1
|
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
|
2
|
Li M, Meindl-Beinker NM, Maenz M, Betge J, Schulte N, Zhan T, Hofheinz RD, Vogel A, Angermeier S, Bolling C, de Wit M, Jakobs R, Karthaus M, Stocker G, Thuss-Patience P, Leidig T, Bauer H, Ebert MP, Haertel N. Functional status and quality of life in older patients with advanced esophageal squamous cell cancer receiving second-line nivolumab ± ipilimumab therapy: A post hoc analysis of the phase 2, multicenter RAMONA study. J Geriatr Oncol 2024; 15:101838. [PMID: 39097500 DOI: 10.1016/j.jgo.2024.101838] [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: 12/13/2023] [Revised: 04/18/2024] [Accepted: 07/19/2024] [Indexed: 08/05/2024]
Abstract
INTRODUCTION The phase 2 RAMONA study demonstrated that second-line nivolumab ± ipilimumab immunotherapy was feasible and effective in older patients with advanced esophageal squamous cell cancer (ESCC). Here, we presented results from functional status (FS) and quality-of-life (QoL) analyses. MATERIALS AND METHODS Patients aged ≥65 years with advanced ESCC and disease progression following first-line therapy were enrolled for study treatment with nivolumab ± ipilimumab. Geriatric assessments (GA) consisting of G8 and GoGo/SlowGo evaluation, and quality of life (QoL) assessments with EORTC QLQ-C30 questionnaires were conducted at baseline and during the treatment. A post hoc analysis was performed to compare therapy efficacy, toxicity, and QoL between age groups (≥70 years vs. <70 years) and functionality groups (G8 > 14 vs. ≤14 and GoGo vs. SlowGo). RESULTS In 66 treated patients with a median age of 70.5 years, older patients had non-inferior overall survival and tumor response compared to younger patients, with no increased treatment-related adverse events. Fitter patients (G8 > 14, GoGo) had a clinically, yet not statistically significant, survival advantage than less fit patients (G8 ≤ 14, SlowGo) patients. Moreover, FS by G8 and GoGo/SlowGo significantly correlated with QoL. Overall, QoL was impaired at baseline but remained stable in all scales over the course of immunotherapy. DISCUSSION The administration of nivolumab ± ipilimumab second-line immunotherapy in older patients with ESCC did not show age-dependent effects and maintained QoL. GA could identify functional deficits and limitations of QoL and should be implemented in the context of immunotherapy. CLINICALTRIALS gov: NCT03416244.
Collapse
Affiliation(s)
- Moying Li
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Nadja M Meindl-Beinker
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Martin Maenz
- Cermed Contract Research GmbH, Friedrichstr. 94, 10117 Berlin, Germany
| | - Johannes Betge
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Nadine Schulte
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Tianzuo Zhan
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Ralf-Dieter Hofheinz
- Mannheim Cancer Center, Outpatient Department, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Arndt Vogel
- Division of Gastroenterology and Hepatology, Toronto General Hospital, Toronto, Canada; Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Hannover Medical School, Hannover, Germany
| | - Stefan Angermeier
- Department of Internal Medicine I, Klinikum Ludwigsburg, Posilipostraße 4, 71640 Ludwigsburg, Germany
| | - Claus Bolling
- Department of Internal Medicine - Hematology and Medical Oncology, Sana Klinikum Offenbach, Starkenburgring 66, 63069, Offenbach am Main, Germany
| | - Maike de Wit
- Department of Internal Medicine - Hematology, Oncology and Palliative Medicine, Vivantes Klinikum Neukölln, Rudower Str. 48, 12351 Berlin, Germany
| | - Ralf Jakobs
- Department of Medicine C, Klinikum der Stadt Ludwigshafen, Bremserstr. 79, 67063, Ludwigshafen, Germany
| | - Meinolf Karthaus
- Department of Hematology, Oncology and Palliative Care, Klinikum Neuperlach, Oskar-Maria-Graf-Ring 51, 81737 München, Germany
| | - Gertraud Stocker
- Department of Medicine (Oncology, Gastroenterology, Hepatology and Pulmonology) and University Cancer Center, University Medicine Leipzig, Liebigstr. 20, 04103 Leipzig, Germany
| | - Peter Thuss-Patience
- Department of Hematology, Medical Oncology and Tumor Immunology, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Tobias Leidig
- CROLLL GmbH, Woernitzstraße 115A, 90449 Nuremberg, Germany
| | - Hans Bauer
- Staburo GmbH, Aschauer Straße 26a, 81549 Munich, Germany
| | - Matthias P Ebert
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Mannheim Institute for Innate Immunity, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Nicolai Haertel
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany.
| |
Collapse
|
3
|
Ebert MP, Fischbach W, Hollerbach S, Höppner J, Lorenz D, Stahl M, Stuschke M, Pech O, Vanhoefer U, Porschen R. S3-Leitlinie Diagnostik und Therapie der Plattenepithelkarzinome und Adenokarzinome des Ösophagus. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:535-642. [PMID: 38599580 DOI: 10.1055/a-2239-9802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Affiliation(s)
- Matthias P Ebert
- II. Medizinische Klinik, Medizinische Fakultät Mannheim, Universitätsmedizin, Universität Heidelberg, Mannheim
- DKFZ-Hector Krebsinstitut an der Universitätsmedizin Mannheim, Mannheim
- Molecular Medicine Partnership Unit, EMBL, Heidelberg
| | - Wolfgang Fischbach
- Deutsche Gesellschaft zur Bekämpfung der Krankheiten von Magen, Darm und Leber sowie von Störungen des Stoffwechsels und der Ernährung (Gastro-Liga) e. V., Giessen
| | | | - Jens Höppner
- Klinik für Allgemeine Chirurgie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Lübeck
| | - Dietmar Lorenz
- Chirurgische Klinik I, Allgemein-, Viszeral- und Thoraxchirurgie, Klinikum Darmstadt, Darmstadt
| | - Michael Stahl
- Klinik für Internistische Onkologie und onkologische Palliativmedizin, Evang. Huyssensstiftung, Evang. Kliniken Essen-Mitte, Essen
| | - Martin Stuschke
- Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Essen, Essen
| | - Oliver Pech
- Klinik für Gastroenterologie und Interventionelle Endoskopie, Krankenhaus Barmherzige Brüder, Regensburg
| | - Udo Vanhoefer
- Klinik für Hämatologie und Onkologie, Katholisches Marienkrankenhaus, Hamburg
| | - Rainer Porschen
- Gastroenterologische Praxis am Kreiskrankenhaus Osterholz, Osterholz-Scharmbeck
| |
Collapse
|
4
|
Angerilli V, Ghelardi F, Nappo F, Grillo F, Parente P, Lonardi S, Luchini C, Pietrantonio F, Ugolini C, Vanoli A, Fassan M. Claudin-18.2 testing and its impact in the therapeutic management of patients with gastric and gastroesophageal adenocarcinomas: A literature review with expert opinion. Pathol Res Pract 2024; 254:155145. [PMID: 38277741 DOI: 10.1016/j.prp.2024.155145] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024]
Abstract
Claudin-18.2 (CLDN18.2) is a member of the tight junction protein family and is a highly selective biomarker with frequent abnormal expression during the occurrence and development of various primary malignant tumors, including gastric cancer (GC) and esophago-gastric junction adenocarcinomas (EGJA). For these reasons, CLDN18.2 has been investigated as a therapeutic target for GC/EGJA malignancies. Recently, zolbetuximab has been proposed as a new standard of care for patients with CLDN18.2-positive, HER2-negative, locally advanced and metastatic GC/EGJA. The use of CLDN18 IHC assays to select patients who might benefit from anti-CLDN18.2 therapy is currently entering clinical practice. In this setting, pathologists play a central role in therapeutic decision-making. Accurate biomarker assessment is essential to ensure the best therapeutic option for patients. In the present review, we provide a comprehensive overview of available evidence on CLDN18.2 testing and its impact on the therapeutic management of patients with GC/EGJA, as well as some practical suggestions for CLDN18.2 staining interpretation and potential pitfalls in the real-world setting.
Collapse
Affiliation(s)
- Valentina Angerilli
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Filippo Ghelardi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Floriana Nappo
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Federica Grillo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Italy.
| | - Paola Parente
- Unit of Pathology, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy.
| | - Sara Lonardi
- Medical Oncology 3, Istituto Oncologico Veneto IOV-IRCCS, Padua, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Clara Ugolini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy; Veneto Institute of Oncology (IOV-IRCCS), Padua, Italy
| |
Collapse
|
5
|
Authors, und die Mitarbeiter der Leitlinienkommission, Collaborators:. S3-Leitlinie Diagnostik und Therapie der Plattenepithelkarzinome und Adenokarzinome des Ösophagus. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2023; 61:e209-e307. [PMID: 37285869 DOI: 10.1055/a-1771-6953] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
|
6
|
YILDIRIM S, Özveren A. Second-Line Chemotherapy in Gastric Cancer: a Retrospective Study. Indian J Surg Oncol 2023; 14:423-427. [PMID: 37324318 PMCID: PMC10267055 DOI: 10.1007/s13193-022-01695-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023] Open
Abstract
Second-line chemotherapy is recommended for patients who have disease progression after first-line chemotherapy and have a good performance status. The aim of our study is thus to determine which chemotherapy regimen is more appropriate for second-line gastric cancer treatment. Patients were included if they met the following inclusion criteria: metastatic gastric adenocarcinoma pathology; no previous treatment for local gastric cancer (surgery, chemotherapy, or radiotherapy); received first-line chemotherapy for metastatic gastric cancer and had the disease progress afterward; had adequate organ functions for second-line chemotherapy; had an Eastern Cooperative Oncology Group (ECOG) score 0-2; and were HER-2 negative. The patients were examined in three groups according to the second-line chemotherapy regimen they received. These three groups were compared in terms of overall and progression-free survival. The three groups were statistically similar in overall survival, which was the primary endpoint of the study; the median overall survival was 5 months in the FOLFIRI group (n = 79), 6.5 months in the platinum-based group (n = 55), and 5.6 months in the taxane-based group (n = 40) (p = 0.554). There was no statistical difference between the groups' progression-free survival either; the median progression-free survival time was 3.43 months in the FOLFIRI group, 4 months in the platinum-based group, and 2.77 months in the taxane-based group (p = 0.546). There was no statistically significant difference between the three irinotecan-based, platinum-based, and taxane-based treatments. According to our study's results, the chemotherapy given in second-line treatment should be decided on an individual basis according to toxicity and cost.
Collapse
Affiliation(s)
- Serkan YILDIRIM
- Medical Oncology Department MD, Başkent University Hospital, Konya, Turkey 42080
| | - Ahmet Özveren
- Medical Oncology Department MD, İzmir Kent Hospital, Izmir, Turkey 35070
| |
Collapse
|
7
|
Lin YT, Liu TX, Chen J, Wang C, Chen Y. Cost-Effectiveness of Nivolumab Immunotherapy vs. Paclitaxel or Docetaxel Chemotherapy as Second-Line Therapy in Advanced Esophageal Squamous Cell Carcinoma in China. Front Public Health 2022; 10:923619. [PMID: 35844891 PMCID: PMC9277084 DOI: 10.3389/fpubh.2022.923619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/09/2022] [Indexed: 12/24/2022] Open
Abstract
This study aimed to evaluate and compare nivolumab's cost-effectiveness with chemotherapy in patients with advanced esophageal squamous cell carcinoma from the Chinese healthcare system perspective. To this end, the researchers utilized a partitioned survival model with three mutually exclusive health stages. The characteristics of the patients used as inclusion and exclusion criteria in this model were the same as those used for patients with advanced esophageal squamous cell carcinoma in the ATTRACTION-3 study. The ATTRACTION-3 trial, which took place between January 7, 2016 and November 12, 2018, also yielded important clinical data. Data on medical and economic preferences were collected from real-world clinical practices. Costs, quality-adjusted life years, and incremental cost-effectiveness ratio were calculated for the two therapy options. The model uncertainty was investigated using a deterministic and probabilistic sensitivity analysis. When compared to chemotherapy, nivolumab was linked with an increase of 0.28 quality-adjusted life years with an increased cost of US$ 36,956.81 per patient in the base case analysis of a hypothetical sample of 419 patients. The incremental cost-effectiveness ratio in the deterministic sensitivity analysis was US$ 132,029.46/quality-adjusted life year, with a 48.02% probability of being cost-effective at willingness-to-pay thresholds of US$ 132,029.22/quality-adjusted life year. The incremental cost-effectiveness ratio remained greater than US$ 80,000/quality-adjusted life year in the deterministic sensitivity analyses. To be more cost-effective and remain below the threshold of 37,653 US$/quality-adjusted life year, which the Chinese population can afford, nivolumab's price would have to be lowered sharply by 53.50%. Nivolumab is clinically beneficial but not cost-effective when compared to chemotherapy. A substantial reduction in nivolumab's drug acquisition cost would be necessary to make it cost-effective for immunotherapy.
Collapse
Affiliation(s)
- Ying-tao Lin
- Department of Drug Clinical Trial Administration Office, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Tian-xiu Liu
- Department of Thoracic Radiotherapy, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Jian Chen
- Department of Surgical Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Chang Wang
- Department of Medical Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Ying Chen
- Department of Drug Clinical Trial Administration Office, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
8
|
Leone AG, Petrelli F, Ghidini A, Raimondi A, Smyth EC, Pietrantonio F. Efficacy and activity of PD-1 blockade in patients with advanced esophageal squamous cell carcinoma: a systematic review and meta-analysis with focus on the value of PD-L1 combined positive score. ESMO Open 2022; 7:100380. [PMID: 35093742 PMCID: PMC8804258 DOI: 10.1016/j.esmoop.2021.100380] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/09/2021] [Accepted: 12/20/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Recently, several randomized controlled trials (RCTs) investigated immunotherapy-based regimens versus chemotherapy alone in patients with advanced esophageal squamous cell carcinoma (ESCC). Here we conducted a systematic review and meta-analysis on the efficacy and activity of programmed cell death protein 1 blockade in these patients, with focus on the value of programmed death-ligand 1 combined positive score (CPS) for selecting patients who may benefit the most. METHODS RCTs investigating treatment with or without immune checkpoint inhibitors for advanced ESCC were selected. The hazard ratio (HR) and the odds ratio were used to compare the treatment effect on survival outcomes and tumor response, respectively, for immunotherapy-based regimens compared with standard chemotherapy, overall and according to geographic region or treatment line. We carried out a subgroup analysis comparing patients with CPS ≥10 or <10 and the evidence for treatment effect was evaluated by interaction test. RESULTS A total of 5257 patients and 10 RCTs were included. Overall, the HR for overall survival benefit with immunotherapy-based regimens was 0.71 [95% confidence interval (CI) 0.66-0.76] compared with chemotherapy alone; such effect was independent from geographical region (Asia versus rest of the world) and treatment line (upfront versus second/further lines). The HR for progression-free survival benefit and the odds ratio for overall response rate increase were 0.78 (95% CI 0.66-0.93) and 1.50 (95% CI 1.22-1.83), respectively. The HR for overall survival benefit with immunotherapy-based treatment was 0.60 (95% CI 0.51-0.70) for CPS ≥10 subgroup versus 0.83 (95% CI 0.69-1.00) for CPS <10 (P for interaction 0.009). CONCLUSIONS Immune checkpoint inhibitors have a consistent benefit in reducing the risk of death for ESCC patients which is dependent on programmed death-ligand 1 CPS status. Further investigations of biomarkers for immunotherapy in the subgroup of patients with CPS <10 are needed.
Collapse
Affiliation(s)
- A G Leone
- Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori di Milano, Milan, Italy
| | - F Petrelli
- Oncology Unit, Medical Sciences Department, ASST Bergamo Ovest, Treviglio, Italy
| | - A Ghidini
- Oncology Unit, Casa di Cura Igea, Milan, Italy
| | - A Raimondi
- Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori di Milano, Milan, Italy
| | - E C Smyth
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - F Pietrantonio
- Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori di Milano, Milan, Italy.
| |
Collapse
|
9
|
Danese M, Gricar J, Abraham P. Real-world outcomes with second-line therapy in advanced esophageal squamous cell carcinoma using SEER-Medicare data. Future Oncol 2022; 18:927-936. [PMID: 35081734 DOI: 10.2217/fon-2021-0460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: To characterize real-world patterns of second-line treatment and outcomes in older patients with advanced/metastatic esophageal squamous cell carcinoma (ESCC). Patients and methods: Patients aged ≥66 years diagnosed with advanced/metastatic ESCC between 2010 and 2015 and followed through 2016 were included in this retrospective analysis using SEER-Medicare data. Results: Of 756 patients with advanced/metastatic ESCC, 104 (14%) received second-line therapy; median duration of treatment was 1.5 months. Median overall survival was 5.7 months for all patients receiving second-line treatment, and 4.5, 5.6 and 8.5 months, respectively, for patients receiving taxane monotherapy, taxane combination therapy and nontaxane therapy. Conclusion: A small proportion of patients with advanced/metastatic ESCC received second-line therapy, which was associated with short duration of treatment and poor overall survival.
Collapse
Affiliation(s)
- Mark Danese
- Outcomes Insights, Inc., Agoura Hills, CA, USA
| | | | | |
Collapse
|
10
|
Lin YT, Chen Y, Liu TX, Kuang F, Huang P. Cost-Effectiveness Analysis of Camrelizumab Immunotherapy versus Docetaxel or Irinotecan Chemotherapy as Second-Line Therapy for Advanced or Metastatic Esophageal Squamous Cell Carcinoma. Cancer Manag Res 2021; 13:8219-8230. [PMID: 34754242 PMCID: PMC8572144 DOI: 10.2147/cmar.s335515] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/24/2021] [Indexed: 01/18/2023] Open
Abstract
Purpose The aim of this study was to assess the cost-effectiveness of camrelizumab immunotherapy versus docetaxel or irinotecan chemotherapy as second-line therapy for advanced esophageal squamous cell carcinoma (ESCC), which was evaluated in the ESCORT trial. Materials and Methods A partitioned survival model was developed to reflect the costs and effectiveness of the ESCORT trial. The clinical efficacy data, safety data, and health-related costs and utilities were derived from published data from clinical trials or health administration departments in China. Adverse event-related costs, drug administration, and other expenses were derived from a single center of Fujian Medical University Cancer Hospital in 2021. All survival analyses were performed with SPSS software. Overall survival was estimated with the Kaplan-Meier method, and progression-free survival was estimated with the life table method. Sensitivity analyses were conducted to assess the uncertainty of the model. Incremental cost, quality-adjusted life years (QALYs), and the incremental cost-effectiveness ratio (ICER) were calculated. Results Camrelizumab therapy had 0.232 QALYs at an incremental cost of USD$9959.44 compared with the chemotherapy group with 0.158 QALYs at an incremental cost of USD$8601.67. The ICER was USD$18393.12/QALY. Probabilistic sensitivity analyses showed that when the willingness-to-pay threshold reached USD$31200/QALY, which is nearly three times the Chinese gross domestic product per capita, camrelizumab had an 80% possibility of being cost-effective versus docetaxel or irinotecan chemotherapy. Conclusion Camrelizumab is a cost-effective option compared with docetaxel or irinotecan chemotherapy in patients with advanced ESCC as second-line therapy in China.
Collapse
Affiliation(s)
- Ying-Tao Lin
- Administration Office of Drug Clinical Trial, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, People's Republic of China
| | - Ying Chen
- Management Office of Science and Technology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Tian-Xiu Liu
- Department of Thoracic Radiotherapy, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Fang Kuang
- Administration Office of Drug Clinical Trial, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, People's Republic of China
| | - Ping Huang
- Administration Office of Drug Clinical Trial, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, People's Republic of China
| |
Collapse
|
11
|
Müller M, Posch F, Kiem D, Barth D, Horvath L, Stotz M, Schaberl-Moser R, Pichler M, Greil R, Jost PJ, Seeber A, Amann A, Schlick K, Gerger A, Riedl JM. Benefit of second-line therapy for advanced esophageal squamous cell carcinoma: a tri-center propensity score analysis. Ther Adv Med Oncol 2021; 13:17588359211039930. [PMID: 34616490 PMCID: PMC8488508 DOI: 10.1177/17588359211039930] [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: 03/25/2021] [Accepted: 07/26/2021] [Indexed: 12/24/2022] Open
Abstract
Background The level of evidence for palliative second-line therapy in advanced esophageal squamous cell carcinoma (aESCC) is limited. This is the first study that reports efficacy data comparing second-line therapy + active symptom control (ASC) versus ASC alone in aESCC. Methods We conducted a tri-center retrospective cohort study (n = 166) including patients with aESCC who had experienced disease progression on palliative first-line therapy. A propensity score model using inverse probability of treatment weighting (IPTW) was implemented for comparative efficacy analysis of overall survival (OS) in patients with second-line + ASC (n = 92, 55%) versus ASC alone (n = 74, 45%). Results The most frequent second-line regimens used were docetaxel (36%) and paclitaxel (18%). In unadjusted primary endpoint analysis, second-line + ASC was associated with significantly longer OS compared with ASC alone [hazard ratio (HR) = 0.49, 95% confidence interval (CI): 0.35-0.69, p < 0.0001]. However, patients in the second-line + ASC group were characterized by more favorable baseline features including a better Eastern Cooperative Oncology Group (ECOG) performance status, a longer first-line treatment duration and lower C-reactive protein levels. After rigorous adjusting for baseline confounders by re-weighting the data with the IPTW the favorable association between second-line and longer OS weakened but prevailed. The median OS was 6.1 months in the second-line + ASC group and 3.2 months in the ASC group, respectively (IPTW-adjusted HR = 0.40, 95% CI: 0.24-0.69, p = 0.001). Importantly, the benefit of second-line was consistent across several clinical subgroups, including patients with ECOG performance status ⩾1 and age ⩾65 years. The most common grade 3 or 4 adverse events associated with palliative second-line therapy were hematological toxicities. Conclusion This real-world study supports the concept that systemic second-line therapy prolongs survival in patients with aESCC.
Collapse
Affiliation(s)
- Moritz Müller
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Florian Posch
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Dominik Kiem
- IIIrd Medical Department of Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectious Disease, Salzburg Cancer Research Institute, Paracelsus Medical University, Salzburg, Austria
| | - Dominik Barth
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Lena Horvath
- Department of Internal Medicine V: Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Stotz
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Renate Schaberl-Moser
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Richard Greil
- IIIrd Medical Department of Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectious Disease, Salzburg Cancer Research Institute, Paracelsus Medical University, Salzburg, Austria
| | - Philipp J Jost
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Andreas Seeber
- Department of Internal Medicine V: Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Arno Amann
- Department of Internal Medicine V: Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Konstantin Schlick
- IIIrd Medical Department of Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectious Disease, Salzburg Cancer Research Institute, Paracelsus Medical University, Salzburg, Austria
| | - Armin Gerger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Jakob M Riedl
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, Graz 8036, Austria
| |
Collapse
|
12
|
Shi J, Zhang X, Li J, Huang W, Wang Y, Wang Y, Qin J. MTA2 sensitizes gastric cancer cells to PARP inhibition by induction of DNA replication stress. Transl Oncol 2021; 14:101167. [PMID: 34280886 PMCID: PMC8313750 DOI: 10.1016/j.tranon.2021.101167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitor olaparib selectively kills cancer cells with BRCA-deficiency and is approved for BRCA-mutated breast, ovarian and pancreatic cancers by FDA. However, phase III study of olaparib failed to show a significant improvement in overall survival in patients with gastric cancer (GC). To discover an effective biomarker for GC patient-selection in olaparib treatment, we analyzed proteomic profiling of 12 GC cell lines. MTA2 was identified to confer sensitivity to olaparib by aggravating olaparib-induced replication stress in cancer cells. Mechanistically, we applied Cleavage Under Targets and Tagmentation assay to find that MTA2 proteins preferentially bind regions of replication origin-associated DNA sequences, which could be enhanced by olaparib treatment. Furthermore, MTA2 was validated here to render cancer cells susceptible to combination of olaparib with ATR inhibitor AZD6738. In general, our study identified MTA2 as a potential biomarker for olaparib sensitivity by aggravating olaparib-induced replication stress.
Collapse
Affiliation(s)
- Jinwen Shi
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiaofeng Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jin'e Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Wenwen Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Yini Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.
| |
Collapse
|
13
|
Sun W, Zou X, Zhang W, Hu S, Ge K. [Clinical efficacy of anlotinib plus S-1 as a second-line therapy for recurrent or metastatic esophageal squamous cell carcinoma]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:250-255. [PMID: 33624599 DOI: 10.12122/j.issn.1673-4254.2021.02.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To investigate the efficacy of anlotinib plus S-1 for treatment of patients with recurrent or metastatic esophageal squamous cell carcinoma with failed first-line chemotherapy. OBJECTIVE Twenty-six patients with recurrent or metastatic esophageal squamous cell carcinoma patients who experienced progression after first-line paclitaxel plus platinum chemotherapy in our hospital between July, 2018 and February, 2020 were enrolled in this study. The patients received oral anlotinib along with S-1 treatment (anlotinib at 12 mg once daily and S-1 at 50 mg twice daily for two weeks; 3 weeks per cycle). The objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS) and adverse effects were evaluated for all the patients. OBJECTIVE No complete remission (CR) was observed in the 26 patients. Partial remission (PR) was achieved in 6 cases, stable disease (SD) in 12 cases, and progressive disease (PD) occurred in 8 cases, with an ORR of 23.1% and a DCR of 69.2% in these patients. The median PFS was 4.5 months (95%CI: 2.7-6.4 months). Univariate analysis showed that the patients with moderate or high tumor differentiation had significantly longer PFS than those with low tumor differentiation (6.1 months vs 1.9 months, P < 0.05). Multivariate analysis suggested that pathological differentiation grade (HR=6.778, 95%CI: 1.997-23.012) was an independent factor for a prolonged PFS. The adverse effects in the patients included mainly fatigue, hypertension and hand-foot syndrome, mostly of grade 1 to 2. OBJECTIVE Patients with recurrent or metastatic esophageal squamous cell carcinoma can benefit from a second-line anlotinib plus S-1 treatment, which has relatively mild adverse effects with a good safety profile.
Collapse
Affiliation(s)
- W Sun
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - X Zou
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - W Zhang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - S Hu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - K Ge
- Depaetment of Oncology, Liyang Hospital of Chinese Medicine, Jiangsu Provincial Hospital of Chinese Medicine Liyang Branch, Liyang 213300, China
| |
Collapse
|
14
|
Jin YQ, Miao DL. Multiomic Analysis of Methylation and Transcriptome Reveals a Novel Signature in Esophageal Cancer. Dose Response 2020; 18:1559325820942075. [PMID: 32728353 PMCID: PMC7364835 DOI: 10.1177/1559325820942075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Epigenetic alterations have been shown to lead to human carcinogenesis. The aim of this study was to perform an integrative analysis to develop an epigenetic signature to predict overall survival (OS) of esophageal cancer. Methods: DNA methylation and messenger RNA expression data of esophageal cancer samples were downloaded from The Cancer Genome Atlas database and were incorporated and analyzed using an R package MethylMix. Functional enrichment analysis of the methylation-related differentially expressed genes (DEGs) was performed. Epigenetic signature and nomogram associated with the OS of esophageal cancer were established by the multivariate Cox model. Results: A total of 71 methylation-related DEGs were identified. Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that these genes were involved in the biological process related to the initiation and progression of esophageal cancer. Two-gene (FAM24B and FAM200A) risk signature for OS was developed by multivariate Cox analysis, of which had high accuracy. The signature is independent of clinicopathological variables and indicated better predictive power than other clinicopathological variables. Moreover, we developed a novel prognostic nomogram based on risk score and 3 clinicopathological factors. Conclusions: Our study indicated possible methylation-related DEGs and established an epigenetic signature, which may provide novel insights for understanding the pathogenesis of esophageal cancer.
Collapse
Affiliation(s)
- Yi-Qi Jin
- Department of Intervention and Vascular Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Dong-Liu Miao
- Department of Intervention and Vascular Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
15
|
Abraham P, Gricar J, Zhang Y, Shankaran V. Real-World Treatment Patterns and Outcomes in Patients Receiving Second-Line Therapy for Advanced/Metastatic Esophageal Squamous Cell Carcinoma. Adv Ther 2020; 37:3392-3403. [PMID: 32533533 PMCID: PMC7467430 DOI: 10.1007/s12325-020-01394-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Currently available second-line (2L) therapies for advanced/metastatic esophageal squamous cell carcinoma (adv/met ESCC) include the taxanes paclitaxel and docetaxel. In clinical trials, such therapies have provided only modest improvements in survival. Few studies have assessed outcomes in routine clinical practice in the USA. We compared real-world clinical outcomes in the US for patients receiving taxane or non-taxane 2L therapy for adv/met ESCC. METHODS The Flatiron Health database was used to identify patients diagnosed with adv/met ESCC (1 January 2011-31 January 2019) who received 2L therapy; index date was date of adv/met diagnosis. Baseline variables and treatment regimens received were identified. Overall survival (OS; 2L start until death or last recorded medical activity) and duration of therapy (DoT; start of 2L therapy until last administration date of 2L therapy) in patients receiving taxane vs. non-taxane-based therapies in the 2L setting were estimated by Kaplan-Meier method. RESULTS There were no clear differences in baseline characteristics between patients who received 2L taxane therapy (n = 37) and 2L non-taxane therapy (n = 49). Median (95% CI) 2L OS was significantly longer with 2L taxanes (7.3 [5.9-11.5] months) vs. 2L non-taxanes (5.1 [2.9-7.6] months); median (95% CI) 2L DoT was 2.1 (1.8-3.0) months vs. 3.3 (2.6-6.7) months, respectively. CONCLUSION Survival was generally poor in patients receiving 2L therapy for adv/met ESCC and was longer in patients receiving 2L taxanes than 2L non-taxane therapy. Efficacious, tolerable therapies for ESCC in the 2L setting are urgently needed.
Collapse
Affiliation(s)
| | - Joe Gricar
- Bristol-Myers Squibb, Lawrenceville, NJ, USA
| | - Ying Zhang
- Bristol-Myers Squibb, Lawrenceville, NJ, USA
| | - Veena Shankaran
- Division of Oncology, Department of Medicine, Seattle Cancer Care Alliance, Seattle, WA, USA.
| |
Collapse
|
16
|
Genetic and epigenetic aberrations of ABCB1 synergistically boost the acquisition of taxane resistance in esophageal squamous cancer cells. Biochem Biophys Res Commun 2020; 526:586-591. [PMID: 32247608 DOI: 10.1016/j.bbrc.2020.03.114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/19/2020] [Indexed: 02/02/2023]
Abstract
Taxanes are applied as potent chemotherapeutic agents in the treatment of patients with esophageal cancer, but their usefulness is limited, partly because of acquisition of chemoresistance. In our previous study, we established three taxane resistant esophageal cancer cell lines; significant ABCB1 upregulations were found in all three. However, the responsible mechanism(s) still remains an open question. In this study, we explored possible mechanisms that might contribute to upregulation of ABCB1 in taxane resistant cells. ABCB1 gene amplification was found in taxane resistant cell line RTE-1P, but expressional upregulation cannot be explained only by gene amplification, because gene amplification is one order of magnitude or less whereas gene expression is more than two orders of magnitude. In the parental TE-1, ABCB1 expression was upregulated after treatment with 5-azadeoxycytidine and/or trichostatin A; epigenetic mechanisms may be deeply involved. ABCB1 has two promoters; a downstream promoter was found to play the dominant role in taxane resistant esophageal cancer cell lines. Analyses of CpG islands demonstrated that taxane resistant cells showed unmethylated CGI whereas parental cells were dominantly methylated. In conclusion, we propose that both the ABCB1 gene amplification and aberrations in epigenetic mechanisms are responsible for acquisition of taxane resistance in esophageal cancer cells.
Collapse
|
17
|
Yang Y, Jia J, Sun Z, Liu C, Xiao Y, Yu J, Du F, Zhang X. Association Between Multiple Lines of Active Therapy and Prognosis in Esophageal Squamous Cell Carcinoma. Cancer Manag Res 2020; 12:2177-2184. [PMID: 32273757 PMCID: PMC7102887 DOI: 10.2147/cmar.s242780] [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: 12/18/2019] [Accepted: 03/15/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose Esophageal squamous cell carcinoma (ESCC) is a critical threat to health and life. More than half of ESCC patients have recurrent or metastatic disease. Most late-stage patients undergo first-line treatment but experience further progression. Many of these patients have good performance status and are able to receive second-line therapy and even further treatments rather than best supportive care. Our analysis aimed to explore whether multiple lines of active treatment are beneficial in ESCC patients. Methods We conducted a retrospective cohort study. Univariate and multivariate survival analyses were used to identify whether the number of active treatment lines was related to prognosis. All analyses and the corresponding survival curves were based on the Cox proportional hazard regression model and the Kaplan–Meier method. Comparisons between groups were conducted using the t-test, chi-square test, and Fisher’s exact test, as applicable. Results Of a total of 138 patients with recurrent or metastatic disease, 66 (61.1%) received one line of active treatment, and 42 (38.9%) patients received two and more lines. Multiple lines of active therapy were statistically significantly associated with better prognosis (crude hazard ratio (HR) (95% confidence interval (CI))=0.21 (0.06–0.73)), even after adjusting for relevant confounders (adjusted HR (95% CI)=0.19 (0.04–0.86)). More grade 3–4 hepatotoxicity patients were observed in the multiple-line treatment group (p=0.033). A small number of patients were cured by palliative management; these patients were more likely to have received both systematic and local treatment than other patients with repeated progression (9/15 versus 40/117, p=0.051). Conclusion Multiple lines of active treatment are related to prolonged survival in recurrent and metastatic ESCC patients, and adverse effects are acceptable. Comprehensive therapy modalities are recommended.
Collapse
Affiliation(s)
- Ying Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), The VIP-II Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), The VIP-II Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhiwei Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), The VIP-II Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chuanling Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), The VIP-II Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanjie Xiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), The VIP-II Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jing Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), The VIP-II Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - Feng Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), The VIP-II Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaodong Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), The VIP-II Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
18
|
Vivaldi C, Catanese S, Massa V, Pecora I, Salani F, Santi S, Lencioni M, Vasile E, Falcone A, Fornaro L. Immune Checkpoint Inhibitors in Esophageal Cancers: are we Finally Finding the Right Path in the Mist? Int J Mol Sci 2020; 21:E1658. [PMID: 32121290 PMCID: PMC7084692 DOI: 10.3390/ijms21051658] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/26/2022] Open
Abstract
Esophageal cancer remains a challenging disease due to limited treatment options and poor prognosis. In recent years, immune checkpoint inhibitors (ICI) have been proven to be safe and effective in the treatment of highly lethal malignancies, such as non-small cell lung cancer and melanoma. Recent clinical trials also showed promising activity in immune checkpoint inhibitors in pretreated advanced esophageal carcinoma and a potentially significant impact on the outcome of selected patients, independently of histology. Combination studies evaluating immunotherapy and chemotherapy and, in localized disease, radiotherapy are in progress and will hopefully confirm their promises in the near future. However, reliable predictive biomarkers are still lacking. Indeed, at present, the role of programmed cell death ligand 1 expression and other factors (such as microsatellite instability and tumor mutational burden) as predictive biomarkers of benefit to immune checkpoint inhibitors is still controversial. Our aim was to explore the rationale of ICIs in esophageal cancer, review the results already available in multiple settings, and investigate future perspectives with single-agent and combination strategies.
Collapse
Affiliation(s)
- Caterina Vivaldi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via savi 10, 56126 Pisa PI, Italy;
| | - Silvia Catanese
- Unit of Medical Oncology, Pisa University Hospital, Via Roma 67, 56126 Pisa, Italy; (S.C.); (V.M.); (I.P.); (F.S.); (M.L.); (E.V.); (L.F.)
| | - Valentina Massa
- Unit of Medical Oncology, Pisa University Hospital, Via Roma 67, 56126 Pisa, Italy; (S.C.); (V.M.); (I.P.); (F.S.); (M.L.); (E.V.); (L.F.)
| | - Irene Pecora
- Unit of Medical Oncology, Pisa University Hospital, Via Roma 67, 56126 Pisa, Italy; (S.C.); (V.M.); (I.P.); (F.S.); (M.L.); (E.V.); (L.F.)
| | - Francesca Salani
- Unit of Medical Oncology, Pisa University Hospital, Via Roma 67, 56126 Pisa, Italy; (S.C.); (V.M.); (I.P.); (F.S.); (M.L.); (E.V.); (L.F.)
| | - Stefano Santi
- Esophageal Surgery Unit, Tuscany Regional Referral Center for the Diagnosis and Treatment of Esophageal Disease, Pisa University Hospital, Via Roma 67, 56126 Pisa, Italy;
| | - Monica Lencioni
- Unit of Medical Oncology, Pisa University Hospital, Via Roma 67, 56126 Pisa, Italy; (S.C.); (V.M.); (I.P.); (F.S.); (M.L.); (E.V.); (L.F.)
| | - Enrico Vasile
- Unit of Medical Oncology, Pisa University Hospital, Via Roma 67, 56126 Pisa, Italy; (S.C.); (V.M.); (I.P.); (F.S.); (M.L.); (E.V.); (L.F.)
| | - Alfredo Falcone
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via savi 10, 56126 Pisa PI, Italy;
| | - Lorenzo Fornaro
- Unit of Medical Oncology, Pisa University Hospital, Via Roma 67, 56126 Pisa, Italy; (S.C.); (V.M.); (I.P.); (F.S.); (M.L.); (E.V.); (L.F.)
| |
Collapse
|
19
|
Hong Y, Ding ZY. PD-1 Inhibitors in the Advanced Esophageal Cancer. Front Pharmacol 2019; 10:1418. [PMID: 31920637 PMCID: PMC6916418 DOI: 10.3389/fphar.2019.01418] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 11/07/2019] [Indexed: 02/05/2023] Open
Abstract
Esophageal cancer (EC) is a lethal disease, and ranks 7th in incidence and 6th in mortality worldwide. Patients are treated with surgery and/or chemoradiotherapy for a curative intent, but for those with advanced diseases systemic chemotherapy and targeted therapy are the mainstay treatment with poor prognosis. For the patients with squamous cell carcinoma and those progressed after chemotherapy, treatment option is even fewer, and effective treatment modalities are urgently needed. Preclinical and clinical studies have found the PD-1/PD-L1 inhibitors activate T lymphocytes, inhibit cancer growth, and improve survival in cancer patients. Multiple PD-1/PD-L1 inhibitors have been approved for the management of a variety of cancers. Interestingly, a large of proportion of EC patients have tumors with PD-L1 expression and high tumor mutation burden. Trials have been performed to evaluate the efficacy and safety of the PD-1/PD-L1 inhibitors in EC patients. This review will summarize the current progress in this field, especially the toxicities associated with these agents.
Collapse
Affiliation(s)
- Ye Hong
- Department of Biotherapy, Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Zhen-Yu Ding
- Department of Biotherapy, Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Kalikawe R, Baba Y, Nomoto D, Okadome K, Miyake K, Eto K, Hiyoshi Y, Nagai Y, Iwatsuki M, Ishimoto T, Iwagami S, Miyamoto Y, Yoshida N, Watanabe M, Baba H. Lysyl oxidase impacts disease outcomes and correlates with global DNA hypomethylation in esophageal cancer. Cancer Sci 2019; 110:3727-3737. [PMID: 31599475 PMCID: PMC6890447 DOI: 10.1111/cas.14214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 10/03/2019] [Accepted: 10/04/2019] [Indexed: 12/29/2022] Open
Abstract
Abnormal function of human body enzymes and epigenetic alterations such as DNA methylation have been shown to lead to human carcinogenesis. Lysyl oxidase (LOX) enzyme has attracted attention due to its involvement in tumor progression in various cancers. The purpose of this study was to clarify the clinical importance of LOX expression and its epigenetic regulation in the pathogenesis of esophageal squamous cell carcinoma (ESCC). Using a database of 284 ESCCs, we examined LOX expression and its prognostic characteristics. The functional role of LOX was assessed by in vitro growth, migration, and invasion assays. The relationship between LOX expression, global DNA hypomethylation (ie, LINE‐1 methylation), and LOX promoter methylation was evaluated by using mRNA expression arrays and pyrosequencing technology. High LOX expression cases had a significantly shorter overall survival and cancer‐specific survival (log‐rank, P < .001). The prognostic effect of LOX expression was not significantly modified by other clinical variables. Silencing and enzymatic inhibition of LOX suppressed growth and reduced the invasion and migration ability of ESCC cell lines along with the downregulation of AKT and MMP2. An integrated gene analysis in tissues and cell lines revealed that LOX was the most highly upregulated gene in LINE‐1 hypomethylated tumors. In vitro, LOX expression was upregulated following DNA demethylation. LOX promoter methylation was not associated with LOX expression. Conclusively LOX expression was associated with poor prognosis in ESCC and was regulated epigenetically by genome‐wide hypomethylation. It could serve as a prognostic biomarker in ESCC patients, and therapeutically targeting LOX could reverse the progression of esophageal cancer.
Collapse
Affiliation(s)
- Rebecca Kalikawe
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Daichi Nomoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuo Okadome
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Keisuke Miyake
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kojiro Eto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yukiharu Hiyoshi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Youhei Nagai
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shiro Iwagami
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto City, Japan
| |
Collapse
|
21
|
Kim EJ, Kim JH, Song HJ, Park SR, Kim YH, Kim HR, Kim SB. Impact of sequential lines of palliative chemotherapy in patients with recurrent/metastatic esophageal squamous cell carcinoma: A retrospective analysis of 107 patients at a single center. Asia Pac J Clin Oncol 2019; 16:e53-e62. [PMID: 31657877 DOI: 10.1111/ajco.13283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 10/04/2019] [Indexed: 01/08/2023]
Abstract
AIM This study was conducted to evaluate the efficacy of palliative chemotherapy by the lines of chemotherapy in recurrent/metastatic esophageal squamous cell carcinoma (ESCC) and to compare the efficacy between the patients with initially metastatic ESCC and those with recurrent/progressed ESCC after curative treatment. MATERIALS AND METHODS All 107 patients who began palliative chemotherapy for recurrent/metastatic ESCC from March 2015 to October 2017 were included, and grouped according to previous treatment: Groups A (previous chemoradiation alone, n = 30), B (previous surgery alone, n = 11), C (previous chemoradiation and surgery, n = 30), and D (initially metastatic or de novo stage IV, n = 36). Groups A, B, and C (pretreated group) and Group D (treatment-naïve group) were reorganized according to treatment history. Overall response rate (ORR) and survival data were retrospectively evaluated for each group, lines of chemotherapy, and chemotherapeutic regimen. RESULTS ORR was 25.2%, 7.3%, and 3.4% in first-, second-, and third-line chemotherapy, respectively. The median progression-free survival (PFS) was 4.7, 2.0, and 2.2 months in first-, second-, third-line chemotherapy, respectively. The median overall survival (OS) after first-line palliative chemotherapy was 10.1 months, and it was not significantly different between pretreated and treatment-naive groups. Previous surgery, good performance, ≥3 lines of chemotherapy, and low C-reactive protein level were linked to a significantly longer OS in multivariate analysis. CONCLUSION Because PFS rapidly declines with advancement of line of chemotherapy, incorporation of effective treatment modalities in early line treatments is crucial in the management of recurrent/metastatic ESCC. If tolerable, continuing advanced lines of chemotherapy may prolong survival.
Collapse
Affiliation(s)
- Eo Jin Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jong Hoon Kim
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ho June Song
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sook Ryun Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yong-Hee Kim
- Department of Thoracic Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyeong Ryul Kim
- Department of Thoracic Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
22
|
Pellino A, Riello E, Nappo F, Brignola S, Murgioni S, Djaballah SA, Lonardi S, Zagonel V, Rugge M, Loupakis F, Fassan M. Targeted therapies in metastatic gastric cancer: Current knowledge and future perspectives. World J Gastroenterol 2019; 25:5773-5788. [PMID: 31636471 PMCID: PMC6801189 DOI: 10.3748/wjg.v25.i38.5773] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/26/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) represents a leading cause of cancer related morbidity and mortality worldwide accounting for more than 1 million of newly diagnosed cases and thousands of deaths every year. In the last decade, the development of targeted therapies and the optimization of already available chemotherapeutic drugs has expanded the available treatment options for advanced GC and granted better survival expectations to the patients. At the same time, global efforts have been undertaken to investigate in detail the genomic and epigenomic heterogeneity of this disease, resulting in the identification of new specific and sensitive predictive and prognostic biomarkers and in innovative molecular classifications based on gene expression profiling. Nonetheless, several randomized studies aimed at exploring new innovative agents, such as immune checkpoint inhibitors, failed to demonstrate clinically meaningful survival advantages. Therefore, it is essential to further improve the molecular characterization of GC subgroups in order to provide researchers and medical oncologists with new tools for patients' selection and stratification in future clinical development programs and subsequent trials. The aim of the present manuscript is to provide a global overview of the recent molecular classifications from The Cancer Genome Atlas and the Asian Cancer Research Group and to present key promising developments in the field of immunotherapy and targeted therapies in metastatic GC.
Collapse
Affiliation(s)
- Antonio Pellino
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua 35100, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35100, Italy
| | - Erika Riello
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua 35100, Italy
- Surgical Pathology & Cytopathology Unit, Department of Medicine, University of Padua, Padua 35100, Italy
| | - Floriana Nappo
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua 35100, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35100, Italy
| | - Stefano Brignola
- Surgical Pathology & Cytopathology Unit, Department of Medicine, University of Padua, Padua 35100, Italy
| | - Sabina Murgioni
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua 35100, Italy
| | | | - Sara Lonardi
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua 35100, Italy
| | - Vittorina Zagonel
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua 35100, Italy
| | - Massimo Rugge
- Surgical Pathology & Cytopathology Unit, Department of Medicine, University of Padua, Padua 35100, Italy
- Veneto Cancer Registry, Padua 35100, Italy
| | - Fotios Loupakis
- Department of Oncology, Veneto Institute of Oncology IOV-IRCCS, Padua 35100, Italy
| | - Matteo Fassan
- Surgical Pathology & Cytopathology Unit, Department of Medicine, University of Padua, Padua 35100, Italy
| |
Collapse
|
23
|
Tong H, Liu Y, Jiang L, Wang J. Multi-Targeting by β-Elemene and Its Anticancer Properties: A Good Choice for Oncotherapy and Radiochemotherapy Sensitization. Nutr Cancer 2019; 72:554-567. [PMID: 31387393 DOI: 10.1080/01635581.2019.1648694] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Several studies have focused on chemical agents, tailored from natural edible products, used to prevent and treat various diseases. β-elemene is a well-known compound derived from Curcuma wenyujin that possesses a wide spectrum of anticancer properties under preclinical and clinical conditions. Several studies have demonstrated its inhibitory effect both in humans and animals with cancers. Numerous in vivo and in vitro experimental models have revealed that β-elemene can modulate multiple molecular pathways involved in carcinogenesis. In general, (1) β-elemene itself can inhibit and kill tumor cells through a variety of mechanisms, and (2) can synergistically enhance the effect of radiotherapy and/or chemotherapy, (3) also can regulate autoimmune in the treatment of tumors. In this article, we critically focused on the available scientific evidence discussing the use of β-elemene in cancer prevention, and its molecular targets and mechanisms of action in different types of cancer. In addition, we have discussed its sources, chemistry, bioavailability, and future research directions.
Collapse
Affiliation(s)
- Hongxuan Tong
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yihua Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.,Department of Traditional Chinese Medicine, Medical College of Xiamen University, Xiamen, China
| | - Lijie Jiang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingshang Wang
- Department of Traditional Chinese Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Liu S, Ren SN, Ding WX, Ge XL, Cao YD, Zhang S, Zhen FX, Sun XC. Concurrent liposomal paclitaxel and cisplatin chemotherapy improved outcomes for locally advanced esophageal squamous cell carcinoma treated with intensity-modulated radiotherapy. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:331. [PMID: 31475201 DOI: 10.21037/atm.2019.06.45] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background To ascertain whether concurrent chemotherapy using liposomal paclitaxel and cisplatin could improve the outcomes of patients with locally advanced esophageal squamous cell carcinoma receiving intensity-modulated radiotherapy (IMRT). Methods A total of 72 patients with locally advanced esophageal squamous cell carcinoma, which were admitted to our hospital from October 2011 to December 2013, were retrospectively analyzed in this study. Results Thirty-six patients (50%) were treated with IMRT alone, while the other 36 patients (50%) were treated by IMRT combined with chemotherapy containing liposomal paclitaxel and cisplatin. Patients treated with chemoradiotherapy showed significantly superior overall survival (OS) and progression-free survival (PFS) compared to patients treated with IMRT alone (median OS: respectively, 29.7 vs. 12.9 months, P=0.0287; median PFS: respectively, 14.0 vs. 6.5 months, P=0.0186). Multivariate Cox analysis confirmed the inclusion of chemotherapy as an independent predictor of favorable OS and PFS. Both chemoradiotherapy and IMRT were well-tolerated in our cohort. Conclusions Chemotherapy improved the prognosis of locally advanced esophageal squamous cell carcinoma treated with IMRT. Large prospective studies are needed to confirm the therapeutic value of IMRT combined with chemotherapy in locally advanced esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Shu Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Sheng-Nan Ren
- Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing 211100, China
| | - Wen-Xiu Ding
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiao-Lin Ge
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yuan-Dong Cao
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Sheng Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Fu-Xi Zhen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xin-Chen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
25
|
Irinotecan plus S-1 versus S-1 in patients with previously treated recurrent or metastatic esophageal cancer (ESWN 01): a prospective randomized, multicenter, open-labeled phase 3 trial. Cancer Commun (Lond) 2019; 39:16. [PMID: 30940189 PMCID: PMC6444575 DOI: 10.1186/s40880-019-0359-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 03/19/2019] [Indexed: 12/18/2022] Open
Abstract
Background The benefit of systemic treatments in esophageal squamous cell carcinoma (ESCC) which has progressed after chemotherapy is still uncertain and optimal regimens based on randomized trials have not yet been established. We aimed to compare the efficacy of irinotecan plus S-1 with S-1 monotherapy in recurrent or metastatic ESCC patients who had resistance to platinum- or taxane-based chemotherapy. Methods We conducted a prospective randomized, multicenter, open-label, phase 3 trial in 15 centers across China. Eligible patients were adults with histologically confirmed recurrent or metastatic ESCC, and were randomly assigned (ratio, 1:1) to receive either irinotecan plus S-1 (intravenous infusion of irinotecan [160 mg/m2] on day 1 and oral S-1 [80–120 mg] on days 1–10, repeated every 14 days) or oral S-1 monotherapy (80–120 mg/day on days 1–14, repeated every 21 days) using a central computerized minimization procedure. The primary endpoint was progression-free survival (PFS). Results Between December 23, 2014 and July 25, 2016, we screened 148 patients and randomly assigned 123 patients to receive either irinotecan plus S-1 regimen (n = 61) or S-1 monotherapy (n = 62). After a median follow-up of 29.2 months (95% confidence interval [CI] 17.5–40.9 months), the median PFS was significantly longer in the irinotecan plus S-1 group than in the S-1 monotherapy group (3.8 months [95% CI 2.9–4.3 months] vs. 1.7 months [95% CI 1.4–2.7 months], hazard ratio = 0.58, 95% CI 0.38–0.86, P = 0.006). The objective response rates were 24.6% in the irinotecan plus S-1 group and 9.7% in the S-1 monotherapy group (P = 0.002). The patients in the irinotecan plus S-1 group presented with increased rates of grade 3–4 leukopenia (16.4% vs. 0%), neutropenia (14.8% vs. 1.6%), and nausea (4.9% vs. 0%). No significant difference in grade 3–4 diarrhea and no treatment-related deaths were observed in both groups. Conclusions The combination of irinotecan with S-1 was similarly tolerable but significantly prolonged PFS compared to S-1 monotherapy as a second- or third-line treatment in patients with recurrent or metastatic ESCC. Clinical Trial Registration NCT02319187. Registered on December 9, 2014 Electronic supplementary material The online version of this article (10.1186/s40880-019-0359-7) contains supplementary material, which is available to authorized users.
Collapse
|
26
|
Meindl-Beinker NM, Betge J, Gutting T, Burgermeister E, Belle S, Zhan T, Schulte N, Maenz M, Ebert MP, Haertel N. A multicenter open-label phase II trial to evaluate nivolumab and ipilimumab for 2nd line therapy in elderly patients with advanced esophageal squamous cell cancer (RAMONA). BMC Cancer 2019; 19:231. [PMID: 30871493 PMCID: PMC6419339 DOI: 10.1186/s12885-019-5446-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/07/2019] [Indexed: 12/16/2022] Open
Abstract
Background Advanced esophageal squamous cell cancer (ESCC) is frequently diagnosed in elderly patients. The impact of 2nd line chemotherapy is poorly defined. Recent data demonstrated effectiveness of checkpoint inhibitors in different squamous cell carcinomas. Therefore, we assess combined nivolumab/ipilimumab as 2nd line therapy in elderly ESCC patients. Methods RAMONA is a multicenter open-label phase II trial. The primary objective is to demonstrate a significant survival benefit of nivolumab/ipilimumab in advanced ESCC compared to historical data of standard chemotherapy. Primary endpoint is therefore overall survival (OS). Major secondary objective is the evaluation of tolerability. Time to QoL deterioration will thus be determined as key secondary endpoint. Further secondary endpoints are tumor response, PFS and safety. We aim to recruit a total of n = 75 subjects that have to be > 65 years old. Eligibility is determined by the geriatric status (G8 screening and Deficit Accumulation Frailty Index (DAFI)). A safety assessment will be performed after a 3 cycle run-in phase of nivolumab (240 mg Q2W) to justify escalation for eligible patients to combined nivolumab (240 mg Q2W) and ipilimumab (1 mg/kg Q6W), while the other patients will remain on nivolumab only. RAMONA also includes translational research sub-studies to identify predictive biomarkers, including PD-1 and PD-L1 evaluation at different time points, establishment of organoid cultures and microbiome analyses for response prediction. Discussion The RAMONA trial aims to implement checkpoint inhibitors for elderly patients with advanced ESCC as second line therapy. Novel biomarkers for checkpoint-inhibitor response are analyzed in extensive translational sub-studies. Trial registration EudraCT Number: 2017–002056-86; NCT03416244, registered: 31.1.2018. Electronic supplementary material The online version of this article (10.1186/s12885-019-5446-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nadja M Meindl-Beinker
- Department of Medicine II, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Johannes Betge
- Department of Medicine II, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Tobias Gutting
- Department of Medicine II, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Elke Burgermeister
- Department of Medicine II, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Sebastian Belle
- Department of Medicine II, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Tianzuo Zhan
- Department of Medicine II, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Nadine Schulte
- Department of Medicine II, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | | | - Matthias P Ebert
- Department of Medicine II, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Nicolai Haertel
- Department of Medicine II, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
27
|
Chen L, You C, Jin X, Zhou L, Huang L, Wang Y. Cytoskeleton-associated protein 4 is a novel serodiagnostic marker for esophageal squamous-cell carcinoma. Onco Targets Ther 2018; 11:8221-8226. [PMID: 30538491 PMCID: PMC6251352 DOI: 10.2147/ott.s183790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Recent years have witnessed significant progress in the treatment of esophageal squamous-cell carcinoma (ESCC); however, the prognosis of ESCC is still unsatisfactory. Bio-markers are required to improve identification of high-risk populations and help management of ESCC. This study was to evaluate the role of serum CKAP4 in ESCC. Methods This longitudinal study recruited 207 ESCC patients and age-/sex-matched healthy controls. Circulating levels of CKAP4 were measured using ELISA kits, while the expression of CKAP4 in esophageal tissue was evaluated using Western blotting. Results Serum CKAP4 levels were higher in ESCC patients (380.2±171.3 pg/mL) than healthy controls (271.8±97.4 pg/mL; P<0.001). The area under the receiver-operating characteristic curve of serum CKAP4 levels to identify the presence of ESCC was 0.675 (95% CI 0.622–0.728; P<0.001). According to Youden’s index, the best cutoff value was 429.1 pg/mL (sensitivity 0.415 and specificity 0.995). Furthermore, after follow-up, multivariate analyses identified that pathological lymph node metastases were the poorest prognostic factor (HR 1.862, 95% CI 1.093–3.173; P=0.022), followed by serum CKAP4 (HR 1.437, 95% CI 1.025–2.014; P=0.035). When stratified by tertiles of serum CKAP4, subjects in the first tertile presented a mean survival time of 75.4 months (95% CI 68.0–81.9), which decreased significantly in the second tertile (73.8 months, 95% CI 61.4–86.3) and the third tertile (59.9 months, 95% CI 49.8–70.0, log-rank χ2=8.235; P=0.016). Conclusion These results suggested that serum CKAP4 could be a potential biomarker for clinical management of ESCC.
Collapse
Affiliation(s)
- Lei Chen
- Department of Oncology, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian, China, .,Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China,
| | - Chuanwen You
- Department of Oncology, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian, China, .,Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China,
| | - Xiaowei Jin
- Department of Oncology, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian, China, .,Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China,
| | - Lei Zhou
- Department of Oncology, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian, China, .,Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China,
| | - Liyou Huang
- Department of Oncology, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian, China, .,Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China,
| | - Yanhua Wang
- Department of Oncology, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian, China, .,Department of Oncology, Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China,
| |
Collapse
|
28
|
Identification of LETM1 as a marker of cancer stem-like cells and predictor of poor prognosis in esophageal squamous cell carcinoma. Hum Pathol 2018; 81:148-156. [DOI: 10.1016/j.humpath.2018.07.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 06/22/2018] [Accepted: 07/03/2018] [Indexed: 11/20/2022]
|
29
|
Effects of Icaritin on the physiological activities of esophageal cancer stem cells. Biochem Biophys Res Commun 2018; 504:792-796. [DOI: 10.1016/j.bbrc.2018.08.060] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 08/06/2018] [Indexed: 01/06/2023]
|
30
|
A novel oral camptothecin analog, gimatecan, exhibits superior antitumor efficacy than irinotecan toward esophageal squamous cell carcinoma in vitro and in vivo. Cell Death Dis 2018; 9:661. [PMID: 29855512 PMCID: PMC5981453 DOI: 10.1038/s41419-018-0700-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/24/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a frequently diagnosed and deadly malignancy with few standard therapeutic options. Camptothecins are considered one of the most promising antitumor drugs. A modified lipophilic analog, gimatecan, was synthesized as a novel oral camptothecin and showed impressive effects in various tumors, but its therapeutic efficacy and mechanisms in ESCC remain unclear. This study investigated the antitumor efficacy and mechanisms of gimatecan in ECSS both in vitro and in vivo. Using ESCC cell lines, cell line-derived xenografts and patient-derived xenografts models, we evaluated gimatecan’s inhibition of tumor growth, and compared its antitumor efficacy with that of irinotecan. Topoisomerase I function and expression were assessed using the DNA relaxation assay and Western blotting, respectively. DNA damage was evaluated by Western blotting. Cell cycle progression and cell apoptosis were assessed using flow cytometry and Western blotting. Gimatecan could significantly suppress tumor growth in vivo and inhibit tumor cell proliferation in vitro, which was superior to irinotecan. Gimatecan suppressed the function and expression of topoisomerase I. It also caused DNA damage and activated the phosphorylation of multiple checkpoint gatekeepers, such as ATM, ATR, BRCA1, H2AX, CHK1, CHK2, and p53. It induced S phase arrest, enhanced the expression of p21WAF1/CIP, and suppressed the expression of CDK2 and cyclin A. Induction of apoptosis was accompanied by increases in Bax, cleaved-caspase 3 activation, cleaved-caspase 9 induction, and a decrease in Bcl-2. The molecular and phenotypic changes induced by gimatecan were stronger than that of irinotecan. In ESCC, gimatecan suppressed the expression and function of topoisomerase I, induced DNA damage and intra-S phase cell cycle arrest, and resulted in apoptosis. And the results suggest that gimatecan has higher potency in inhibiting ESCC tumor growth than irinotecan, providing a rational novel therapeutic strategy for future clinical evaluation.
Collapse
|
31
|
Liw PX, Wen YW, Tsai CY, Chang HK, Tseng CK, Hung TM, Chao YK. Pretreatment clinical stage predicts locoregional recurrence in patients with esophageal cancer who achieved a complete clinical response to chemoradiotherapy. J Thorac Cardiovasc Surg 2018; 155:2233-2242.e2. [DOI: 10.1016/j.jtcvs.2017.12.082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 09/16/2017] [Accepted: 12/17/2017] [Indexed: 01/14/2023]
|
32
|
Nakamura K, Baba Y, Kosumi K, Harada K, Shigaki H, Miyake K, Kiyozumi Y, Ohuchi M, Kurashige J, Ishimoto T, Iwatsuki M, Sakamoto Y, Yoshida N, Watanabe M, Nakao M, Baba H. UHRF1 regulates global DNA hypomethylation and is associated with poor prognosis in esophageal squamous cell carcinoma. Oncotarget 2018; 7:57821-57831. [PMID: 27507047 PMCID: PMC5295392 DOI: 10.18632/oncotarget.11067] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 07/19/2016] [Indexed: 01/27/2023] Open
Abstract
Background Global DNA hypomethylation contributes to oncogenesis through various mechanisms. The level of long interspersed nucleotide element-1 (LINE- 1) methylation is considered a surrogate marker of global DNA methylation, and is attracting interest as a good predictor of cancer prognosis. However, the mechanism how LINE-1 (global DNA) methylation is controlled in cancer cells remains to be fully elucidated. Ubiquitin-like with PHD and RING finger domain 1 (UHRF1) plays a crucial role in DNA methylation. UHRF1 is overexpressed in many cancers, and UHRF1 overexpression may be a mechanism underlying DNA hypomethylation in cancer cells. Nonetheless, the relationship between UHRF1, LINE-1 methylation level, and clinical outcome in esophageal squamous cell carcinoma (ESCC) remains unclear. Results In ESCC cell lines, vector-mediated UHRF1 overexpression caused global DNA (LINE-1) hypomethylation and, conversely, UHRF1 knockdown using siRNA increased the global DNA methylation level. In ESCC tissues, UHRF1 expression was significantly associated with LINE-1 methylation levels. Furthermore, UHRF1 overexpression correlated with poor prognosis in our cohort of 160 ESCC patients. Materials and Methods The relationships between UHRF1 expression and LINE-1 methylation level (i.e., global DNA methylation level) were investigated using ESCC tissues and cell lines. In addition, we examined the correlation between UHRF1 expression, LINE-1 methylation, and clinical outcome in patients with ESCC. Conclusions Our results suggest that UHRF1 is a key epigenetic regulator of DNA methylation and might be a potential target for cancer treatment.
Collapse
Affiliation(s)
- Kenichi Nakamura
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Keisuke Kosumi
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Kazuto Harada
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Hironobu Shigaki
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Keisuke Miyake
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yuki Kiyozumi
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Mayuko Ohuchi
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Junji Kurashige
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yasuo Sakamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Mitsuyoshi Nakao
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
33
|
Katsaounis P, Kotsakis A, Kentepozidis N, Polyzos A, Bakogeorgos M, Koinis F, Vamvakas L, Vardakis N, Kalbakis K, Boukovinas I, Varthalitis II, Prinarakis E, Georgoulias V, Souglakos J. Nab-paclitaxel as second-line treatment in advanced gastric cancer: a multicenter phase II study of the Hellenic Oncology Research Group. Ann Gastroenterol 2017; 31:65-70. [PMID: 29333068 PMCID: PMC5759614 DOI: 10.20524/aog.2017.0215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 10/09/2017] [Indexed: 01/05/2023] Open
Abstract
Background This study evaluated the safety and efficacy of nab-paclitaxel as second-line treatment in patients with advanced gastric adenocarcinoma. Methods Thirty-nine pretreated patients [33 with taxane-based regimens (docetaxel, cisplatin, and fluorouracil)] and 6 with combination of fluoropyrimidines plus cisplatin with locally advanced inoperable and metastatic gastric and gastroesophageal junction adenocarcinoma were treated with weekly nab-paclitaxel (150 mg/m2 d1, d8, d15 in cycles of 28 days). Results Partial response (PR) was documented in nine patients (23.1%; 95% confidence interval 10.1-37.2%), stable disease (SD) in 11 (28.2%) and disease progression in 18 (46.2%). The disease control rate (SD + PR + complete response) was 51.3%. Grade 3 and 4 neutropenia occurred in 10.2% and 5.1% of patients, respectively; grade 3 anemia in 5.1%; grade 3 neurotoxicity in 5.1%; and grade 2 pain in 5.1%. The median progression-free survival was 3.0 months (range 0.3-13.6) and the median overall survival 6.8 months (range 0.3-22). Conclusion Nab-paclitaxel as second-line treatment in locally advanced inoperable or metastatic gastric and gastroesophageal junction carcinoma is an active chemotherapy regimen with a manageable toxicity profile and merits further evaluation.
Collapse
Affiliation(s)
- Panagiotis Katsaounis
- Department of Medical Oncology, IASO General Hospital, Athens (Papangiotis Katsaounis, Vassilis Georgoulias), Greece
| | - Athanasios Kotsakis
- Department of Medical Oncology, University General Hospital of Heraklion, Crete (Athanasios Kotsakis, Filippos Koinis, Lambros Vamvbakas, Nikolaos Vardakis, Kostas Kalbakis, John Souglakos), Greece
| | - Nikolaos Kentepozidis
- Department of Medical Oncology, 251 Air Force General Hospital, Athens (Nikolaos Kentepozidis, Marios Bakogeorgos), Greece
| | - Aris Polyzos
- Department of Medicine, Medical School, University of Athens, Laikon General Hospital, Athens (Aris Polyzos), Greece
| | - Marios Bakogeorgos
- Department of Medical Oncology, 251 Air Force General Hospital, Athens (Nikolaos Kentepozidis, Marios Bakogeorgos), Greece
| | - Filippos Koinis
- Department of Medical Oncology, University General Hospital of Heraklion, Crete (Athanasios Kotsakis, Filippos Koinis, Lambros Vamvbakas, Nikolaos Vardakis, Kostas Kalbakis, John Souglakos), Greece
| | - Lambros Vamvakas
- Department of Medical Oncology, University General Hospital of Heraklion, Crete (Athanasios Kotsakis, Filippos Koinis, Lambros Vamvbakas, Nikolaos Vardakis, Kostas Kalbakis, John Souglakos), Greece
| | - Nikolaos Vardakis
- Department of Medical Oncology, University General Hospital of Heraklion, Crete (Athanasios Kotsakis, Filippos Koinis, Lambros Vamvbakas, Nikolaos Vardakis, Kostas Kalbakis, John Souglakos), Greece
| | - Kostas Kalbakis
- Department of Medical Oncology, University General Hospital of Heraklion, Crete (Athanasios Kotsakis, Filippos Koinis, Lambros Vamvbakas, Nikolaos Vardakis, Kostas Kalbakis, John Souglakos), Greece
| | - Ioannis Boukovinas
- Department of Medical Oncology, Bioclinic of Thessaloniki (Ioannis Boukovinas), Greece
| | - Ioannis I Varthalitis
- Department of Medical Oncology, Henry Dunant Hospital, Athens (Ioannis I. Varthalitis), Greece
| | | | - Vassilis Georgoulias
- Department of Medical Oncology, IASO General Hospital, Athens (Papangiotis Katsaounis, Vassilis Georgoulias), Greece
| | - John Souglakos
- Department of Medical Oncology, University General Hospital of Heraklion, Crete (Athanasios Kotsakis, Filippos Koinis, Lambros Vamvbakas, Nikolaos Vardakis, Kostas Kalbakis, John Souglakos), Greece
| |
Collapse
|
34
|
Circulating Tumor Cells Detected by the Expression of Cancer Stem Cell Markers CD90 and CD44 in Patients With Esophageal Cancer. Int Surg 2017. [DOI: 10.9738/intsurg-d-16-00022.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background
Epithelial cell adhesion molecule (EpCAM) is a marker for circulating tumor cells (CTCs) in various types of cancer. Cell surface antigens, such as CD90 and CD44, have been reported to be cancer stem cell (CSC) markers in esophageal squamous cell carcinoma (ESCC). The aim of this study was to assess the use of CD90 and CD44 as markers to identify clinically significant CTC subpopulations in ESCC.
Methods
We collected 3 mL of peripheral blood from 10 ESCC patients and 10 healthy volunteers to detect combined expression of EpCAM, CD90, and CD40 using flow cytometry.
Results
The number of EpCAM-positive cell counts (average ± SD) in the patients was significantly higher than healthy volunteers (29.1 ± 35.9 and 2.3 ± 2.5, P = 0.001). The proportions (average ±SD) of CD90- and CD44-positive cells in EpCAM-positive cells were 45.7% ± 42.4% and 98.7% ± 2.7%, respectively. EpCAM-positive/CD44-positive CTC counts, which was equivalent to EpCAM-positive CTC counts, correlated with pathologic V factors in the resected primary tumors (P > 0.01). EpCAM-positive/CD90-positive CTC counts, but not EpCAM-positive/CD90-negative CTC counts, correlated with pathologic V factors in the resected primary tumors (P = 0.01). Our results suggested that combined expression of EpCAM and CD90 may useful to detect CTC subsets, which have highly metastatic features in ESCC. CD44, on the other hand, is equivalent to EpCAM as a marker to detect CTCs in ESCC.
Collapse
|
35
|
Saigí M, Oliva M, Aliste L, Calvo M, Hormigo G, Serra Ò, Boladeras A, Farran L, Robles J, Creus G, Paúles MJ, Gornals JB, de Lama E, Borràs JM, Sala N, Galán M. Clinical relevance of histologic subtypes in locally advanced esophageal carcinoma treated with pre-operative chemoradiotherapy: Experience of a monographic oncologic centre. PLoS One 2017; 12:e0184737. [PMID: 28931046 PMCID: PMC5607166 DOI: 10.1371/journal.pone.0184737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/30/2017] [Indexed: 12/13/2022] Open
Abstract
Background Locally advanced esophageal carcinoma (LAEC) represents less than 30% of all diagnosed esophageal carcinoma worldwide. The standard of care for resectable tumours consists of preoperative chemoradiotherapy (CRT) followed by surgery. Despite the curative intent, the prognosis is still poor mainly due to relapse. A multidisciplinary approach is required in order to optimize the therapeutic strategy and follow-up. Differences in outcomes between the two main histological subtypes, adenocarcinoma (ADC) and squamous cell carcinoma (SCC), have been reported. Nevertheless, the heterogeneity in trials design and data available have hampered the achievement of clear conclusions. The purpose of this study is to report the outcomes from a cohort of patients with LAEC treated with a multidisciplinary approach and to remark the differences observed between the two main histologic subtypes and their clinical implications. Methods We retrospectively reviewed 100 patients diagnosed with LAEC that were treated with preoperative CRT at our institution and integrated centres. Histopathological characteristics and toxicities during treatment were recorded. Patterns of recurrence at the first relapse were analysed. Survival curves were plotted using the Kaplan Meier method and multivariate Cox proportional hazards models were used. Results Among the patients who received preoperative CRT, 83% underwent surgery. The median overall survival (mOS) was 31.7 months, 26.9 months for ADC and 45.5 for SCC (p-value = 0.33). In the multivariate Cox regression analysis, ypN+ was the only factor that negatively influenced in OS (OR = 4.1, p-value = 0.022). Patterns of recurrence differed according to histologic subtype. Distant relapse was more frequent in ADC (62%), whereas locoregional relapse was higher in SCC (50%) (p-value = 0.027). Second line therapeutic strategies could be offered to 50% of those patients who relapsed. Conclusions Differences in outcomes and recurrence pattern could be observed between the two main histologic subtypes of LAEC. A better molecular characterization, adapted therapeutic regimens and follow up strategies should be adopted in order to improve survival of these patients.
Collapse
Affiliation(s)
- Maria Saigí
- Department of Medical Oncology, Institut Català Oncologia (ICO), IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
- Gastroesophageal Tumours Functional Unit (UTEG), Institut Català d’Oncologia- Hospital Universitari de Bellvitge- IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- * E-mail:
| | - Marc Oliva
- Department of Medical Oncology, Institut Català Oncologia (ICO), IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
- Gastroesophageal Tumours Functional Unit (UTEG), Institut Català d’Oncologia- Hospital Universitari de Bellvitge- IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Luisa Aliste
- Evaluation Unit, Cancer Plan, Department of Health, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Mariona Calvo
- Department of Medical Oncology, Institut Català Oncologia (ICO), IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
- Gastroesophageal Tumours Functional Unit (UTEG), Institut Català d’Oncologia- Hospital Universitari de Bellvitge- IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Gloria Hormigo
- Department of Medical Oncology, Institut Català Oncologia (ICO), IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
- Gastroesophageal Tumours Functional Unit (UTEG), Institut Català d’Oncologia- Hospital Universitari de Bellvitge- IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Òlbia Serra
- Department of Medical Oncology, Institut Català Oncologia (ICO), IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
- Gastroesophageal Tumours Functional Unit (UTEG), Institut Català d’Oncologia- Hospital Universitari de Bellvitge- IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Anna Boladeras
- Gastroesophageal Tumours Functional Unit (UTEG), Institut Català d’Oncologia- Hospital Universitari de Bellvitge- IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Department of Radiation Oncology, Institut Català Oncologia (ICO), IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Leandre Farran
- Gastroesophageal Tumours Functional Unit (UTEG), Institut Català d’Oncologia- Hospital Universitari de Bellvitge- IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Digestive Surgery Department, Hospital Universitari de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Javier Robles
- Gastroesophageal Tumours Functional Unit (UTEG), Institut Català d’Oncologia- Hospital Universitari de Bellvitge- IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Nuclear Medecine Department, Institut del Diagnòstic Imatge (IDI), Hospital Universitari Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Gloria Creus
- Gastroesophageal Tumours Functional Unit (UTEG), Institut Català d’Oncologia- Hospital Universitari de Bellvitge- IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Clinical Nutrition Unit, Hospital Universitari de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Ma José Paúles
- Gastroesophageal Tumours Functional Unit (UTEG), Institut Català d’Oncologia- Hospital Universitari de Bellvitge- IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Pathology Department, Hospital Universitari de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Joan B. Gornals
- Gastroesophageal Tumours Functional Unit (UTEG), Institut Català d’Oncologia- Hospital Universitari de Bellvitge- IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Gastroenterology Department, Hospital Universitari de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Eugenia de Lama
- Gastroesophageal Tumours Functional Unit (UTEG), Institut Català d’Oncologia- Hospital Universitari de Bellvitge- IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Radiology Department, Hospital Universitari de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Josep Ma Borràs
- Department of Clinical Sciences, University of Barcelona, IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Núria Sala
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program and Translational Research Laboratory, Institut Català d’Oncologia (ICO)-IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Maica Galán
- Department of Medical Oncology, Institut Català Oncologia (ICO), IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
- Gastroesophageal Tumours Functional Unit (UTEG), Institut Català d’Oncologia- Hospital Universitari de Bellvitge- IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
36
|
Petty RD, Dahle-Smith A, Stevenson DAJ, Osborne A, Massie D, Clark C, Murray GI, Dutton SJ, Roberts C, Chong IY, Mansoor W, Thompson J, Harrison M, Chatterjee A, Falk SJ, Elyan S, Garcia-Alonso A, Fyfe DW, Wadsley J, Chau I, Ferry DR, Miedzybrodzka Z. Gefitinib and EGFR Gene Copy Number Aberrations in Esophageal Cancer. J Clin Oncol 2017; 35:2279-2287. [PMID: 28537764 DOI: 10.1200/jco.2016.70.3934] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024] Open
Abstract
Purpose The Cancer Esophagus Gefitinib trial demonstrated improved progression-free survival with the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor gefitinib relative to placebo in patients with advanced esophageal cancer who had disease progression after chemotherapy. Rapid and durable responses were observed in a minority of patients. We hypothesized that genetic aberration of the EGFR pathway would identify patients benefitting from gefitinib. Methods A prespecified, blinded molecular analysis of Cancer Esophagus Gefitinib trial tumors was conducted to compare efficacy of gefitinib with that of placebo according to EGFR copy number gain (CNG) and EGFR, KRAS, BRAF, and PIK3CA mutation status. EGFR CNG was determined by fluorescent in situ hybridization (FISH) using prespecified criteria and EGFR FISH-positive status was defined as high polysomy or amplification. Results Biomarker data were available for 340 patients. In EGFR FISH-positive tumors (20.2%), overall survival was improved with gefitinib compared with placebo (hazard ratio [HR] for death, 0.59; 95% CI, 0.35 to 1.00; P = .05). In EGFR FISH-negative tumors, there was no difference in overall survival with gefitinib compared with placebo (HR for death, 0.90; 95% CI, 0.69 to 1.18; P = .46). Patients with EGFR amplification (7.2%) gained greatest benefit from gefitinib (HR for death, 0.21; 95% CI, 0.07 to 0.64; P = .006). There was no difference in overall survival for gefitinib versus placebo for patients with EGFR, KRAS, BRAF, and PIK3CA mutations, or for any mutation versus none. Conclusion EGFR CNG assessed by FISH appears to identify a subgroup of patients with esophageal cancer who may benefit from gefitinib as a second-line treatment. Results of this study suggest that anti-EGFR therapies should be investigated in prospective clinical trials in different settings in EGFR FISH-positive and, in particular, EGFR-amplified esophageal cancer.
Collapse
Affiliation(s)
- Russell D Petty
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Asa Dahle-Smith
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - David A J Stevenson
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Aileen Osborne
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Doreen Massie
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Caroline Clark
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Graeme I Murray
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Susan J Dutton
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Corran Roberts
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Irene Y Chong
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Wasat Mansoor
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Joyce Thompson
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Mark Harrison
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Anirban Chatterjee
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Stephen J Falk
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Sean Elyan
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Angel Garcia-Alonso
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - David Walter Fyfe
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Jonathan Wadsley
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Ian Chau
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - David R Ferry
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| | - Zosia Miedzybrodzka
- Russell D. Petty, University of Dundee; Asa Dahle-Smith, Ninewells Hospital and Medical School, Dundee; David A.J. Stevenson, Aileen Osborne, Doreen Massie, Caroline Clark, Zosia Miedzybrodzka, and Graeme I. Murray, University of Aberdeen, Aberdeen; Susan J. Dutton and Corran Roberts, Centre for Statistics in Medicine, University of Oxford, Oxford; Mark Harrison, Mount Vernon Hospital, Northwood; Irene Y. Chong and Ian Chau, Royal Marsden Hospital, London and Surrey; Wasat Mansoor, Christie Hospital, Manchester; Joyce Thompson, Birmingham Heartland Hospital, Heart of England National Health Service Trust, Birmingham; Anirban Chatterjee, Royal Shrewsbury Hospital, Shrewsbury; Stephen J. Falk, Bristol Oncology Centre, Bristol; Sean Elyan, Cheltenham General Hospital, Cheltenham; Angel Garcia-Alonso, Clan Clwyd Hospital, Rhyl; David Walter Fyfe, Furness General Hospital, Furness; Jonathan Wadsley, Weston Park Hospital, Sheffield, United Kingdom; and David R. Ferry, Eli Lilly and Company, Bridgewater, NJ
| |
Collapse
|
37
|
|
38
|
Kojima H, Okumura T, Yamaguchi T, Miwa T, Shimada Y, Nagata T. Enhanced cancer stem cell properties of a mitotically quiescent subpopulation of p75NTR-positive cells in esophageal squamous cell carcinoma. Int J Oncol 2017; 51:49-62. [PMID: 28534989 PMCID: PMC5467780 DOI: 10.3892/ijo.2017.4001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/27/2017] [Indexed: 12/14/2022] Open
Abstract
Mitotically quiescent cancer stem cells (CSCs) possess higher malignant potential than other CSCs, indicating their higher contribution to therapeutic resistance than that of other CSCs. In esophageal squamous cell carcinoma (ESCC), p75 neurotrophin receptor (p75NTR) is expressed in a candidate CSC population showing high tumorigenicity and chemoresistance. In the present study, we isolated and characterized quiescent CSCs population in ESCC based on p75NTR expression and cell cycle status. Expression of p75NTR and Ki-67 in ESCC cell lines (KYSE cells) and surgically resected ESCC specimens was detected by performing immunocytochemical analysis. p75NTR-positive KYSE cells were fractionated into quiescent and proliferating cells by performing flow cytometry with a fluorescent DNA-staining dye to determine their CSC phenotype. Immunocytochemical analysis showed that 21.8 and 36.5% of the p75NTR-positive cells were Ki-67-negative (G0), which accounted for 11.4 and 15.7% of cells in KYSE-30 and KYSE-140 cell lines, respectively. Flow cytometric cell sorting showed that p75NTR-positive cells in the G0-G1 phase (p75NTR-positive/G0-1 cells) but not in the S-G2-M phase (p75NTR-positive/S-G2-M cells) showed strong expression of stem cell-related genes Nanog, BMI-1, and p63; high colony formation ability; high tumorigenicity in a mouse xenograft model; and strong chemoresistance against cisplatin because of the expression of drug resistance genes ABCG2 and ERCC1. Label-retention assay showed that 3.4% p75NTR-positive cells retained fluorescent cell-tracing dye, but p75NTR-negative cells did not. Immunohistochemical analysis of ESCC specimens showed p75NTR expression in 39 of 95 (41.1%) patients, with a median of 13.2% (range, 3.0-80.1%) p75NTR-positive/Ki-67-negative cells, which were found to be associated with poorly differentiated histology. Our results suggest that p75NTR-positive/G0-1 cells represent quiescent CSCs in ESCC and indicate that these cells can be used as targets to investigate molecular processes regulating CSC phenotype and to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Hirofumi Kojima
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama city, Toyama 930-0194, Japan
| | - Tomoyuki Okumura
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama city, Toyama 930-0194, Japan
| | - Tetsuji Yamaguchi
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama city, Toyama 930-0194, Japan
| | - Takeshi Miwa
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama city, Toyama 930-0194, Japan
| | - Yutaka Shimada
- Department of Nanobio Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takuya Nagata
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama city, Toyama 930-0194, Japan
| |
Collapse
|
39
|
Abstract
INTRODUCTION Esophageal cancer (EC) is the eighth most common cancer in the world, and the prognosis of EC is still poor. Although immunotherapy has been developed in melanoma and lung cancer, it is also expected to show efficacy in EC. Currently, several clinical trials are ongoing to evaluate the safety and efficacy of immunotherapies, immune checkpoint inhibitors, adoptive T cell transfer, and therapeutic cancer vaccines in EC. Areas covered: This review provides an overview and the status of immunotherapy in EC. Clinical significance of molecules related immune checkpoints, especially PD-1 and PD-L1 is presented and the designs, results and future directions of clinical trials using immunotherapy in EC are provided. Expert opinion: To bring immunotherapy to the forefront of treatment for EC, it is necessary to select patients who can obtain a high efficacy of immunotherapy and to also elucidate the correct timing for administration. Moreover, combination therapies of immunotherapy with existing chemotherapy or radiation or other immunotherapy with different mechanisms of action must be evaluated to achieve excellent outcomes in patients with EC.
Collapse
Affiliation(s)
- Tomokazu Tanaka
- a Department of Surgery , Saga University Faculty of Medicine , Saga , Japan
| | - Jun Nakamura
- a Department of Surgery , Saga University Faculty of Medicine , Saga , Japan
| | - Hirokazu Noshiro
- a Department of Surgery , Saga University Faculty of Medicine , Saga , Japan
| |
Collapse
|
40
|
Wang R, Sumarpo A, Saiki Y, Chen N, Sunamura M, Horii A. ABCB1 Is Upregulated in Acquisition of Taxane Resistance: Lessons from Esophageal Squamous Cell Carcinoma Cell Lines. TOHOKU J EXP MED 2017; 240:295-301. [PMID: 27941276 DOI: 10.1620/tjem.240.295] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Esophageal cancer is one of the common malignancies worldwide, particularly in eastern African and Asian countries including Japan. Taxane (paclitaxel or docetaxel) is one of the effective chemotherapeutic reagents for patients with esophageal cancer, but acquisition of chemoresistance frequently occurs; this is one of the most frequent causes for therapeutic failure. In this study, we established three taxane resistant esophageal squamous cell carcinoma cell lines and explored possible mechanisms for the acquisition of chemoresistance. Microarray analyses indicated that the ABCB1 (ATP binding cassette subfamily B member 1) gene was significantly upregulated in taxane resistant esophageal cancer cell lines. Moreover, we found that siRNA mediated ABCB1 knockdown successfully restored drug sensitivity in both paclitaxel and docetaxel resistant esophageal cancer cell lines. In conclusion, we propose that ABCB1 might play a pivotal role in acquisition of taxane resistance and could be a promising target for treatment of patients with esophageal cancer after acquisition of taxane resistance.
Collapse
Affiliation(s)
- Ruobing Wang
- Department of Molecular Pathology, Tohoku University School of Medicine
| | | | | | | | | | | |
Collapse
|
41
|
Suzuki S, Yokobori T, Altan B, Hara K, Ozawa D, Tanaka N, Sakai M, Sano A, Sohda M, Bao H, Fukuchi M, Miyazaki T, Kaira K, Asao T, Kuwano H. High stathmin 1 expression is associated with poor prognosis and chemoradiation resistance in esophageal squamous cell carcinoma. Int J Oncol 2017; 50:1184-1190. [PMID: 28350065 DOI: 10.3892/ijo.2017.3899] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 02/24/2017] [Indexed: 01/12/2023] Open
Abstract
Stathmin 1 (STMN1) is a major cytosolic phosphoprotein regulating microtubule dynamics, thereby playing an important role in cancer progression and resistance to microtubule-binding anticancer agents. We assessed the prognostic significance of STMN1 expression and STMN1-associated resistance to docetaxel and radiation in esophageal squamous cell carcinoma (ESCC) patients. STMN1 expression was evaluated by immunohistochemistry in 172 surgical specimens. The association of STMN1 expression with chemoradiation resistance using docetaxel was examined by comparing expression in 15 biopsy specimens obtained before neoadjuvant therapy to histological grades of post-therapy surgically resected tumors. We also evaluated the effects of STMN1 on sensitivity to docetaxel and radiation in ESCC cell lines. High STMN1 immunoexpression was significantly associated with tumor depth, lymph node metastasis, lymphatic invasion and venous invasion. Survival rates were significantly lower in ESCC patients with high STMN1 expression than in those with low STMN1 expression. Multivariable analysis showed that high STMN1 expression was an independent factor for poor survival. High STMN1 expression was also associated with poor response to neoadjuvant chemoradiotherapy using docetaxel. Knockdown of STMN1 expression enhanced ESCC cell line sensitivity to docetaxel and radiation. STMN1 appears critical for ESCC invasiveness and predicts an unfavorable prognosis in ESCC.
Collapse
Affiliation(s)
- Shigemasa Suzuki
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Takehiko Yokobori
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Bolag Altan
- Department of Oncology Clinical Development, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Keigo Hara
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Daigo Ozawa
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Naritaka Tanaka
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Makoto Sakai
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Akihiko Sano
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Makoto Sohda
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Halin Bao
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Minoru Fukuchi
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Tatsuya Miyazaki
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Kyoichi Kaira
- Department of Oncology Clinical Development, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Takayuki Asao
- Big Data Center for Integrative Analysis, Gunma University Initiative for Advance Research (GIAR), Gunma, Japan
| | - Hiroyuki Kuwano
- Department of General Surgical Science, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
42
|
Ojima T, Nakamori M, Nakamura M, Katsuda M, Hayata K, Matsumura S, Iwahashi M, Yamaue H. Phase I/II study of divided-dose docetaxel, cisplatin and fluorouracil for patients with recurrent or metastatic squamous cell carcinoma of the esophagus. Dis Esophagus 2017; 30:1-7. [PMID: 26725778 DOI: 10.1111/dote.12450] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Squamous cell carcinoma of the esophagus (SCCE) has a poor prognosis compared with other gastrointestinal cancers. Many patients present with locoregional unresectable or metastatic disease at the time of diagnosis. For these patients with metastatic esophageal cancer, chemotherapy is generally indicated. The aim of this phase I/II study was to evaluate the efficacy and safety of the combined use of docetaxel, cisplatin (CDDP) and 5-fluorouracil (5-FU)(DCF) in patients with recurrent/metastatic SCCE. This study adopted divided doses of docetaxel and CDDP in order to reduce the toxicities of the treatment. The dose of docetaxel was escalated using the following protocol in the phase I stage: level 1, 30 mg/m2; level 2, 35 mg/m2 and level 3, 40 mg/m2, which was intravenously infused for 2 hours on days 1 and 8. CDDP was administered at a dose of 12 mg/m2 infused for 4 hours on days 1-5. The 5-FU was administered at a dose of 600 mg/m2 continuously infused from day 1 to 5. This regimen was repeated every 4 weeks. The study subjects were nine patients (phase I) and 48 patients (phase II). The recommended dose was determined as level 3 in phase I. In the phase II stage, the overall response rate was 62.5%, with a complete response rate of 12.5%. The median progression-free survival was 6 months, and the median overall survival was 13 months. Grade 3/4 toxicities of leukopenia, neutropenia and febrile neutropenia occurred in 64.6%, 68.8% and 14.6% of the patients, while grade 3/4 non-hematological toxicities were relatively rare. No treatment-related death was recorded. This modified DCF regimen with divided doses can be a tolerable and useful regimen of definitive chemotherapy for unresectable SCCE because of its high efficacy, although adequate care for severe neutropenia must be administered.
Collapse
Affiliation(s)
- T Ojima
- Second Department of Surgery, Wakayama Medical University, School of Medicine, Wakayama, Japan
| | - M Nakamori
- Second Department of Surgery, Wakayama Medical University, School of Medicine, Wakayama, Japan
| | - M Nakamura
- Second Department of Surgery, Wakayama Medical University, School of Medicine, Wakayama, Japan
| | - M Katsuda
- Second Department of Surgery, Wakayama Medical University, School of Medicine, Wakayama, Japan
| | - K Hayata
- Second Department of Surgery, Wakayama Medical University, School of Medicine, Wakayama, Japan
| | - S Matsumura
- Second Department of Surgery, Wakayama Medical University, School of Medicine, Wakayama, Japan
| | - M Iwahashi
- Second Department of Surgery, Wakayama Medical University, School of Medicine, Wakayama, Japan
| | - H Yamaue
- Second Department of Surgery, Wakayama Medical University, School of Medicine, Wakayama, Japan
| |
Collapse
|
43
|
Nakamura K, Yoshida N, Baba Y, Kosumi K, Uchihara T, Kiyozumi Y, Ohuchi M, Ishimoto T, Iwatsuki M, Sakamoto Y, Watanabe M, Baba H. Elevated preoperative neutrophil-to-lymphocytes ratio predicts poor prognosis after esophagectomy in T1 esophageal cancer. Int J Clin Oncol 2017; 22:469-475. [PMID: 28097441 DOI: 10.1007/s10147-017-1090-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/10/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND The neutrophil-to-lymphocyte ratio (NLR) has been reported to predict the prognosis of various malignant tumors, including esophageal cancer. However, no previous reports have supported the use of the preoperative NLR as an independent prognostic marker focused on superficial (T1) esophageal cancer. The aim of this study was to elucidate the prognostic impact of the preoperative NLR in T1 esophageal cancer. METHODS This retrospective study recruited 245 consecutive patients with T1 esophageal cancer who underwent subtotal esophagectomy between 2005 and 2016. The relationship between the preoperative NLR and clinicopathological characteristics was analyzed. RESULTS The preoperative NLR was significantly higher in male patients (p = 0.029), patients with T1b esophageal cancer (p = 0.0274), and patients with venous vessel invasion (p = 0.0082). In the Kaplan-Meier analysis, the elevated preoperative NLR was significantly associated with a poorer disease-free survival (p < 0.0001) and overall survival (p = 0.0004). In the multivariate Cox model, the elevated preoperative NLR was an independent prognostic marker for both disease-free survival (p = 0.0013) and overall survival (p = 0.0027). CONCLUSION An elevated preoperative NLR predicts poor prognosis in T1 esophageal cancer, suggesting the utility of the NLR as an easily measurable and generally available independent prognostic marker.
Collapse
Affiliation(s)
- Kenichi Nakamura
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto, Kumamoto, 860-8556, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto, Kumamoto, 860-8556, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto, Kumamoto, 860-8556, Japan
| | - Keisuke Kosumi
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto, Kumamoto, 860-8556, Japan
| | - Tomoyuki Uchihara
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto, Kumamoto, 860-8556, Japan
| | - Yuki Kiyozumi
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto, Kumamoto, 860-8556, Japan
| | - Mayuko Ohuchi
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto, Kumamoto, 860-8556, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto, Kumamoto, 860-8556, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto, Kumamoto, 860-8556, Japan
| | - Yasuo Sakamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto, Kumamoto, 860-8556, Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Kumamoto, Kumamoto, 860-8556, Japan.
| |
Collapse
|
44
|
Hatogai K, Fujii S, Kojima T, Daiko H, Doi T, Ohtsu A, Ochiai A, Takiguchi Y, Yoshino T. Concordance between PIK3CA mutations in endoscopic biopsy and surgically resected specimens of esophageal squamous cell carcinoma. BMC Cancer 2017; 17:36. [PMID: 28068934 PMCID: PMC5220610 DOI: 10.1186/s12885-016-3041-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 12/20/2016] [Indexed: 01/01/2023] Open
Abstract
Background PIK3CA mutations are expected to be potential therapeutic targets for esophageal squamous cell carcinoma (ESCC). We aimed to clarify the concordance between PIK3CA mutations detected in endoscopic biopsy specimens and corresponding surgically resected specimens. Methods We examined five hotspot mutations in the PIK3CA gene (E542K, E545K, E546K, H1047R, and H1047L) in formalin-fixed and paraffin-embedded tissue sections of paired endoscopic biopsy and surgically resected specimens from 181 patients undergoing curative resection for ESCC between 2000 and 2011 using a Luminex technology-based multiplex gene mutation detection kit. Results Mutation analyses were successfully performed for both endoscopic biopsy and surgically resected specimens in all the cases. A PIK3CA mutation was detected in either type of specimen in 13 cases (7.2%, 95% confidence interval: 3.9–12.0). The overall concordance rate, positive predictive value, and negative predictive value were 98.3% (178/181), 90.9% (10/11), and 98.8% (168/170), respectively. Among patients with a PIK3CA mutation detected in both types of specimens, the concordance between PIK3CA mutation genotypes was 100%. There were three cases with a discordant mutation status between the types of specimens (PIK3CA mutation in surgically resected specimen and wild-type in biopsy specimen in two cases, and the opposite pattern in one case), suggesting possible intratumoral heterogeneity in the PIK3CA mutation status. Conclusions The PIK3CA mutation status was highly concordant between endoscopic biopsy and surgically resected specimens from the same patient, suggesting that endoscopic biopsy specimens can be clinically used to detect PIK3CA mutations in patients with ESCC.
Collapse
Affiliation(s)
- Ken Hatogai
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.,Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan.,Department of Medical Oncology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| | - Satoshi Fujii
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan.
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Hiroyuki Daiko
- Department of Esophageal Surgery, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Toshihiko Doi
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Atsushi Ohtsu
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.,Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Atsushi Ochiai
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Yuichi Takiguchi
- Department of Medical Oncology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| |
Collapse
|
45
|
Okumura T, Yamaguchi T, Watanabe T, Nagata T, Shimada Y. Clinical Relevance of a Candidate Stem Cell Marker, p75 Neurotrophin Receptor (p75NTR) Expression in Circulating Tumor Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 994:247-254. [PMID: 28560678 DOI: 10.1007/978-3-319-55947-6_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite advances in its diagnosis and multimodal therapies, the prognosis of esophageal squamous cell carcinoma (ESCC) patients remains poor, because of high incidences of metastasis . Recent reports suggested that circulating tumor stem cells (CTSCs), rather than circulating tumor cells (CTCs), were more accurate diagnostic marker for metastasis, because tumor stem cells or cancer stem cells (CSCs) are more responsible for metastasis through processes such as epithelial mesenchymal transition (EMT) and tumor initiation. A neurotrophin receptor p75 (p75NTR) is expressed in a candidate CSC s in ESCC, which possess enhanced tumorigenicity along with strong expression of EMT-related genes. Our recent report using two-color flow cytometry demonstrated that CTC counts based on a combined expression of epithelial cell adhesion molecule (EpCAM) and p75NTR was significantly higher in peripheral blood samples of ESCC patients than healthy controls. In addition, EpCAM + p75NTR+, but not EpCAM + p75NTR- CTC counts, correlated with clinically diagnosed distant metastasis and pathological venous invasion in surgically resected primary ESCC tumors. Malignant cytology of the isolated EpCAM + p75NTR+ cells was microscopically confirmed as well. These results demonstrated that EpCAM + p75NTR+ CTC count was a more accurate diagnostic marker than EpCAM+ CTC count, suggesting the highly metastatic potential of CTCs with p75NTR expression.Investigation using the isolated EpCAM + p75NTR+ CTCs to assess their stem cell properties may shed light on their roles in tumor metastasis in ESCC.Further investigations based on large-scale prospective studies with long term follow up may provide us with evidences for its clinical use.
Collapse
Affiliation(s)
- Tomoyuki Okumura
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama City, Toyama, 930-0194, Japan.
| | - Tetsuji Yamaguchi
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama City, Toyama, 930-0194, Japan
| | - Toru Watanabe
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama City, Toyama, 930-0194, Japan
| | - Takuya Nagata
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama City, Toyama, 930-0194, Japan
| | - Yutaka Shimada
- Department of Nanobio Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimo-Adachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| |
Collapse
|
46
|
Okumura T, Yamaguchi T, Watanabe T, Nagata T, Shimada Y. Flow Cytometric Detection of Circulating Tumor Cells Using a Candidate Stem Cell Marker, p75 Neurotrophin Receptor (p75NTR). Methods Mol Biol 2017; 1634:211-217. [PMID: 28819854 DOI: 10.1007/978-1-4939-7144-2_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The most widely studied detection for circulating tumor cells (CTCs) in peripheral blood of cancer patients has been based on immunomagnetic enrichment using antibodies against epithelial cell adhesion molecule (EpCAM), which is overexpressed in epithelial cells. A neurotrophin receptor p75 (p75NTR) is expressed in a candidate stem cell fraction in esophageal squamous cell carcinoma (ESCC), which shows significantly higher colony formation, enhanced tumor formation in mice, along with strong expression of epithelial mesenchymal transition-related genes. Here, we describe a method to detect CTCs in ESCC based on the combined expression of EpCAM and p75NTR using flow cytometry, demonstrating the feasibility of expression analysis of multiple cell surface markers in viable cells.
Collapse
Affiliation(s)
- Tomoyuki Okumura
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama City, Toyama, 930-0194, Japan.
| | - Tetsuji Yamaguchi
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama City, Toyama, 930-0194, Japan
| | - Toru Watanabe
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama City, Toyama, 930-0194, Japan
| | - Takuya Nagata
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, 2630 Sugitani, Toyama City, Toyama, 930-0194, Japan
| | - Yutaka Shimada
- Department of Nanobio Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimo-Adachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| |
Collapse
|
47
|
SEOM Clinical Guideline for the diagnosis and treatment of esophageal cancer (2016). Clin Transl Oncol 2016; 18:1179-1186. [PMID: 27900538 PMCID: PMC5138258 DOI: 10.1007/s12094-016-1577-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 11/11/2016] [Indexed: 12/17/2022]
Abstract
Esophageal cancer (EC) is an aggressive tumor that represents the 6th most common cause of cancer death worldwide. The estimated incidence in Spain is 2090 cases/year. Two main pathological subtypes exist, squamous cell carcinoma and adenocarcinoma. The main differences between them are localization and underlying factors which are the principal cause of the recent incidence changes observed in west countries. Staging techniques and treatment options which combine surgery, chemotherapy and radiotherapy, reflected the high complexity of the EC management. An undeniably multidisciplinary approach is, therefore, required. In this guide, we review the status of current diagnosis and treatment, define evidence and propose recommendations.
Collapse
|
48
|
Nomura M, Iwasa S, Tsushima T, Kato K, Yasui H, Boku N, Muto M, Muro K. Active salvage chemotherapy versus best supportive care for patients with recurrent or metastatic squamous cell carcinoma of the esophagus refractory or intolerable to fluorouracil, platinum, and taxane. Cancer Chemother Pharmacol 2016; 78:1209-1216. [PMID: 27778070 DOI: 10.1007/s00280-016-3179-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/19/2016] [Indexed: 11/28/2022]
Abstract
PURPOSE The survival benefit of chemotherapy (CTx) compared with best supportive care (BSC) is unclear in patients with recurrent or metastatic esophageal squamous cell carcinoma (R/M-ESCC) previously treated with fluorouracil (FU), platinum (PT), and taxane (TAX). PATIENTS AND METHODS The data of 283 consecutive patients with R/M-ESCC, who could not tolerate or whose tumor was refractory to all three agents (FU, PT, and TAX) between March 2003 and November 2014, were retrospectively reviewed. Patient selection criteria were as follows: age 20-75 years, performance status 0-2, and preserved organ function. Prognostic factors for overall survival (OS), defined as survival time from the last administration of all three agents, were analyzed using the Kaplan-Meier method and Cox proportional hazards model. RESULTS Of the 283 patients, 147 received BSC and 136 received CTx (PT-containing regimen 37, investigative new drugs 28, irinotecan-containing regimen 29, TAX alone 25, FU alone 16, others 4). Most patients' tumors were refractory to all three agents. The median OS of BSC and CTx patients was 4.2 and 7.8 months (hazard ratio 0.41; 95% confidence interval 0.31-0.54), respectively. Multivariate analysis with or without propensity score matching indicated that BSC, metastatic sites other than lung, liver, or bone, and hemoglobin <10 g/dL were associated with poor prognosis. CONCLUSION CTx might provide a survival benefit over BSC even for patients with R/M-ESCC who cannot tolerate or whose tumor is refractory to all three agents.
Collapse
Affiliation(s)
- Motoo Nomura
- Department of Clinical Oncology, Aichi Cancer Center Hospital, 1-1 Kanokoden Chikusa-ku, Nagoya, Aichi, 464-8681, Japan. .,Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Satoru Iwasa
- Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Kyoto, 104-0045, Japan
| | - Takahiro Tsushima
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, 1007 Shimonogakubo, Nagaizumi-cho, Sunto-gun, Shizuoka, 411-8777, Japan
| | - Ken Kato
- Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Kyoto, 104-0045, Japan
| | - Hirofumi Yasui
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, 1007 Shimonogakubo, Nagaizumi-cho, Sunto-gun, Shizuoka, 411-8777, Japan
| | - Narikazu Boku
- Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Kyoto, 104-0045, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, 1-1 Kanokoden Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| |
Collapse
|
49
|
Liu Y, Xiong Z, Beasley A, D'Amico T, Chen XL. Personalized and targeted therapy of esophageal squamous cell carcinoma: an update. Ann N Y Acad Sci 2016; 1381:66-73. [PMID: 27399176 PMCID: PMC5083215 DOI: 10.1111/nyas.13144] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/16/2016] [Accepted: 05/23/2016] [Indexed: 12/20/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a deadly disease that requires extensive research. In this review, we update recent progress in the research area of targeted therapy for ESCC. SOX2 and its associated proteins (e.g., ΔNP63α), which regulate lineage survival of ESCC cells, are proposed as therapeutic targets. It is believed that targeting the lineage-survival mechanism may be more effective than targeting other mechanisms. With the advent of a new era of personalized targeted therapy, there is a need to move from the tumor-centric model into an organismic model.
Collapse
Affiliation(s)
- Yongjing Liu
- Department of Cardiothoracic Surgery, 105th Hospital of PLA, Hefei, Anhui Province, China
- Division of Thoracic Surgery, Duke University Medical Center, Durham, North Carolina
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina
| | - Zhaohui Xiong
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina
| | - Andrea Beasley
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina
| | - Thomas D'Amico
- Division of Thoracic Surgery, Duke University Medical Center, Durham, North Carolina
| | - Xiaoxin Luke Chen
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina.
- Division of Gastroenterology and Hepatology, Department of Medicine, Center for Esophageal Disease and Swallowing, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
50
|
Personalized Kampo Medicine Facilitated Both Cytotoxic T Lymphocyte Response and Clinical Benefits Induced by Personalized Peptide Vaccination for Advanced Esophageal Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:5929525. [PMID: 27703488 PMCID: PMC5040795 DOI: 10.1155/2016/5929525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/18/2016] [Indexed: 11/17/2022]
Abstract
We retrospectively evaluated if personalized Kampo medicine (PKM) could facilitate CTL responses and clinical benefits induced by personalized peptide vaccination (PPV), in which HLA-matched vaccines were selected and administered based on the preexisting host immunity, for advanced esophageal cancer (aEC) patients. Among 34 aEC patients entered in the clinical study, 23 patients received PKM and PPV without (n = 12) or with chemotherapy (n = 11), while the remaining 11 patients did not receive PKM but received PPV without (n = 6) or with chemotherapy (n = 5), respectively. Incidence of adverse events was significantly lower or higher in PKM and PPV arm (n = 23) or PPV and chemotherapy arm (n = 16) as compared to that of the counter arm (n = 11 or 18), respectively. Postvaccination PBMCs from the patients undergoing PKM and PPV showed significantly higher CTL responses as compared to the counter arm. The median progression-free survival (PFS) or median survival time (MST) of 34 patients was 2.9 or 7.6 months, respectively. The combination therapy in PPV and PKM arm, but not that in PPV and chemotherapy arm, significantly (P = 0.02) prolonged MST. These results could warrant a next step of prospective clinical study of PKM and PPV for aEC patients.
Collapse
|