1
|
Roth JA, Kratochvil D, Dorman S, Bernauer M. Cost-efficiency and expanded access modeling of conversion to biosimilar bevacizumab in metastatic colorectal and non-squamous non-small cell lung cancer in Medicare. J Med Econ 2025; 28:378-386. [PMID: 40035265 DOI: 10.1080/13696998.2025.2474884] [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: 01/06/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Biosimilars to originator bevacizumab (Avastin), such as bevacizumab-bvzr (Zirabev), can deliver substantial savings and/or expanded access to biologic therapy for patients with metastatic colorectal (mCRC) and non-squamous non-small cell lung cancer (mNSCLC). The objective of this study is to explore the cost-efficiency and budget-neutral expanded access of bevacizumab-bvzr in mCRC and mNSCLC in Medicare. METHODS We developed a Medicare payer perspective simulation model of patients treated for mCRC and mNSCLC to estimate cost-savings from converting bevacizumab (originator) to bevacizumab-bvzr or alternative biosimilars such as bevacizumab-awwb, -maly, and -abcd. The target patient population receiving annual first-line systemic therapy was calculated using Medicare enrollment data, SEER cancer incidence rates in patients age ≥65, and an assumption that 39.3% and 77.2% of new diagnoses receive systemic therapy for mCRC and mNSCLC respectively based on recent evidence. 76.0% of systemically treated mCRC patients and 11.4% of incident mNSCLC patients were expected to be treated with bevacizumab-based regimens based on recent evidence. Costs were derived from the 2024 Average Sales Price (ASP). Results include per-patient per-month (PPPM) cost savings (vs. originator), total monthly savings in the cohort, and number needed to convert (NNC) to biosimilar to fund the treatment of an additional 100 patients. RESULTS PPPM savings from conversion to bevacizumab-bvzr were $4,205 in mCRC and $8,410 in mNSCLC. In 100% conversion scenarios, full cohort monthly savings were $27,664,432 in mCRC (n = 6,579) and $32,319,323 in mNSCLC (n = 3,843), respectively. At 100% conversion, monthly savings from biosimilar conversion could fund up to 13,887 additional mCRC patient-months of treatment with bevacizumab-bvzr + FOLFOX, and up to 8,959 additional mNSCLC patient-months of treatment with bevacizumab-bvzr + paclitaxel + carboplatin. In mCRC and mNSCLC the biosimilar NNC from the originator was 47 and 43, respectively. The biosimilar NNC from other biosimilars ranged from 60-4,564 and 55-4,422 for mCRC and NSCLC, respectively. CONCLUSION In the first cost-efficiency and expanded access study of biosimilar bevacizumab in mCRC and mNSCLC, we find that bevacizumab-bvzr-based regimens can result in substantial cost savings relative to originator-based first line treatment in Medicare. These cost savings could be reinvested to treat a substantial number of additional patients with mCRC or mNSCLC, or fund other costs of care in Medicare, on a budget-neutral basis.
Collapse
Affiliation(s)
- Joshua A Roth
- Pfizer, New York, NY, USA
- School of Pharmacy, University of Washington, Seattle, WA, USA
| | | | | | | |
Collapse
|
2
|
Gastón DC, Evelin B, Josefina PD, Lorena M, Melchora FC, Angélica RM, Adrián MM. Bevacizumab and cytostatics induce oxidative changes in the submandibular gland of male rats. Arch Oral Biol 2025; 174:106248. [PMID: 40220572 DOI: 10.1016/j.archoralbio.2025.106248] [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: 10/21/2024] [Revised: 02/26/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025]
Abstract
OBJECTIVE To investigate whether the administration of bevacizumab in combination with cytostatics could cause an oxidative response in the submandibular gland of an animal model. MATERIALS AND METHODS 48 Male Wistar rats (350-400 g) were used. Group 1: Control; Group 2: 5-Flourouracil + Leucovorin calcium (Intraperitoneal injection for five consecutive days with 20 mg/kg+10 mg/kg); Group 3: Bevacizumab, two intraperitoneal doses of 0.2 mg/kg on days 1 and 15; Group 4: Oxaliplatin, one intraperitoneal dose of 25 mg/kg on days 1 and 15; Group 5: Bevacizumab+Oxaliplatin+ 5-Fluorouracil+Leucovorin calcium, single intraperitoneal dose of 20 mg/kg, 10 mg/kg, 0.2 mg/kg, and 25 mg/kg on days 1 and 15; Group 6: pair-fed group without drugs. In submandibular gland homogenates, Uric Acid, Lipid Peroxides, Aqueous Peroxides, and Superoxide Dismutase activity were measured. RESULTS Uric Acid in Groups 1, 3, and 6 were higher than in cytostatic groups (p < 0.02, 0.02, and 0.001). Lipid Peroxides and Aqueus Peroxides were similar. Superoxide Dismutase Activity was higher in Groups 2, 3, 4, 5, and 6 compared to Group 1 (p < 0.0001). In Group 3, Superoxide Dismutase Activity was lower than in cytostatic-treated groups but higher than Group 6 (p < 0.001). Group 2 showed higher activity compared to Groups 3 and 6 (p < 0.0001). CONCLUSIONS Cytostatic treatment exerted a pro-oxidant effect at the acinar level. Conversely, bevacizumab may promote an antioxidant glandular response. Further research into these treatments' effects on the stomatognathic system is crucial for improving quality of life in patients undergoing anti-tumour therapy.
Collapse
Affiliation(s)
| | - Bachmeier Evelin
- Chair of Physiology, Faculty of Dentistry, National University of Córdoba, Argentina
| | | | - Moine Lorena
- Chair of Physiology, Faculty of Dentistry, National University of Córdoba, Argentina
| | | | - Rivoira María Angélica
- Chair of Biochemistry and Molecular Biology, Faculty of Medical Sciences, National University of Cordoba, Argentina
| | - Mazzeo Marcelo Adrián
- Chair of Physiology, Faculty of Dentistry, National University of Córdoba, Argentina.
| |
Collapse
|
3
|
Soler G, Grávalos C, Rivera F, Safont MJ, Virgili AC, Valladares-Ayerbes M, Ferreiro-Monteagudo R, Ortiz-Morales MJ, Polo-Marques E, Kornusova-Bersheva I, Soustre E, Yao W, Élez E. Predictive factors of efficacy for FTD/TPI plus bevacizumab in refractory metastatic colorectal cancer patients: a subanalysis of the Spanish cohort from the SUNLIGHT phase III trial. Clin Transl Oncol 2025:10.1007/s12094-025-03907-z. [PMID: 40338282 DOI: 10.1007/s12094-025-03907-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/13/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND The addition of bevacizumab to trifluridine-tipiracil (FTD/TPI) therapy improves outcomes in patients with refractory metastatic colorectal cancer (CRC). However, predictive factors of efficacy for FTD/TPI when used in combination with bevacizumab are not yet fully recognized. METHODS Patients included in the Spanish cohort of the SUNLIGHT trial were evaluated. The primary endpoint of this post-hoc exploratory subanalysis was overall survival (OS). Secondary endpoints included progression-free survival (PFS), disease control rate (DCR), safety and the assessment of potential predictive factors of efficacy for FTD/TPI plus bevacizumab, such as age, KRAS mutational status, the incidence of neutropenia, patient prognosis, and prior administration of bevacizumab. RESULTS A total of 115 patients were analysed, 58 receiving FTD/TPI and 57 FTD/TPI plus bevacizumab. The median OS was 8.5 vs. 10.6 months (p = 0.254), respectively, and the median PFS was 2.4 vs. 4.7 months (p < 0.0001), respectively. Severe neutropenia affected 20% more patients in the experimental arm than in the control arm (51% vs. 31%, respectively). The univariate analysis showed that a benefit of adding bevacizumab to FTD/TPI in terms of PFS was observed in all previously defined patient subgroups and was accompanied by DCR of 69% for FTD/TPI plus bevacizumab vs. 45% in arm of FTD/TPI alone. CONCLUSIONS The addition of bevacizumab to FTD/TPI tends to improve OS probably due to the reduced number of patients included in this cohort and significantly improved PFS in all patient subgroups with metastatic CRC. Prospective studies are needed to confirm these results and to find out additional predictive factors that help us to discriminate which patients would benefit most from FTD/TPI plus bevacizumab.
Collapse
Affiliation(s)
- Gemma Soler
- Medical Oncology Department, Institut Catalá d'Oncologia (ICO), Badalona·Applied Research Group in Oncology (B·ARGO), Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), De Can Ruti Camí de les Escoles s/n, 08916, Badalona, Barcelona, Spain.
| | | | - Fernando Rivera
- Marqués de Valdecilla University Hospital, IDIVAL, Santander, Spain
| | - María José Safont
- General University Hospital Consortium of Valencia, Valencia University, Valencia, Spain
| | | | | | | | | | | | | | - Elsa Soustre
- Department of Medical Affairs, Servier Laboratories, Madrid, Spain
| | - Weiyu Yao
- Department of Biostatistics, Servier Pharmaceuticals LLC, Boston, MA, USA
| | - Elena Élez
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
4
|
Lecomte T, Giraudeau B, Phelip JM, Tournigand C, Ducreux M, Tougeron D, Lepage C, Mineur L, Laplaige P, Desgrippes R, Artru P, Borg C, Jary M, Bouché O, Metges JP, Guimbaud R, Aparicio T, Foubert F, Hautefeuille V, Muller M, Bouhier-Leporrier K, Darrius R, Lobet S, Monmousseau F, Bejan-Angoulvant T, Paintaud G, Ternant D. Bevacizumab-based chemotherapy adaptive to pharmacokinetic of bevacizumab in first-line treatment of patients with unresectable metastatic colorectal cancer: A double-blind, multicenter, randomized phase III trial study (PHARBEVACOL trial). Dig Liver Dis 2025; 57:624-630. [PMID: 40044552 DOI: 10.1016/j.dld.2025.02.002] [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: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 04/27/2025]
Abstract
Bevacizumab shows inter-individual pharmacokinetic variability, with an exposure-response relationship in metastatic colorectal cancer (mCRC) patients. This study explores whether a double dose of bevacizumab, compared to a standard dose, increases efficacy in mCRC patients treated with bevacizumab-based chemotherapy as first-line therapy and who have a low initial trough concentration of bevacizumab. PHARBEVACOL is a multicenter, randomized, double-blind, two-parallel group trial. All patients will receive first-line bi-weekly 5 mg/kg bevacizumab-based chemotherapy and those with low initial bevacizumab concentrations (≤15.5 mg/L) will be randomized to either continue the standard dose (5 mg/kg every 14 days) or receive a double dose (10 mg/kg every 14 days). The primary objective is to evaluate the effect of doubling dose on progression-free survival (PFS). During a screening phase, the first serum trough concentration will be measured on day 14, before the second infusion of bevacizumab. We hypothesize a 40 % PFS in the control group at 9 months versus 60 % in the study group, corresponding to a hazard ratio of 0.56. With 80 % power, a 5 % two-sided type I error, and a minimum 12-month follow-up, 116 patients need to be included. Since only 50 % of screened patients will be eligible for randomization, approximately 244 patients will be screened. Recruitment is scheduled to begin in February 2025.
Collapse
Affiliation(s)
- Thierry Lecomte
- Service d'Hépato-gastroentérologie et de cancérologie digestive, CHU de Tours, Tours, France; INSERM UMR 1069, N2COx "Niche, Nutrition, Cancer and Oxidative Metabolism", Université de Tours, Tours, France.
| | | | - Jean-Marc Phelip
- Service d'Hépato-gastroentérologie et de cancérologie digestive, CHU Saint Etienne, Saint Etienne, France
| | | | - Michel Ducreux
- Service d'Oncologie digestive, Institut Gustave Roussy, Villejuif, France
| | - David Tougeron
- Service d'Hépato-gastroentérologie, CHU de Poitiers, Poitiers, France
| | - Côme Lepage
- Service d'Hépato-gastroentérologie et de cancérologie digestive, CHU de Dijon, Dijon, France
| | | | | | - Romain Desgrippes
- Service d'Hépato-gastroentérologie et de cancérologie digestive, CH de Saint-Malo, Saint-Malo, France
| | | | - Christophe Borg
- Service d'Oncologie Médicale, CHU de Besançon, Besançon, France
| | - Marine Jary
- Service de chirurgie digestive, CHU Estaing, Clermont-Ferrand, France
| | - Olivier Bouché
- Service d'Hépato-gastroentérologie et de cancérologie digestive, CHU de Reims, Reims, France
| | | | - Rosine Guimbaud
- Service d'oncologie médicale, CHU de Toulouse, Toulouse, France
| | - Thomas Aparicio
- Service d'Hépato-gastroentérologie et de cancérologie digestive, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Fanny Foubert
- Service d'Hépato-gastroentérologie et de cancérologie digestive, CHU de Nantes, Nantes, France
| | - Vincent Hautefeuille
- Service d'Hépato-gastroentérologie et de cancérologie digestive, CHU d'Amiens, Amiens, France
| | - Marie Muller
- Service d'Hépato-gastroentérologie et de cancérologie digestive, CHU de Nancy, Nancy, France
| | | | - Rémi Darrius
- Service d'Hépato-gastroentérologie et de cancérologie digestive, CH de Colmar, Colmar, France
| | - Sarah Lobet
- INSERM UMR 1069, N2COx "Niche, Nutrition, Cancer and Oxidative Metabolism", Université de Tours, Tours, France
| | | | - Théodora Bejan-Angoulvant
- Service de Pharmacologie Médicale, CHU de Tours, Tours, France; UMR 1327, ISCHEMIA, Membrane signalling and inflammation in reperfusion injuries, INSERM, Université de Tours, Tours, France; Plateforme Recherche, Centre Pilote de suivi Biologique des traitements par Anticorps, CHU de Tours, Tours, France
| | - Gilles Paintaud
- Service de Pharmacologie Médicale, CHU de Tours, Tours, France; UMR 1327, ISCHEMIA, Membrane signalling and inflammation in reperfusion injuries, INSERM, Université de Tours, Tours, France; Plateforme Recherche, Centre Pilote de suivi Biologique des traitements par Anticorps, CHU de Tours, Tours, France
| | - David Ternant
- Service de Pharmacologie Médicale, CHU de Tours, Tours, France; UMR 1327, ISCHEMIA, Membrane signalling and inflammation in reperfusion injuries, INSERM, Université de Tours, Tours, France; Plateforme Recherche, Centre Pilote de suivi Biologique des traitements par Anticorps, CHU de Tours, Tours, France
| |
Collapse
|
5
|
Pumpalova YS. Systemic Therapy for Metastatic Colon Cancer: New Frontiers. Clin Colon Rectal Surg 2025; 38:229-236. [PMID: 40291998 PMCID: PMC12020547 DOI: 10.1055/s-0044-1787826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
We have made steady gains in improving overall survival in patients with metastatic, unresectable, colon cancer in the last 5 to 10 years. The backbone of systemic treatment for most patients remains combination chemotherapy, but the field is becoming increasingly biomarker driven, with exciting new targeted therapies on the horizon. This review is organized in sections corresponding to currently relevant biomarkers in colon cancer and will summarize first-, second-, and third-line standard of care for metastatic, unresectable, colon cancer. The last section is intended to introduce the reader to promising agents and novel therapeutic strategies currently under investigation.
Collapse
Affiliation(s)
- Yoanna S. Pumpalova
- Division of Hematology and Oncology, Department of Internal Medicine, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
6
|
Ogawa C, Yatabe M, Nagayama Y, Nakadate K, Adachi S, Inoue M. Impact of bevacizumab preparation conditions on inline-filter blockage. J Oncol Pharm Pract 2025:10781552251336176. [PMID: 40270130 DOI: 10.1177/10781552251336176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
PurposeThis study aimed to investigate the effects of preparation conditions and external factors, particularly ultraviolet (UV) exposure, on inline-filter clogging in liquid monoclonal antibody (l-mAb) formulations. Bevacizumab (BmAb) was used as a model drug to address the critical question of how UV exposure impacts protein stability and filter performance during clinical administration.MethodsBevacizumab solutions were exposed to UV-C (254 nm) light to evaluate its effects on protein stability and filter clogging. Scanning electron microscopy (SEM) was used to visualize protein aggregates on the filter surface. Dynamic light scattering (DLS) analysis measured particle size distribution and average particle size. To assess the effectiveness of light shielding, additional samples were prepared under conditions that minimized UV exposure.ResultsUV-C exposure induced significant precipitate formation, with SEM images revealing protein aggregates on the filter surface. DLS analysis showed a broader particle size distribution and an increase in average particle size (18.1 nm) in UV-exposed solutions compared to non-exposed controls. Light shielding effectively suppresses precipitate formation, maintaining the stability of BmAb solutions and preventing inline-filter clogging.ConclusionUV-C destabilizes BmAb solutions, leading to protein aggregation and inline-filter clogging. Light shielding was identified as an effective strategy to maintain the stability and safety of l-mAb formulations, particularly in settings where prolonged exposure to environmental light is possible. These findings emphasize the importance of implementing protective measures against UV exposure and call for further research on additional environmental factors and optimized protocols to enhance l-mAb formulation reliability.
Collapse
Affiliation(s)
- Chiaki Ogawa
- Department of Pharmacy, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Megumi Yatabe
- Department of Pharmacy, National Hospital Organization, Saitama Hospital, Saitama, Japan
| | - Yoshiyuki Nagayama
- Department of Pharmacy, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | | | | | | |
Collapse
|
7
|
Ren J, Yan G, Yang L, Kong L, Guan Y, Sun H, Liu C, Liu L, Han Y, Wang X. Cancer chemoprevention: signaling pathways and strategic approaches. Signal Transduct Target Ther 2025; 10:113. [PMID: 40246868 PMCID: PMC12006474 DOI: 10.1038/s41392-025-02167-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025] Open
Abstract
Although cancer chemopreventive agents have been confirmed to effectively protect high-risk populations from cancer invasion or recurrence, only over ten drugs have been approved by the U.S. Food and Drug Administration. Therefore, screening potent cancer chemopreventive agents is crucial to reduce the constantly increasing incidence and mortality rate of cancer. Considering the lengthy prevention process, an ideal chemopreventive agent should be nontoxic, inexpensive, and oral. Natural compounds have become a natural treasure reservoir for cancer chemoprevention because of their superior ease of availability, cost-effectiveness, and safety. The benefits of natural compounds as chemopreventive agents in cancer prevention have been confirmed in various studies. In light of this, the present review is intended to fully delineate the entire scope of cancer chemoprevention, and primarily focuses on various aspects of cancer chemoprevention based on natural compounds, specifically focusing on the mechanism of action of natural compounds in cancer prevention, and discussing in detail how they exert cancer prevention effects by affecting classical signaling pathways, immune checkpoints, and gut microbiome. We also introduce novel cancer chemoprevention strategies and summarize the role of natural compounds in improving chemotherapy regimens. Furthermore, we describe strategies for discovering anticancer compounds with low abundance and high activity, revealing the broad prospects of natural compounds in drug discovery for cancer chemoprevention. Moreover, we associate cancer chemoprevention with precision medicine, and discuss the challenges encountered in cancer chemoprevention. Finally, we emphasize the transformative potential of natural compounds in advancing the field of cancer chemoprevention and their ability to introduce more effective and less toxic preventive options for oncology.
Collapse
Affiliation(s)
- Junling Ren
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Guangli Yan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
| | - Ling Kong
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Yu Guan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Hui Sun
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
| | - Chang Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Lei Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Ying Han
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Xijun Wang
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China.
| |
Collapse
|
8
|
Ozaydin S, Dogan S, Ozaydin IY, Aycicek A, Ata R, Mahmut Z, Guvenc U, Besik C, Demirali O. Colorectal cancer in children: an evaluation of the existing literature based on the 11-year experience of a single center. Pediatr Surg Int 2025; 41:116. [PMID: 40232502 PMCID: PMC12000104 DOI: 10.1007/s00383-025-06020-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
PURPOSE Children's risk of developing colorectal cancer (CRC) is relatively low. In this report, we present our experience with CRC in the pediatric age group, together with an extensive review of the literature. METHODS Between the years 2013 and 2024, a total of five patients diagnosed with CRC underwent treatment at the Department of Pediatric Surgery in our tertiary hospital. A retrospective evaluation was conducted on patients' charts, encompassing demographics, admission symptoms, patient and family histories, laboratory and radiologic findings, operative and pathology reports, genetic and molecular study results, treatment protocols, and follow-up data. RESULTS There were three males and two females, with a mean age of 13.5 ± 2.5 years. The primary sites were the sigmoid and rectosigmoid. Two patients were diagnosed with mucinous adenocarcinoma based on histopathological examination. Among the three remaining patients, one presented with signet ring cells, one displayed moderately differentiated adenocarcinoma characteristics, and one exhibited well-differentiated adenocarcinoma characteristics. At the time of the most recent follow-up, two patients have demonstrated survival. CONCLUSION The two main factors contributing to poor survival in pediatric CRC were concluded to be at an advanced stage during diagnosis and having an aggressive histologic subtype. Including CRC in the preliminary diagnosis list is essential for an early diagnosis in the pediatric age group.
Collapse
Affiliation(s)
- Seyithan Ozaydin
- Department of Pediatric Surgery, Kanuni Sultan Suleyman Training and Research Hospital/Health Sciences University, Istanbul, Turkey.
| | - Sumeyra Dogan
- Department of Pediatric Radiology, Kanuni Sultan Suleyman Training and Research Hospital/Health Sciences University, Istanbul, Turkey
| | - Ipek Yildiz Ozaydin
- Department of Pathology, Kanuni Sultan Suleyman Training and Research Hospital/Health Sciences University, Istanbul, Turkey
| | - Ali Aycicek
- Department of Pediatric Oncology, Kanuni Sultan Suleyman Training and Research Hospital/Health Sciences University, Istanbul, Turkey
| | - Rabia Ata
- Department of Pediatric Surgery, Kanuni Sultan Suleyman Training and Research Hospital/Health Sciences University, Istanbul, Turkey
| | - Zahit Mahmut
- Department of Pediatric Surgery, Kanuni Sultan Suleyman Training and Research Hospital/Health Sciences University, Istanbul, Turkey
| | - Unal Guvenc
- Department of Pediatric Surgery, Kanuni Sultan Suleyman Training and Research Hospital/Health Sciences University, Istanbul, Turkey
| | - Cemile Besik
- Department of Pediatric Surgery, Kanuni Sultan Suleyman Training and Research Hospital/Health Sciences University, Istanbul, Turkey
| | - Oyhan Demirali
- Department of Pediatric Surgery, Kanuni Sultan Suleyman Training and Research Hospital/Health Sciences University, Istanbul, Turkey
| |
Collapse
|
9
|
Yildirim ME, Karadurmuş N, Ökten İN, Türk HM, Urakçı Z, Arslan Ç, Çelik S, Dane F, Şendur MAN, Bilir C, Karabulut B, Cicin İ, Çubukçu E, Karaca M, Ozcelik M, Artaç M, Tanrikulu E, Alacacioglu A, Açıkgöz Ö, Öven B, Geredeli Ç, Çil T, Harputluoğlu H, Kefeli U, Bozkurt O, Tural D, Sakin A, Yalçın Ş, Gumus M. Real-world treatment outcomes from nationwide Onco-colon Turkey registry in RAS wild-type patients treated with biologics second-line mCRC. J Oncol Pharm Pract 2025; 31:404-411. [PMID: 38613329 DOI: 10.1177/10781552241241004] [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: 04/14/2024]
Abstract
Backgrounds and ObjectivesColorectal cancer is one of the leading causes of mortality both globally and in our country. In Turkey, we conducted a multicenter investigation into the effectiveness of second-line treatments and real-life data for patients with RAS wild-type metastatic colorectal cancer (NCT04757311).Materials and MethodsIn this retrospective analysis, records from 28 centers were collected, and histopathological, molecular, and clinical characteristics were documented. Patients were categorized into groups based on their second-line biological treatments: anti-EGFR (Group A and Group B, panitumumab and cetuximab) and anti-VEGF (Group C, bevacizumab and aflibercept). They were then compared within these groups.ResultsA total of 588 patients with documented RAS wild-type status were evaluated. The median OS was 15.7, 14.3 and 14.7 months in Group A, Group B and Group C, respectively (p = 0.764). The median PFS of the patients in second-line setting that received panitumumab, cetuximab and bevacizumab/aflibercept were 7.8, 6.6 and 7.4 months, respectively (p = 0.848).ConclusionAccording to the results of our real-life data study, there is no significant difference in efficiency between the combination of biological agent and chemotherapy used in the second-line treatments.
Collapse
Affiliation(s)
- Mahmut Emre Yildirim
- Medical Oncology Department, İstanbul Dr. Lütfi Kırdar Kartal City Hospital, Istanbu, Türkiye
| | - Nuri Karadurmuş
- Medical Oncology Department, Gulhane Training and Research Hospital, Ankara, Türkiye
| | - İlker Nihat Ökten
- Medical Oncology Department, Medeniyet University Goztepe Training and Research Hospital, Istanbul, Türkiye
| | - Hacı Mehmet Türk
- Department of Medical Oncology, Bezmialem Vakif University Faculty of Medicine, Istanbul, Türkiye
| | - Zuhat Urakçı
- Department of Medical Oncology, Dicle University Medical Faculty, Diyarbakir, Türkiye
| | - Çağatay Arslan
- Medical Oncology, Bahcesehir Universitesi Tip Fakultesi, Istanbul, Türkiye
| | - Sinemis Çelik
- Medical Oncology Department, Istanbul Oncology Hospital, Istanbul, Türkiye
| | - Faysal Dane
- Department of Internal Medicine, Division of Medical Oncology, Marmara University School of Medicine, Istanbul, Türkiye
| | | | - Cemil Bilir
- Medical Oncology Department, Sakarya University Training and Research Hospital, Sakarya, Türkiye
| | - Bülent Karabulut
- Medical Oncology, Ege University Faculty of Medicine, Izmir, Türkiye
| | - İrfan Cicin
- Department of Internal Medicine, Division of Oncology, Trakya University Faculty of Medicine, Edirne, Türkiye
| | - Erdem Çubukçu
- Faculty of Medicine, Medical Oncology, Uludag University, Bursa, Türkiye
| | - Mustafa Karaca
- Medical Oncology Department, Antalya Training and Research Hospital, Antalya, Türkiye
| | - Melike Ozcelik
- Department of Oncology, Umraniye Training and Research Hospital, Istanbul, Türkiye
| | - Mehmet Artaç
- Department of Medical Oncology, Necmettin Erbakan University Medical Faculty, Konya, Türkiye
| | - Eda Tanrikulu
- Medical Oncology, Istanbul Haydarpasa Numune Training and Research Hospital, Istanbul, Türkiye
| | - Ahmet Alacacioglu
- Medical Oncology Department, Ministry of Health İzmir Katip Çelebi University Atatürk Education and Research Hospital, Izmir, Türkiye
| | - Özgür Açıkgöz
- Medical Oncology Department, Istanbul Medipol University, İstanbul, Türkiye
| | - Başak Öven
- Medical Oncology Department, Yeditepe University Hospital, Istanbul, Türkiye
| | - Çağlayan Geredeli
- Department of Medical Oncology, Okmeydani Training and Research Hospital, Istanbul, Türkiye
| | - Timucin Çil
- Department of Medical Oncology, University of Health Sciences, Adana City Education and Research Hospital, Adana, Türkiye
| | | | - Umut Kefeli
- Medical Oncology, Kocaeli University School of Medicine, Kocaeli, Türkiye
| | - Oktay Bozkurt
- Medical Oncology Department, Erciyes Universitesi, Kayseri, Türkiye
| | - Deniz Tural
- Medical Oncology, Istanbul Bakirkoy Dr Sadi Konuk Training and Research Hospital, Istanbul, Türkiye
| | - Abdullah Sakin
- Department of Medical Oncology, Yuzuncu Yil University, Van, Türkiye
| | - Şuayip Yalçın
- Department of Medical Oncology, Hacettepe University Faculty of Medicine, Ankara, Türkiye
| | - Mahmut Gumus
- Department of Medical Oncology, Istanbul Medeniyet University Faculty of Medicine, Istanbul, Türkiye
| |
Collapse
|
10
|
Vonica RC, Butuca A, Morgovan C, Pumnea M, Cipaian RC, Frum A, Dobrea CM, Vonica-Tincu AL, Pacnejer AM, Ghibu S, Batar F, Gligor FG. Bevacizumab-Insights from EudraVigilance Database on the Assessments of the Safety Profile of Monoclonal Antibodies Used as Targeted Cancer Treatment. Pharmaceuticals (Basel) 2025; 18:501. [PMID: 40283938 PMCID: PMC12030381 DOI: 10.3390/ph18040501] [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: 03/10/2025] [Revised: 03/22/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Worldwide, colon cancer is a major cause of cancer-related mortality, with an increasing incidence influenced by genetic, environmental, and lifestyle factors. Despite advances in diagnosis and personalized treatments, challenges remain in improving patient prognosis, particularly in metastatic colorectal cancer (mCRC). Bevacizumab (BEV), a monoclonal antibody, is widely used in colorectal cancer treatment. This study aimed to analyze adverse events associated with BEV compared with other therapies based on data from the EudraVigilance (EV) database. Methods: A descriptive and disproportionality analysis was conducted on signals reported in the EV database related to BEV. The study included comparisons with other antineoplastic treatments, such as chemotherapy, targeted therapy, and immunotherapy. Patient demographics, severity of adverse drug reactions (ADRs), and distribution patterns were analyzed to assess the safety profile of BEV in colorectal cancer treatment. Results: The majority of the signals for BEV were from patients aged 18-64 years (39.42%) and 65-85 years (34.08%). Hypertension, thromboembolism, proteinuria, and gastrointestinal disorders have been the most frequently reported. Serious ADRs, including gastrointestinal perforations, hemorrhage, and arterial thromboembolism, were observed in 93.74% of Individual Case Safety Reports. BEV was associated with a higher likelihood of vascular and endocrine disorders compared with chemotherapy and other targeted therapies. Immunotherapy was linked to increased immunological ADRs, while BEV demonstrated fewer immune-related toxicities. Conclusions: Continuous monitoring is necessary to optimize patient management, particularly in elderly patients or those with cardiovascular comorbidities. Understanding BEV's safety profile allows for better personalization of treatment strategies, minimizing risks while enhancing therapeutic outcomes.
Collapse
Affiliation(s)
- Razvan Constantin Vonica
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Anca Butuca
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Claudiu Morgovan
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Manuela Pumnea
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Remus Calin Cipaian
- Clinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania;
- County Clinical Emergency Hospital of Sibiu, 2–4 Corneliu Coposu Str., 550245 Sibiu, Romania
| | - Adina Frum
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Carmen Maximiliana Dobrea
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Andreea Loredana Vonica-Tincu
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Aliteia-Maria Pacnejer
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timişoara, Romania
| | - Steliana Ghibu
- Department of Pharmacology, Physiology and Pathophysiology, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Florina Batar
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Felicia Gabriela Gligor
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| |
Collapse
|
11
|
Chitoran E, Rotaru V, Stefan DC, Gullo G, Simion L. Blocking Tumoral Angiogenesis VEGF/VEGFR Pathway: Bevacizumab-20 Years of Therapeutic Success and Controversy. Cancers (Basel) 2025; 17:1126. [PMID: 40227654 PMCID: PMC11988089 DOI: 10.3390/cancers17071126] [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: 01/07/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
The "angiogenesis switch"-defined as the active process by which solid tumors develop their own circulation-plays an important role in both tumoral growth and propagation. As the malignant tumor grows and reaches a critical size, the metabolic needs as a function of an ever-increasing distance to the nearest emergent blood vessel, can no longer be covered by the microenvironment of the peritumoral tissue. Although a relatively discrete process, the "angiogenic switch" acts as a limiting stage of tumoral development present from the avascular hyperplasia phase to the vascularized neoplastic phase, providing support for tumor expansion and metastasis. Over time, research has focused on blocking the angiogenetic pathways (such as VEGF/VEGFR signaling axis) leading to the development of targeted therapeutic agents such as Bevacizumab. Objectives: We conducted a review of the molecular principles of tumoral angiogenesis and we tried to follow the history of Bevacizumab from its first approval for human usage 20 years ago to current days, focusing on the impact this agent had in solid tumor therapy. A comprehensive review of clinical trials pertaining to Bevacizumab (from the era of the preclinic trials leading to approval for human usage, to the more recent randomized trial focusing on combination targeted therapy) further details the role of this drug. We aimed to establish if this ancient drug continues to have a place in modern oncology. Conclusions: Bevacizumab, one of the first drugs targeting tumoral microenvironment, remains one of the most important oncologic agents blocking the VEGF/VEGFR angiogenic pathway. otherwise, history of 20 years marked by numerous controversies (ranging from methodological errors of clinical trials to withdrawal of approval for human usage in breast cancer patients, from discussions about severe side effects to resistance to therapy and limited efficacity), Bevacizumab continues to provide an optimal therapeutic option for many solid tumors that previously had little to no means of treatment, improving otherwise bleak outcomes. Even in the era of personalized precision oncology, Bevacizumab continues to be a key element in many therapeutic regimens both as monotherapy and in combination with newer targeted agents.
Collapse
Affiliation(s)
- Elena Chitoran
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Vlad Rotaru
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Daniela-Cristina Stefan
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Giuseppe Gullo
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, University of Palermo, 90146 Palermo, Italy
| | - Laurentiu Simion
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| |
Collapse
|
12
|
Chen Y, Zheng Y, Wu J, Ye R, Jia H, Zhou Z, Chen W, Xu L, Zhang Y, Zheng M. Neoadjuvant chemotherapy combined with bevacizumab for resectable colorectal liver metastasis with risk factors for recurrence: a multicenter real-world study. Ther Adv Med Oncol 2025; 17:17588359251328457. [PMID: 40151550 PMCID: PMC11948564 DOI: 10.1177/17588359251328457] [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] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Background Currently, guidelines prohibit the addition of targeted drugs in neoadjuvant chemotherapy (NAC) for initially resectable colorectal liver metastasis (CRLM). Objective Nevertheless, efficacy data of NAC combined with bevacizumab (Bev) for initially resectable CRLM with risk factors for recurrence (RFR) are lacking. Designs We conducted a multicenter real-world cohort study to retrospectively analyze the efficacy and feasibility of NAC combined with Bev for CRLM with RFR. Methods The patients were divided into the NAC alone group and NAC combined with the Bev group. We designated progression-free survival (PFS), objective response rate (ORR), and overall survival (OS) as the outcomes. Kaplan-Meier, Cox proportional hazards regression models, and subgroup analysis were utilized. RFR was a clinical risk score of 3-5. Subgroup analysis was applied to explore which subgroup was more suitable for NAC combined with Bev. Results Between 2015 and 2020, this multicenter real-world study encompassed 335 CRLM patients from six medical centers who underwent curative hepatectomy following NAC. Two hundred seventeen patients were in the NAC alone group, and 118 received NAC combined with Bev. The NAC alone group exhibited an ORR of 51.15%, compared to 66.95% in the NAC combined with Bev (p = 0.005). The R0 resection rates achieved 91.71% for the NAC alone group and 94.92% for the NAC combined with Bev (p = 0.276). Three-year PFS rate was 27.6% for NAC alone and 41.5% for the NAC combined with the Bev group (p = 0.006). Furthermore, the 3-year OS was calculated to be 57.0% for the NAC alone and 66.7% for the NAC combined with Bev patients (p = 0.079). Conclusion For initially resectable CRLM patients with RFR, NAC combined with Bev exhibited a higher ORR and longer PFS. Chinese clinical trial registry ChiCTR2400082966.
Collapse
Affiliation(s)
- Yizhen Chen
- Fujian Key Laboratory of Geriatrics Diseases, Department of Geriatric Medicine, Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, Fujian, China
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Yuanyuan Zheng
- Fujian Key Laboratory of Geriatrics Diseases, Department of Geriatric Medicine, Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, Fujian, China
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Jia Wu
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Rong Ye
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Hangdong Jia
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhenyuan Zhou
- Anorectal Surgical Department, Hangzhou Red Cross Hospital, Hangzhou, Zhenjiang, China
| | - Weijie Chen
- Department of Hepatobiliary Pancreatic Surgery, The First Hospital of PuTian City, Putian, Fujian, China
| | - Linwei Xu
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Yuhua Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Ming Zheng
- Fujian Key Laboratory of Geriatrics Diseases, Department of Geriatric Medicine, Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, School of Medicine, Fuzhou University, Fuzhou, Fujian 350001, China
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian 350001, China
| |
Collapse
|
13
|
Razzaq S, Fatima I, Moafian Z, Rahdar A, Fathi-Karkan S, Kharaba Z, Shirzad M, Khan A, Pandey S. Nanomedicine innovations in colon and rectal cancer: advances in targeted drug and gene delivery systems. Med Oncol 2025; 42:113. [PMID: 40097759 DOI: 10.1007/s12032-025-02670-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Nanotechnology has revolutionized cancer diagnostics and therapy, offering unprecedented possibilities to overcome the constraints of conventional treatments. This study provides a detailed overview of the current progress and difficulties in the creation of nanostructured materials, with a specific emphasis on their use in drug and gene delivery systems. The study examines tactics that attempt to improve the effectiveness and safety of chemotherapeutic drugs such as doxorubicin (Dox) by focusing on the potential of antibody-drug conjugates and functionalized nanoparticles. Moreover, it clarifies the challenges encountered in administering nanoparticles orally for gastrointestinal treatments, emphasizing the crucial physicochemical properties that affect their behavior in the gastrointestinal system. This study highlights the transformational potential of nanostructured materials in precision oncology by examining advanced breakthroughs such cell membrane-camouflaged nanoparticles and inorganic nanoparticles designed for gastrointestinal disorders. The text investigates the processes involved in the absorption of nanoparticles and their destruction in lysosomes, revealing the many methods in which enterocytes take up these particles. This study strongly supports the use of advanced nanoparticle-based methods to reduce the harmful effects on the whole body and improve the effectiveness of therapy, based on a thorough examination of current experiments on animals and humans. The main objective of this paper is to provide a fundamental comprehension that will stimulate more investigation and practical use in the field of cancer nanomedicine, advancing its boundaries.
Collapse
Affiliation(s)
- Sobia Razzaq
- School of Pharmacy, University of Management and Technology, Lahore, Punjab, Pakistan
| | - Iqra Fatima
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Zeinab Moafian
- Department of Chemistry and Biochemistry, University of Delaware, Newark, USA
| | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol, 538-98615, Iran.
| | - Sonia Fathi-Karkan
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran.
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 9414974877, Iran.
| | - Zelal Kharaba
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, UAE
| | - Maryam Shirzad
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ahmad Khan
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| | - Sadanand Pandey
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, 173229, Himachal Pradesh, India.
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk, 38541, Republic of Korea.
| |
Collapse
|
14
|
Liu B, Polack M, Coudray N, Claudio Quiros A, Sakellaropoulos T, Le H, Karimkhan A, Crobach ASLP, van Krieken JHJM, Yuan K, Tollenaar RAEM, Mesker WE, Tsirigos A. Self-supervised learning reveals clinically relevant histomorphological patterns for therapeutic strategies in colon cancer. Nat Commun 2025; 16:2328. [PMID: 40057490 PMCID: PMC11890774 DOI: 10.1038/s41467-025-57541-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
Self-supervised learning (SSL) automates the extraction and interpretation of histopathology features on unannotated hematoxylin-eosin-stained whole slide images (WSIs). We train an SSL Barlow Twins encoder on 435 colon adenocarcinoma WSIs from The Cancer Genome Atlas to extract features from small image patches (tiles). Leiden community detection groups tiles into histomorphological phenotype clusters (HPCs). HPC reproducibility and predictive ability for overall survival are confirmed in an independent clinical trial (N = 1213 WSIs). This unbiased atlas results in 47 HPCs displaying unique and shared clinically significant histomorphological traits, highlighting tissue type, quantity, and architecture, especially in the context of tumor stroma. Through in-depth analyses of these HPCs, including immune landscape and gene set enrichment analyses, and associations to clinical outcomes, we shine light on the factors influencing survival and responses to treatments of standard adjuvant chemotherapy and experimental therapies. Further exploration of HPCs may unveil additional insights and aid decision-making and personalized treatments for colon cancer patients.
Collapse
Affiliation(s)
- Bojing Liu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Karolinska, Sweden
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY, USA
| | - Meaghan Polack
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Nicolas Coudray
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | | | - Theodore Sakellaropoulos
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY, USA
| | - Hortense Le
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY, USA
| | - Afreen Karimkhan
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | | | - J Han J M van Krieken
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ke Yuan
- Department of Computing Science, University of Glasgow, Glasgow, United Kingdom
- School of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Rob A E M Tollenaar
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Wilma E Mesker
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Aristotelis Tsirigos
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
15
|
Wang F, Chen G, Zhang Z, Yuan Y, Wang Y, Gao Y, Sheng W, Wang Z, Li X, Yuan X, Cai S, Ren L, Liu Y, Xu J, Zhang Y, Liang H, Wang X, Zhou A, Ying J, Li G, Cai M, Ji G, Li T, Wang J, Hu H, Nan K, Wang L, Zhang S, Li J, Xu R. The Chinese Society of Clinical Oncology (CSCO): Clinical guidelines for the diagnosis and treatment of colorectal cancer, 2024 update. Cancer Commun (Lond) 2025; 45:332-379. [PMID: 39739441 PMCID: PMC11947620 DOI: 10.1002/cac2.12639] [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: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 01/02/2025] Open
Abstract
The 2024 updates of the Chinese Society of Clinical Oncology (CSCO) Clinical Guidelines for the diagnosis and treatment of colorectal cancer emphasize standardizing cancer treatment in China, highlighting the latest advancements in evidence-based medicine, healthcare resource access, and precision medicine in oncology. These updates address disparities in epidemiological trends, clinicopathological characteristics, tumor biology, treatment approaches, and drug selection for colorectal cancer patients across diverse regions and backgrounds. Key revisions include adjustments to evidence levels for intensive treatment strategies, updates to regimens for deficient mismatch repair (dMMR)/ microsatellite instability-high (MSI-H) patients, proficient mismatch repair (pMMR)/ microsatellite stability (MSS) patients who have failed standard therapies, and rectal cancer patients with low recurrence risk. Additionally, recommendations for digital rectal examination and DNA polymerase epsilon (POLE)/ DNA polymerase delta 1 (POLD1) gene mutation testing have been strengthened. The 2024 CSCO Guidelines are based on both Chinese and international clinical research, as well as expert consensus, ensuring their relevance and applicability in clinical practice, while maintaining a commitment to scientific rigor, impartiality, and timely updates.
Collapse
Affiliation(s)
- Feng Wang
- Department of Medical OncologySun Yat‐sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical SciencesGuangzhouGuangdongP. R. China
| | - Gong Chen
- Department of Colorectal SurgerySun Yat‐sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerGuangzhouGuangdongP. R. China
| | - Zhen Zhang
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiP. R. China
| | - Ying Yuan
- Department of Medical OncologyThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Yi Wang
- Department of RadiologyPeking University People's HospitalBeijingP. R. China
| | - Yuan‐Hong Gao
- Department of Radiation OncologySun Yat‐sen University Cancer Centre, The State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Weiqi Sheng
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiP. R. China
| | - Zixian Wang
- Department of Medical OncologySun Yat‐sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical SciencesGuangzhouGuangdongP. R. China
| | - Xinxiang Li
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiP. R. China
| | - Xianglin Yuan
- Department of OncologyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubeiP. R. China
| | - Sanjun Cai
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiP. R. China
| | - Li Ren
- Department of General SurgeryZhongshan HospitalFudan UniversityShanghaiP. R. China
| | - Yunpeng Liu
- Department of Medical OncologyThe First Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Jianmin Xu
- Department of General SurgeryZhongshan HospitalFudan UniversityShanghaiP. R. China
| | - Yanqiao Zhang
- Department of OncologyHarbin Medical University Cancer HospitalHarbinHeilongjiangP. R. China
| | - Houjie Liang
- Department of OncologySouthwest HospitalThird Military Medical University (Army Medical University)ChongqingP. R. China
| | - Xicheng Wang
- Department of Gastrointestinal OncologyCancer Medical Center, Peking Union Medical College HospitalChinese Academy of Medical SciencesBeijingChina
| | - Aiping Zhou
- Department of Medical OncologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Jianming Ying
- Department of PathologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| | - Guichao Li
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiP. R. China
| | - Muyan Cai
- Department of PathologySun Yat‐sen University Cancer Center, The State Key Laboratory of Oncology in South ChinaGuangzhouGuangdongP. R. China
| | - Gang Ji
- Department of Gastrointestinal SurgeryXijing HospitalAir Force Military Medical UniversityXi'anShaanxiP. R. China
| | - Taiyuan Li
- Department of General SurgeryThe First Affiliated Hospital of Nanchang UniversityNanchangJiangxiP. R. China
| | - Jingyu Wang
- Department of RadiologyThe First Hospital of Jilin UniversityChangchunJilinP. R. China
| | - Hanguang Hu
- Department of Medical OncologyThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Kejun Nan
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiP. R. China
| | - Liuhong Wang
- Department of RadiologySecond Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Suzhan Zhang
- Department of Colorectal SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangP. R. China
| | - Jin Li
- Department of Medical OncologyShanghai GoBroad Cancer HospitalChina Pharmaceutical UniversityShanghaiP. R. China
| | - Rui‐Hua Xu
- Department of Medical OncologySun Yat‐sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat‐sen University, Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical SciencesGuangzhouGuangdongP. R. China
| |
Collapse
|
16
|
Pat Fong W, Li ZJ, Ren C, Guan WL, Zuo MX, Zhang TQ, Li BK, Zheng Y, Wu XJ, Ding PR, Chen G, Pan ZZ, Yuan YF, Tan Q, Wang ZQ, Li YH, Wang DS. Percutaneous hepatic artery infusion chemotherapy with oxaliplatin and fluoropyrimidines in treatment-resistant colorectal cancer patients with unresectable liver metastases: a retrospective cohort study. HPB (Oxford) 2025; 27:289-298. [PMID: 39668070 DOI: 10.1016/j.hpb.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Subsequent lines of therapy for chemotherapy-resistant metastatic colorectal cancer (CRC) have shown limited efficacy. Herein, we retrospectively investigated the efficacy and safety of hepatic artery infusion chemotherapy (HAIC) using oxaliplatin plus 5-FU/FUDR in patients with unresectable colorectal liver metastases (CRLM) who progressed following standard chemotherapy regimens. METHODS From March 2017 to April 2023, CRC patients with unresectable CRLM who progressed following standard chemotherapy and subsequently received HAIC oxaliplatin plus 5-FU/FUDR were evaluated. Objective response rate (ORR), disease control rate (DCR), median depth of tumor response (DpR), no evidence of disease (NED) rate, progression-free survival (PFS), overall survival (OS), and safety were assessed. RESULTS A total of 21 patients who progressed after a median of two (range: 1-4) lines of standard systemic chemotherapy were included. The ORR and DCR were 28.6 % and 95.2 %, respectively, with six patients reaching partial response. Additionally, the median DpR was 10.6 %, and seven patients underwent successful conversion surgery. Stratification revealed significantly better PFS in patients with liver-limited metastases compared to those with concurrent hepatic and extrahepatic metastases (P = 0.0003). CONCLUSION HAIC oxaliplatin plus 5-FU/FUDR is a robust regimen for treatment-resistant CRC patients with unresectable CRLM, particularly those with liver-limited disease.
Collapse
Affiliation(s)
- William Pat Fong
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zi-Jing Li
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chao Ren
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wen-Long Guan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Meng-Xuan Zuo
- Department of Minimally Invasive & Interventional Therapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Tian-Qi Zhang
- Department of Minimally Invasive & Interventional Therapy, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Bin-Kui Li
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yun Zheng
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiao-Jun Wu
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Pei-Rong Ding
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Gong Chen
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zhi-Zhong Pan
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yun-Fei Yuan
- Department of Hepatobiliary Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qiong Tan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zhi-Qiang Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yu-Hong Li
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - De-Shen Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
17
|
Li Y, Liu F, Cai Q, Deng L, Ouyang Q, Zhang XHF, Zheng J. Invasion and metastasis in cancer: molecular insights and therapeutic targets. Signal Transduct Target Ther 2025; 10:57. [PMID: 39979279 PMCID: PMC11842613 DOI: 10.1038/s41392-025-02148-4] [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: 05/09/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
The progression of malignant tumors leads to the development of secondary tumors in various organs, including bones, the brain, liver, and lungs. This metastatic process severely impacts the prognosis of patients, significantly affecting their quality of life and survival rates. Research efforts have consistently focused on the intricate mechanisms underlying this process and the corresponding clinical management strategies. Consequently, a comprehensive understanding of the biological foundations of tumor metastasis, identification of pivotal signaling pathways, and systematic evaluation of existing and emerging therapeutic strategies are paramount to enhancing the overall diagnostic and treatment capabilities for metastatic tumors. However, current research is primarily focused on metastasis within specific cancer types, leaving significant gaps in our understanding of the complex metastatic cascade, organ-specific tropism mechanisms, and the development of targeted treatments. In this study, we examine the sequential processes of tumor metastasis, elucidate the underlying mechanisms driving organ-tropic metastasis, and systematically analyze therapeutic strategies for metastatic tumors, including those tailored to specific organ involvement. Subsequently, we synthesize the most recent advances in emerging therapeutic technologies for tumor metastasis and analyze the challenges and opportunities encountered in clinical research pertaining to bone metastasis. Our objective is to offer insights that can inform future research and clinical practice in this crucial field.
Collapse
Affiliation(s)
- Yongxing Li
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA
- Graduate School of Biomedical Science, Cancer and Cell Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lijun Deng
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
18
|
Padmanabhan C, Nussbaum DP, D'Angelica M. Surgical Management of Colorectal Cancer Liver Metastases. Hematol Oncol Clin North Am 2025; 39:1-24. [PMID: 39510667 DOI: 10.1016/j.hoc.2024.08.011] [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
Approximately 50% of colorectal cancer patients develop liver metastases. Hepatic metastases represent the most common cause of colorectal cancer-related mortality. Metastasectomy, if possible, represents the most effective treatment strategy; 20% of patients will be cured and more than 50% survive at least 5 years. Nuances to treatment planning hinge on whether patients present with resectable disease upfront, whether the future liver remnant is adequate, and whether the primary tumor, if present, is colon versus rectal in origin. This article discusses considerations impacting our approach to patients with colorectal liver metastases and the role for various multimodal treatment options.
Collapse
Affiliation(s)
- Chandrasekhar Padmanabhan
- Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, C-1272, New York, NY 10065, USA
| | - Daniel P Nussbaum
- Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, C-1272, New York, NY 10065, USA
| | - Michael D'Angelica
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, C-898, New York, NY 10065, USA.
| |
Collapse
|
19
|
Alexander HR, Devi-Chou V. Hepatic Perfusion for Diffuse Metastatic Cancer to the Liver: Open and Percutaneous Techniques. Hematol Oncol Clin North Am 2025; 39:177-190. [PMID: 39510672 DOI: 10.1016/j.hoc.2024.08.007] [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
The management of patients with diffuse liver metastases remains a significant clinical challenge. In many cancer patients, metastatic disease may be isolated to the liver or the liver may be the dominant site of progressive metastatic cancer. In this setting, progression of disease in the liver generally is the most significant cause of morbidity and mortality.
Collapse
Affiliation(s)
- H Richard Alexander
- Department of Surgery, Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 195 Little Albany Street, Room 2009, New Brunswick, NJ 08901, USA.
| | - Virginia Devi-Chou
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
Fourrier T, Truntzer C, Peroz M, Derangère V, Vincent J, Bengrine-Lefèvre L, Hennequin A, Palmier R, Orry D, Rabel T, Ghiringhelli F. Factors Influencing the Duration of Maintenance Therapy in Metastatic Colorectal Cancer. Cancers (Basel) 2024; 17:88. [PMID: 39796718 PMCID: PMC11720154 DOI: 10.3390/cancers17010088] [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: 10/17/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Metastatic colorectal cancer (mCRC) is mainly treated with 5-Fluoro-Uracil (5-FU), Oxaliplatin and Irinotecan chemotherapies and anti-Epidermal Growth Factor Receptor (EGFR) or anti-Vascular Endothelial Growth Factor (VEGF) targeted therapies. Due to chemotherapy-related toxicity, patients receive induction treatment to achieve tumour response followed by maintenance therapy with less cytotoxic molecules or a chemotherapy-free interval to reduce chemotherapy-related toxicity. In this study, the aim was to determine the patient, cancer and treatment factors that influence the duration of maintenance therapy (DMT). METHODS We collected retrospective data on a cohort of 133 patients treated at the Centre Georges François Leclerc (CGFL) cancer centre in Dijon between March 2014 and June 2022. Patients had unresectable or potentially resectable diseases. They received first-line induction treatment with chemotherapy and/or targeted therapy and maintenance treatment, defined as the interruption of at least one chemotherapy agent. RESULTS In the multivariate analysis, age (HR: 1.02, 95% CI 1.00-1.04, p = 0.031), N2 nodal status (HR: 1.78, 95% CI 1.09-2.89, p = 0.021) and the presence of peritoneal metastases (HR: 2.05, 95% CI 1.25-3.36, p = 0.004), as well as baseline carcino-embryonic antigen (CEA) level (HR: 1.10, 95% CI 1.00-1.20, p = 0.052), were significantly associated to poor DMT. Local treatment of liver metastases also significantly reduced the DMT (HR: 0.49, 95% CI 0.28-0.86, p = 0.013). In our cohort, induction triplet chemotherapy significantly increased the CEA delta (70% vs. 44%, p = 0.047) compared to doublet chemotherapy and led to a higher rate of liver surgery (40% vs. 21%, p = 0.014) and a trend for a higher rate of local treatment of metastases (62% vs. 45%, p = 0.059). CONCLUSIONS Duration of maintenance therapy is determined by the initial patient and colorectal cancer characteristics. However, it is significantly increased by local treatment of liver metastases. By reducing the tumour burden, a triplet induction chemotherapy regimen increases the rate of liver metastase resection.
Collapse
Affiliation(s)
- Théo Fourrier
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Caroline Truntzer
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- INSERM UMR1231 Research Center, University of Burgundy, 21000 Dijon, France
| | - Morgane Peroz
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Valentin Derangère
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- INSERM UMR1231 Research Center, University of Burgundy, 21000 Dijon, France
| | - Julie Vincent
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Leila Bengrine-Lefèvre
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Audrey Hennequin
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Rémi Palmier
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - David Orry
- Department of Surgical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Thomas Rabel
- Department of Surgical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - François Ghiringhelli
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- INSERM UMR1231 Research Center, University of Burgundy, 21000 Dijon, France
| |
Collapse
|
26
|
Qiu MZ, Bai Y, Wang J, Gu K, Yang M, He Y, Yi C, Jin Y, Liu B, Wang F, Chen YK, Dai W, Jiang Y, Huang C, Xu RH, Luo HY. Addition of SHR-1701 to first-line capecitabine and oxaliplatin (XELOX) plus bevacizumab for unresectable metastatic colorectal cancer. Signal Transduct Target Ther 2024; 9:349. [PMID: 39676137 DOI: 10.1038/s41392-024-02063-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
This phase 2/3 trial (NCT04856787) assessed the efficacy and safety of SHR-1701, a bifunctional protein targeting PD-L1 and TGF-β, in combination with BP102 (a bevacizumab biosimilar) and XELOX (capecitabine plus oxaliplatin) as a first-line treatment for unresectable metastatic colorectal cancer (mCRC). In this phase 2 study, a total of 62 patients with untreated, histologically confirmed colorectal adenocarcinoma and no prior systemic therapy for metastatic disease were enrolled. Patients received SHR-1701 (30 mg/kg), bevacizumab (7.5 mg/kg), and oxaliplatin (130 mg/m2) intravenously on day 1, along with oral capecitabine (1 g/m2 twice daily) on days 1-14 of 21-day cycles. Up to eight induction cycles were administered, followed by maintenance therapy for responders or those with stable disease. The primary endpoints were safety and objective response rate (ORR) per RECIST v1.1. The combination achieved an ORR of 59.7% and a disease control rate (DCR) of 83.9%. Median progression-free survival (PFS) was 10.3 months (95% CI: 8.3-13.7), with 6- and 12-month PFS rates of 77.2% and 41.3%, respectively. The estimated 12-month overall survival (OS) rate was 67.7%. Grade ≥3 treatment-related adverse events (TRAEs) were reported in 59.7% of patients, with anemia and neutropenia (8.1% each) being the most common. Retrospective DNA sequencing revealed that high tumor mutational burden, neo-antigens, and SBS15 enrichment correlated with better responses. Elevated baseline lactate dehydrogenase was linked to shorter PFS. SHR-1701 combined with XELOX and bevacizumab demonstrated a manageable safety profile and potent antitumor activity in unresectable mCRC.
Collapse
Affiliation(s)
- Miao-Zhen Qiu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, PR China.
| | - Yuxian Bai
- Department of Gastroenterology 1, Harbin Medical University Cancer Hospital, Harbin, PR China
| | - Jufeng Wang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, PR China
| | - Kangsheng Gu
- Oncology Ward 1, The First Affiliated Hospital of Anhui Medical University, Hefei, PR China
| | - Mudan Yang
- Gastroenterology Ward (2), Shanxi Provincial Cancer Hospital, Taiyuan, PR China
| | - Yifu He
- Medical Oncology Ward 1, Anhui Provincial Cancer Hospital, Hefei, PR China
| | - Cheng Yi
- Abdominal Oncology, West China Hospital of Sichuan University, Chengdu, PR China
| | - Yongdong Jin
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science & Technology of China, Chengdu, PR China
| | - Bo Liu
- Gastroenterology Ward 3, Cancer Hospital Affiliated to Shandong First Medical University, Jinan, PR China
| | - Feng Wang
- Oncology Department 1, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Yu-Kun Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, PR China
| | - Wei Dai
- Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, PR China
| | - Yingyi Jiang
- Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, PR China
| | - Chuanpei Huang
- Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, PR China
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, PR China.
| | - Hui-Yan Luo
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, PR China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, PR China.
| |
Collapse
|
27
|
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
|
28
|
Roazzi L, Patelli G, Bencardino KB, Amatu A, Bonazzina E, Tosi F, Amoruso B, Bombelli A, Mariano S, Stabile S, Porta C, Siena S, Sartore-Bianchi A. Ongoing Clinical Trials and Future Research Scenarios of Circulating Tumor DNA for the Treatment of Metastatic Colorectal Cancer. Clin Colorectal Cancer 2024; 23:295-308. [PMID: 38519391 DOI: 10.1016/j.clcc.2024.02.001] [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: 09/14/2023] [Revised: 01/04/2024] [Accepted: 02/11/2024] [Indexed: 03/24/2024]
Abstract
Liquid biopsy using circulating tumor DNA (ctDNA) has emerged as a minimally invasive, timely approach to provide molecular diagnosis and monitor tumor evolution in patients with cancer. Since the molecular landscape of metastatic colorectal cancer (mCRC) is substantially heterogeneous and dynamic over space and time, ctDNA holds significant advantages as a biomarker for this disease. Numerous studies have demonstrated that ctDNA broadly recapitulates the molecular profile of the primary tumor and metastases, and have mainly focused on the genotyping of RAS and BRAF, that is propaedeutic for anti-EGFR treatment selection. However, ctDNA soon broadened its scope towards the assessment of early tumor response, as well as the identification of drug resistance biomarkers to drive potential molecular actionability. In this review article, we provide an overview of the current state-of-the-art of this methodology and its applications, focusing on ongoing clinical trials that employ ctDNA to prospectively guide treatment in patients with mCRC.
Collapse
Affiliation(s)
- Laura Roazzi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giorgio Patelli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy; IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Katia Bruna Bencardino
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alessio Amatu
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Erica Bonazzina
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federica Tosi
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Brunella Amoruso
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy; Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Anna Bombelli
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Sara Mariano
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Stefano Stabile
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Camillo Porta
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy; Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy.
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Division of Clinical Research and Innovation, Grande Ospedale Metropolitano Niguarda, Milan, Italy.
| |
Collapse
|
29
|
Vaz-Salgado MA, Albarran V, Pozas J, Ferreiro R, Pachón V, Longo F, Rodriguez M, Barreto E, Earl J. Single nucleotide polymorphisms: impact on susceptibility to chemotherapy in patients with colorectal cancer. Future Sci OA 2024; 10:2428077. [PMID: 39576003 PMCID: PMC11587850 DOI: 10.1080/20565623.2024.2428077] [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: 02/19/2024] [Accepted: 10/11/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Single-nucleotide polymorphisms (SNPs) in enzyme-coding genes play a role in susceptibility to anti-cancer therapy. MATERIALS & METHODS A prospective study was performed of the relationship between enzyme activity and treatment response, drug toxicity and hypersensitivity reactions in 51 patients with colorectal cancer treated with fluoropyrimidine-based chemotherapy. SNP analysis was performed in 22 enzyme-coding genes with a previously described role in treatment efficacy. RESULTS SLC6 and MTHR enzyme activity was related with rates of progressive disease, GSTP1 activity with anti-EGFR antibodies-related skin toxicity, CYP3A5 and MTHR with chemotherapy dose reduction, CYP2B6, IL10, MTHR and TYMS activity with the risk of drug hypersensitivity reactions. CONCLUSION Pharmacogenetics is a valuable predictive marker in oncology, related to chemotherapy treatment response, toxicity and hypersensitivity.
Collapse
Affiliation(s)
- M. Angeles Vaz-Salgado
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
| | - Victor Albarran
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Javier Pozas
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Reyes Ferreiro
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
| | - Vanessa Pachón
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Federico Longo
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
| | - Mercedes Rodriguez
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
| | - Emma Barreto
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
- University of Alcalá, Madrid, Spain
| | - Julie Earl
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid, Spain
- The Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
| |
Collapse
|
30
|
Underwood PW, Pawlik TM. Precision Medicine for Metastatic Colorectal Cancer: Where Do We Stand? Cancers (Basel) 2024; 16:3870. [PMID: 39594824 PMCID: PMC11593240 DOI: 10.3390/cancers16223870] [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: 10/07/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Metastatic colorectal cancer is a leading cause of cancer-related death across the world. The treatment paradigm has shifted away from systemic chemotherapy alone to include targeted therapy and immunotherapy. The past two decades have been characterized by increased investigation into molecular profiling of colorectal cancer. These molecular profiles help physicians to better understand colorectal cancer biology among patients with metastatic disease. Additionally, improved data on genetic pathways allow for specific therapies to be targeted at the underlying molecular profile. Investigation of the EGFR, VEGF, HER2, and other pathways, as well as deficient mismatch repair, has led to the development of multiple targeted therapies that are now utilized in the National Comprehensive Cancer Network guidelines for colon and rectal cancer. While these new therapies have contributed to improved survival for metastatic colorectal cancer, long-term survival remains poor. Additional investigation to understand resistance to targeted therapy and development of new targeted therapy is necessary. New therapies are under development and are being tested in the preclinical and clinical settings. The aim of this review is to provide a comprehensive evaluation of molecular profiling, currently available therapies, and ongoing obstacles in the field of colorectal cancer.
Collapse
Affiliation(s)
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center, 395 W. 12th Ave., Suite 670, Columbus, OH 43210, USA;
| |
Collapse
|
31
|
Miyashita K, Shioi S, Kajitani T, Koi Y, Shimokawa M, Makiyama A, Oda S, Esaki T. More subtle microsatellite instability better predicts fluorouracil insensitivity in colorectal cancer patients. Sci Rep 2024; 14:27257. [PMID: 39516234 PMCID: PMC11549419 DOI: 10.1038/s41598-024-77770-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Microsatellite instability (MSI) is now widely used as an indispensable biomarker. However, the relationship between MSI-H (high) and defective DNA mismatch repair (MMR) is not as straightforward as has been expected. Genome-edited cells carrying Lynch syndrome mutations do not exhibit drastic MSI typical in MSI-H (i.e. Type B) but more subtle MSI (i.e. Type A). In this study, we explored a connection between Type A MSI and 5-fluorouracil (5-FU) resistance in colorectal cancer patients. Using our precision and high-resolution MSI assay technique, tumour microsatellites were analysed in 30 colorectal cancer patients treated with FOLFOX or CAPOX. Among 30 tumours, eleven (37%) were judged as Type A MSI-positive. In Type A MSI+ tumours, the patient response to fluoropyrimidine and oxaliplatin was significantly poor (Fisher's exact test, p = 0.021). Accordingly, median PFS and OS were significantly poor in Type A+ patients (log-rank test, p < 0.001/p = 0.009). Type A MSI was an independent predictor of patient prognosis in this pilot cohort (Cox regression analysis, p = 0.003). Thus, more subtle Type A MSI better predicts fluoropyrimidine insensitivity in colorectal cancer patients, which may shed light on a hitherto overlooked connection between the MSI phenotypes and drug resistance in human cancer.
Collapse
Affiliation(s)
- Kaname Miyashita
- Cancer Genetics Laboratory, Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, 811-1395, Japan
- Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, Japan
- Department of Hematology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Seijiro Shioi
- Cancer Genetics Laboratory, Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, 811-1395, Japan
- Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Tatsuhiro Kajitani
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yumiko Koi
- Department of Breast Oncology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Mototsugu Shimokawa
- Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, Japan
- Cancer Biostatistics Laboratory, Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, Japan
| | | | - Shinya Oda
- Cancer Genetics Laboratory, Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, 811-1395, Japan.
- Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, Japan.
| | - Taito Esaki
- Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, Japan
- Department of Gastrointestinal and Medical Oncology, NHO Kyushu Cancer Center, Fukuoka, Japan
| |
Collapse
|
32
|
Zwart K, Braat M, van der Baan F, May A, Roodhart J, Al-Toma D, Otten J, Los M, Oostergo T, Fijneman R, van Dodewaard-de Jong J, Punt C, Meijer G, Lagendijk J, Koopman M, Intven M, Bol G. Feasibility and safety of single-fraction sub-ablative radiotherapy with systemic therapy in colorectal cancer patients with ≤ 10 metastases: A multicenter pilot study (NCT05375708). Clin Transl Radiat Oncol 2024; 49:100874. [PMID: 39885896 PMCID: PMC11780375 DOI: 10.1016/j.ctro.2024.100874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/16/2024] [Accepted: 10/05/2024] [Indexed: 02/01/2025] Open
Abstract
Colorectal cancer patients with ≤10 unresectable metastases were treated with single-fraction sub-ablative radiotherapy in addition to standard of care systemic therapy in a single-arm, open-label, multicenter, pilot study (SIRIUS) to assess feasibility and safety. Results indicate that radiotherapy combined with systemic therapy is feasible and safe in this population.
Collapse
Affiliation(s)
- K. Zwart
- Department of Medical Oncology, Imaging & Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - M.N.G.J.A. Braat
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - F.H. van der Baan
- Department of Medical Oncology, Imaging & Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - A.M. May
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - J.M.L. Roodhart
- Department of Medical Oncology, Imaging & Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - D. Al-Toma
- Department of Medical Oncology, Imaging & Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - J.M.M.B. Otten
- Department of Oncology, Meander Medical Centre, Amersfoort, the Netherlands
| | - M. Los
- Department of Oncology, St. Antonius, Nieuwegein, the Netherlands
| | - T. Oostergo
- Department of Oncology, Diakonessenhuis, Utrecht, the Netherlands
| | - R.J.A. Fijneman
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - C.J.A Punt
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - G. Meijer
- Department of Radiotherapy, Imaging & Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - J.J.W. Lagendijk
- Department of Radiotherapy, Imaging & Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - M. Koopman
- Department of Medical Oncology, Imaging & Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - M. Intven
- Department of Radiotherapy, Imaging & Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - G.M. Bol
- Department of Medical Oncology, Imaging & Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Radiotherapy, Imaging & Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
33
|
Herrera-Quintana L, Vázquez-Lorente H, Silva RCMC, Olivares-Arancibia J, Reyes-Amigo T, Pires BRB, Plaza-Diaz J. The Role of the Microbiome and of Radiotherapy-Derived Metabolites in Breast Cancer. Cancers (Basel) 2024; 16:3671. [PMID: 39518108 PMCID: PMC11545256 DOI: 10.3390/cancers16213671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
The gut microbiome has emerged as a crucial player in modulating cancer therapies, including radiotherapy. In the case of breast cancer, the interplay between the microbiome and radiotherapy-derived metabolites may enhance therapeutic outcomes and minimize adverse effects. In this review, we explore the bidirectional relationship between the gut microbiome and breast cancer. We explain how gut microbiome composition influences cancer progression and treatment response, and how breast cancer and its treatments influence microbiome composition. A dual role for radiotherapy-derived metabolites is explored in this article, highlighting both their therapeutic benefits and potential hazards. By integrating genomics, metabolomics, and bioinformatics tools, we present a comprehensive overview of these interactions. The study provides real-world insight through case studies and clinical trials, while therapeutic innovations such as probiotics, and dietary interventions are examined for their potential to modulate the microbiome and enhance treatment effectiveness. Moreover, ethical considerations and patient perspectives are discussed, ensuring a comprehensive understanding of the subject. Towards revolutionizing treatment strategies and improving patient outcomes, the review concludes with future research directions. It also envisions integrating microbiome and metabolite research into personalized breast cancer therapy.
Collapse
Affiliation(s)
- Lourdes Herrera-Quintana
- Department of Physiology, Schools of Pharmacy and Medicine, University of Granada, 18071 Granada, Spain; (L.H.-Q.); (H.V.-L.)
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, 18016 Granada, Spain
| | - Héctor Vázquez-Lorente
- Department of Physiology, Schools of Pharmacy and Medicine, University of Granada, 18071 Granada, Spain; (L.H.-Q.); (H.V.-L.)
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, 18016 Granada, Spain
| | | | - Jorge Olivares-Arancibia
- AFySE Group, Research in Physical Activity and School Health, School of Physical Education, Faculty of Education, Universidad de Las Américas, Santiago 7500975, Chile;
| | - Tomás Reyes-Amigo
- Physical Activity Sciences Observatory (OCAF), Department of Physical Activity Sciences, Universidad de Playa Ancha, Valparaíso 2360072, Chile;
| | - Bruno Ricardo Barreto Pires
- Biometry and Biophysics Department, Institute of Biology Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, RJ, Brazil;
| | - Julio Plaza-Diaz
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- School of Health Sciences, Universidad Internacional de La Rioja, Avenida de la Paz, 137, 26006 Logroño, Spain
| |
Collapse
|
34
|
Masuda T, Funakoshi T, Horimatsu T, Yamamoto S, Matsubara T, Masui S, Nakagawa S, Ikemi Y, Yanagita M, Muto M, Terada T, Yonezawa A. Low serum concentrations of bevacizumab and nivolumab owing to excessive urinary loss in patients with proteinuria: a case series. Cancer Chemother Pharmacol 2024; 94:615-622. [PMID: 38456954 DOI: 10.1007/s00280-024-04659-3] [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: 12/26/2023] [Accepted: 02/18/2024] [Indexed: 03/09/2024]
Abstract
PURPOSE Proteinuria can cause interindividual variability in the pharmacokinetics of therapeutic antibodies and may affect therapeutic efficacy. Here, we measured the serum and urinary concentrations of bevacizumab (BV) and nivolumab (NIVO) in patients with proteinuria and reported a case series of these patients. METHODS Thirty-two cancer patients who received BV every 3 weeks or NIVO every 2 weeks between November 2020 and September 2021 at Kyoto University Hospital were enrolled in this study. The serum and urinary concentrations of BV and NIVO were measured using liquid chromatography-tandem mass spectrometry. RESULTS We divided the BV-treated patients and the NIVO-treated patients into two groups based on the urine protein-creatinine ratio (UPCR): UPCR 1 g/g or higher (BV, n = 9; NIVO, n = 3) and UPCR less than 1 g/g (BV, n = 14; NIVO, n = 6). Serum concentrations of the therapeutic antibodies adjusted by their doses were significantly lower in both BV- and NIVO-treated patients with UPCR 1 g/g or higher compared to those with less than 1 g/g. In patients with UPCR 1 g/g or higher, urinary concentrations of the therapeutic antibodies adjusted by their serum concentrations and urinary creatinine concentrations tended to increase. CONCLUSION This case-series study suggests a possibility of reduction in serum concentrations of BV and NIVO in patients with proteinuria by urinary excretion of these drugs.
Collapse
Affiliation(s)
- Takashi Masuda
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Taro Funakoshi
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Horimatsu
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinya Yamamoto
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Matsubara
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sho Masui
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
- Division of Integrative Clinical Pharmacology, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato- ku, Tokyo, 105-8512, Japan
| | - Shunsaku Nakagawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Yasuaki Ikemi
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Terada
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Atsushi Yonezawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan.
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan.
- Division of Integrative Clinical Pharmacology, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato- ku, Tokyo, 105-8512, Japan.
| |
Collapse
|
35
|
Lee Y, Lee E, Roh TH, Kim SH. Bevacizumab Alone Versus Bevacizumab Plus Irinotecan in Patients With Recurrent Glioblastoma: A Nationwide Population-Based Study. J Korean Med Sci 2024; 39:e244. [PMID: 39228184 PMCID: PMC11372412 DOI: 10.3346/jkms.2024.39.e244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/10/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND For treating recurrent glioblastoma, for which there is no established treatment, the antiangiogenic antibody, bevacizumab, is used alone or with irinotecan. This study was aimed at comparing the survival of patients with recurrent glioblastoma receiving bevacizumab monotherapy and those receiving bevacizumab plus irinotecan combination therapy (B+I) by using a nationwide population-based dataset. METHODS Patients matching the International Classification of Diseases code C71.x were screened from the Health Insurance Review and Assessment Service database. From January 2008 to November 2021, patients who underwent surgery or biopsy and subsequent standard concurrent chemoradiation with temozolomide were included. Among them, those who received bevacizumab monotherapy or B+I were selected. Demographic characteristics, inpatient stay, prescription frequency, survival outcomes, and steroid prescription duration were compared between these two groups. RESULTS Eight hundred and forty-six patients who underwent surgery or biopsy and received concurrent chemoradiotherapy with temozolomide were included. Of these, 450 and 396 received bevacizumab monotherapy and B+I, respectively. The corresponding median overall survival from the initial surgery was 22.60 months (95% confidence interval [CI], 20.50-24.21) and 20.44 months (95% CI, 18.55-22.60; P = 0.508, log-rank test). The B+I group had significantly more bevacizumab prescriptions (median 5 times; BEV group: median 3 times). Cox analysis, based on the postsurgery period, revealed that male sex (hazard ratio [HR], 1.28; P = 0.002), older age (HR, 1.01; P = 0.042), and undergoing biopsy instead of surgery (HR, 1.79; P < 0.0001) were significantly associated with decreased survival. Fewer radiotherapy cycles correlated with improved survival outcomes (HR, 0.63; P = 0.001). Cox analysis, conducted from the start of chemotherapy including bevacizumab, showed that male sex was the only variable significantly associated with decreased survival (HR, 1.18; P = 0.044). CONCLUSION We found no significant difference in overall survival between the bevacizumab monotherapy and B+I groups. Considering the additional potential toxicity associated with irinotecan, bevacizumab monotherapy could be a suitable treatment option for treating recurrent glioblastoma.
Collapse
Affiliation(s)
- Yeonhu Lee
- Department of Neurosurgery, Ajou University School of Medicine, Suwon, Korea
| | - Eunyoung Lee
- Department of Neurology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Tae Hoon Roh
- Department of Neurosurgery, Ajou University School of Medicine, Suwon, Korea.
| | - Se-Hyuk Kim
- Department of Neurosurgery, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
36
|
Flynn A, Galettis P, Gurney H, Michael M, Desar I, Westerdijk K, Schneider J, Martin J. Therapeutic drug monitoring in anticancer agents: perspectives of Australian medical oncologists. Intern Med J 2024; 54:1458-1464. [PMID: 38767393 DOI: 10.1111/imj.16415] [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: 09/22/2023] [Accepted: 04/20/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND In the development of anticancer agents for solid tumours, body surface area continues to be used to personalise dosing despite minimal evidence for its use over other dosing strategies. With the development of tyrosine kinase inhibitors and other oral targeted anticancer agents, dosing using therapeutic drug monitoring (TDM) is now utilised in many health systems but has had limited uptake in Australia. AIM To determine attitudes and barriers to the implementation of TDM among Australian oncologists. METHODS A comprehensive questionnaire was developed by the Dutch Pharmacology Oncology Group from semistructured interviews of stakeholders. Seventy-nine questions across seven domains were developed with three free-text responses. This was rationalised to 17 questions with three free-text responses for Australian medical oncologists who identified limited experience with TDM. RESULTS Fifty-seven responses were received, with 49 clinicians (86%) identifying limited experience of performing TDM in daily practice. Clinicians were positive (62-91% agree/strongly agree across seven questions) about the advantages of TDM. There was a mixed response for cost-effectiveness and scientific evidence being a barrier to implementation, but strong agreement that prospective studies were needed (75% agreed or strongly agreed); that national treatment guidelines would enable practice (80%) and that a 'pharmacology of oncolytics' education programme would be useful (96%) to provide knowledge for dose individualisation. CONCLUSION Despite the limited experience of TDM in oncology in Australia, medical oncologists appear positive about the potential benefit to their patients. We have identified three barriers to implementation that could be targeted for increased adoption of TDM in oncology in Australia.
Collapse
Affiliation(s)
- Alexandra Flynn
- University of Newcastle, Newcastle, New South Wales, Australia
| | - Peter Galettis
- University of Newcastle, Newcastle, New South Wales, Australia
| | - Howard Gurney
- Macquarie University Hospital, Sydney, New South Wales, Australia
| | - Michael Michael
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ingrid Desar
- Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Kim Westerdijk
- Radboud University Medical Centre, Nijmegen, the Netherlands
| | | | - Jennifer Martin
- University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
37
|
Cohen R, Raeisi M, Chibaudel B, Shi Q, Yoshino T, Zalcberg JR, Adams R, Cremolini C, Van Cutsem E, Heinemann V, Tabernero J, Punt CJA, Arnold D, Hurwitz HI, Douillard JY, Venook AP, Saltz LB, Maughan TS, Kabbinavar F, Bokemeyer C, Grothey A, Mayer RJ, Kaplan R, Tebbutt NC, Randolph Hecht J, Giantonio BJ, Díaz-Rubio E, Sobrero AF, Peeters M, Koopman M, Goldberg RM, Andre T, de Gramont A. Prognostic value of liver metastases in colorectal cancer treated by systemic therapy: An ARCAD pooled analysis. Eur J Cancer 2024; 207:114160. [PMID: 38896997 DOI: 10.1016/j.ejca.2024.114160] [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: 04/20/2024] [Revised: 05/27/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND The liver is the most frequent site of metastases in colorectal cancer (CRC). This study aimed to assess the response rate and survival outcomes in metastatic CRC patients with non-liver metastases (NLM) compared to those with liver metastases (LM) across different lines of treatment. METHODS A total of 17,924 mCRC patients included in 26 trials from the ARCAD CRC database were analyzed. The analysis was conducted based on the presence or absence of LM across different treatment groups: chemotherapy (CT) alone, CT + anti-VEGF, CT + anti-EGFR in KRAS wild-type tumors, within the first-line (1 L) and second-line (2 L), and patients enrolled in third-line (≥3 L) trials treated with trifluridine/tipiracil or regorafenib or placebo. The endpoints were overall survival (OS), progression-free survival (PFS), and overall response rate (ORR). RESULTS Out of the 17,924 patients, 14,066 had LM (30.6 % with only liver involvement and 69.4 % with liver and other metastatic sites), while 3858 patients had NLM. In the CT alone and CT + anti-VEGF subgroups, NLM patients showed better OS and PFS in the 1 L and 2 L settings. However, in the CT + anti-EGFR 1 L and 2 L subgroups, there was no significant difference in OS and PFS between NLM and LM patients. In the ≥ 3 L subgroups, better OS and PFS were observed in NLM patients. ORRs were higher in LM patients than in NLM patients across all cohorts treated in the 1 L and only in the anti-EGFR cohort in the 2 L. CONCLUSION LM is a poor prognostic factor for mCRC increasing from 1 L to ≥ 3 L except for patients in 1 L and 2 L receiving CT+anti-EGFR. These data justify using LM as a stratification factor in future trials for patients with unresectable mCRC.
Collapse
Affiliation(s)
- Romain Cohen
- Sorbonne University, Department of Medical Oncology, Hôpital Saint-Antoine, AP-HP, and INSERM UMRS 938, Équipe Instabilité des Microsatellites et Cancer, Équipe Labellisée par la Ligue Nationale Contre le Cancer, SIRIC CURAMUS, Centre de recherche Saint Antoine, Paris, France
| | | | - Benoist Chibaudel
- Department of Medical Oncology, Franco-British Hospital, Fondation Cognacq-Jay, Levallois-Perret, France
| | - Qian Shi
- Department of Quantitative Health Science, Mayo Clinic, Rochester, USA
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - John R Zalcberg
- Monash University School of Public Health and Preventive Medicine and Department of Medical Oncology, Alfred Health, Melbourne, VIC, Australia
| | - Richard Adams
- Cardiff University and Velindre Cancer Centre, Cardiff, United Kingdom
| | - Chiara Cremolini
- Department of Translational Research and New Technologies, University of Pisa, Pisa, Italy
| | - Eric Van Cutsem
- Department of Gastrointestinal and Liver Diseases, Digestive Oncology Unit, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| | - Volker Heinemann
- Department of Medical Oncology, LMU Hospital, University of Munich, Munich, Germany
| | - Josep Tabernero
- Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), IOB-Quiron, Barcelona, Spain
| | | | - Dirk Arnold
- Asklepios Tumorzentrum Hamburg, Asklepios Klinik Altona, Hamburg, Germany
| | | | - Jean-Yves Douillard
- Medical Oncology Department, Integrated Centers for Oncology Nantes, Nantes, France
| | - Alan P Venook
- Department of Medicine, The University of California San Francisco, San Francisco, CA, USA
| | | | | | | | - Carsten Bokemeyer
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | - Niall C Tebbutt
- Department of Medical Oncology, Olivia Newton-John Cancer, Wellness and Research Centre, Austin Health, Heidelberg, VIC, Australia
| | - J Randolph Hecht
- David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, USA
| | | | | | | | | | - Miriam Koopman
- University Medical Center Utrecht, Utrecht, the Netherlands
| | - Richard M Goldberg
- Department of Medicine, West Virginia University Cancer Institute, Morgantown, USA
| | - Thierry Andre
- Sorbonne University, Department of Medical Oncology, Hôpital Saint-Antoine, AP-HP, and INSERM UMRS 938, Équipe Instabilité des Microsatellites et Cancer, Équipe Labellisée par la Ligue Nationale Contre le Cancer, SIRIC CURAMUS, Centre de recherche Saint Antoine, Paris, France; ARCAD Foundation, Paris, France
| | - Aimery de Gramont
- ARCAD Foundation, Paris, France; Department of Medical Oncology, Franco-British Hospital, Levallois-Perret, France
| |
Collapse
|
38
|
Ota M, Taniguchi K, Hori M, Katanoda K, Nakata K, Miyashiro I, Matsuda T, Lee S, Ito Y. Trends in patterns of treatment and survival of colorectal cancer patients using cancer registry data in Japan: 1995-2015. Cancer Sci 2024; 115:2786-2794. [PMID: 38715379 PMCID: PMC11309936 DOI: 10.1111/cas.16210] [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: 02/06/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 08/10/2024] Open
Abstract
Recent advances in treating colorectal cancer (CRC) have increased the importance of multidisciplinary treatment. This study aimed to clarify trends in the treatment and survival of CRC using population-based cancer registry data in Japan. We analyzed the survival of CRC cases diagnosed from 1995 through 2015 from a population-based cancer registry of six prefectures. The year of diagnosis was classified into five periods, and the trends in the detailed categorization of treatments and survival were identified. We calculated net survival and excess hazard of death from cancer using data on 256,590 CRC patients. The use of laparoscopic surgery has been increasing since 2005 and accounts for the largest proportion of treatment types in the most recent period. Net survival of CRC patients diagnosed after 2005 remained high for laparoscopic surgery and endoscopic surgery (endoscopic mucosal resection or endoscopic submucosal dissection). There was an upward trend in treatment with chemotherapy in addition to open and laparoscopic surgery. Using the excess hazard ratio at the regional stage since 2005, there has been a significant improvement in survival in the younger age group and the rectum cancer group. By type of treatment, there was a tendency toward significant improvement in the open surgery + chemotherapy group. We clarified the trends in treating CRC and the associated trends in survival. Continuous survey based on population-based data helps monitor the impact of developments in treatment.
Collapse
Affiliation(s)
- Masato Ota
- Department of General and Gastroenterological SurgeryOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
- Center for Medical Research & Development, Division of Translational ResearchOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| | - Kohei Taniguchi
- Translational Research ProgramOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| | - Megumi Hori
- School of NursingUniversity of ShizuokaShizuoka CityJapan
| | - Kota Katanoda
- National Cancer Center Institute for Cancer ControlTokyoJapan
| | - Kayo Nakata
- Cancer Control Center, Osaka International Cancer InstituteOsakaJapan
| | - Isao Miyashiro
- Cancer Control Center, Osaka International Cancer InstituteOsakaJapan
| | | | - Sang‐Woong Lee
- Department of General and Gastroenterological SurgeryOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| | - Yuri Ito
- Center for Medical Research & Development, Division of Translational ResearchOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| |
Collapse
|
39
|
Chitoran E, Rotaru V, Ionescu SO, Gelal A, Capsa CM, Bohiltea RE, Mitroiu MN, Serban D, Gullo G, Stefan DC, Simion L. Bevacizumab-Based Therapies in Malignant Tumors-Real-World Data on Effectiveness, Safety, and Cost. Cancers (Basel) 2024; 16:2590. [PMID: 39061228 PMCID: PMC11274419 DOI: 10.3390/cancers16142590] [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: 06/30/2024] [Revised: 07/08/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Overall, it is estimated that more than 3,500,000 patients have received Bevacizumab as part of systemic oncologic treatment. Bevacizumab and its biosimilars are currently marketed in over 130 countries. Given the wide usage of Bevacizumab in current oncological practice, it is very important to compare the "real-world" results to those obtained in controlled clinical trials. This study aims to describe the clinical experience of using Bevacizumab in a large cohort of cancer patients in "non-controlled real-world" conditions with regard to effectiveness, safety, and cost of therapy. METHODS For this purpose, we conducted an open, observational, retrospective study involving all patients treated for solid malignant tumors in the Bucharest Institute of Oncology with "Prof. Dr. Al. Trestioreanu" with Bevacizumab-based systemic therapy, between 2017 and 2021. RESULTS The study consisted of 657 treatment episodes in 625 patients (F/B = 1.62/1, with a median age of 57.6 years) which were treated for malignant tumors (majority colorectal, non-small cell lung, ovarian, and breast cancer). First-line treatment was administered in 229 patients, and the rest received Bevacizumab as second or subsequent lines of treatment. The overall response rate to Bevacizumab-based therapies was around 60-65% across all indication except for subsequent treatment lines in colorectal and ovarian cancers, where lower values were recorded (27.1%, and 31.5% respectively). Median PFS for the entire cohort was 8.2 months (95% CI 6.8-9.6), and the median OS was 13.2 months (95% CI 11.5-14.9). Usual bevacizumab-related toxicities were observed, including bleeding, hypertension, wound-healing complications, gastrointestinal perforation, other types of fistulas, septic complications, and thromboembolic events. Although the clinical benefits are undeniable, the addition of Bevacizumab to standard chemotherapy increased the overall treatment cost by 213%. CONCLUSIONS Bevacizumab remains a high-cost therapy, but it can add to clinical benefits (like overall survival, progression-free survival, and response rate) when used in conjunction with standard chemotherapy. Similar results as those presented in various controlled trials are observable even on unselected cohorts of patients in the uncontrolled conditions of "real-world" oncological practice. Off-label usage is encountered in clinical practice, and this aspect should be monitored given the potential adverse effects of the therapy.
Collapse
Affiliation(s)
- Elena Chitoran
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Vlad Rotaru
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Sinziana-Octavia Ionescu
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Aisa Gelal
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Cristina-Mirela Capsa
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Radiology Department, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| | - Roxana-Elena Bohiltea
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Obstetrics and Gynecology Department, “Filantropia” Clinical Hospital, 011132 Bucharest, Romania
| | - Madalina-Nicoleta Mitroiu
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Obstetrics and Gynecology Department, “Filantropia” Clinical Hospital, 011132 Bucharest, Romania
| | - Dragos Serban
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Surgery Department 4, Bucharest University Emergency Hospital, 050098 Bucharest, Romania
| | - Giuseppe Gullo
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, University of Palermo, 90146 Palermo, Italy
| | - Daniela-Cristina Stefan
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Laurentiu Simion
- Medicine School, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
| |
Collapse
|
40
|
Jiang Y, Zhao M, Tang W, Zheng X. Comparison of systemic treatments for previously treated patients with unresectable colorectal liver metastases: a systematic review and network meta-analysis. Front Oncol 2024; 14:1293598. [PMID: 39050571 PMCID: PMC11266080 DOI: 10.3389/fonc.2024.1293598] [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: 09/13/2023] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Background There is limited evidence of comparative results among different treatments for patients with unresectable colorectal liver metastases (CRLM) who have failed at least one line of previous systemic therapy. We aimed to compare the efficacy of systemic treatments among these patients through this investigation. Methods We collected randomized controlled trials (RCTs) reported in English up until July 2023, from databases including PubMed, Embase, Cochrane Library, ClinicalTrials.gov, and prominent conference databases, for this Bayesian network meta-analysis. Phase II or III trials that evaluated at least two therapeutic regimens were included. Primary outcome was overall survival (OS), secondary outcome was progression-free survival (PFS). Hazards ratios (HRs) with 95% confidence intervals (CIs) were used as effect size. Subgroup analysis was performed based on metastatic sites. The current systematic review protocol was registered on PROSPERO (CRD42023420498). Results 30 RCTs were included, with a total of 13,511 patients. Compared to chemotherapy, multi-targeted therapy (HR 0.57, 95% CI 0.37-0.87) and targeted therapy plus chemotherapy (HR 0.78, 95% CI 0.67-0.91) show significant advantages. Targeted therapy (HR 0.92, 95% CI 0.54-1.57) and local treatment plus chemotherapy (HR 1.03, 95% CI 0.85-1.23) had comparable performance. For patients with liver metastases, TAS-102 plus bevacizumab, aflibercept plus fluorouracil-based combination chemotherapy (CTFU), and bevacizumab plus capecitabine-based combination chemotherapy (CTCA) showed the best outcomes in terms of OS. Bevacizumab plus intensified CTFU, bevacizumab plus CTCA, and HAI followed by single-agent chemotherapy (SingleCT) performed the best regarding PFS. For patients with liver-limited metastases, aflibercept plus CTFU is the optimal choice in OS. For PFS, the best options were HAI followed by SingleCT, aflibercept plus CTFU, and panitumumab plus CTFU. For patients with multiple-site metastases, the best treatments were TAS-102 plus bevacizumab, bevacizumab plus CTCA, bevacizumab plus CTFU, and aflibercept plus CTFU. Conclusion Multi-targeted therapy and targeted therapy plus chemotherapy are the best treatment mechanisms. TAS-102 plus bevacizumab is superior in OS, the combination of anti-VEGF drugs like bevacizumab and aflibercept with standard chemotherapy is the preferred option for CRLM patients.
Collapse
Affiliation(s)
- Yunlin Jiang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
- Graduate School of Nanjing University of Chinese Medicine, Nanjing, China
| | - Mingye Zhao
- Department of Pharmacoeconomics, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wenxi Tang
- Department of Pharmacoeconomics, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xueping Zheng
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
41
|
Sugumar K, Stitzel H, Wu V, Bajor D, Chakrabarti S, Conces M, Henke L, Lumish M, Mahipal A, Mohamed A, Winter JM, Hardacre JM, Ammori JB, Selfridge JE, Ocuin LM. Outcomes of Hepatic Artery-Based Therapies and Systemic Multiagent Chemotherapy in Unresectable Colorectal Liver Metastases: A Systematic Review and Meta-analysis. Ann Surg Oncol 2024; 31:4413-4426. [PMID: 38502296 PMCID: PMC11164761 DOI: 10.1245/s10434-024-15187-y] [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: 01/25/2024] [Accepted: 03/01/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Treatment of unresectable colorectal liver metastases (UCRLM) includes locoregional and systemic therapy. A comprehensive analysis capturing long-term outcomes of these treatment options has not been performed. OBJECTIVE A systematic review and meta-analysis was performed to calculate pooled outcomes of hepatic artery infusion with systemic chemotherapy (HAI-S), transarterial chemoembolization with systemic chemotherapy (TACE-S), transarterial radioembolization with systemic chemotherapy (TARE-S), doublet (FOLFOX, FOLFIRI), and triplet chemotherapy (FOLFOXIRI). METHODS Outcomes included overall survival (OS), progression-free survival (PFS), rate of conversion to resection (CTR), and response rate (RR). RESULTS A total of 32, 7, 9, and 14 publications were included in the HAI-S, TACE-S, and TARE-S chemotherapy arms. The 6/12/24/36-month OS estimates for HAI-S, TACE-S, TARE-S, FOLFOX, FOLFIRI, and FOLFOXIRI were 97%/80%/54%/35%, 100%/83%/40%/14%, 82%/61%/34%/21%, 96%/83%/53%/36%, and 96%/93%/72%/55%. Similarly, the 6/12/24/36-month PFS estimates were 74%/44%/19%/14%, 66%/20%/9%/3%, 57%/23%/10%/3%, 69%/30%/12%/7%, and 88%/55%/18%/11%. The corresponding CTR and RR rates were 31, 20%, unmeasurable (TARE-S), 35, 53; and 49, 45, 45, 50, 80%, respectively. The majority of chemotherapy studies included first-line therapy and liver-only metastases, whereas most HAI-S studies were pretreated. On subgroup analysis in first-line setting with liver-only metastases, the HAI-S arm had comparable outcomes to FOLFOXIRI and outperformed doublet chemotherapy regimens. Although triplet chemotherapy appeared to outperform other arms, high toxicity and inclusion of potentially resectable patients must be considered while interpreting results. CONCLUSIONS HAI-S and multiagent chemotherapy are effective therapies for UCRLM. To make definitive conclusions, a randomized trial with comparable patient characteristics and line of therapy will be required. The upcoming EA2222 PUMP trial may help to address this question.
Collapse
Affiliation(s)
- Kavin Sugumar
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Henry Stitzel
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Victoria Wu
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - David Bajor
- Division of Hematology/Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, case Western Reserve University, Cleveland, OH, USA
| | - Sakti Chakrabarti
- Division of Hematology/Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, case Western Reserve University, Cleveland, OH, USA
| | - Madison Conces
- Division of Hematology/Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, case Western Reserve University, Cleveland, OH, USA
| | - Lauren Henke
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | - Melissa Lumish
- Division of Hematology/Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, case Western Reserve University, Cleveland, OH, USA
| | - Amit Mahipal
- Division of Hematology/Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, case Western Reserve University, Cleveland, OH, USA
| | - Amr Mohamed
- Division of Hematology/Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, case Western Reserve University, Cleveland, OH, USA
| | - Jordan M Winter
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jeffrey M Hardacre
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - John B Ammori
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jennifer E Selfridge
- Division of Hematology/Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, case Western Reserve University, Cleveland, OH, USA
| | - Lee M Ocuin
- Division of Surgical Oncology, Department of Surgery, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
| |
Collapse
|
42
|
Regan SN, Hendren S, Krauss JC, Crysler OV, Cuneo KC. Treatment of Locally Recurrent Rectal Cancer: A Review. Cancer J 2024; 30:264-271. [PMID: 39042778 DOI: 10.1097/ppo.0000000000000728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
ABSTRACT Up to 10% of patients with locally advanced rectal cancer will experience locoregional recurrence. In the setting of prior surgery and often radiation and chemotherapy, these represent uniquely challenging cases. When feasible, surgical resection offers the best chance for oncologic control yet risks significant morbidity. Studies have consistently indicated that a negative surgical resection margin is the strongest predictor of oncologic outcomes. Chemoradiation is often recommended to increase the chance of an R0 resection, and in cases of close/positive margins, intraoperative radiation/brachytherapy can be utilized. In patients who are not surgical candidates, radiation can provide symptomatic relief. Ongoing phase III trials are aiming to address questions regarding the role of reirradiation and induction multiagent chemotherapy regimens in this population.
Collapse
Affiliation(s)
| | | | - John C Krauss
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Oxana V Crysler
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | | |
Collapse
|
43
|
Nakagawa Y, Sozu T. Improvement of Midpoint Imputation for Estimation of Median Survival Time for Interval-Censored Time-to-Event Data. Ther Innov Regul Sci 2024; 58:721-729. [PMID: 38598082 DOI: 10.1007/s43441-024-00640-7] [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: 05/12/2023] [Accepted: 03/08/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Progression-free survival (PFS) is used to evaluate treatment effects in cancer clinical trials. Disease progression (DP) in patients is typically determined by radiological testing at several scheduled tumor-assessment time points. This produces a discrepancy between the true progression time and the observed progression time. When the observed progression time is considered as the true progression time, a positively biased PFS is obtained for some patients, and the estimated survival function derived by the Kaplan-Meier method is also biased. METHODS While the midpoint imputation method is available and replaces interval-censored data with midpoint data, it unrealistically assumes that several DPs occur at the same time point when several DPs are observed within the same tumor-assessment interval. We enhanced the midpoint imputation method by replacing interval-censored data with equally spaced timepoint data based on the number of observed interval-censored data within the same tumor-assessment interval. RESULTS The root mean square error of the median of the enhanced method is almost always smaller than that of the midpoint imputation regardless of the tumor-assessment frequency. The coverage probability of the enhanced method is close to the nominal confidence level of 95% in most scenarios. CONCLUSION We believe that the enhanced method, which builds upon the midpoint imputation method, is more effective than the midpoint imputation method itself.
Collapse
Affiliation(s)
- Yuki Nakagawa
- Biometrics Department, Chugai Pharmaceutical Co., Ltd., 2-1-1 Nihonbashi-Muromachi, Chuo-Ku, Tokyo, 103-8324, Japan.
- Department of Management Science, Graduate School of Engineering, Tokyo University of Science, Tokyo, Japan.
| | - Takashi Sozu
- Department of Information and Computer Technology, Faculty of Engineering, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
44
|
Mazard T, Mollevi C, Loyer EM, Léger J, Chautard R, Bouché O, Borg C, Armand-Dujardin P, Bleuzen A, Assenat E, Lecomte T. Prognostic value of the tumor-to-liver density ratio in patients with metastatic colorectal cancer treated with bevacizumab-based chemotherapy. A post-hoc study of the STIC-AVASTIN trial. Cancer Imaging 2024; 24:77. [PMID: 38886836 PMCID: PMC11181627 DOI: 10.1186/s40644-024-00722-7] [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: 12/14/2023] [Accepted: 06/10/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND The Response Evaluation Criteria in Solid Tumors (RECIST) are often inadequate for the early assessment of the response to cancer therapy, particularly bevacizumab-based chemotherapy. In a first cohort of patients with colorectal cancer liver metastases (CRLM), we showed that variations of the tumor-to-liver density (TTLD) ratio and modified size-based criteria determined using computed tomography (CT) data at the first restaging were better prognostic criteria than the RECIST. The aims of this study were to confirm the relevance of these radiological biomarkers as early predictors of the long-term clinical outcome and to assess their correlation with contrast-enhanced ultrasound (CEUS) parameters in a new patient cohort. METHODS In this post-hoc study of the multicenter STIC-AVASTIN trial, we retrospectively reviewed CT data of patients with CRLM treated with bevacizumab-based regimens. We determined the size, density and TTLD ratio of target liver lesions at baseline and at the first restaging and also performed a morphologic evaluation according to the MD Anderson criteria. We assessed the correlation of these parameters with progression-free survival (PFS) and overall survival (OS) using the log-rank test and a Cox proportional hazard model. We also examined the association between TTLD ratio and quantitative CEUS parameters. RESULTS This analysis concerned 79 of the 137 patients included in the STIC-AVASTIN trial. PFS and OS were significantly longer in patients with tumor size reduction > 15% at first restaging, but were not correlated with TTLD ratio variations. However, PFS was longer in patients with TTLD ratio > 0.6 at baseline and first restaging than in those who did not reach this threshold. In the multivariate analysis, only baseline TTLD ratio > 0.6 was a significant survival predictor. TTLD ratio > 0.6 was associated with improved perfusion parameters. CONCLUSIONS Although TTLD ratio variations did not correlate with the long-term clinical outcomes, TTLD absolute values remained a good predictor of survival at baseline and first restaging, and may reflect tumor microvascular features that might influence bevacizumab-based treatment efficiency. TRIAL REGISTRATION NCT00489697, registration number of the STIC-AVASTIN trial.
Collapse
Affiliation(s)
- Thibault Mazard
- Medical Oncology Department, Montpellier Cancer Institute (ICM), University of Montpellier, Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, 208 avenue des apothicaires, Parc Euromédecine, Montpellier Cedex 5, Montpellier, 34298, France.
| | - Caroline Mollevi
- Institute Desbrest of Epidemiology and Public Health, University of Montpellier, INSERM, Cancer Institute of Montpellier, Montpellier, France
| | - Evelyne M Loyer
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Julie Léger
- INSERM CIC 1415, CHRU de Tours, Tours Cedex 9, 37044, France
| | - Romain Chautard
- Department of Hepatogastroenterology and Digestive Oncology, UMR INSERM U 1069, Hôpital Trousseau, CHRU de Tours, Université de Tours, Tours Cedex 9, 37044, France
| | - Olivier Bouché
- Department of Hepatogastroenterology, Hôpital Robert Debré, CHU de Reims, Avenue Général Koenig, Reims Cedex, 51092, France
| | - Christophe Borg
- Department of Medical Oncology, Hôpital Jean Minjoz, CHRU de Besançon, 3 Boulevard Alexandre Fleming, Besançon, 25000, France
| | | | - Aurore Bleuzen
- Department of Radiology, CHRU de Tours, Tours Cedex 9, 37044, France
| | - Eric Assenat
- Medical Oncology Department, Montpellier Cancer Institute (ICM), University of Montpellier, CHU Montpellier, Montpellier, France
| | - Thierry Lecomte
- Department of Hepatogastroenterology and Digestive Oncology, UMR INSERM U 1069, Hôpital Trousseau, CHRU de Tours, Université de Tours, Tours Cedex 9, 37044, France
| |
Collapse
|
45
|
Wirsik NM, Appel PC, Braun A, Strowitzki MJ, Schleussner N, Nienhüser H, Schneider M, Schmidt T. Inhibition of the Renin-Angiotensin System Improves Response to Neoadjuvant Therapy in Patients With Liver Metastasis of Colorectal Cancers. J Surg Res 2024; 298:176-184. [PMID: 38621351 DOI: 10.1016/j.jss.2024.03.013] [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: 08/02/2023] [Revised: 02/01/2024] [Accepted: 03/16/2024] [Indexed: 04/17/2024]
Abstract
INTRODUCTION Renin-angiotensin-aldosterone system inhibitors (RAAS-I) have been shown to prolong overall survival in patients with liver metastasized colorectal cancer in combination with antiangiogenic treatment. The effects of RAAS-I combined with neoadjuvant chemotherapy on colorectal cancer liver metastasis remain unexplored. We aimed to study the response of patients undergoing liver resection to RAAS-I in combination with neoadjuvant therapy to elucidate their potential benefits. METHODS Between February 2005 and May 2012, 62 patients fulfilled the inclusion criteria for distant metastasis (cM1) and comparable computed tomography or magnetic resonance tomography scans in the Picture Archiving Communication System of our center before and after neoadjuvant chemotherapy. Follow-up data and clinicopathological characteristics were collected from a prospective database and retrospectively investigated. The chemotherapeutic response to liver metastasis was evaluated according to the Response Evaluation Criteria in Solid Tumors criteria 1.1. RESULTS Comparing the average reduction of measured lesions, a significant response to chemotherapy was detected in the patients receiving RAAS-I (n = 24) compared to those who did not (n = 38) (P = 0.031). Interestingly, the effect was more distinctive when the size reduction was compared between high responses with more than 50% size reduction of all measured lesions (P = 0.011). In the subgroup analysis of patients receiving bevacizumab treatment, high responses to chemotherapy were observed only in the RAAS-I cohort (28.6% versus 0%, P = 0.022). CONCLUSIONS For neoadjuvantly treated patients, concomitant antihypertensive treatment with RAAS-I showed a higher total size reduction of liver metastasis as a sign of treatment response, especially in combination with antiangiogenic treatment with bevacizumab.
Collapse
Affiliation(s)
- Naita M Wirsik
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany; Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany
| | - Pia C Appel
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexander Braun
- Department of Radiology, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | - Henrik Nienhüser
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Schneider
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Schmidt
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany; Department of General, Visceral, Cancer and Transplant Surgery, University Hospital of Cologne, Cologne, Germany.
| |
Collapse
|
46
|
González NS, Marchese PV, Baraibar I, Ros J, Salvà F, Rodríguez M, Salvà C, Vaghi C, Alcaraz A, García A, Tabernero J, Élez E. Epidermal growth factor receptor antagonists in colorectal cancer: emerging strategies for precision therapy. Expert Opin Investig Drugs 2024; 33:613-625. [PMID: 38775361 DOI: 10.1080/13543784.2024.2349287] [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: 02/23/2024] [Accepted: 04/25/2024] [Indexed: 06/25/2024]
Abstract
INTRODUCTION The global prevalence of colorectal cancer highlights the need to enhance treatment strategies for improved patient outcomes. The pivotal role of epidermal growth factor receptor (EGFR) signaling in regulating cellular processes for this disease pinpoints its value as a therapeutic target, despite the emergence of resistance mechanisms over time. AREAS COVERED This review discusses the clinical evidence supporting the use of EGFR inhibitors in molecularly-selected patients based on molecular characteristics (notably BRAF V600E and KRAS G12C) including combination approaches targeting different points in in the signaling pathway, as well as strategies such as EGFR inhibitor rechallenge. The role of HER2 inhibitors and emerging approaches such as bispecific antibodies are also reviewed. EXPERT OPINION Recently, inhibitors targeting the KRAS G12C variant have emerged, albeit with modest monotherapy activity compared to other tumor types, emphasizing the influence of histologic origins on the EGFR signaling pathway. Integration of EGFR inhibitors into precision medicine has facilitated tailored therapies addressing resistance mechanisms. Patient selection for EGFR inhibitor rechallenge guided by ctDNA findings is crucial, with ongoing investigations exploring novel combinations to enhance EGFR blockade, highlighting the transformative potential of precision medicine in shaping the future of mCRC treatment toward personalized and targeted approaches.
Collapse
Affiliation(s)
- Nadia Saoudi González
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | | | - Iosune Baraibar
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Javier Ros
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Francesc Salvà
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Marta Rodríguez
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Clara Salvà
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Caterina Vaghi
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Adriana Alcaraz
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ariadna García
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Josep Tabernero
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Elena Élez
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Vall d'Hebron Hospital Campus, Barcelona, Spain
| |
Collapse
|
47
|
Kinos S, Hagman H, Halonen P, Soveri LM, O'Reilly M, Pfeiffer P, Frödin JE, Sorbye H, Heervä E, Liposits G, Kallio R, Ålgars A, Ristamäki R, Salminen T, Bärlund M, Shah CH, McDermott R, Röckert R, Flygare P, Kwakman J, Teske A, Punt C, Glimelius B, Österlund P. Detailed analysis of metastatic colorectal cancer patients who developed cardiotoxicity on another fluoropyrimidine and switched to S-1 treatment (subgroup analysis of the CardioSwitch-study). Acta Oncol 2024; 63:248-258. [PMID: 38698698 PMCID: PMC11332541 DOI: 10.2340/1651-226x.2024.24023] [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: 11/21/2023] [Accepted: 02/16/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND AND PURPOSE The CardioSwitch-study demonstrated that patients with solid tumors who develop cardiotoxicity on capecitabine or 5-fluorouracil (5-FU) treatment can be safely switched to S-1, an alternative fluoropyrimidine (FP). In light of the European Medicines Agency approval of S-1 in metastatic colorectal cancer (mCRC), this analysis provides more detailed safety and efficacy information, and data regarding metastasectomy and/or local ablative therapy (LAT), on the mCRC patients from the original study. MATERIALS AND METHODS This retrospective cohort study was conducted at 12 European centers. The primary endpoint was recurrence of cardiotoxicity after switch. For this analysis, safety data are reported for 78 mCRC patients from the CardioSwitch cohort (N = 200). Detailed efficacy and outcomes data were available for 66 mCRC patients. RESULTS Data for the safety of S-1 in mCRC patients were similar to the original CardioSwitch cohort and that expected for FP-based treatment, with no new concerns. Recurrent cardiotoxicity (all grade 1) with S-1-based treatment occurred in 4/78 (5%) mCRC patients; all were able to complete FP treatment. Median progression-free survival from initiation of S-1-based treatment was 9.0 months and median overall survival 26.7 months. Metastasectomy and/or LAT was performed in 33/66 (50%) patients, and S-1 was successfully used in recommended neoadjuvant/conversion or adjuvant-like combination regimens and schedules as for standard FPs. INTERPRETATION S-1 is a safe and effective FP alternative when mCRC patients are forced to discontinue 5-FU or capecitabine due to cardiotoxicity and can be safely used in the standard recommended regimens, settings, and schedules.
Collapse
Affiliation(s)
- Sampsa Kinos
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland; Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Helga Hagman
- Department of Oncology, Skåne University Hospital, Malmö, Sweden
| | - Päivi Halonen
- Department of Oncology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Leena-Maija Soveri
- Department of Oncology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Mary O'Reilly
- Department of Oncology, St Vincent's University Hospital and University College Dublin, Dublin, Ireland
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Jan-Erik Frödin
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Halfdan Sorbye
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Eetu Heervä
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Gabor Liposits
- Department of Oncology, Regional Hospital West Jutland, Hjørring, Denmark
| | - Raija Kallio
- Department of Oncology, Oulu University and University Hospital, Oulu, Finland
| | - Annika Ålgars
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Raija Ristamäki
- Department of Oncology, Turku University Hospital and University of Turku, Turku, Finland
| | - Tapio Salminen
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland; Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Maarit Bärlund
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland; Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Carl-Henrik Shah
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Ray McDermott
- Department of Oncology, St Vincent's University Hospital and University College Dublin, Dublin, Ireland
| | | | - Petra Flygare
- Department of Oncology, Sundsvall Hospital, Sundsvall, Sweden
| | - Johannes Kwakman
- Department of Medical Oncology, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Arco Teske
- Department of Cardiology, University Medical Centre, Utrecht University, Utrecht, The Netherlands
| | - Cornelis Punt
- Depatment of Epidemiology, Jules Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherland
| | | | - Pia Österlund
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland; Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland; Department of Oncology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland; Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden; rTema Cancer, Department of GI-cancer, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
48
|
Kato K, Noda T, Kobayashi S, Sasaki K, Iwagami Y, Yamada D, Tomimaru Y, Takahashi H, Uemura M, Asaoka T, Shimizu J, Doki Y, Eguchi H. KLK10 derived from tumor endothelial cells accelerates colon cancer cell proliferation and hematogenous liver metastasis formation. Cancer Sci 2024; 115:1520-1535. [PMID: 38475666 PMCID: PMC11093189 DOI: 10.1111/cas.16144] [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: 09/29/2023] [Revised: 02/06/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Tumor endothelial cells (TECs), which are thought to be structurally and functionally different from normal endothelial cells (NECs), are increasingly attracting attention as a therapeutic target in hypervascular malignancies. Although colorectal liver metastasis (CRLM) tumors are hypovascular, inhibitors of angiogenesis are a key drug in multidisciplinary therapy, and TECs might be involved in the development and progression of cancer. Here, we analyzed the function of TEC in the CRLM tumor microenvironment. We used a murine colon cancer cell line (CT26) and isolated TECs from CRLM tumors. TECs showed higher proliferation and migration than NECs. Coinjection of CT26 and TECs yielded rapid tumor formation in vivo. Immunofluorescence analysis showed that coinjection of CT26 and TECs increased vessel formation and Ki-67+ cells. Transcriptome analysis identified kallikrein-related peptide 10 (KLK10) as a candidate target. Coinjection of CT26 and TECs after KLK10 downregulation with siRNA suppressed tumor formation in vivo. TEC secretion of KLK10 decreased after KLK10 downregulation, and conditioned medium after KLK10 knockdown in TECs suppressed CT26 proliferative activity. Double immunofluorescence staining of KLK10 and CD31 in CRLM tissues revealed a significant correlation between poor prognosis and positive KLK10 expression in TECs and tumor cells. On multivariate analysis, KLK10 expression was an independent prognostic factor in disease-free survival. In conclusion, KLK10 derived from TECs accelerates colon cancer cell proliferation and hematogenous liver metastasis formation. KLK10 in TECs might offer a promising therapeutic target in CRLM.
Collapse
Affiliation(s)
- Kazuya Kato
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Takehiro Noda
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Kazuki Sasaki
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Yoshifumi Iwagami
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Daisaku Yamada
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Yoshito Tomimaru
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Hidenori Takahashi
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Mamoru Uemura
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Tadafumi Asaoka
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Junzo Shimizu
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of MedicineOsaka UniversityOsakaJapan
| |
Collapse
|
49
|
Yang W, Chen D, Niu Y, Wu G, Huang Z, Bi X, Zhao H, Che X, Sun Y. FOLFOXIRI plus cetuximab as conversion therapy for unresectable RAS/BRAF wild-type left-sided colorectal cancer with liver-limited metastases: a prospective dual-center pilot study. Front Oncol 2024; 14:1375906. [PMID: 38638850 PMCID: PMC11024419 DOI: 10.3389/fonc.2024.1375906] [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: 01/24/2024] [Accepted: 03/20/2024] [Indexed: 04/20/2024] Open
Abstract
Purpose To explore the efficacy and safety of FOLFOXIRI plus cetuximab regimen as conversion therapy for patients with unresectable RAS/BRAF wild-type colorectal liver-limited metastases (CLM). Patients and methods This was a dual-center, phase II trial with the rate of no evidence of disease (NED) achieved as the primary endpoint. All enrolled patients with initially unresectable left-sided RAS/BRAF wild-type colorectal liver-limited metastases received a modified FOLFOXIRI plus cetuximab regimen as conversion therapy. Results Between October 2019 and October 2021, fifteen patients were enrolled. Nine patients (60%) achieved NED. The overall response rate (ORR) was 92.9%, and the disease control rate (DCR) was 100%. The median relapse-free survival (RFS) was 9 (95% CI: 0-20.7) months. The median progression-free survival (PFS) was 13.0 months (95% CI: 5.7-20.5), and the median overall survival (OS) was not reached. The most frequently occurring grade 3-4 adverse events were neutropenia (20%), peripheral neurotoxicity (13.3%), diarrhea (6.7%), and rash acneiform (6.7%). Conclusion The FOLFOXIRI plus cetuximab regimen displayed tolerable toxicity and promising anti-tumor activity in terms of the rate of NED achieved and response rate in patients with initially unresectable left-sided RAS/BRAF wild-type CLM. This regimen merits further investigation.
Collapse
Affiliation(s)
- Wenwei Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dong Chen
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yaru Niu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guifu Wu
- Department of Medical Oncology, Beijing Chaoyang District Sanhuan Cancer Hospital, Beijing, China
| | - Zhangkan Huang
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Xinyu Bi
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong Zhao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Che
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongkun Sun
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
50
|
Pan QZ, Zhao JJ, Liu L, Zhang DS, Wang LP, Hu WW, Weng DS, Xu X, Li YZ, Tang Y, Zhang WH, Li JY, Zheng X, Wang QJ, Li YQ, Xiang T, Zhou L, Yang SN, Wu C, Huang RX, He J, Du WJ, Chen LJ, Wu YN, Xu B, Shen Q, Zhang Y, Jiang JT, Ren XB, Xia JC. XELOX (capecitabine plus oxaliplatin) plus bevacizumab (anti-VEGF-A antibody) with or without adoptive cell immunotherapy in the treatment of patients with previously untreated metastatic colorectal cancer: a multicenter, open-label, randomized, controlled, phase 3 trial. Signal Transduct Target Ther 2024; 9:79. [PMID: 38565886 PMCID: PMC10987514 DOI: 10.1038/s41392-024-01788-2] [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: 09/11/2023] [Revised: 01/30/2024] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Fluoropyrimidine-based combination chemotherapy plus targeted therapy is the standard initial treatment for unresectable metastatic colorectal cancer (mCRC), but the prognosis remains poor. This phase 3 trial (ClinicalTrials.gov: NCT03950154) assessed the efficacy and adverse events (AEs) of the combination of PD-1 blockade-activated DC-CIK (PD1-T) cells with XELOX plus bevacizumab as a first-line therapy in patients with mCRC. A total of 202 participants were enrolled and randomly assigned in a 1:1 ratio to receive either first-line XELOX plus bevacizumab (the control group, n = 102) or the same regimen plus autologous PD1-T cell immunotherapy (the immunotherapy group, n = 100) every 21 days for up to 6 cycles, followed by maintenance treatment with capecitabine and bevacizumab. The main endpoint of the trial was progression-free survival (PFS). The median follow-up was 19.5 months. Median PFS was 14.8 months (95% CI, 11.6-18.0) for the immunotherapy group compared with 9.9 months (8.0-11.8) for the control group (hazard ratio [HR], 0.60 [95% CI, 0.40-0.88]; p = 0.009). Median overall survival (OS) was not reached for the immunotherapy group and 25.6 months (95% CI, 18.3-32.8) for the control group (HR, 0.57 [95% CI, 0.33-0.98]; p = 0.043). Grade 3 or higher AEs occurred in 20.0% of patients in the immunotherapy group and 23.5% in the control groups, with no toxicity-associated deaths reported. The addition of PD1-T cells to first-line XELOX plus bevacizumab demonstrates significant clinical improvement of PFS and OS with well tolerability in patients with previously untreated mCRC.
Collapse
Affiliation(s)
- Qiu-Zhong Pan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Jing-Jing Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Liang Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, PR China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, PR China
- Department of Biotherapy/Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Dong-Sheng Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Li-Ping Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Wen-Wei Hu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China
| | - De-Sheng Weng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Xiang Xu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, PR China
| | - Yi-Zhuo Li
- Imaging Diagnosis and Interventional Center, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Yan Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Wei-Hong Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, PR China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, PR China
- Department of Biotherapy/Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Jie-Yao Li
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China
| | - Qi-Jing Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Yong-Qiang Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Tong Xiang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Li Zhou
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, PR China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, PR China
- Department of Biotherapy/Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Shuang-Ning Yang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Chen Wu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China
| | - Rong-Xing Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Jia He
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Wei-Jiao Du
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, PR China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, PR China
- Department of Biotherapy/Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Lu-Jun Chen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China
| | - Yue-Na Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China
| | - Bin Xu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China
| | - Qiong Shen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China
| | - Yi Zhang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Jing-Ting Jiang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, PR China.
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, 213003, PR China.
- Institute for Cell Therapy of Soochow University, Changzhou, Jiangsu, 213003, PR China.
| | - Xiu-Bao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, PR China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, 300060, PR China.
- Department of Biotherapy/Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China.
| | - Jian-Chuan Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China.
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, 510060, PR China.
| |
Collapse
|