
|
61
|
Boland PM, Lenz HJ, Ciombor KK, Florou V, Pishvaian MJ, Cusnir M, Cohen D, Guo JY, Tang M, Rajagopalan P, Wiley SE, Ghalie RG, Hochster HS. A Phase 1b study of the OxPhos inhibitor ME-344 with bevacizumab in refractory metastatic colorectal cancer. Invest New Drugs 2025; 43:60-68. [PMID: 39725778 PMCID: PMC11868331 DOI: 10.1007/s10637-024-01489-1] [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: 11/06/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
Antiangiogenic drugs may cause vascular normalization and correct hypoxia in tumors, shifting cells to mitochondrial respiration as the primary source of energy. In turn, the addition of an inhibitor of mitochondrial respiration to antiangiogenic therapy holds potential to induce synthetic lethality. This study evaluated the mitochondrial inhibitor ME-344 in combination with bevacizumab in patients with refractory metastatic colorectal cancer (mCRC). Patients were eligible if they had disease progression after standard therapies, adequate hematologic, hepatic and renal function, and no contraindications to bevacizumab. ME-344 was administered intravenously on days 1, 8 and 15 and bevacizumab on days 1 and 15 of 28-day cycles until disease progression or intolerance. The primary efficacy endpoint was progression-free survival (PFS) at week 16. In the 23 patients enrolled, the median age was 58 years, median number of prior lines of therapy was 4, and median interval from last therapy was 3 months. The most common adverse events (all grades/grade ≥ 3) were fatigue (48%/13%), abdominal pain (35%/4%), diarrhea (30%/4%) and constipation (30%/0%). No patient had an objective response; 9 patients (39%) achieved stable disease. The 16-week PFS was 30.6% (95% confidence interval [CI]: 12.2-51.3), the median PFS was 1.9 months (95% CI: 1.6-4.7), and the median overall survival was 6.7 months (95% CI: 3.4-not reached). ME-344 plus bevacizumab was well tolerated. Disease control was limited in this heavily pretreated patient population. Additional investigations in earlier lines are indicated, and extended-release ME-344 formulations may provide longer drug exposure to maximize benefit. (Trial registration number ClinicalTrials.gov NCT05824559. Registration date 22 March 2022).
Collapse
Affiliation(s)
- Patrick M Boland
- Division of Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.
- Rutgers Cancer Institute of New Jersey, 195 Little Albany St, New Brunswick, NJ, 08901, USA.
| | - Heinz-Josef Lenz
- Division of Medical Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Kristen K Ciombor
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Vaia Florou
- Division of Oncology, Huntsman Cancer Institute of the University of Utah, Salt Lake City, UT, USA
| | - Michael J Pishvaian
- Department of Medicine, Johns Hopkins Kimmel Cancer Center, Washington, DC, USA
| | - Michael Cusnir
- Division of Hematology & Oncology, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Deirdre Cohen
- Department of Hematology Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessie Y Guo
- Rutgers Cancer Institute of New Jersey, 195 Little Albany St, New Brunswick, NJ, 08901, USA
| | - Min Tang
- Statbeyond Consulting, Irvine, CA, USA
| | | | | | | | - Howard S Hochster
- Division of Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
62
|
Gehrels AM, Vissers PA, Sprangers MA, Fijnheer NM, Pijnappel EN, van der Geest LG, Cirkel GA, de Vos-Geelen J, Homs MY, Creemers GJ, Stommel MW, Daamen LA, Besselink MG, Wilmink JW, van Laarhoven HW. Longitudinal health-related quality of life in patients with pancreatic cancer stratified by treatment: a nationwide cohort study. EClinicalMedicine 2025; 80:103068. [PMID: 39902316 PMCID: PMC11788757 DOI: 10.1016/j.eclinm.2024.103068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 02/05/2025] Open
Abstract
Background Pancreatic adenocarcinoma (PAC) has a poor prognosis and substantially impairs health-related quality of life (HRQoL). Large studies on longitudinal HRQoL in patients with PAC, taking patient treatment into account, are lacking. This study aimed to investigate HRQoL over time in patients with PAC undergoing various treatments. Methods This nationwide cohort study included patients diagnosed with PAC between 2015 and 2020. Data were collected from the Dutch Pancreatic Cancer Project (PACAP) and the Netherlands Cancer Registry. Patients were categorized into four groups based on treatment modality: resection (R-PAC), chemotherapy for localized disease (C-PAC), chemotherapy for metastatic disease (M1-C-PAC), and best supportive care (BSC). HRQoL was assessed using the EORTC QLQ-C30 and -PAN26 questionnaires at baseline, during treatment, and at 0-3 months and 3-6 months after treatment. Linear mixed models were used to analyze changes in HRQoL over time, with clinically relevant changes defined as a minimal mean difference of 10 points in absolute scores and reported with 95% confidence intervals. Findings Overall, 1496 patients were included (673 [45.0%] female), of whom 675 (45.1%) in R-PAC, 319 (21.3%) in C-PAC, 340 patients (22.7%) in M1-C-PAC, and 162 (10.8%) in BSC group. In R-PAC, hepatic symptoms and health care satisfaction improved while role and social functioning deteriorated and eating related problems, side effects and fear of future health increased during treatment. In C-PAC, insomnia, pancreatic pain, hepatic symptoms decreased while diarrhea, side effects and fear of future health increased. In M1-C-PAC, pain, insomnia, pancreatic pain, hepatic symptoms, ascites and constipation decreased, sexuality improved while fear of future health and side effects increased. In BSC, hepatic symptoms decreased and flatulence increased. Interpretation This nationwide study identified specific improvements and deteriorations in various HRQoL domains during 6 months follow-up. This information may be valuable in the clinical setting to inform patients on potential outcomes of the course of HRQoL during various treatment strategies. Funding None.
Collapse
Affiliation(s)
- Anne M. Gehrels
- Department of Medical Oncology, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - Pauline A.J. Vissers
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
- Department of Surgery, RadboudUMC, Nijmegen, the Netherlands
| | - Mirjam A.G. Sprangers
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
- Department of Medical Psychology, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
| | - Nienke M. Fijnheer
- Department of Medical Oncology, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - Esther N. Pijnappel
- Department of Medical Oncology, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - Lydia G. van der Geest
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, the Netherlands
| | - Geert A. Cirkel
- Department of Medical Oncology, Regional Academic Cancer Center Utrecht, Meander Medical Center Amersfoort, University Medical Center, Utrecht, the Netherlands
| | - Judith de Vos-Geelen
- Division of Medical Oncology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Marjolein Y.V. Homs
- Department of Medical Oncology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Geert- Jan Creemers
- Department of Medical Oncology, Catharina Hospital, Eindhoven, the Netherlands
| | | | - Lois A. Daamen
- Department of Surgery, Regional Academic Cancer Center Utrecht, University Medical Center Utrecht and St. Antonius Hospital Nieuwegein, the Netherlands
- Division of Imaging and Oncology, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Marc G. Besselink
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
- Amsterdam UMC Location University of Amsterdam, Department of Surgery, Amsterdam, the Netherlands
| | - Johanna W. Wilmink
- Department of Medical Oncology, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| | - Hanneke W.M. van Laarhoven
- Department of Medical Oncology, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, the Netherlands
| |
Collapse
|
63
|
Yonatan ER, Ruby R, Prasetya A, Arifin ES. Evaluation of fruquintinib's efficacy and safety in refractory metastatic colorectal cancer: a systematic review and meta-analysis of phase II and III randomized controlled trials. Clin J Gastroenterol 2025; 18:11-22. [PMID: 39704756 DOI: 10.1007/s12328-024-02087-7] [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: 10/20/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Metastatic colorectal cancer (mCRC) remains a significant cause of mortality despite advancements in treatments. Fruquintinib, a potent VEGFR inhibitor, has shown promise as an advanced therapy for mCRC. This review evaluates the efficacy and safety of fruquintinib compared to placebo in patients with refractory mCRC, focusing on Phase II and III trials. METHOD This study was conducted using the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. A literature search was performed using PubMed, EBSCOHost, ProQuest, and Google Scholar. The analysis of overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), and safety was pooled using hazard ratios (HR) and risk ratios (RR) in RevMan 5.4 software. RESULTS Three RCTs comprising 1,178 patients were included. Fruquintinib significantly improved OS (HR: 0.66; 95% CI 0.57-0.76) and PFS (HR: 0.30; 95% CI 0.26-0.35) compared to placebo. ORR (RR: 5.91; 95% CI 1.09-32.16) and DCR (RR: 3.83; 95% CI 3.00-4.90) were also higher with fruquintinib. Grade 3 or higher adverse events were more frequent with fruquintinib (RR: 1.31; 95% CI 1.02-1.70). No significant difference was observed in serious adverse events or treatment-related deaths. CONCLUSION Fruquintinib significantly improves survival and tumor response in patients with refractory mCRC. While associated with an increased risk of high-grade adverse events, fruquintinib remains a viable and relatively safe treatment option for mCRC patients. Further studies are needed to confirm its efficacy and safety across diverse populations.
Collapse
Affiliation(s)
- Eric Ricardo Yonatan
- Faculty of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Special Capital Region of Jakarta, Pluit Raya Street No. 2, North Jakarta, 14440, Indonesia.
| | - Rivaldi Ruby
- Faculty of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Special Capital Region of Jakarta, Pluit Raya Street No. 2, North Jakarta, 14440, Indonesia
| | - Alver Prasetya
- Faculty of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Special Capital Region of Jakarta, Pluit Raya Street No. 2, North Jakarta, 14440, Indonesia
| | - Erlangga Saputra Arifin
- Faculty of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Special Capital Region of Jakarta, Pluit Raya Street No. 2, North Jakarta, 14440, Indonesia
| |
Collapse
|
64
|
Melhorn P, Raderer M, Kiesewetter B. Selecting systemic treatment for metastatic neuroendocrine tumors of the lung-current evidence and clinical implications. Cancer Treat Rev 2025; 133:102878. [PMID: 39787793 DOI: 10.1016/j.ctrv.2024.102878] [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: 09/20/2024] [Revised: 12/15/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
Neuroendocrine tumors (NET) of the lung are a slowly growing subtype of lung cancer that has a different treatment paradigm than aggressive and more common forms of lung neuroendocrine neoplasms (NEN) like small cell lung cancer (SCLC). Current guidelines for metastatic lung NET advocate a handful of treatment options, including somatostatin analogs (SSA), everolimus, temozolomide- or platin-based chemotherapy, and peptide receptor radionuclide therapy (PRRT). However, there is no clear treatment sequence, and the therapy of choice may depend on several factors such as tumor grade / growth rate, tumor burden / symptoms, disease progression status, and somatostatin receptor (SSTR) expression. In order to tailor treatment to each individual patient, the latest scientific findings and patient-specific clinical features must be considered together. This review critically evaluates the available evidence with regards to relevant patient characteristics, inclusion and exclusion criteria, and outcome metrics of clinical trials given the presumed natural disease course. Specific patient subgroups with an unmet therapeutic need are identified and discussed in the context of ongoing clinical trials.
Collapse
Affiliation(s)
- Philipp Melhorn
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Austria
| | - Markus Raderer
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Austria
| | - Barbara Kiesewetter
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Austria.
| |
Collapse
|
65
|
He L, Cheng X, Gu Y, Zhou C, Li Q, Zhang B, Cheng X, Tu S. Fruquintinib Combined With PD-1 Inhibitors for the Treatment of the Patients With Microsatellite Stability Metastatic Colorectal Cancer: Real-World Data. Clin Oncol (R Coll Radiol) 2025; 38:103700. [PMID: 39700765 DOI: 10.1016/j.clon.2024.103700] [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: 11/10/2023] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024]
Abstract
AIMS Programmed death-1 (PD-1) or programmed death-ligand 1 (PD-L1) inhibitors have shown limited effectiveness in patients with microsatellite stable (MSS) metastatic colorectal cancer (mCRC). Combining anti-angiogenesis inhibitors with PD-1 inhibitors has the potential to reverse the immunosuppressive tumour microenvironment, synergistically enhancing the anti-tumour immune response in MSS mCRC. The goal is to present real-world data that prove the clinical efficacy and safety of fruquintinib combined with PD-1 inhibitors in MSS mCRC. MATERIALS AND METHODS We conducted a real-world retrospective study in patients with MSS mCRC who received treatment with fruquintinib combined with PD-1 inhibitors between May 2019 and March 2023 in our centre. RESULTS Seventy seven patients with MSS mCRC received fruquintinib combined with PD-1 inhibitors. In total, 5.2% of patients (4/77) achieved a partial response (PR), while 50.6% (39/77) had a stable disease (SD). Notably, three lesions achieving PR were all lung metastases and the overall disease control rate (DCR) reached 55.8% (43/77). Median progression-free survival (PFS) and overall survival (OS) reached 5.1 months (95% CI: 3.6-6.7) and 14.6 months (95% CI: 9.6-15.6), respectively. Multivariate Cox analysis showed that prior treatment without vascular endothelial growth factor (VEGF) inhibitors was significantly associated with PFS and OS (p < 0.05). Further analysis indicated that total- or polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) significantly decreased after treatment (P = 0.039), especially in the PR/SD group (P = 0.003). Most adverse events included abdominal pain, rash, oedema, diarrhoea, and immunotherapy-associated hypothyroidism, yet symptoms were controllable. CONCLUSION Our results provided additional evidence that patients with MSS mCRC could benefit from the combination of fruquintinib and PD-1 inhibitors, especially those with lung metastases or without prior treatment with VEGF inhibitors. The detection of MDSCs may be an immune indicator for predicting of the combined therapy.
Collapse
Affiliation(s)
- L He
- State Key Laboratory of Systems Medicine for Cancer, Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - X Cheng
- State Key Laboratory of Systems Medicine for Cancer, Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Y Gu
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - C Zhou
- State Key Laboratory of Systems Medicine for Cancer, Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Q Li
- State Key Laboratory of Systems Medicine for Cancer, Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - B Zhang
- State Key Laboratory of Systems Medicine for Cancer, Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - X Cheng
- State Key Laboratory of Systems Medicine for Cancer, Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - S Tu
- State Key Laboratory of Systems Medicine for Cancer, Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
66
|
Connell LC, Kemeny NE. Intraarterial Chemotherapy for Liver Metastases. Hematol Oncol Clin North Am 2025; 39:143-159. [PMID: 39510670 DOI: 10.1016/j.hoc.2024.08.005] [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: 11/15/2024]
Abstract
Colorectal cancer (CRC) is one of the leading cancers globally in terms of both incidence and cancer-related mortality. Liver metastatic disease is the main prognostic driver for patients with CRC. The management options for liver metastatic CRC continue to evolve, particularly with the incorporation of locoregional therapies into the treatment paradigm. Hepatic arterial infusion (HAI) chemotherapy is one such liver directed approach used with the goal of converting patients to liver resection, reducing the risk of recurrence, treating recurrent disease, and most importantly improving overall survival. This article summarizes the role of HAI chemotherapy in the treatment of liver metastatic CRC.
Collapse
Affiliation(s)
- Louise C Connell
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, 10th floor, New York, NY 10065, USA
| | - Nancy E Kemeny
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, 10th floor, New York, NY 10065, USA.
| |
Collapse
|
67
|
Ji P, Chen T, Li C, Zhang J, Li X, Zhu H. Comprehensive review of signaling pathways and therapeutic targets in gastrointestinal cancers. Crit Rev Oncol Hematol 2025; 206:104586. [PMID: 39653094 DOI: 10.1016/j.critrevonc.2024.104586] [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: 05/31/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024] Open
Abstract
Targeted therapy, the milestone in the development of human medicine, originated in 2004 when the FDA approved the first targeted agent bevacizumab for colorectal cancer treatment. This new development has resulted from drug developers moving beyond traditional chemotherapy, and several trials have popped up in the last two decades with an unprecedented speed. Specifically, EGF/EGFR, VEGF/VEGFR, HGF/c-MET, and Claudin 18.2 therapeutic targets have been developed in recent years. Some targets previously thought to be undruggable are now being newly explored, such as the RAS site. However, the efficacy of targeted therapy is extremely variable, especially with the emergence of new drugs and the innovative use of traditional targets for other tumors in recent years. Accordingly, this review provides an overview of the major signaling pathway mechanisms and recent advances in targeted therapy for gastrointestinal cancers, as well as future perspectives.
Collapse
Affiliation(s)
- Pengfei Ji
- Department of Thoracic Surgery, West China Hospital, Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China
| | - Tingting Chen
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Chao Li
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Jinyuan Zhang
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Xiao Li
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| |
Collapse
|
68
|
Guo NT, Ni J, Wei GL, Huo JG. Progress in research of hand-foot skin reactions related to antineoplastic drugs. Shijie Huaren Xiaohua Zazhi 2025; 33:27-33. [DOI: 10.11569/wcjd.v33.i1.27] [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: 10/21/2024] [Revised: 11/25/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
Hand-foot syndrome (HFS)/hand-foot skin reactions (HFSR) is the most common toxic skin reaction in tumor treatment, especially in anti-cancer treatment of malignant digestive system tumors, having a great impact on patients' anti-tumor therapy, quality of life, and physical and mental health. It has been found that patients with HFS/HFSR are often the dominant population who obtain survival benefit from anti-tumor treatment. At present, the pathogenesis of HFSR is not clear and there is no effective intervention measures available in the clinic. In this paper, we review the clinical characteristics, incidence, pathogenesis, risk factors, and intervention measures for HFSR, as well as its relationship with anti-tumor efficacy, in order to provide reference for further research.
Collapse
Affiliation(s)
- Nai-Ting Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
- Guang'anmen Hospital Jinan Branch of China Academy of Chinese Medical Sciences (Jinan Municipal Hospital of Traditional Chinese Medicine), Jinan 250012, Shandong Province, China
| | - Jing Ni
- Lishui District Hospital of Traditional Chinese Medicine, Nanjing 211200, Jiangsu Province, China
| | - Guo-Li Wei
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
| | - Jie-Ge Huo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
| |
Collapse
|
69
|
Guo NT, Ni J, Wei GL, Huo JG. Progress in research of hand-foot skin reactions related to anti-digestive system tumor drug treatment. Shijie Huaren Xiaohua Zazhi 2025; 33:21-27. [DOI: 10.11569/wcjd.v33.i1.21] [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: 10/21/2024] [Revised: 11/25/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
Hand-foot syndrome (HFS)/hand-foot skin reactions (HFSR) is the most common toxic skin reaction in tumor treatment, especially in anti-cancer treatment of malignant digestive system tumors, having a great impact on patients' anti-tumor therapy, quality of life, and physical and mental health. It has been found that patients with HFS/HFSR are often the dominant population who obtain survival benefit from anti-tumor treatment. At present, the pathogenesis of HFSR is not clear and there is no effective intervention measures available in the clinic. In this paper, we review the clinical characteristics, incidence, pathogenesis, risk factors, and intervention measures for HFSR, as well as its relationship with anti-tumor efficacy, in order to provide reference for further research.
Collapse
Affiliation(s)
- Nai-Ting Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
- Guang'anmen Hospital Jinan Branch of China Academy of Chinese Medical Sciences (Jinan Municipal Hospital of Traditional Chinese Medicine), Jinan 250012, Shandong Province, China
| | - Jing Ni
- Lishui District Hospital of Traditional Chinese Medicine, Nanjing 211200, Jiangsu Province, China
| | - Guo-Li Wei
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
| | - Jie-Ge Huo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, Jiangsu Province, China
| |
Collapse
|
70
|
Muñoz-Mármol AM, Meléndez B, Hernandez A, Sanz C, Domenech M, Arpí-Llucia O, Gut M, Esteve A, Esteve-Codina A, Parra G, Carrato C, Aldecoa I, Mallo M, Pineda E, Alameda F, de la Iglesia N, Martinez-Balibrea E, Martinez-Cardús A, Estival-Gonzalez A, Balana C. Multikinase Treatment of Glioblastoma: Evaluating the Rationale for Regorafenib. Cancers (Basel) 2025; 17:375. [PMID: 39941744 PMCID: PMC11816343 DOI: 10.3390/cancers17030375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/07/2025] [Accepted: 01/10/2025] [Indexed: 02/16/2025] Open
Abstract
We explored the rationale for treating glioblastoma (GBM) with regorafenib. In 103 newly diagnosed GBM patients, we assessed mutations, copy number variants (CNVs), fusions, and overexpression in 46 genes encoding protein kinases (PKs) potentially targeted by regorafenib or its metabolites and performed a functional enrichment analysis to assess their implications in angiogenesis. We analyzed regorafenib's binding inhibitory activity and target affinity for these 46 PKs and focused on a subset of 18 genes inhibited by regorafenib at clinically achievable concentrations and on 19 genes involved in angiogenesis. Putative oncogenic alterations were defined as oncogenic/likely oncogenic mutations, oncogenic fusions, CNVs > 5, and/or gene overexpression. Regorafenib did not target all 46 PKs. For the 46-gene set, 40 genes (86.9%) and 73 patients (70.8%) harbored at least one alteration in genes encoding targetable PKs, but putative oncogenic alterations were present in only 34 patients (33%). In the 18-gene set, 18 genes (100%) and 48 patients (46.6%) harbored alterations, but putative oncogenic alterations were detected in only 26 patients (25.2%). Thirty patients (29.1%) had oncogenic alterations in the 18-gene set and/or in angiogenesis-related genes. Around 33% of patients had oncogenic alterations in any of the 46 potential targets. Additionally, the suboptimal dosing of regorafenib, due to its poor penetration of the blood-brain barrier, may reduce the likelihood of effectively targeting certain PKs. Future use of multi-target drugs must be guided by a thorough understanding of target presence, effective inhibition, and the drug's ability to reach brain tumors at adequate concentrations.
Collapse
Affiliation(s)
- Ana Maria Muñoz-Mármol
- Pathology Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (A.M.M.-M.); (C.S.); (C.C.)
| | - Bárbara Meléndez
- Molecular Pathology Research Unit, Hospital Universitario de Toledo, 45005 Toledo, Spain;
| | - Ainhoa Hernandez
- Medical Oncology, Institut Catala d’Oncologia (ICO), 08916 Badalona, Spain; (A.H.); (M.D.); (A.E.)
- Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), 08916 Badalona, Spain;
- CARE Program, Germans Trias i Pujol Research Institute (IGTP), Ctra de Can Ruti, Cami de les Escoles s/n, 08916 Badalona, Spain;
| | - Carolina Sanz
- Pathology Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (A.M.M.-M.); (C.S.); (C.C.)
| | - Marta Domenech
- Medical Oncology, Institut Catala d’Oncologia (ICO), 08916 Badalona, Spain; (A.H.); (M.D.); (A.E.)
- Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), 08916 Badalona, Spain;
- CARE Program, Germans Trias i Pujol Research Institute (IGTP), Ctra de Can Ruti, Cami de les Escoles s/n, 08916 Badalona, Spain;
| | - Oriol Arpí-Llucia
- Cancer Research Program, Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain;
| | - Marta Gut
- Centro Nacional de Análisis Genómico, Universitat de Barcelona (UB), C/Baldiri Reixac 4, 08028 Barcelona, Spain; (M.G.); (A.E.-C.); (G.P.)
| | - Anna Esteve
- Medical Oncology, Institut Catala d’Oncologia (ICO), 08916 Badalona, Spain; (A.H.); (M.D.); (A.E.)
- Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), 08916 Badalona, Spain;
- CARE Program, Germans Trias i Pujol Research Institute (IGTP), Ctra de Can Ruti, Cami de les Escoles s/n, 08916 Badalona, Spain;
| | - Anna Esteve-Codina
- Centro Nacional de Análisis Genómico, Universitat de Barcelona (UB), C/Baldiri Reixac 4, 08028 Barcelona, Spain; (M.G.); (A.E.-C.); (G.P.)
| | - Genis Parra
- Centro Nacional de Análisis Genómico, Universitat de Barcelona (UB), C/Baldiri Reixac 4, 08028 Barcelona, Spain; (M.G.); (A.E.-C.); (G.P.)
| | - Cristina Carrato
- Pathology Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (A.M.M.-M.); (C.S.); (C.C.)
| | - Iban Aldecoa
- Department of Pathology, Biomedical Diagnostic Centre (CDB) and Neurological Tissue Bank of the Biobank-IDIBAPS, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain;
| | - Mar Mallo
- Unidad de Microarrays, Institut de Recerca Contra la Leucèmia Josep Carreras (IJC), ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Spain;
| | - Estela Pineda
- Medical Oncology, Hospital Clínic, Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain;
| | - Francesc Alameda
- Pathology Department, Neuropathology Unit, Hospital del Mar, Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain;
| | - Nuria de la Iglesia
- IrsiCaixa AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain;
| | - Eva Martinez-Balibrea
- CARE Program, Germans Trias i Pujol Research Institute (IGTP), Ctra de Can Ruti, Cami de les Escoles s/n, 08916 Badalona, Spain;
- ProCURE Program, Catalan Institute of Oncology, Ctra. de Can Ruti, Camí de les Escoles s/n, 08916 Badalona, Spain
| | - Anna Martinez-Cardús
- Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), 08916 Badalona, Spain;
- CARE Program, Germans Trias i Pujol Research Institute (IGTP), Ctra de Can Ruti, Cami de les Escoles s/n, 08916 Badalona, Spain;
| | - Anna Estival-Gonzalez
- Medical Oncology, Hospital Universitario Insular de Gran Canaria, 35016 Las Palmas de Gran Canaria, Spain;
| | - Carmen Balana
- Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), 08916 Badalona, Spain;
| |
Collapse
|
71
|
Unseld M, Kühberger S, Graf R, Beichler C, Braun M, Dandachi N, Heitzer E, Prager GW. Circulating tumor DNA (ctDNA) trajectories predict survival in trifluridine/tipiracil-treated metastatic colorectal cancer patients. Mol Oncol 2025. [PMID: 39840713 DOI: 10.1002/1878-0261.13755] [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: 12/15/2023] [Revised: 06/18/2024] [Accepted: 10/10/2024] [Indexed: 01/23/2025] Open
Abstract
Late-line treatment in metastatic colorectal cancer (mCRC) can improve prognosis. However, not every patient has a benefit and may experience severe side effects. Thus, predictive/prognostic biomarkers are urgently needed. We investigated the prognostic role of circulating tumor DNA (ctDNA) in mCRC patients before and during treatment with trifluridine/tipiracil (FTD/TPI). This noninterventional translational biomarker phase II study enrolled 30 mCRC patients (60% male, 40% female). Using a 77-gene panel, ctDNA was detectable in 90% of patients. Tumor levels were assessed based on aneuploidy (ichorCNA) as well as the highest variant allele frequency, and correlated with overall survival (OS). Uni- and multivariate survival analyses were performed with clinical variables. Longitudinal changes in tumor levels over time were analyzed with linear mixed and joint models. The median OS was 8.1 months, with a recorded disease control rate of 30%. High ctDNA levels (≥ 5%) were associated with inferior survival after undergoing FTD/TPI therapy. Elevated tumor level trajectories over time were associated with higher risks of death. Therefore, ctDNA can help identify patients who are unlikely to benefit significantly from this treatment in late-stage disease, thus preventing unnecessary treatments and their associated side effects, ultimately enhancing quality of life.
Collapse
Affiliation(s)
- Matthias Unseld
- Division of Palliative Care, Department of Medicine I, Medical University of Vienna, Austria
| | - Stefan Kühberger
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Austria
- Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Medical University of Graz, Austria
| | - Ricarda Graf
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Austria
| | - Christine Beichler
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Austria
| | - Markus Braun
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Austria
| | - Nadia Dandachi
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Austria
- Research Unit for Epigenetic and Genetic Cancer Biomarkers, Medical University of Graz, Austria
| | - Ellen Heitzer
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Austria
- Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Medical University of Graz, Austria
| | - Gerald W Prager
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Austria
| |
Collapse
|
72
|
Pinto C, Lonardi S, Maiello E, Martinelli E, Prisciandaro M, Salvatore L, Sartore-Bianchi A, Scartozzi M, Aprile G, Cremolini C, Sobrero A. Trifluridine/tipiracil regimen in combination with bevacizumab for metastatic colorectal cancer in the third line: an expert opinion. Front Oncol 2025; 14:1502185. [PMID: 39911824 PMCID: PMC11794989 DOI: 10.3389/fonc.2024.1502185] [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: 09/26/2024] [Accepted: 10/31/2024] [Indexed: 02/07/2025] Open
Abstract
The prolongation of survival along with the preservation of quality of life, possibly avoiding harmful cumulative toxicities, is the primary therapeutic aim for patients with metastatic colorectal cancer (mCRC) in the third-line setting. Several therapeutic options are now available, although some differences across countries in drug approval and the optimal therapeutic sequencing associated with each peculiar patient subgroup represent a clinical challenge for oncologists. Among various options, the SUNLIGHT trial showed how the combination of trifluridine/tipiracil (FTD/TPI) with bevacizumab is effective with an easily manageable toxicity profile compared to FTD/TPI alone. Of note, the efficacy is confirmed independently from KRAS mutational status and also for patients who had breaks in anti-vascular endothelial growth factor (anti-VEGF) therapy. Herein, we describe the current state of the art in the landscape of treatments after the second progression in mCRC. Based on a critical review of the literature aimed to guide clinicians in their daily decision-making, we point out that the combination of FTD/TPI with bevacizumab produces a clinical benefit in unselected mCRC patients. Therefore, the FTD/TPI plus bevacizumab regimen can represent a new standard of care for the treatment of patients with refractory mCRC who have progressed after two lines of therapy.
Collapse
Affiliation(s)
- Carmine Pinto
- Oncologia Medica, Comprehensive Cancer Centre, AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Evaristo Maiello
- Oncology Unit, Foundation IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Erika Martinelli
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania “L. Vanvitelli “, Napoli, Italy
| | - Michele Prisciandaro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Lisa Salvatore
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
- Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Andrea Sartore-Bianchi
- Division of Clinical Research and Innovation, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
- Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy
| | - Mario Scartozzi
- Oncologia Medica, Azienda Ospedaliero Universitaria e Università degli Studi di Cagliari, Cagliari, Italy
| | - Giuseppe Aprile
- Department of Medical Oncology, AULSS8 Berica, Vicenza, Italy
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alberto Sobrero
- Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
73
|
Begagic E, Vranic S, Sominanda A. The role of interleukin 17 in cancer: a systematic review. Carcinogenesis 2025; 46:bgae079. [PMID: 39673782 DOI: 10.1093/carcin/bgae079] [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: 09/07/2024] [Revised: 12/08/2024] [Accepted: 12/13/2024] [Indexed: 12/16/2024] Open
Abstract
Interleukin 17 (IL17) is a cytokine involved in immune regulation and has been increasingly recognized for its role in cancer progression. This systematic review aims to integrate data on IL17's role in various tumors to better understand its implications for cancer prognosis and treatment. The review included 105 studies (27.6% experimental and 72.4% clinical). Clinical studies involved 9266 patients: 31.2% males, 60.0% females, and 8.8% with undefined gender. IL17A and IL17 were the most studied subtypes (36.2% and 33.3%, respectively). Breast cancer (26.7%), colorectal carcinoma (13.3%), and hematologic malignancies (10.5%) were the most researched neoplasms. IL17A promoted tumor growth in breast cancer and correlated with poor outcomes in colorectal, breast, and lung cancers. IL17 also played a significant role in immune modulation in gliomas and other tumors. IL17A significantly influences tumor growth and prognosis across various cancers, with notable roles in immune modulation and poor outcomes in multiple cancer types.
Collapse
Affiliation(s)
- Emir Begagic
- Department of Neurosurgery, Cantonal Hospital Zenica, Crkvice 67, 72000 Zenica, Bosnia and Herzegovina
| | - Semir Vranic
- Department of Pathology, College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Ajith Sominanda
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| |
Collapse
|
74
|
Thibaudin M, Roussot N, Burlot C, Schmitt A, Vincent J, Tharin Z, Bengrine L, Bellio H, Bertaut A, Hampe L, Daumoine S, Rederstorff E, Peroz M, Huppe T, Derangère V, Rageot D, Simard J, Truntzer C, Fumet JD, Ghiringhelli F. Safety and efficacy of trifluridine/tipiracil +/- bevacizumab plus XB2001 (anti-IL-1α antibody): a single-center phase 1 trial. Signal Transduct Target Ther 2025; 10:22. [PMID: 39820336 PMCID: PMC11739593 DOI: 10.1038/s41392-024-02116-4] [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: 09/02/2024] [Revised: 11/26/2024] [Accepted: 12/25/2024] [Indexed: 01/19/2025] Open
Abstract
In the tumour microenvironment, IL-1α promotes neoangiogenesis, matrix remodelling, tumour proliferation, chemoresistance, and metastases. Highly expressed in human colorectal cancers, IL-1α is associated with poor prognosis. XB2001, a fully human monoclonal antibody neutralizing IL-1α, was evaluated for safety and preliminary efficacy with trifluridine/tipiracil (FTD/TPI) and bevacizumab in metastatic colorectal cancer patients previously treated with oxaliplatin- and irinotecan-based chemotherapies. This single institution, phase 1 study used a 3 + 3 design to assess XB2001 at doses of 250 mg, 500 mg and 1000 mg every 14 days, associated with FTD/TPI 35 mg/m² (days 1-5 and 8-12, every 28 days) (NCT05201352). The Maximum Tolerated Dose of XB2001 + FTD/TPI was then associated in combination with bevacizumab (5 mg/kg, days 1 and 15). Safety, efficacy, pharmacokinetics and pharmacodynamics were assessed. Seventeen patients (median age: 67.4 years) were enroled. No patient exhibited dose-limiting toxicity at any dose. The most common treatment-related adverse events (TRAE) of any grade (G) were diarrhoea (35.3%), nausea (47.1%) and anaemia (35.3%). G3-4 TRAE were neutropenia (17.6%) hypertension and infection (5.9% each). The RP2D (recommended phase 2 dose) of XB2001 was 1000 mg. The disease control rate was 76%, with 23% of patients achieving an objective response, including one complete response. Response and longer progression-free survival were associated with a decrease in serum IL-6 levels during therapy. High intratumoral IL-1α expression at baseline and CD8/PD-L1 infiltration are associated with a better progression-free survival. The combination of XB2001 with FTD/TPI and bevacizumab is feasible and safe, and showed encouraging clinical activity in chemotherapy-resistant mCRC.
Collapse
Affiliation(s)
- Marion Thibaudin
- Centre de Recherche INSERM Center for Translational and Molecular Medicine, 21000, Dijon, France.
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc Equipe Labellisée Ligue Contre le Cancer, 21000, Dijon, France.
- University of Bourgogne Franche-Comté, 21000, Dijon, France.
- Genetic and Immunology Medical Institute, Dijon, France.
| | - Nicolas Roussot
- Centre de Recherche INSERM Center for Translational and Molecular Medicine, 21000, Dijon, France
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc Equipe Labellisée Ligue Contre le Cancer, 21000, Dijon, France
- University of Bourgogne Franche-Comté, 21000, Dijon, France
- Genetic and Immunology Medical Institute, Dijon, France
- Department of Medical Oncology, Centre Georges-François Leclerc, 1 rue du Professeur Marion, 21000, Dijon, France
| | - Chloé Burlot
- Pharmacy Department, Centre Georges-François Leclerc, 1 rue Pr Marion, 21079, Dijon Cedex, France
| | - Antonin Schmitt
- Pharmacy Department, Centre Georges-François Leclerc, 1 rue Pr Marion, 21079, Dijon Cedex, France
| | - Julie Vincent
- Department of Medical Oncology, Centre Georges-François Leclerc, 1 rue du Professeur Marion, 21000, Dijon, France
| | - Zoé Tharin
- Department of Medical Oncology, Centre Georges-François Leclerc, 1 rue du Professeur Marion, 21000, Dijon, France
| | - Leila Bengrine
- Department of Medical Oncology, Centre Georges-François Leclerc, 1 rue du Professeur Marion, 21000, Dijon, France
| | - Hélène Bellio
- Department of Medical Oncology, Centre Georges-François Leclerc, 1 rue du Professeur Marion, 21000, Dijon, France
| | - Aurélie Bertaut
- Methodolgy and biostatistics unit, GF Leclerc Center, Dijon, France
| | - Léa Hampe
- Centre de Recherche INSERM Center for Translational and Molecular Medicine, 21000, Dijon, France
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc Equipe Labellisée Ligue Contre le Cancer, 21000, Dijon, France
| | - Susy Daumoine
- Centre de Recherche INSERM Center for Translational and Molecular Medicine, 21000, Dijon, France
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc Equipe Labellisée Ligue Contre le Cancer, 21000, Dijon, France
| | - Emilie Rederstorff
- Clinical research center, Centre Georges-François Leclerc, Dijon, France
| | - Morgane Peroz
- Centre de Recherche INSERM Center for Translational and Molecular Medicine, 21000, Dijon, France
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc Equipe Labellisée Ligue Contre le Cancer, 21000, Dijon, France
| | - Titouan Huppe
- Centre de Recherche INSERM Center for Translational and Molecular Medicine, 21000, Dijon, France
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc Equipe Labellisée Ligue Contre le Cancer, 21000, Dijon, France
| | - Valentin Derangère
- Centre de Recherche INSERM Center for Translational and Molecular Medicine, 21000, Dijon, France
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc Equipe Labellisée Ligue Contre le Cancer, 21000, Dijon, France
- Genetic and Immunology Medical Institute, Dijon, France
- Department of Medical Oncology, Centre Georges-François Leclerc, 1 rue du Professeur Marion, 21000, Dijon, France
| | - David Rageot
- Centre de Recherche INSERM Center for Translational and Molecular Medicine, 21000, Dijon, France
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc Equipe Labellisée Ligue Contre le Cancer, 21000, Dijon, France
- Genetic and Immunology Medical Institute, Dijon, France
| | | | - Caroline Truntzer
- Centre de Recherche INSERM Center for Translational and Molecular Medicine, 21000, Dijon, France
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc Equipe Labellisée Ligue Contre le Cancer, 21000, Dijon, France
| | - Jean David Fumet
- Centre de Recherche INSERM Center for Translational and Molecular Medicine, 21000, Dijon, France
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc Equipe Labellisée Ligue Contre le Cancer, 21000, Dijon, France
- University of Bourgogne Franche-Comté, 21000, Dijon, France
- Genetic and Immunology Medical Institute, Dijon, France
- Department of Medical Oncology, Centre Georges-François Leclerc, 1 rue du Professeur Marion, 21000, Dijon, France
| | - Francois Ghiringhelli
- Centre de Recherche INSERM Center for Translational and Molecular Medicine, 21000, Dijon, France.
- Cancer Biology Transfer Platform, Centre Georges-François Leclerc Equipe Labellisée Ligue Contre le Cancer, 21000, Dijon, France.
- University of Bourgogne Franche-Comté, 21000, Dijon, France.
- Genetic and Immunology Medical Institute, Dijon, France.
- Department of Medical Oncology, Centre Georges-François Leclerc, 1 rue du Professeur Marion, 21000, Dijon, France.
| |
Collapse
|
75
|
Zhan T, Betge J, Schulte N, Dreikhausen L, Hirth M, Li M, Weidner P, Leipertz A, Teufel A, Ebert MP. Digestive cancers: mechanisms, therapeutics and management. Signal Transduct Target Ther 2025; 10:24. [PMID: 39809756 PMCID: PMC11733248 DOI: 10.1038/s41392-024-02097-4] [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: 06/29/2024] [Revised: 10/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Cancers of the digestive system are major contributors to global cancer-associated morbidity and mortality, accounting for 35% of annual cases of cancer deaths. The etiologies, molecular features, and therapeutic management of these cancer entities are highly heterogeneous and complex. Over the last decade, genomic and functional studies have provided unprecedented insights into the biology of digestive cancers, identifying genetic drivers of tumor progression and key interaction points of tumor cells with the immune system. This knowledge is continuously translated into novel treatment concepts and targets, which are dynamically reshaping the therapeutic landscape of these tumors. In this review, we provide a concise overview of the etiology and molecular pathology of the six most common cancers of the digestive system, including esophageal, gastric, biliary tract, pancreatic, hepatocellular, and colorectal cancers. We comprehensively describe the current stage-dependent pharmacological management of these malignancies, including chemo-, targeted, and immunotherapy. For each cancer entity, we provide an overview of recent therapeutic advancements and research progress. Finally, we describe how novel insights into tumor heterogeneity and immune evasion deepen our understanding of therapy resistance and provide an outlook on innovative therapeutic strategies that will shape the future management of digestive cancers, including CAR-T cell therapy, novel antibody-drug conjugates and targeted therapies.
Collapse
Affiliation(s)
- Tianzuo Zhan
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johannes Betge
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Schulte
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Dreikhausen
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Hirth
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moying Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philip Weidner
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Antonia Leipertz
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Teufel
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
76
|
Yoshida Y, Takahashi M, Komine K, Taniguchi S, Yamada H, Sasaki K, Umegaki S, Kawamura Y, Kasahara Y, Ouchi K, Imai H, Saijo K, Shirota H, Takenaga N, Ishioka C. Correlation between Efficacy and Cardiovascular Adverse Events in Patients with Advanced Solid Cancer Who Received VEGF Pathway Inhibitors: Hypertension within the First Eight Weeks Is Associated with Favorable Outcomes of Patients Treated with VEGF Pathway Inhibitors. Intern Med 2025; 64:177-185. [PMID: 38866528 PMCID: PMC11802217 DOI: 10.2169/internalmedicine.3373-23] [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: 12/21/2023] [Accepted: 04/10/2024] [Indexed: 06/14/2024] Open
Abstract
Objective Many vascular endothelial growth factor (VEGF) pathway inhibitors are used in the treatment of patients with various advanced cancers; however, treatments induce cardiovascular adverse events (CVAEs), such as hypertension, heart failure, arrhythmia, arterial or venous embolism, and hemorrhage. Some studies have suggested a correlation between efficacy and CVAEs; however, further evidence is required. This study evaluated real-world data concerning the frequency and degree of CVAEs and possible associations between CVAEs and efficacy in such patients. Methods We analyzed CVAEs observed in 294 patients with advanced cancer who were treated with ramucirumab, regorafenib, pazopanib, sunitinib, or sorafenib. Results CVAEs of any grade and proteinuria within 8 weeks after the initiation of VEGF pathway inhibitors (early) or during the treatment period (total period) were observed in 72-85% and 77-92% of the patients, respectively. The progression-free survival (PFS) of patients with a CVAE of grade ≥1 in the early period was favorable compared with the PFS of those who had no CVAE (median, 4.9 vs. 3.5 months, p=0.016, log-rank test). Furthermore, the PFS of patients with a CVAE grade ≥3 in the early period was favorable compared to that of those with CVAEs of grades 0-2. Taken together, a higher degree of CVAE was correlated with favorable patient outcomes. Conclusion This study revealed the frequency and degree of CVAEs in patients with solid cancers who received VEGF pathway inhibitors in a real-world setting and added evidence regarding the correlation between CVAEs and efficacy of VEGF pathway inhibitors.
Collapse
Affiliation(s)
- Yuya Yoshida
- Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Japan
- Department of Medical Oncology, Tohoku University Hospital, Japan
| | - Masanobu Takahashi
- Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Japan
- Department of Medical Oncology, Tohoku University Hospital, Japan
| | - Keigo Komine
- Department of Medical Oncology, Tohoku University Hospital, Japan
| | - Sakura Taniguchi
- Department of Medical Oncology, Tohoku University Hospital, Japan
| | - Hideharu Yamada
- Department of Medical Oncology, Tohoku University Hospital, Japan
| | - Keiju Sasaki
- Department of Medical Oncology, Tohoku University Hospital, Japan
| | - Sho Umegaki
- Department of Medical Oncology, Tohoku University Hospital, Japan
| | | | - Yuki Kasahara
- Department of Medical Oncology, Tohoku University Hospital, Japan
- Department of Clinical Oncology, Institute of Development, Aging and Cancer, Tohoku University, Japan
| | - Kota Ouchi
- Department of Medical Oncology, Tohoku University Hospital, Japan
| | - Hiroo Imai
- Department of Medical Oncology, Tohoku University Hospital, Japan
| | - Ken Saijo
- Department of Medical Oncology, Tohoku University Hospital, Japan
| | - Hidekazu Shirota
- Department of Medical Oncology, Tohoku University Hospital, Japan
| | - Noriko Takenaga
- Department of Medical Oncology, Tohoku University Hospital, Japan
| | - Chikashi Ishioka
- Department of Clinical Oncology, Tohoku University Graduate School of Medicine, Japan
- Department of Medical Oncology, Tohoku University Hospital, Japan
| |
Collapse
|
77
|
Omori Y, Matsukuma S, Kawa M, Ishimitsu K, Kawaoka T, Akiyama N, Tokuno K, Fujita Y, Sato S, Yamamoto S. Prognostic impact and risk factors of severe neutropenia in the early phase of treatment with trifluridine-tipiracil for metastatic colorectal cancer patients: a single-center retrospective study. Int J Colorectal Dis 2025; 40:11. [PMID: 39800823 PMCID: PMC11725538 DOI: 10.1007/s00384-024-04798-2] [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] [Accepted: 12/26/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE We aimed to identify the risk factors for severe neutropenia in the early phase of trifluridine-tipiracil (FTD/TPI) treatment, and their impact on overall survival (OS). METHODS This single-center retrospective study included patients with unresectable metastatic colorectal cancer who were treated with FTD/TPI. The primary endpoint was OS, and the secondary endpoint was severe neutropenia during the first and second cycles of FTD/TPI. We assessed the association between outcomes and potential confounders using multivariate analysis. RESULTS Of the 77 total patients, 33 developed severe neutropenia during the first and second treatment cycles. In Cox hazard analysis, the independent factors associated with OS were neutropenia ≥ grade 1 during cycles 1 and 2 (adjusted hazard ratio 0.43; 95% confidence interval (CI) 0.21-0.87), combined treatment with bevacizumab (0.47; 95% CI 0.27-0.83), number of metastatic organs ≥ 3 (2.15; 95% CI 1.22-3.82), and time since diagnosis of metastasis until commencement of FTD/TPI < 18 months (1.94; 95% CI 1.13-3.33). Severe neutropenia during cycles 1 and 2 was not associated with OS (0.75; 0.44-1.27). The risk of severe neutropenia adjusted for initial dose reduction was defined as renal impairment with creatinine clearance (Ccr) of < 60 ml/min (adjusted odds ratio, 4.67; 95% CI, 1.38-15.80) and absolute neutrophil count (per 1000/μl, 0.47; 0.27-0.81). CONCLUSION The neutropenia ≥ grade 1 during cycles 1 and 2 of FTD/TPI is a predictor of favorable outcomes; however, the effect of severe neutropenia on OS was not clear. Renal impairment was also associated with severe neutropenia.
Collapse
Affiliation(s)
- Yoshiro Omori
- Department of Pharmacy, Japan Community Healthcare Organization Tokuyama Central Hospital, 1-1 Koda-Cho, Shunan, Yamaguchi, 745-0822, Japan
| | - Satoshi Matsukuma
- Department of Surgery, Japan Community Healthcare Organization Tokuyama Central Hospital, 1-1 Koda-Cho, Shunan, Yamaguchi, 745-0822, Japan.
| | - Mikiko Kawa
- Department of Pharmacy, Japan Community Healthcare Organization Tokuyama Central Hospital, 1-1 Koda-Cho, Shunan, Yamaguchi, 745-0822, Japan
| | - Kazuki Ishimitsu
- Department of Pharmacy, Japan Community Healthcare Organization Tokuyama Central Hospital, 1-1 Koda-Cho, Shunan, Yamaguchi, 745-0822, Japan
| | - Toru Kawaoka
- Department of Surgery, Japan Community Healthcare Organization Tokuyama Central Hospital, 1-1 Koda-Cho, Shunan, Yamaguchi, 745-0822, Japan
| | - Norio Akiyama
- Department of Surgery, Japan Community Healthcare Organization Tokuyama Central Hospital, 1-1 Koda-Cho, Shunan, Yamaguchi, 745-0822, Japan
| | - Kazuhisa Tokuno
- Department of Surgery, Japan Community Healthcare Organization Tokuyama Central Hospital, 1-1 Koda-Cho, Shunan, Yamaguchi, 745-0822, Japan
| | - Yuji Fujita
- Department of Surgery, Japan Community Healthcare Organization Tokuyama Central Hospital, 1-1 Koda-Cho, Shunan, Yamaguchi, 745-0822, Japan
| | - Shinya Sato
- Department of Pharmacy, Japan Community Healthcare Organization Tokuyama Central Hospital, 1-1 Koda-Cho, Shunan, Yamaguchi, 745-0822, Japan
| | - Shigeru Yamamoto
- Department of Surgery, Japan Community Healthcare Organization Tokuyama Central Hospital, 1-1 Koda-Cho, Shunan, Yamaguchi, 745-0822, Japan
| |
Collapse
|
78
|
Liu P, Xiao J, Xiao J, Zhou J. Metformin and the risk of malignant tumors of digestive system: a mendelian randomization study. Diabetol Metab Syndr 2025; 17:6. [PMID: 39773528 PMCID: PMC11705776 DOI: 10.1186/s13098-024-01573-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Observational studies suggest that metformin may reduce the risk of malignant tumors of the digestive system (MTDS), but these findings are often confounded by bias and unmeasured variables. Recent meta-analyses have questioned these associations, emphasizing the need for robust causal inference. METHODS Mendelian randomization (MR) was used to evaluate the causal relationship between metformin and MTDS. Genetic variants associated with metformin's molecular targets were selected from GTEx, eQTLGen, and UK Biobank and validated using genetic colocalization to ensure instrument validity. GWAS summary statistics for MTDS, encompassing up to 314,193 controls and 6,847 colorectal cancer cases, were obtained from FinnGen and EBI. The primary analysis employed the inverse-variance weighted (IVW) method, supplemented by MR-Egger, weighted median, and weighted mode analyses. Bonferroni correction was applied to adjust for multiple testing across 14 cancer types. RESULTS Genetically proxied metformin use was associated with an increased risk of colorectal cancer (OR = 2.38, 95%CI = 1.38-4.09, P = 0.0018) and related subtypes. No causal relationship was found for hepatocellular carcinoma, gastric cancer, pancreatic cancer, or other digestive system cancers. The robustness of these findings was supported by sensitivity analyses, which indicated no significant pleiotropy, and leave-one-out tests. CONCLUSION This study provides robust genetic evidence that metformin use increases the risk of colorectal cancer, challenging its role as a preventive agent for digestive cancers. These findings emphasize the need for clinicians to carefully evaluate the risks and benefits of metformin, particularly in populations at higher risk for colorectal cancer. Future research should focus on delineating the mechanisms underlying this association to optimize the use of metformin in clinical practice.
Collapse
Affiliation(s)
- Ping Liu
- Department of Radiation Oncology and Hunan Key Laboratory of Translational Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, HunanCancer Hospital, Changsha, China
| | - Junqi Xiao
- Cancer center, The Second Affiliated Hospital, Hengyang Medical School, University of South China, No.35 of Jiefang Avenue, Hengyang City, P. R. China
| | - Jinghuang Xiao
- Department of Radiation Oncology and Hunan Key Laboratory of Translational Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, HunanCancer Hospital, Changsha, China
| | - Jumei Zhou
- Department of Radiation Oncology and Hunan Key Laboratory of Translational Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, HunanCancer Hospital, Changsha, China.
| |
Collapse
|
79
|
Wang J, Liao L, Miao B, Yang B, Li B, Ma X, Fitz A, Wu S, He J, Zhang Q, Ji S, Jin G, Zhang J, Cao Y, Wang H, Qin W, Sun C, Bernards R, Wang C. Deciphering the role of the MALT1-RC3H1 axis in regulating GPX4 protein stability. Proc Natl Acad Sci U S A 2025; 122:e2419625121. [PMID: 39739814 PMCID: PMC11725786 DOI: 10.1073/pnas.2419625121] [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: 09/24/2024] [Accepted: 11/25/2024] [Indexed: 01/02/2025] Open
Abstract
Ferroptosis, a unique form of iron-dependent cell death triggered by lipid peroxidation accumulation, holds great promise for cancer therapy. Despite the crucial role of GPX4 in regulating ferroptosis, our understanding of GPX4 protein regulation remains limited. Through FACS-based genome-wide CRISPR screening, we identified MALT1 as a regulator of GPX4 protein. Inhibition of MALT1 expression enhances GPX4 ubiquitination-mediated degradation by up-regulating the E3 ubiquitin ligase RC3H1. Using both rescue assays and functional genetic screening, we demonstrate that pharmacologically targeting MALT1 triggers ferroptosis in liver cancer cells. Moreover, we show that targeting MALT1 synergizes with sorafenib or regorafenib to induce ferroptosis across multiple cancer types. These findings elucidate the modulatory effects of the MALT1-RC3H1 axis on GPX4 stability, revealing a molecular mechanism that could be exploited to induce ferroptosis for cancer therapy.
Collapse
Affiliation(s)
- Jun Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Long Liao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Beiping Miao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
- German Cancer Research Center, Division Immune Regulation in Cancer, Heidelberg69120, Germany
| | - Bo Yang
- Division of Reproduction and Genetics, First Affiliated Hospital of University of Science and Technology of China, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei230027, China
| | - Botai Li
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Xuhui Ma
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam1066 CX, The Netherlands
| | - Annika Fitz
- German Cancer Research Center, Division Immune Regulation in Cancer, Heidelberg69120, Germany
| | - Shanshan Wu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Jia He
- Department of Medical Oncology, Shanghai Tenth People’s Hospital, Tongji University, School of Medicine, Shanghai200072, China
| | - Qianqian Zhang
- National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200025, China
| | - Shuyi Ji
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai200032, China
| | - Guangzhi Jin
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200336, China
| | - Jianming Zhang
- Institute of Translational Medicine, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai200240, China
| | - Ying Cao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Hui Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| | - Chong Sun
- German Cancer Research Center, Division Immune Regulation in Cancer, Heidelberg69120, Germany
| | - René Bernards
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam1066 CX, The Netherlands
| | - Cun Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200032, China
| |
Collapse
|
80
|
Ai L, Li Q, Zhang S, Dong Y, Yang M, Li J, Pan Y, Yuan Y, Yi S, Wang J, Cheng Y, Feng J, Gao S, Wang X, Qu S, Zhang X, Lu J, Xiu P, Wang S, Yang X, Yu Y, Liu T. Famitinib plus camrelizumab in patients with advanced colorectal cancer: Data from a multicenter, basket study. Innovation (N Y) 2025; 6:100745. [PMID: 39872476 PMCID: PMC11763884 DOI: 10.1016/j.xinn.2024.100745] [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: 03/09/2024] [Accepted: 11/26/2024] [Indexed: 01/30/2025] Open
Abstract
Concurrent inhibition of angiogenesis and immune checkpoints represents a potent therapeutic approach. We conducted a phase 2, multicenter, basket study to assess the efficacy and safety of combination therapy of famitinib (anti-angiogenic agent) plus camrelizumab (PD-1 antagonist) in patients with metastatic solid tumors across 11 cohorts (this study was registered at Clinicaltrials.gov [NCT04346381]). This report focuses on the cohort of patients with metastatic or advanced colorectal cancer. Eligible patients, who had previously received ≥2 lines of systemic treatments for their metastatic disease, were treated with famitinib (20 mg once daily) in combination with camrelizumab (200 mg intravenously every 3 weeks). The primary endpoint was the objective response rate, with secondary endpoints encompassing progression-free survival, overall survival, duration of response, safety and exploratory biomarkers. A total of 44 patients were enrolled and treated. With a median follow-up time of 9.46 months (range 2.0-22.5 months), objective responses were observed in 6 patients (13.6%; 95% confidence interval [CI], 5.2%-27.4%), all of whom had rectal cancer. The median duration of response is 6.2 months (95% CI, 2.3-10.6 months). Median progression-free survival was 3.3 months (95% CI, 2.1-4.1 months), and median overall survival was 10.9 months (95% CI, 7.6-15.2 months). Among the 44 patients, 29 (65.9%) experienced grade 3 or 4 treatment-related adverse events, predominantly hypertension and proteinuria. In conclusion, the combination of famitinib and camrelizumab demonstrates promising antitumor activity with a manageable safety profile in metastatic colorectal cancer patients. Further research is warranted to confirm and extend these findings.
Collapse
Affiliation(s)
- Luoyan Ai
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Qian Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Shilong Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Yu Dong
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Mudan Yang
- The Second Ward of Gastroenterology Department, Shanxi Province Cancer Hospital, Taiyuan 030000, China
| | - Jin Li
- Department of Oncology, Shanghai East Hospital, Shanghai 200120, China
| | - Yueyin Pan
- Oncology Chemotherapy Department, the First Affiliated Hospital of USTC Hospital/Anhui Provicinal Hospital, Hefei 230001, China
| | - Ying Yuan
- Department of Medical Oncology, the Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou 310022, China
| | - Shanyong Yi
- Department of Medical Oncology, Zhengzhou Central Hospital-Department of Medical Oncology, Zhengzhou 450000, China
| | - Junsheng Wang
- Department of Medical Oncology, AnYang Cancer Hospital, Anyang 455000, China
| | - Ying Cheng
- Department of Medical Oncology, JiLin Cancer Hospital, Changchun 130000, China
| | - Jifeng Feng
- Department of Medical Oncology, Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Shegan Gao
- Department of Medical Oncology, The First Affiliated Hospital of Henan University of Science & Technology-Cancer Hospital, Luoyang 471003, China
| | - Xicheng Wang
- Department of Oncology, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Song Qu
- Department of Radiotherapy, Guangxi Medical University Affiliated Tumor Hospital, Nanning 530021, China
| | - Xizhi Zhang
- Oncology Department, Clinical Medical College, Yangzhou University, Yangzhou 225001, China
| | - Jin Lu
- Department of Abdominal Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to School of Medicine, UESTC, Chengdu 610000, China
| | - Peng Xiu
- Clinical Research & Development, Jiangsu Hengrui Medicine Co., Ltd, Shanghai 201203, China
| | - Shuni Wang
- Clinical Research & Development, Jiangsu Hengrui Medicine Co., Ltd, Shanghai 201203, China
| | - Xinfeng Yang
- Clinical Research & Development, Jiangsu Hengrui Medicine Co., Ltd, Shanghai 201203, China
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital Fudan University, Shanghai 200032, China
| |
Collapse
|
81
|
Hossain MA. A comprehensive review of targeting RAF kinase in cancer. Eur J Pharmacol 2025; 986:177142. [PMID: 39577552 DOI: 10.1016/j.ejphar.2024.177142] [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: 08/25/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024]
Abstract
RAF kinases, particularly the BRAF isoform, play a crucial role in the MAPK/ERK signaling pathway, regulating key cellular processes such as proliferation, differentiation, and survival. Dysregulation of this pathway often caused by mutations in the BRAF gene or alterations in upstream regulators like Ras and receptor tyrosine kinases contributes significantly to cancer development. Mutations, such as BRAF-V600E, are present in a variety of malignancies, with the highest prevalence in melanoma. Targeted therapies against RAF kinases have achieved substantial success, especially in BRAF-V600E-mutant melanomas, where inhibitors like vemurafenib and dabrafenib have demonstrated remarkable efficacy, leading to improved patient outcomes. These inhibitors have also shown clinical benefits in cancers such as thyroid and colorectal carcinoma, although to a lesser extent. Despite these successes, therapeutic resistance remains a major hurdle. Resistance mechanisms, including RAF dimerization, feedback reactivation of the MAPK pathway, and paradoxical activation of ERK signaling, often lead to diminished efficacy over time, resulting in disease progression or even secondary malignancies. In response, current research is focusing on novel therapeutic strategies, including combination therapies that target multiple components of the pathway simultaneously, such as MEK inhibitors used in tandem with RAF inhibitors. Additionally, next-generation RAF inhibitors are being developed to address resistance and enhance therapeutic specificity. This review discusses the clinical advancements in RAF-targeted therapies, with a focus on ongoing efforts to overcome therapeutic resistance and enhance outcomes for cancer patients. It also underscores the persistent challenges in effectively targeting RAF kinase in oncology.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
82
|
Sen T, Dotsu Y, Corbett V, Puri S, Sen U, Boyle TA, Mack P, Hirsch F, Aljumaily R, Naqash AR, Sukrithan V, Karim NA. Pulmonary neuroendocrine neoplasms: the molecular landscape, therapeutic challenges, and diagnosis and management strategies. Lancet Oncol 2025; 26:e13-e33. [PMID: 39756451 DOI: 10.1016/s1470-2045(24)00374-7] [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: 01/29/2024] [Revised: 05/20/2024] [Accepted: 06/25/2024] [Indexed: 01/07/2025]
Abstract
Lung neuroendocrine neoplasms are a group of diverse, heterogeneous tumours that range from well-differentiated, low-grade neuroendocrine tumours-such as typical and atypical carcinoids-to high-grade, poorly differentiated aggressive malignancies, such as large-cell neuroendocrine carcinoma (LCNEC) and small-cell lung cancer (SCLC). While the incidence of SCLC has decreased, the worldwide incidence of other pulmonary neuroendocrine neoplasms has been increasing over the past decades. In addition to the standard histopathological classification of lung neuroendocrine neoplasms, the introduction of molecular and sequencing techniques has led to new advances in understanding the biology of these diseases and might influence future classifications and staging that can subsequently improve management guidelines in the adjuvant or metastatic settings. Due to the rarity of neuroendocrine neoplasms, there is a paucity of prospective studies that focus on the lungs, especially in rare, well-differentiated carcinoids and LCNECs. In contrast with the success of targeted therapies in non-small-cell lung cancer (NSCLC), high-grade neuroendocrine carcinomas of the lung often only have a few specific targetable gene alterations. Optimal therapy for LCNECs is not well defined and treatment recommendations are based on extrapolating guidelines for the management of patients with SCLC and NSCLC. This Review explores the epidemiology, diagnosis, and staging of lung neuroendocrine neoplasms to date. In addition, we focus on the evolving molecular landscape and biomarkers, ranging from tumour phenotypes to functional imaging studies and novel molecular biomarkers. We outline the various clinical outcomes, challenges, the treatment landscape, ongoing clinical trials, and future directions.
Collapse
Affiliation(s)
- Triparna Sen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Yosuke Dotsu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Virginia Corbett
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sonam Puri
- Division of Clinical Oncology, The Huntsman Cancer Institute at The University of Utah, Salt Lake City, UT, USA
| | - Utsav Sen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Phil Mack
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fred Hirsch
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raid Aljumaily
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Abdul Rafeh Naqash
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Vineeth Sukrithan
- Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | | |
Collapse
|
83
|
Tang YC, Ou JJ, Hsu SC, Huang CH, Lin LM, Chang HH, Wang YH, Huang ZT, Sun M, Liu KJ, Hung YM, Lai CY, Shih C, Chen CT, Chang JY, Hsieh HP, Jiaang WT, Kuo CC. Advancing precision therapy for colorectal cancer: Developing clinical indications for multi-target kinase inhibitor BPR1J481 using patient-derived xenograft models. Pharmacol Res 2025; 211:107556. [PMID: 39709137 DOI: 10.1016/j.phrs.2024.107556] [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: 06/17/2024] [Revised: 12/04/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
The large and rapid increase in the incidence and mortality of colorectal cancer (CRC) demonstrates the urgent need for new drugs with higher efficacy to treat CRC. However, the lack of applicable and reliable preclinical models significantly hinders the progress of drug development. Patient-derived xenograft (PDX) models are currently considered reliable in vivo preclinical models for predicting drug efficacy in cancer patients. This study successfully uses the CRC PDX model to develop clinical indications for the new multi-target kinase inhibitor BPR1J481 and demonstrated the anti-cancer mechanism and competitive advantages of this drug candidate. The results demonstrate that BPR1J481 exhibits significant anticancer efficacy by inducing apoptosis in CRC PDX tumor tissues and corresponding PDX-derived CRC cells. Through kinase competitive binding and kinase activity assays, we discover that BPR1J481 effectively inhibits SRC kinase activity by directly binding to its active site. The reduction in SRC phosphorylation observed in CRC PDX tumor tissues and derived cells upon treatment with BPR1J481 further validates its inhibitory potential. Furthermore, the decrease in viable cells after SRC knockout and the poorer prognosis observed in patients with higher SRC expression, emphasizes the critical significance and clinical relevance of SRC in CRC. Additionally, BPR1J481 exhibits robust anti-angiogenic effects by suppressing VEGF- and PDGF-induced endothelial cell proliferation, migration, and capillary-like tube formation through inhibition of VEGFR2 and PDGFRβ phosphorylation. Remarkably, BPR1J481 appears to demonstrate greater efficacy against CRC compared to regorafenib. These findings highlight the therapeutic potential of BPR1J481 for patients with CRC.
Collapse
Affiliation(s)
- Ya-Chu Tang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Jing-Jim Ou
- Department of Surgery, Chang Bing Show Chwan Memorial Hospital, Changhua County 505029, Taiwan
| | - Shu-Ching Hsu
- Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Chih-Hsiang Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Li-Mei Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Hsin-Huei Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Yi-Hsin Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Zih-Ting Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Manwu Sun
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Ko-Jiunn Liu
- National Institute of Cancer Research, National Health Research Institutes, Tainan City 704016, Taiwan
| | - Yi-Mei Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan City 704016, Taiwan
| | - Chi-Yun Lai
- Pathology Core Laboratory, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Chuan Shih
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Jang-Yang Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan; Taipei Medical University Hospital, College of Medicine, Taipei Medical University, Taipei City 110301, Taiwan; Taipei Cancer Center, Taiwan; TMU Research Center of Cancer Translational Medicine, 110301, Taiwan
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Weir-Torn Jiaang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan.
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan.
| |
Collapse
|
84
|
Yamaguchi K, Tsuchihashi K, Ueno S, Uehara K, Taguchi R, Ito M, Isobe T, Imajima T, Kitazono T, Tanoue K, Ohmura H, Akashi K, Baba E. Efficacy of pembrolizumab in microsatellite-stable, tumor mutational burden-high metastatic colorectal cancer: genomic signatures and clinical outcomes. ESMO Open 2025; 10:104108. [PMID: 39765187 PMCID: PMC11758824 DOI: 10.1016/j.esmoop.2024.104108] [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: 09/10/2024] [Revised: 12/01/2024] [Accepted: 12/09/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Pembrolizumab, an immune checkpoint inhibitor (ICI), shows significant survival benefits in patients with microsatellite instability-high (MSI-H) metastatic colorectal cancer (mCRC), but its efficacy in microsatellite-stable (MSS) mCRC is limited. Although ICIs are effective in tumor mutational burden-high (TMB-H) solid tumors, the impact on MSS-TMB-H mCRC, a rare subset within MSS mCRC, remains unclear. MATERIALS AND METHODS We conducted a retrospective analysis using clinical and genomic data from the Center for Cancer Genomics and Advanced Therapeutics (C-CAT) repository in Japan. Patients with MSS-TMB-H mCRC who underwent tissue-based comprehensive genomic profiling and were treated with pembrolizumab or other later-line therapies were included. Pembrolizumab's efficacy was compared with that of trifluridine/tipiracil (FTD/TPI) and regorafenib. Genomic profiles of MSS-TMB-H, MSI-H-TMB-H, and MSS-TMB-low (TMB-L) CRCs were analyzed across 71 cancer-related genes. RESULTS Among 127 TMB-H mCRC cases treated with pembrolizumab in the C-CAT repository, 77 were MSS and 50 were MSI-H. Pembrolizumab showed significantly shorter time to treatment failure (TTF) and overall survival (OS) in patients with MSS-TMB-H mCRC compared with those with MSI-H-TMB-H mCRC [median TTF 2.0 versus 10.6 months; hazard ratio (HR) 4.79, 95% confidence interval (CI) 2.65-8.64, median OS 4.5 versus 33.6 months; HR 9.86, 95% CI 3.93-24.77, both P < 0.0001]. Among MSS-TMB-H mCRC patients, 19 received pembrolizumab, 73 received FTD/TPI (±bevacizumab), and 18 received regorafenib as their first later-line therapy. Pembrolizumab showed significantly shorter TTF and OS compared with FTD/TPI (median TTF 1.6 versus 4.1 months; HR 2.66, 95% CI 1.41-5.02, P = 0.0017, median OS 5.4 versus 13.8 months; HR 2.42, 95% CI, 1.09-5.38, P = 0.025). Genomic analysis of 6737 CRCs revealed that MSS-TMB-H CRCs harbored fewer pathogenic alterations than MSI-H-TMB-H CRCs but had a profile similar to MSS-TMB-L CRCs. CONCLUSIONS Pembrolizumab may be less effective than FTD/TPI in later-line treatment of MSS-TMB-H mCRC, potentially due to genomic similarities between MSS-TMB-H and MSS-TMB-L CRC, suggesting the need for alternative therapeutic strategies in this subgroup.
Collapse
Affiliation(s)
- K Yamaguchi
- Department of Hematology, Oncology, and Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan; Department of Medical Education, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - K Tsuchihashi
- Department of Hematology, Oncology, and Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - S Ueno
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - K Uehara
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - R Taguchi
- Department of Hematology, Oncology, and Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - M Ito
- Department of Hematology, Oncology, and Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - T Isobe
- Department of Hematology, Oncology, and Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan; Department of Comprehensive Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - T Imajima
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - T Kitazono
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - K Tanoue
- Department of Hematology, Oncology, and Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - H Ohmura
- Department of Hematology, Oncology, and Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan; Department of Comprehensive Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - K Akashi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - E Baba
- Department of Hematology, Oncology, and Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan; Department of Comprehensive Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
85
|
Li Y, Wu W, Yao J, Wang S, Wu X, Yan J. Patient-Derived Tumor Organoids: A Platform for Precision Therapy of Colorectal Cancer. Cell Transplant 2025; 34:9636897251314645. [PMID: 39953837 PMCID: PMC11829288 DOI: 10.1177/09636897251314645] [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: 10/31/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 02/17/2025] Open
Abstract
Colorectal cancer (CRC) represents a significant cause of cancer-related mortality on a global scale. It is a highly heterogeneous cancer, and the response of patients to homogeneous drug therapy varies considerably. Patient-derived tumor organoids (PDTOs) represent an optimal preclinical model for cancer research. A substantial body of evidence from numerous studies has demonstrated that PDTOs can accurately predict a patient's response to different drug treatments. This article outlines the utilization of PDTOs in the management of CRC across a range of therapeutic contexts, including postoperative adjuvant chemotherapy, palliative chemotherapy, neoadjuvant chemoradiotherapy, targeted therapy, third-line and follow-up treatment, and the treatment of elderly patients. This article delineates the manner in which PDTOs can inform therapeutic decisions at all stages of CRC, thereby assisting clinicians in selecting treatment options and reducing the risk of toxicity and resistance associated with clinical drugs. Moreover, it identifies shortcomings of existing PDTOs, including the absence of consistent criteria for assessing drug sensitivity tests, the lack of vascular and tumor microenvironment models, and the high cost of the technology. In conclusion, despite their inherent limitations, PDTOs offer several advantages, including rapid culture, a high success rate, high consistency, and high throughput, which can be employed as a personalized treatment option for CRC. The use of PDTOs in CRC allows for the prediction of responses to different treatment modalities at various stages of disease progression. This has the potential to reduce adverse drug reactions and the emergence of resistance associated with clinical drugs, facilitate evidence-based clinical decision-making, and guide CRC patients in the selection of personalized medications, thereby advancing the individualized treatment of CRC.
Collapse
Affiliation(s)
- Yiran Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Wei Wu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Jiaxin Yao
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Suidong Wang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Xiufeng Wu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, P.R. China
| | - Jun Yan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| |
Collapse
|
86
|
Ciracì P, Studiale V, Taravella A, Antoniotti C, Cremolini C. Late-line options for patients with metastatic colorectal cancer: a review and evidence-based algorithm. Nat Rev Clin Oncol 2025; 22:28-45. [PMID: 39558030 DOI: 10.1038/s41571-024-00965-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 11/20/2024]
Abstract
Over the past few years, several novel systemic treatments have emerged for patients with treatment-refractory metastatic colorectal cancer, thus making selection of the most effective later-line therapy a challenge for medical oncologists. Over the past decade, regorafenib and trifluridine-tipiracil were the only available drugs and often provided limited clinical benefit compared to best supportive care. Results from subsequent practice-changing trials opened several novel therapeutic avenues, both for unselected patients (such as trifluridine-tipiracil plus bevacizumab or fruquintinib) and for subgroups defined by the presence of actionable alterations in their tumours (such as HER2-targeted therapies or KRASG12C inhibitors) or with no acquired mechanisms of resistance to the previously received targeted agents in circulating tumour DNA (such as retreatment with anti-EGFR antibodies). In this Review, we provide a comprehensive overview of advances in the field over the past few years and offer a practical perspective on translation of the most relevant results into the daily management of patients with metastatic colorectal cancer using an evidence-based algorithm. Finally, we discuss some of the most appealing ongoing areas of research and highlight approaches with the potential to further expand the therapeutic armamentarium.
Collapse
Affiliation(s)
- Paolo Ciracì
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Vittorio Studiale
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Ada Taravella
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Carlotta Antoniotti
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Chiara Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy.
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.
| |
Collapse
|
87
|
Anbari K, Ghanadi K. Colorectal Cancer: Risk Factors, Novel Approaches in Molecular Screening and Treatment. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2025; 14:576-605. [PMID: 40123590 PMCID: PMC11927155 DOI: 10.22088/ijmcm.bums.14.1.576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 07/23/2024] [Indexed: 03/25/2025]
Abstract
By 2040 the burden of colorectal cancer will increase to 3.2 million new cases per year and 1.6 million deaths per year. This highlights the importance of improving preventive measures and treatment strategies. This piece concisely overviews the latest therapeutic and diagnostic approaches for colorectal cancer. In 2019, factors such as low milk intake, smoking, insufficient calcium consumption, and alcohol use had a significant impact on colorectal cancer DALYs worldwide. A comprehensive search was conducted in December 2023 using keywords related to drugs, therapeutic agents, colorectal cancer, diagnostic methods, epidemiology, and novel therapeutic approaches in the PubMed and Scopus databases. Initially, 325 articles were identified based on titles, abstracts, and publication dates. After removing duplicates, 170 unique articles were included. Medications like Nimotuzumab, Cetuximab, and Panitumumab target the Epidermal Growth Factor Receptor (EGFR), which EGF activates. HER2, activated by ligands, is the focus of drugs like Trastuzumab and Pertuzumab. The PD-1/PD-L1 and CTLA-4 pathways, as the immune checkpoints, which involve T cells, are targeted by medications like Ipilimumab. Adoptive cell therapy, including CAR-T cell therapy, TCR modification, and enhancing T cell activity through tumor-infiltrating lymphocytes, is used to combat cancer cell growth. In medical advancements, adoptive cell transfer therapy (ACT) and exosomes in the tumor immune microenvironment (TME) are notable treatment methods that boost the immune system. HIF1A-AS1, CRNDE-h, NEAT1, ZFAS1, and GAS5, along with IGFBP-2, have demonstrated significant CRC diagnostic capacity. Compared to CRC patients with low HIF1A-AS1 expression, individuals with high expression levels were linked to a worse 5-year survival rate.
Collapse
Affiliation(s)
- Khatereh Anbari
- Social Determinants of Health Research Center, Lorestan University of Medical Science, Khorramabad, Iran.
| | - Koroush Ghanadi
- Internal Department, School of Medicine, Lorestan University of Medical Science, Khorramabad, Iran.
| |
Collapse
|
88
|
Liu J, Lu J, Wu L, Zhang T, Wu J, Li L, Tai Z, Chen Z, Zhu Q. Targeting tumor-associated macrophages: Novel insights into immunotherapy of skin cancer. J Adv Res 2025; 67:231-252. [PMID: 38242529 PMCID: PMC11725115 DOI: 10.1016/j.jare.2024.01.013] [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: 11/13/2023] [Revised: 12/19/2023] [Accepted: 01/11/2024] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND The incidence of skin cancer is currently increasing, and conventional treatment options inadequately address the demands of disease management. Fortunately, the recent rapid advancement of immunotherapy, particularly immune checkpoint inhibitors (ICIs), has ushered in a new era for numerous cancer patients. However, the efficacy of immunotherapy remains suboptimal due to the impact of the tumor microenvironment (TME). Tumor-associated macrophages (TAMs), a major component of the TME, play crucial roles in tumor invasion, metastasis, angiogenesis, and immune evasion, significantly impacting tumor development. Consequently, TAMs have gained considerable attention in recent years, and their roles have been extensively studied in various tumors. However, the specific roles of TAMs and their regulatory mechanisms in skin cancer remain unclear. AIM OF REVIEW This paper aims to elucidate the origin and classification of TAMs, investigate the interactions between TAMs and various immune cells, comprehensively understand the precise mechanisms by which TAMs contribute to the pathogenesis of different types of skin cancer, and finally discuss current strategies for targeting TAMs in the treatment of skin cancer. KEY SCIENTIFIC CONCEPTS OF OVERVIEW With a specific emphasis on the interrelationship between TAMs and skin cancer, this paper posits that therapeutic modalities centered on TAMs hold promise in augmenting and harmonizing with prevailing clinical interventions for skin cancer, thereby charting a novel trajectory for advancing the landscape of immunotherapeutic approaches for skin cancer.
Collapse
Affiliation(s)
- Jun Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Jiaye Lu
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Ling Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Tingrui Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Junchao Wu
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Lisha Li
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China.
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China.
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai 200443, China; Shanghai Engineering Research Center of Topical Chinese Medicine, 1278 Baode Road, Shanghai 200443, China.
| |
Collapse
|
89
|
Kang DW, Lynn PB, Wang L, Zhou S, Shen C. Cost-Effectiveness of Fruquintinib for Refractory Metastatic Colorectal Cancer in the USA. PHARMACOECONOMICS - OPEN 2025; 9:93-101. [PMID: 39377863 PMCID: PMC11718036 DOI: 10.1007/s41669-024-00529-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND The FRESCO-2 trial established the efficacy and safety of fruquintinib in patients with refractory metastatic colorectal cancer. However, its cost-effectiveness in the US context is not well documented. OBJECTIVE This study evaluates the cost-effectiveness of fruquintinib versus placebo for this patient population from the perspective of US payers. METHODS We developed a partitioned survival model on the basis of patient-level data reconstructed from the survival curves of the FRESCO-2 trial. Parametric estimation was conducted to estimate long-term clinical outcomes and medical costs over a lifetime horizon. Cost inputs and utilities were sourced from public data and previous literature. We used a discount rate of 3.0% per year for both clinical outcomes and costs. We adopted an incremental cost-effectiveness ratio (ICER) threshold of US$100,000 per quality-adjusted life-year (QALY) gained. We performed sensitivity and scenario analyses to examine the robustness of cost-effectiveness results. RESULTS Fruquintinib treatment resulted in incremental gains of 0.108 life years (LYs) and 0.073 QALYs compared with the placebo, at an additional cost of US$112,294, primarily driven by medication expenses. The ICER for fruquintinib versus placebo was calculated at US$1,037,855 per LY and US$1,546,619 per QALY gained, exceeding the predefined cost-effectiveness threshold. The cost-effectiveness results were robust across all sensitivity and scenario analyses. CONCLUSION AND RELEVANCE Despite the survival benefit, fruquintinib was not cost-effective compared with the placebo in patients with refractory metastatic colorectal cancer in the US setting, on the basis of the conventional willingness-to-pay threshold. Our findings may provide a basis for informing the pricing and reimbursement decisions regarding fruquintinib.
Collapse
Affiliation(s)
- Dong-Won Kang
- Department of Surgery, Penn State College of Medicine, The Pennsylvania State University, 500 University Drive, H151, Hershey, PA, 17033-0850, USA
| | - Patricio B Lynn
- Department of Surgery, Penn State College of Medicine, The Pennsylvania State University, 500 University Drive, H151, Hershey, PA, 17033-0850, USA
| | - Li Wang
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Shouhao Zhou
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Chan Shen
- Department of Surgery, Penn State College of Medicine, The Pennsylvania State University, 500 University Drive, H151, Hershey, PA, 17033-0850, USA.
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
90
|
Todo M, Shirotake S, Kaneko GO, Okamoto K, Sugasawa M, Nakahira M, Ishikawa S, Wakui N, Makino Y, Saeki T. Proactive Symptom Monitoring by Pharmacists Using a Cancer Patient Support App in Out-patient Oral Multi-kinase Inhibitor Therapy: Feasibility Study of a Small Patient Population. In Vivo 2025; 39:440-451. [PMID: 39740869 PMCID: PMC11705115 DOI: 10.21873/invivo.13847] [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: 10/12/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND/AIM Measures to control adverse events (AEs) in the use of oral multi-kinase inhibitors (OMI) are important for the continuation of treatment. PATIENTS AND METHODS In this study, oncology pharmacists monitored symptoms of patients receiving outpatient therapy with OMIs in real-time using a smartphone Web app for the early detection/early treatment of AEs. This feasibility study evaluated the effects of using the app in 10 patients compared with data from 10 patients who did not use the app. RESULTS In the app group, grade 3 AEs were reported in all patients on the day of their occurrence. In contrast, in the no-app group, it took 1-22 days for pharmacists to detect these AEs, among whom 2 patients needed emergency consultations and admissions due to grade 3 AEs. In the app group, 1 patient had an emergency consultation, without admission. The percentage of patients requiring 10 minutes or more for symptom checking during the interview before the physician's examination was significantly lower (p=0.001), and the frequency of telephone calls was also significantly lower (p=0.029) in the app group compared to the no app group. CONCLUSION Using the Web app facilitates the early detection of AEs, contributing to reducing the need for emergency consultations and admissions, and minimizing the time pharmacists spend confirming symptoms with patients.
Collapse
Affiliation(s)
- Maki Todo
- Departments of Pharmacy, Saitama Medical University, Saitama International Medical Center, Saitama, Japan
| | - Suguru Shirotake
- Departments of Uro-Oncology, Saitama Medical University, Saitama International Medical Center, Saitama, Japan
| | - G O Kaneko
- Departments of Uro-Oncology, Saitama Medical University, Saitama International Medical Center, Saitama, Japan
| | - Kojun Okamoto
- Departments of Gastroenterological Surgery, Saitama Medical University, Saitama International Medical Center, Saitama, Japan
| | - Masashi Sugasawa
- Departments of Head and Neck Oncology, and Saitama Medical University, Saitama International Medical Center, Saitama, Japan
| | - Mitsuhiko Nakahira
- Departments of Head and Neck Oncology, and Saitama Medical University, Saitama International Medical Center, Saitama, Japan
| | - Shiho Ishikawa
- Departments of Pharmacy, Saitama Medical University, Saitama International Medical Center, Saitama, Japan
| | - Noriko Wakui
- Departments of Breast Oncology, Saitama Medical University, Saitama International Medical Center, Saitama, Japan
| | - Yoshinori Makino
- Departments of Pharmacy, Saitama Medical University, Saitama International Medical Center, Saitama, Japan
| | - Toshiaki Saeki
- Departments of Breast Oncology, Saitama Medical University, Saitama International Medical Center, Saitama, Japan
| |
Collapse
|
91
|
Tünbekici S, Yuksel HC, Acar C, Sahin G, Orman S, Majidova N, Coskun A, Seyyar M, Dilek MS, Kara M, Dıslı AK, Demir T, Kolkıran N, Sahbazlar M, Demırcıler E, Kuş F, Aytac A, Menekse S, Yucel H, Biter S, Koseci T, Unsal A, Ozveren A, Sevınc A, Goker E, Gürsoy P. Regorafenib Treatment for Recurrent Glioblastoma Beyond Bevacizumab-Based Therapy: A Large, Multicenter, Real-Life Study. Cancers (Basel) 2024; 17:46. [PMID: 39796675 PMCID: PMC11718784 DOI: 10.3390/cancers17010046] [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: 11/26/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES In the REGOMA trial, regorafenib demonstrated an overall survival advantage over lomustine, and it has become a recommended treatment for recurrent glioblastoma in guidelines. This study aimed to evaluate the effectiveness and safety of regorafenib as a third-line treatment for patients with recurrent glioblastoma who progressed while taking bevacizumab-based therapy. METHODS This retrospective, multicenter study in Turkey included 65 patients treated between 2021 and 2023 across 19 oncology centers. The main inclusion criteria were histologically confirmed isocitrate dehydrogenase (IDH)-wildtype glioblastoma, progression after second-line bevacizumab-based treatment, and an Eastern Cooperative Oncology Group (ECOG) performance status score of ≤2. Patients received regorafenib 160 mg once daily for the first 3 weeks of each 4-week cycle. RESULTS The median age of the patients was 53 years (18-67 years), with a median progression-free survival of 2.5 months (95% Confidence Interval: 2.23-2.75) and a median overall survival of 4.1 months (95% CI: 3.52-4.68). The median overall survival was improved in patients who received subsequent therapy after regorafenib treatment compared with those who did not (p = 0.022). Progression-free survival was longer in patients with ECOG 0-1 than in those with ECOG 2 (p = 0.042). The safety profile was consistent with that of the REGOMA trial, with no drug-related deaths observed. CONCLUSIONS Regorafenib shows good efficacy and safety as a third-line treatment for recurrent glioblastoma after bevacizumab-based therapy. This study supports the use of regorafenib and emphasizes the need for further randomized studies to validate its role and optimize treatment strategies.
Collapse
Affiliation(s)
- Salih Tünbekici
- Department of Medical Oncology, Ege University Medical Faculty, Izmir 35040, Turkey; (H.c.Y.); (C.A.); (G.S.); (P.G.)
| | - Haydar cagatay Yuksel
- Department of Medical Oncology, Ege University Medical Faculty, Izmir 35040, Turkey; (H.c.Y.); (C.A.); (G.S.); (P.G.)
| | - Caner Acar
- Department of Medical Oncology, Ege University Medical Faculty, Izmir 35040, Turkey; (H.c.Y.); (C.A.); (G.S.); (P.G.)
| | - Gökhan Sahin
- Department of Medical Oncology, Ege University Medical Faculty, Izmir 35040, Turkey; (H.c.Y.); (C.A.); (G.S.); (P.G.)
| | - Seval Orman
- Department of Medical Oncology, Kartal Dr. Lütfi Kirdar City Hospital, Health Science University, Cevizli, D-100 Güney Yanyol, Cevizli Mevkii No:47, Kartal, Istanbul 34865, Turkey;
| | - Nargiz Majidova
- Department of Medical Oncology, School of Medicine, Marmara University, Istanbul 34899, Turkey;
| | - Alper Coskun
- Department of Medical Oncology, Uludağ University, Bursa 16059, Turkey;
| | - Mustafa Seyyar
- Department of Medical Oncology, Gaziantep City Hospital, Gaziantep 27470, Turkey;
| | - Mehmet sıddık Dilek
- Medical Oncology, Medical School, Dicle University, Diyarbakir 21280, Turkey;
| | - Mahmut Kara
- Department of Medical Oncology, Yuzuncu Yil University Faculty of Medicine, Van 65090, Turkey;
| | - Ahmet Kursat Dıslı
- Department of Medical Oncology, Erciyes University Faculty of Medicine, Kayseri 38030, Turkey;
| | - Teyfik Demir
- Department of Medical Oncology, Ondokuz Mayis University Faculty of Medicine, Samsun 55270, Turkey;
| | - Nagihan Kolkıran
- Department of Medical Oncology, Celal Bayar University Faculty of Medicine, Manisa 45030, Turkey; (N.K.); (M.S.)
| | - Mustafa Sahbazlar
- Department of Medical Oncology, Celal Bayar University Faculty of Medicine, Manisa 45030, Turkey; (N.K.); (M.S.)
| | - Erkut Demırcıler
- Department of Medical Oncology, 9 Eylül University Faculty of Medicine, Izmir 35220, Turkey;
| | - Fatih Kuş
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara 06230, Turkey;
| | - Ali Aytac
- Department of Medical Oncology, Mehmet Akif İnan Training and Research Hospital, Sanlıurfa 63040, Turkey;
| | - Serkan Menekse
- Department of Medical Oncology, Manisa City Hospital, Manisa 45040, Turkey;
| | - Hakan Yucel
- Department of Medical Oncology, School of Medicine, Gaziantep University, Gaziantep 27580, Turkey;
| | - Sedat Biter
- Department of Medical Oncology, Cukurova University, Adana 01790, Turkey; (S.B.); (T.K.)
| | - Tolga Koseci
- Department of Medical Oncology, Cukurova University, Adana 01790, Turkey; (S.B.); (T.K.)
| | - Ahmet Unsal
- Department of Medical Oncology, Gumushane State Hospital, Gumushane 29000, Turkey;
| | - Ahmet Ozveren
- Medical Oncology, Department MD, İzmir Kent Hospital, Izmir 35620, Turkey;
| | - Alper Sevınc
- Medical Oncology, Medical Park Gaziantep Hospital, Gaziantep 27090, Turkey;
| | - Erdem Goker
- Department of Medical Oncology, Ege University Medical Faculty, Izmir 35040, Turkey; (H.c.Y.); (C.A.); (G.S.); (P.G.)
| | - Pınar Gürsoy
- Department of Medical Oncology, Ege University Medical Faculty, Izmir 35040, Turkey; (H.c.Y.); (C.A.); (G.S.); (P.G.)
| |
Collapse
|
92
|
Saha S, Ghosh S, Ghosh S, Nandi S, Nayak A. Unraveling the complexities of colorectal cancer and its promising therapies - An updated review. Int Immunopharmacol 2024; 143:113325. [PMID: 39405944 DOI: 10.1016/j.intimp.2024.113325] [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: 05/04/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Colorectal cancer (CRC) continues to be a global health concern, necessitating further research into its complex biology and innovative treatment approaches. The etiology, pathogenesis, diagnosis, and treatment of colorectal cancer are summarized in this thorough review along with recent developments. The multifactorial nature of colorectal cancer is examined, including genetic predispositions, environmental factors, and lifestyle decisions. The focus is on deciphering the complex interactions between signaling pathways such as Wnt/β-catenin, MAPK, TGF-β as well as PI3K/AKT that participate in the onset, growth, and metastasis of CRC. There is a discussion of various diagnostic modalities that span from traditional colonoscopy to sophisticated molecular techniques like liquid biopsy and radiomics, emphasizing their functions in early identification, prognostication, and treatment stratification. The potential of artificial intelligence as well as machine learning algorithms in improving accuracy as well as efficiency in colorectal cancer diagnosis and management is also explored. Regarding therapy, the review provides a thorough overview of well-known treatments like radiation, chemotherapy, and surgery as well as delves into the newly-emerging areas of targeted therapies as well as immunotherapies. Immune checkpoint inhibitors as well as other molecularly targeted treatments, such as anti-epidermal growth factor receptor (anti-EGFR) as well as anti-vascular endothelial growth factor (anti-VEGF) monoclonal antibodies, show promise in improving the prognosis of colorectal cancer patients, in particular, those suffering from metastatic disease. This review focuses on giving readers a thorough understanding of colorectal cancer by considering its complexities, the present status of treatment, and potential future paths for therapeutic interventions. Through unraveling the intricate web of this disease, we can develop a more tailored and effective approach to treating CRC.
Collapse
Affiliation(s)
- Sayan Saha
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Shreya Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Suman Ghosh
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India
| | - Sumit Nandi
- Department of Pharmacology, Gupta College of Technological Sciences, Asansol, West Bengal 713301, India
| | - Aditi Nayak
- Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F, Nilgunj Rd, Sahid Colony, Panihati, Kolkata, West Bengal 700114, India.
| |
Collapse
|
93
|
Qu F, Wu S, Yu W. Progress of Immune Checkpoint Inhibitors Therapy for pMMR/MSS Metastatic Colorectal Cancer. Onco Targets Ther 2024; 17:1223-1253. [PMID: 39735789 PMCID: PMC11681808 DOI: 10.2147/ott.s500281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/12/2024] [Indexed: 12/31/2024] Open
Abstract
Immunotherapy is one of the research hotspots in colorectal cancer field in recent years. The colorectal cancer patients with mismatch repair-deficient (dMMR) or high microsatellite instability (MSI-H) are the primary beneficiaries of immunotherapy. However, the vast majority of colorectal cancers are mismatch repair proficient (pMMR) or microsatellite stability (MSS), and their immune microenvironment is characterized by "cold tumors" that are generally insensitive to single immunotherapy based on immune checkpoint inhibitors (ICIs). Studies have shown that some pMMR/MSS colorectal cancer patients regulate the immune microenvironment by combining other treatments, such as multi-target tyrosine kinase inhibitors, anti-vascular endothelial growth factor (VEGF) monoclonal antibodies, chemotherapy, radiotherapy, anti-epithelial growth factor receptor (EGFR) monoclonal antibodies, and mitogen-activated protein kinase (MAPK) signaling pathway inhibitors and oncolytic viruses, etc. to transform "cold tumor" into "hot tumor", thereby improving the response to immunotherapy. In addition, screening for potential prognostic biomarkers can also enrich the population benefiting from immunotherapy for microsatellite stable colorectal cancer. Therefore, in pMMR or MSS metastatic colorectal cancer (mCRC), the optimization of immunotherapy regimens and the search for effective efficacy prediction biomarkers are currently important research directions. In this paper, we review the progress of efficacy of immunotherapy (mainly ICIs) in pMMR /MSS mCRC, challenges and potential markers, in order to provide research ideas for the development of immunotherapy for mCRC.
Collapse
Affiliation(s)
- Fanjie Qu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| | - Shuang Wu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| | - WeiWei Yu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| |
Collapse
|
94
|
Liang B, Tang M, Huang C, Yang Y, He Y, Liao S, Shen W. A Systematic Review and Meta-Analysis of the Efficacy and Safety of Regorafenib in the Treatment of Metastatic Colorectal Cancer. J Gastrointest Cancer 2024; 56:36. [PMID: 39710828 DOI: 10.1007/s12029-024-01158-9] [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] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND AND OBJECTIVE Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide. Despite advances in treatment, metastatic colorectal cancer (mCRC) remains a significant challenge due to its heterogeneity and resistance to therapy. Regorafenib, a multikinase inhibitor, can inhibit tumor progression through multiple mechanisms, thereby improving patient prognosis. It has emerged as a potential treatment option for mCRC patients who have progressed on standard therapies. This systematic review and meta-analysis aims to evaluate the efficacy and safety of Regorafenib in this patient population, synthesizing data from clinical trials to provide a comprehensive understanding of its role in mCRC treatment. METHODS A systematic literature search was conducted via the PubMed, Web of Science (WOS), and Embase databases from January 2012 to December 2024. Studies were included if they were randomized controlled trials (RCTs) or clinical trials that reported outcomes of regorafenib treatment in mCRC patients, including overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and disease control rate (DCR). Secondary outcomes included the incidence of serious adverse events (SAEs). OS refers to the length of time from the start of treatment until the death of the patient from any cause, while mortality specifically denotes the number of deaths occurring within the study period. Data were extracted by two independent reviewers using a standardized form. The meta-analysis was performed using RevMan 5.0 statistical software. RESULTS A total of 5,082 articles were retrieved, and ultimately, 9 eligible studies involving a total of 2,823 patients were included. All 9 included studies reported OS and PFS. In these mCRC patients, the dose of regorafenib was usually 160 mg daily. The meta results indicated that the OS of patients in the regorafenib group was significantly different [MD = 1.33, 95% CI (0.33, 2.33), P = 0.009]. Eight studies reported the ORR of the disease [OR = 1.13, 95% CI (0.73, 1.76), P = 0.57]. Five studies reported the DCR, and the DCR of patients in the regorafenib group was significantly different from that of patients in the control group [OR = 3.45, 95% CI (2.04, 5.84), P < 0.00001]. The incidence of SAEs (> grade 3) was reported in all 9 included studies [OR = 2.48, 95% CI (1.29, 4.73), P = 0.006]. CONCLUSION In this systematic review of prospective trials, regorafenib resulted in improved OS with manageable adverse effects for patients with advanced mCRC. Still, considering the safety, future research should focus on investigating the dose optimization of regorafenib, as well as predictive biomarkers for therapeutic efficacy.
Collapse
Affiliation(s)
- Bingjun Liang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Shenzhen University (People's Hospital of Shenzhen Baoan District), Shenzhen, 518100, China
| | - Ming Tang
- Radiotherapy Department, The Second Affiliated Hospital of Shenzhen University (People's Hospital of Shenzhen Baoan District), Shenzhen, 518100, China
| | - Chao Huang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Shenzhen University (People's Hospital of Shenzhen Baoan District), Shenzhen, 518100, China.
| | - Yidian Yang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Shenzhen University (People's Hospital of Shenzhen Baoan District), Shenzhen, 518100, China
| | - Yue He
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Shenzhen University (People's Hospital of Shenzhen Baoan District), Shenzhen, 518100, China
| | - Shengrong Liao
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Shenzhen University (People's Hospital of Shenzhen Baoan District), Shenzhen, 518100, China
| | - Weizeng Shen
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Shenzhen University (People's Hospital of Shenzhen Baoan District), Shenzhen, 518100, China
| |
Collapse
|
95
|
He C, Chi J, Du Z, Zhuang Z, Li L. Case report: Durable response from tegafur/gimeracil/oteracil (S-1) combined with fruquintinib and sintilimab as a third-line treatment for MSS metastatic colorectal cancer with a BRAF V600E mutation. Front Oncol 2024; 14:1468532. [PMID: 39759155 PMCID: PMC11695214 DOI: 10.3389/fonc.2024.1468532] [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: 07/22/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Patients with microsatellite stable (MSS) metastatic colorectal cancer (mCRC) who fail first- and second-line treatments face significant challenges in third-line therapy, where monotherapies often yield poor outcomes and limited survival benefits. The prognosis is particularly poor for mCRC with the unique molecular subtype of BRAF V600E mutation. This report describes sustained benefits from a third-line treatment regimen (SFS) combining tegafur/gimeracil/oteracil (S-1), fruquintinib, and sintilimab in a patient with BRAF V600E-mutated MSS mCRC. A 23-year-old woman was admitted with dizziness, and enhanced computed tomography (CT) and colonoscopy revealed colon cancer. Based on pathological and genetic testing, the final diagnosis was colon adenocarcinoma with lymph node and liver metastases (cT3N1M1, stage IVc, BRAF-V600E(+), MSS type). Following progressive disease (PD) after FOLFOX chemotherapy and surgery, the patient received 40 cycles of the SFS regimen (S-1 60 mg bid po d1-14 + fruquintinib 3 mg qd d1-21 + sintilimab 200 mg ivd q3w), achieving stable disease (SD). At the most recent follow-up, the patient has remained in sustained remission for over 3 years. The SFS regimen may be an attractive therapeutic strategy for patients with BRAF V600E-mutated MSS mCRC, warranting further evaluation in a larger patient cohort. We have registered a related clinical study (registration number: ChiCTR2300079188) and hope that the results will bring new hope for patients with MSS mCRC.
Collapse
Affiliation(s)
- Chunxia He
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical College, University of Guangzhou Traditional Chinese Medicine, Guangzhou, China
| | - Jiaxin Chi
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangdong, Guangzhou, China
| | - Zhihua Du
- Department of Pathology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical College, University of Guangzhou Traditional Chinese Medicine, Guangzhou, China
| | - Zhenjie Zhuang
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical College, University of Guangzhou Traditional Chinese Medicine, Guangzhou, China
| | - Liuning Li
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical College, University of Guangzhou Traditional Chinese Medicine, Guangzhou, China
| |
Collapse
|
96
|
Pham TD, Becker JH, Metropulos AE, Mubin N, Spaulding C, Bentrem DJ, Munshi HG. Regorafenib induces DNA damage and enhances PARP inhibitor efficacy in pancreatic ductal carcinoma. BMC Cancer 2024; 24:1562. [PMID: 39707244 DOI: 10.1186/s12885-024-13334-y] [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: 04/27/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND There is increasing interest in enhancing the response of the PARP inhibitor olaparib, which is currently approved for pancreatic ductal adenocarcinoma (PDAC) patients with defects in DNA damage repair associated with germline BRCA1/2 mutations. Moreover, agents that can mimic these defects in the absence of germline BRCA1/2 mutations are an area of active research in hopes of increasing the number of patients eligible for treatment with PARP inhibitors. The extent to which regorafenib, an FDA-approved tyrosine kinase inhibitor, can be used to enhance the efficacy of PARP inhibitors in PDAC cells without known BRCA1/2 mutations remains to be investigated. METHODS Comet assay, cell cycle analysis, western blotting, and immunofluorescent detection of H2AXS139 were used to evaluate the extent to which regorafenib induces DNA damage in PDAC cell lines. The effects of regorafenib, either alone or in combination with PARPi inhibitors, on PDAC cell death were assessed by Annexin V/PI co-staining assay in cell lines and by immunohistochemistry staining for cleaved caspase-3 in mouse tumors and in ex vivo slice cultures of human PDAC tumors. Flow cytometry-based analysis was used to evaluate the ability of regorafenib to reprogram PDAC tumor microenvironment. RESULTS We now show that regorafenib, a tyrosine-kinase inhibitor with efficacy in several gastrointestinal malignancies, can enhance the response of olaparib in pancreatic cancer. While regorafenib induces DNA damage and limits the ability of PDAC cells to resolve the damage, regorafenib by itself does not induce apoptosis. However, regorafenib in combination with olaparib further induces DNA damage in vitro, in tumor-bearing mice, and in ex vivo slice cultures of human PDAC tumors, resulting in increased apoptosis compared to olaparib alone. Notably, we show that the efficacy of the combination treatment is not dependent on cytolytic T cells. CONCLUSIONS Together, these findings demonstrate that regorafenib can attenuate DNA damage response and potentiate the efficacy of PARP inhibitors in PDAC tumors.
Collapse
Affiliation(s)
- Thao D Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.
| | - Jeffrey H Becker
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Anastasia E Metropulos
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Nida Mubin
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Christina Spaulding
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - David J Bentrem
- Jesse Brown VA Medical Center, Chicago, IL, USA
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hidayatullah G Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Jesse Brown VA Medical Center, Chicago, IL, USA.
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.
| |
Collapse
|
97
|
Cruz-Diaz WE, Paitan V, Medina J, Flores R, Haro-Varas J, Mantilla R, Castro-Oliden V. Temozolomide and capecitabine regimen as first-line treatment in advanced gastroenteropancreatic neuroendocrine tumors at a Latin American reference center. World J Gastrointest Oncol 2024; 16:4675-4684. [PMID: 39678797 PMCID: PMC11577364 DOI: 10.4251/wjgo.v16.i12.4675] [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: 07/22/2024] [Revised: 08/31/2024] [Accepted: 09/19/2024] [Indexed: 11/12/2024] Open
Abstract
BACKGROUND Numerous studies have indicated that the temozolomide and capecitabine regimen (TEMCAP) exhibits a certain level of efficacy in treating advanced, well-differentiated gastroenteropancreatic neuroendocrine tumors (GEP-NET). However, published data from Peru are limited. We hypothesize that this regimen could be a viable therapeutic option for advanced GEP-NET in the Peruvian population. AIM To evaluate overall survival (OS) in patients diagnosed with advanced GEP-NET treated with TEMCAP at the Instituto Nacional de Enfermedades Neoplásicas (INEN) in Lima-Perú. METHODS A retrospective review was conducted to identify patients with GEP-NEN treated with the TEMCAP regimen between 2011 and 2021 at the INEN. A total of thirty-eight patients were included in the final analysis: Thirty-five received TEMCAP as a first-line treatment, and three as a second-line treatment. The primary objective was to evaluate OS. The efficacy and safety of TEMCAP were assessed until the occurrence of unacceptable toxicity or disease progression. Survival outcomes were estimated using the Kaplan-Meier method. RESULTS The median age of the patients was 52 years (range 24-77 years), and 53.3% were female. The most common symptoms at diagnosis were abdominal pain in 31 patients (81.6%). Primary tumors included 12 in the rectum (31.6%), 11 in the pancreas (28.9%), 3 in the ileum (7.9%), 2 in the mesentery (5.3%), 2 in the small intestine (5.3%), 1 in the appendix (2.6%), 1 in the stomach (2.6%) and 6 cases of liver metastasis of unknown primary (15.8%). Five were neuroendocrine tumors (NET) G1 (13.2%), 33 were NET G2 (86.8%), five had Ki67 < 3% (13.2%), and 33 had Ki67 between 3% and 20% (86.8%). TEMCAP was administered to 35 (92.1%) patients as first-line treatment. OS at 12, 36, and 60 months was estimated in 80%, 66%, and 42%, respectively, with a median OS of 49 months. CONCLUSION TEMCAP therapy is a viable first-line option regarding efficacy and tolerability in areas where standard therapy is inaccessible.
Collapse
Affiliation(s)
| | - Victor Paitan
- Department of Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15038, Perú
| | - Jersinho Medina
- Department of Pathology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15038, Perú
| | - Raymundo Flores
- Department of Radiology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15038, Perú
| | - Juan Haro-Varas
- Department of Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15038, Perú
| | - Raul Mantilla
- Department of Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15038, Perú
| | - Victor Castro-Oliden
- Department of Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima 15038, Perú
| |
Collapse
|
98
|
Badran O, Cohen I, Bar-Sela G. The Impact of Iron on Cancer-Related Immune Functions in Oncology: Molecular Mechanisms and Clinical Evidence. Cancers (Basel) 2024; 16:4156. [PMID: 39766056 PMCID: PMC11674619 DOI: 10.3390/cancers16244156] [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: 11/09/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Iron metabolism plays a dual role in cancer, serving as an essential nutrient for cellular functions and a potential catalyst for tumor growth and immune evasion. Here, we cover the complex interplay between iron levels within the serum or in the microenvironment and cancer therapy, focusing on how iron deficiency and overload can impact immune function, tumor progression, and treatment efficacy. On the one hand, we highlight iron deficiency as a factor of primary immune responses and its adverse effects on anti-cancer immunotherapy efficacy. On the other hand, we also stress the impact of iron overload as an essential factor contributing to tumor growth, creating a suppressive tumor microenvironment that hinders immune checkpoint inhibitor immunotherapy. Overall, we emphasize the necessity of the personalized management of iron levels in oncology patients as a critical element in treatment optimization to achieve favorable outcomes. Based on these considerations, we believe that close and careful monitoring and the tailored balancing of iron supplementation strategies should be the subject of further clinical studies, and routine iron management should be implemented in oncology clinical practice and integrated into cancer therapy protocols.
Collapse
Affiliation(s)
- Omar Badran
- Department of Oncology, Emek Medical Center, Afula 1834111, Israel; (O.B.); (I.C.)
- Technion Integrated Cancer Center, Faculty of Medicine, Technion, Haifa 3525422, Israel
| | - Idan Cohen
- Department of Oncology, Emek Medical Center, Afula 1834111, Israel; (O.B.); (I.C.)
| | - Gil Bar-Sela
- Department of Oncology, Emek Medical Center, Afula 1834111, Israel; (O.B.); (I.C.)
- Technion Integrated Cancer Center, Faculty of Medicine, Technion, Haifa 3525422, Israel
| |
Collapse
|
99
|
Ros J, Ucha JM, Garcia-Galea E, Gomez P, Martini G, Balconi F, Comas R, Alonso V, Rodriguez M, Baraibar I, Salva F, Saoudi N, Alcaraz A, Garcia A, Tabernero J, Elez E. Real-World Data of Patients with BRAF V600E-Mutated Metastatic Colorectal Cancer Treated with Trifluridine/Tipiracil. Cancers (Basel) 2024; 16:4140. [PMID: 39766040 PMCID: PMC11674344 DOI: 10.3390/cancers16244140] [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: 11/12/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND For patients with refractory metastatic colorectal cancer (mCRC), trifluridine/tipiracil (FTD-TPI) has been associated with a significant improvement in overall survival (OS). However, data are lacking regarding the activity of FTD-TPI in patients with BRAF-mutated mCRC. METHODS This retrospective, multicenter, international cohort included patients with BRAF-mutated mCRC treated with FTD-TPI in a real-life setting in Spain and Italy. Survival analysis was performed using Kaplan-Meier methods and Cox proportional hazard models and according to established prognostic groups: good prognosis characteristics (GPC; < 3 metastatic sites and time from metastases to FTD-TPI ≥ 18 months) and poor prognosis characteristics (PPC; ≥ 3 metastatic sites or time from metastases to FTD-TPI < 18 months). RESULTS In the 26 patients included, the median age was 61 years, 13 (50%) were female, and 20 (77%) had an Eastern Cooperative Oncology Group (ECOG) performance status of 1. Fourteen (56%) patients had right-sided tumors, six (23%) had microsatellite instability tumors, and thirteen (50%) had liver metastases. Median progression-free survival was 2.3 months (95% CI 2.0-3.2), and median OS (mOS) was 6.6 months (95% CI 4.4-12.0). mOS was 7.6 vs. 4.2 months (HR 1.64, 95% CI 0.65-4.10, p = 0.3) for GPC and PPC patients, respectively. Exploratory analyses identified ECOG as the only feature associated with survival. The most frequent grade 3-4 adverse events were neutropenia (8%), anemia (8%), and asthenia (4%). CONCLUSIONS Patients with BRAF mutant mCRC achieved modest benefits with FTD-TPI; however, patients with GPC and ECOG 0 achieved longer OS compared with those with PPC or ECOG 1-2, thus warranting further exploration in prospective cohorts.
Collapse
Affiliation(s)
- Javier Ros
- Medical Oncology, Vall d’Hebron University Hospital, 08035 Barcelona, Spain
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Jose Maria Ucha
- Medical Oncology, Vall d’Hebron University Hospital, 08035 Barcelona, Spain
| | | | - Pablo Gomez
- Medical Oncology, Miguel Servet Hospital, 50009 Zaragoza, Spain
| | - Giulia Martini
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Francesca Balconi
- Medical Oncology, University Hospital, University of Cagliari, 09124 Cagliari, Italy
| | - Raquel Comas
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Vicente Alonso
- Medical Oncology, Miguel Servet Hospital, 50009 Zaragoza, Spain
| | - Marta Rodriguez
- Medical Oncology, Vall d’Hebron University Hospital, 08035 Barcelona, Spain
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Iosune Baraibar
- Medical Oncology, Vall d’Hebron University Hospital, 08035 Barcelona, Spain
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Francesc Salva
- Medical Oncology, Vall d’Hebron University Hospital, 08035 Barcelona, Spain
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Nadia Saoudi
- Medical Oncology, Vall d’Hebron University Hospital, 08035 Barcelona, Spain
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain
| | | | - Ariadna Garcia
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Josep Tabernero
- Medical Oncology, Vall d’Hebron University Hospital, 08035 Barcelona, Spain
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain
| | - Elena Elez
- Medical Oncology, Vall d’Hebron University Hospital, 08035 Barcelona, Spain
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain
| |
Collapse
|
100
|
Hsiao KY, Chen HP, Rau KM, Liu KW, Shia BC, Chang WS, Liang HY, Hsieh MC. Association between sidedness and survival among chemotherapy refractory metastatic colorectal cancer patients treated with trifluridine/tipiracil or regorafenib. Oncologist 2024; 29:e1669-e1679. [PMID: 39245044 PMCID: PMC11630785 DOI: 10.1093/oncolo/oyae235] [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: 12/23/2023] [Accepted: 08/05/2024] [Indexed: 09/10/2024] Open
Abstract
BACKGROUND The impact of sidedness on survival of later-line treatment in patients with metastatic colorectal cancer (mCRC) is undetermined. This study aimed to investigate the association between sidedness and survival among chemotherapy refractory patients with mCRC treated with trifluridine/tipiracil (TAS-102) or regorafenib or both. PATIENTS AND METHODS Patients with mCRC treated with TAS-102 or regorafenib between 2015 and 2020 was retrospectively collected. Patients were stratified into TAS-102 first and regorafenib first, then subdivided into TAS-102 followed by regorafenib (T-R) and regorafenib followed by TAS-102 (R-T) groups. The oncologic outcomes were presented with time-to-treatment failure (TTF) and overall survival (OS). RESULTS After matching, 376 TAS-102 patients and 376 regorafenib patients were included for outcomes comparison. TTF had insignificant differences while OS was significantly different between TAS-102 and regorafenib groups. Median TTF and OS were 1.9 months versus 2.0 months (P = .701) and 9.1 months versus 7.0 months (P = .008) in TAS-102 and regorafenib, respectively. The OS benefits were consistent regardless primary tumor location. Subgroup analysis with 174 T-R patients and 174 R-T patients was investigated for treatment sequences. TTF and OS had significant differences in both groups. Median TTF and OS were 8.5 months versus 6.3 months (P = .001) and 14.4 months versus 12.6 months (P = .035) in T-R and R-T groups, respectively. The TTF and OS benefits were persisted regardless primary tumor location. CONCLUSION TAS-102 first provided a better survival benefit in chemotherapy refractory patients with mCRC across all sidedness. Further prospective studies are warranted to validate our conclusions.
Collapse
Affiliation(s)
- Kai-Yuan Hsiao
- Artificial Intelligence Development Center, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Hsin-Pao Chen
- Division of Colon and Rectum Surgery, Department of Surgery, E-Da Hospital, Kaohsiung, Taiwan
- College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Kun-Ming Rau
- College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Department of Hematology-Oncology, E-Da Cancer Hospital, Kaohsiung, Taiwan
| | - Kuang-Wen Liu
- Division of Colon and Rectum Surgery, Department of Surgery, E-Da Hospital, Kaohsiung, Taiwan
- College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Ben-Chang Shia
- Artificial Intelligence Development Center, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Wei-Shan Chang
- Artificial Intelligence Development Center, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Hao-Yun Liang
- College of Medicine, I-Shou University, Kaohsiung, Taiwan
- EMMT Systems Corporation, Director, Taiwan
| | - Meng-Che Hsieh
- College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Department of Hematology-Oncology, E-Da Cancer Hospital, Kaohsiung, Taiwan
| |
Collapse
|