1
|
Hagi T, Shiraishi O, Nakanishi T, Kohda M, Hiraki Y, Kato H, Yasuda A, Shinkai M, Imano M, Yasuda T. Utility of Initial Tumor Reduction as a Prognostic Factor in Esophageal Squamous Cell Cancer Patients Undergoing Neoadjuvant Chemotherapy Followed by Surgery : A Retrospective Cohort Study. Ann Surg Oncol 2024; 31:5064-5074. [PMID: 38664331 DOI: 10.1245/s10434-024-15314-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/01/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND While a neoadjuvant chemotherapy regimen using docetaxel, cisplatin, and 5-fluorouracil (NAC-DCF) is considered the standard treatment for locally advanced esophageal cancer (EC) in Japan, a reliable marker for early prediction of treatment efficacy remains unclear. We investigated the utility of the tumor response after a first course of NAC-DCF as a post-surgery survival predictor in patients with EC. METHODS We enrolled 150 consecutive patients who underwent NAC-DCF followed by surgery for EC between September 2009 and January 2019. The initial tumor reduction (ITR), defined as the percentage decrease in the shorter diameter of the tumor after the first course of NAC-DCF, was evaluated using computed tomography. We analyzed the relationship between ITR, clinicopathological parameters, and survival. RESULTS The median ITR was 21.07% (range -11.45 to 50.13%). The optimal cut-off value for ITR for predicting prognosis was 10% (hazard ratio [HR] 3.30, 95% confidence interval [CI] 1.98-5.51), based on univariate logistic regression analyses for recurrence-free survival (RFS). Compared with patients with ITR <10%, patients with ITR ≥10% showed a significantly higher proportion of ypM0 (80.0% vs. 92.5%) and responders in terms of overall clinical response (50.0% vs. 80.8%). Multivariate analysis for RFS revealed that ypN2-3 (HR 2.78, 95% CI 1.67-4.62), non-response in terms of overall clinical response (HR 1.87, 95% CI 1.10-3.18), and ITR <10% (HR 2.48, 95% CI 1.42-4.32) were independent prognostic factors. CONCLUSIONS Tumor response after the first course of NAC-DCF may be a good predictor of survival in patients with EC who underwent NAC-DCF plus surgery.
Collapse
Affiliation(s)
- Takaomi Hagi
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Osamu Shiraishi
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan.
| | - Tomoya Nakanishi
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Masashi Kohda
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Yoko Hiraki
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Hiroaki Kato
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Atsushi Yasuda
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Masayuki Shinkai
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Motohiro Imano
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Takushi Yasuda
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| |
Collapse
|
2
|
Brosda S, Aoude LG, Bonazzi VF, Patel K, Lonie JM, Belle CJ, Newell F, Koufariotis LT, Addala V, Naeini MM, Pearson JV, Krause L, Waddell N, Barbour AP. Spatial intra-tumour heterogeneity and treatment-induced genomic evolution in oesophageal adenocarcinoma: implications for prognosis and therapy. Genome Med 2024; 16:90. [PMID: 39020404 PMCID: PMC11253399 DOI: 10.1186/s13073-024-01362-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 07/09/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Oesophageal adenocarcinoma (OAC) is a highly heterogeneous cancer with poor survival. Standard curative treatment is chemotherapy with or without radiotherapy followed by oesophagectomy. Genomic heterogeneity is a feature of OAC and has been linked to treatment resistance. METHODS Whole-genome sequencing data from 59 treatment-naïve and 18 post-treatment samples from 29 OAC patients was analysed. Twenty-seven of these were enrolled in the DOCTOR trial, sponsored by the Australasian Gastro-Intestinal Trials Group. Two biopsies from each treatment-naïve tumour were assessed to define 'shared' (between both samples) and 'private' (present in one sample) mutations. RESULTS Mutational signatures SBS2/13 (APOBEC) and SBS3 (BRCA) were almost exclusively detected in private mutation populations of treatment-naïve tumours. Patients presenting these signatures had significantly worse disease specific survival. Furthermore, mutational signatures associated with platinum-based chemotherapy treatment as well as high platinum enrichment scores were only detected in post-treatment samples. Additionally, clones with high putative neoantigen binding scores were detected in some treatment-naïve samples suggesting immunoediting of clones. CONCLUSIONS This study demonstrates the high intra-tumour heterogeneity in OAC, as well as indicators for treatment-induced changes during tumour evolution. Intra-tumour heterogeneity remains a problem for successful treatment strategies in OAC.
Collapse
Affiliation(s)
- Sandra Brosda
- Frazer Institute, The University of Queensland, 37 Kent Street, Woolloongabba, QLD, 4102, Australia.
| | - Lauren G Aoude
- Frazer Institute, The University of Queensland, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Vanessa F Bonazzi
- Frazer Institute, The University of Queensland, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Kalpana Patel
- Frazer Institute, The University of Queensland, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - James M Lonie
- Frazer Institute, The University of Queensland, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Clemence J Belle
- Frazer Institute, The University of Queensland, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Felicity Newell
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | | | - Venkateswar Addala
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Marjan M Naeini
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia
| | - John V Pearson
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Lutz Krause
- Frazer Institute, The University of Queensland, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
- Microba Life Sciences, Brisbane, QLD, 4000, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Andrew P Barbour
- Frazer Institute, The University of Queensland, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
- Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia
| |
Collapse
|
3
|
Deboever N, Jones CM, Yamashita K, Ajani JA, Hofstetter WL. Advances in diagnosis and management of cancer of the esophagus. BMJ 2024; 385:e074962. [PMID: 38830686 DOI: 10.1136/bmj-2023-074962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Esophageal cancer is the seventh most common malignancy worldwide, with over 470 000 new cases diagnosed each year. Two distinct histological subtypes predominate, and should be considered biologically separate disease entities.1 These subtypes are esophageal adenocarcinoma (EAC) and esophageal squamous cell carcinoma (ESCC). Outcomes remain poor regardless of subtype, with most patients presenting with late stage disease.2 Novel strategies to improve early detection of the respective precursor lesions, squamous dysplasia, and Barrett's esophagus offer the potential to improve outcomes. The introduction of a limited number of biologic agents, as well as immune checkpoint inhibitors, is resulting in improvements in the systemic treatment of locally advanced and metastatic esophageal cancer. These developments, coupled with improvements in minimally invasive surgical and endoscopic treatment approaches, as well as adaptive and precision radiotherapy technologies, offer the potential to improve outcomes still further. This review summarizes the latest advances in the diagnosis and management of esophageal cancer, and the developments in understanding of the biology of this disease.
Collapse
Affiliation(s)
- Nathaniel Deboever
- Department of Thoracic and Cardiovascular Surgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher M Jones
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Kohei Yamashita
- Department of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Wayne L Hofstetter
- Department of Thoracic and Cardiovascular Surgery, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
4
|
Day F, Sridharan S, Johnson C, Quah GT, Mallesara G, Kumar M, Poulter AL, Morrison A, van der Westhuizen A, Fraser A, Oldmeadow C, Martin J. Esophageal chemoradiotherapy with concurrent nivolumab: Pilot results in the palliative treatment of oligometastatic disease. Asia Pac J Clin Oncol 2024; 20:416-422. [PMID: 38512856 DOI: 10.1111/ajco.14057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 03/07/2024] [Indexed: 03/23/2024]
Abstract
AIMS Many patients diagnosed with esophageal cancer have dysphagia from their primary tumor and de novo metastatic disease. The purpose of this study was to test the safety and efficacy of nivolumab given concurrently with hypofractionated chemoradiotherapy to patients with oligometastatic and obstructing esophageal tumors. METHODS Patients were enrolled in a planned single-arm, phase 2 clinical trial. Eligible participants had previously untreated oligometastatic (≤5 metastases on fludeoxyglucose-18 positron emission tomography scan outside the primary tumor radiotherapy field) esophageal or gastroesophageal carcinoma, dysphagia, and Eastern Cooperative Oncology Group performance status 0-1. Treatment was with 2 weeks of concurrent hypofractionated radiotherapy (30 Gy/10#) to the primary tumor, weekly carboplatin AUC2, weekly paclitaxel 50 mg/m2, and q2weekly nivolumab 240 mg, followed by nivolumab 480 mg continuing q4weekly until disease progression or 24 months total. A single metastasis was treated with stereotactic radiotherapy (SBRT) (24 Gy/3#) in week 7. RESULTS Five patients were recruited before trial closure to new participants for logistical reasons. Existing participants continued treatment per protocol as a pilot study at one center. All five patients completed chemoradioimmunotherapy and SBRT. All patients derived an improvement in their dysphagia. Two patients completed 24 months of nivolumab without disease progression. Grade 3 adverse events (AEs) occurred in 3 patients, however, there were no grade 4 AEs, AEs due to SBRT, or AEs of special interest as defined by the protocol. CONCLUSION Pilot results from five patients at one center found that treatment was well tolerated and effective for dysphagia relief. The efficacy of hypofractionated chemoradiotherapy with concurrent checkpoint inhibition should be tested in a multicentre study.
Collapse
Affiliation(s)
- Fiona Day
- Department of Medical Oncology, Calvary Mater Newcastle, Waratah, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | - Swetha Sridharan
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
- Department of Radiation Oncology, Calvary Mater Newcastle, Waratah, Australia
| | - Catherine Johnson
- Department of Medical Oncology, Calvary Mater Newcastle, Waratah, Australia
| | - Gaik T Quah
- Department of Medical Oncology, Calvary Mater Newcastle, Waratah, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | - Girish Mallesara
- Department of Medical Oncology, Calvary Mater Newcastle, Waratah, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | - Mahesh Kumar
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
- Department of Radiation Oncology, Calvary Mater Newcastle, Waratah, Australia
| | | | - Anthony Morrison
- Department of Medical Oncology, Calvary Mater Newcastle, Waratah, Australia
| | - Andre van der Westhuizen
- Department of Medical Oncology, Calvary Mater Newcastle, Waratah, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | - Allison Fraser
- Department of Medical Oncology, Calvary Mater Newcastle, Waratah, Australia
- Department of Radiation Oncology, Calvary Mater Newcastle, Waratah, Australia
| | - Christopher Oldmeadow
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
- Clinical Research Design, Information Technology and Statistical Support Unit, Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Jarad Martin
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
- Department of Radiation Oncology, Calvary Mater Newcastle, Waratah, Australia
| |
Collapse
|
5
|
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
|
6
|
Song J, Zhang H, Jian J, Chen H, Zhu X, Xie J, Xu X. The Prognostic Significance of Lymph Node Ratio for Esophageal Cancer: A Meta-Analysis. J Surg Res 2023; 292:53-64. [PMID: 37586187 DOI: 10.1016/j.jss.2023.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 08/18/2023]
Abstract
INTRODUCTION This meta-analysis aimed to investigate the prognostic significance of positive lymph node ratio (LNR) in patients with esophageal cancer. MATERIALS AND METHODS The meta-analysis following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. We conducted a systematic search of relevant literature published until April 2022 in PubMed, EMBASE, and the Cochrane Library. The primary and secondary outcomes were overall survival (OS) and disease-free survival (DFS), with corresponding hazard ratios (HR) and 95% confidence intervals (CI). The included studies were subgrouped based on age, study area, adjuvant therapy, sensitivity analysis, and assessment of publication bias. We analyzed and discussed the results. RESULTS We included 21 studies with 29 cohorts and 11,849 patients. The Newcastle-Ottawa Scale scores of the included studies were no less than six, indicating high research quality. The combined results of HR and 95% CI showed that patients with esophageal cancer with a lower LNR had better OS (HR, 2.58; 95% CI, 2.15-3.11; P < 0.001) and DFS (HR, 3.07; 95% CI, 1.85-5.10; P < 0.001). The subgroup analysis suggested that geographic region, age, and adjuvant therapy affected OS. When any cohort was excluded, no significant changes were observed in the pooled HR of the OS group, indicating reliable and robust results. Egger's and Begg's tests showed no potential publication bias in the studies that used OS as an outcome measurement index, indicating reliable results. Sensitivity analyses and assessments of publication bias (<10) were not performed because of an insufficient number of DFS studies. CONCLUSION Patients with a lower positive LNR had a higher survival rate, suggesting that positive LNR may be a promising predictor of EC prognosis in esophageal cancer. After radical resection of esophageal cancer, the ratio of the number of dissected lymph nodes to the number of positive lymph nodes in patients with esophageal cancer should be considered to accurately evaluate the prognosis.
Collapse
Affiliation(s)
- Jiannan Song
- Department of Thoracic Surgery, Anhui Chest Hospital, Thoracic Clinical College of Anhui Medical University, Hefei, Anhui, China
| | - Heng Zhang
- Department of Thoracic Surgery, Anhui Chest Hospital, Thoracic Clinical College of Anhui Medical University, Hefei, Anhui, China
| | - Junling Jian
- Department of Thoracic Surgery, Anhui Chest Hospital, Thoracic Clinical College of Anhui Medical University, Hefei, Anhui, China
| | - Hai Chen
- Department of Thoracic Surgery, Anhui Chest Hospital, Thoracic Clinical College of Anhui Medical University, Hefei, Anhui, China
| | - Xiaodong Zhu
- Department of Thoracic Surgery, Anhui Chest Hospital, Thoracic Clinical College of Anhui Medical University, Hefei, Anhui, China
| | - Jianfeng Xie
- Department of Thoracic Surgery, Anhui Chest Hospital, Thoracic Clinical College of Anhui Medical University, Hefei, Anhui, China
| | - Xianquan Xu
- Department of Thoracic Surgery, Anhui Chest Hospital, Thoracic Clinical College of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
7
|
Su F, Yang X, Yin J, Shen Y, Tan L. Validity of Using Pathological Response as a Surrogate for Overall Survival in Neoadjuvant Studies for Esophageal Cancer: A Systematic Review and Meta-analysis. Ann Surg Oncol 2023; 30:7461-7471. [PMID: 37400616 DOI: 10.1245/s10434-023-13778-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/04/2023] [Indexed: 07/05/2023]
Abstract
BACKGROUND Pathological response is a critical factor in predicting long-term survival of patients with esophageal cancer after preoperative therapy. However, the validity of using pathological response as a surrogate for overall survival (OS) for esophageal cancer has not yet been established. In this study, a literature-based meta-analysis was conducted to evaluate pathological response as a proxy endpoint for survival in esophageal cancer. METHODS Three databases were systematically searched to identify relevant studies investigating neoadjuvant treatment for esophageal cancer. The correlation between pathological complete response (pCR) and OS were assessed using a weighted multiple regression analysis at the trial level, and the coefficient of determination (R2) was calculated. The research design and histological subtypes were considered in the performance of subgroup analysis. RESULTS In this meta-analysis, a total of 40 trials, comprising 43 comparisons and 55,344 patients were qualified. The surrogacy between pCR and OS was moderate (R2 = 0.238 in direct comparison, R2 = 0.500 for pCR reciprocals, R2 = 0.541 in log settings). pCR could not serve as an ideal surrogate endpoint in randomized controlled trials (RCTs) (R2 = 0.511 in direct comparison, R2 = 0.460 for pCR reciprocals, R2 = 0.523 in log settings). A strong correlation was observed in studies comparing neoadjuvant chemoradiotherapy and neoadjuvant chemotherapy (R2 = 0.595 in direct comparison, R2 = 0.840 for pCR reciprocals, R2 = 0.800 in log settings). CONCLUSIONS A lack of surrogacy of pathological response for long-term survival at trial level is established in this study. Hence, caution should be exercised when using pCR as the primary endpoint in neoadjuvant studies for esophageal cancer.
Collapse
Affiliation(s)
- Feng Su
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Xinyu Yang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Jun Yin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Yaxing Shen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China.
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| |
Collapse
|
8
|
Lonie JM, Brosda S, Bonazzi VF, Aoude LG, Patel K, Brown I, Sharma S, Lampe G, Addala V, Koufariotis LT, Wood S, Waddell N, Dolcetti R, Barbour AP. The oesophageal adenocarcinoma tumour immune microenvironment dictates outcomes with different modalities of neoadjuvant therapy - results from the AGITG DOCTOR trial and the cancer evolution biobank. Front Immunol 2023; 14:1220129. [PMID: 37965317 PMCID: PMC10642165 DOI: 10.3389/fimmu.2023.1220129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/14/2023] [Indexed: 11/16/2023] Open
Abstract
A plateau in treatment effect can be seen for the current 'one-size-fits-all' approach to oesophageal adenocarcinoma (OAC) management using neoadjuvant chemoradiotherapy (nCRT) or chemotherapy (nCT). In OAC, the tumour microenvironment (TME) is largely immunosuppressed, however a subgroup of patients with an immune-inflamed TME exist and show improved outcomes. We aimed to understand the overall immune-based mechanisms underlying treatment responses and patient outcomes in OAC, and in relation to neoadjuvant therapy modality. This study included 107 patients; 68 patients were enrolled in the Australian Gastro-Intestinal Trials Group sponsored DOCTOR Trial, and 38 patients were included from the Cancer Evolution Biobank. Matched pre-treatment and post-treatment tumour biopsies were used to perform multi-modality analysis of the OAC TME including NanoString mRNA expression analysis, multiplex and single colour immunohistochemistry (IHC), and peripheral blood mononuclear cell analysis of tumour-antigen specific T cell responses. Patients with the best clinicopathological outcomes and survival had an immune-inflamed TME enriched with anti-tumour immune cells and pathways. Those with the worst survival showed a myeloid T regulatory cell enriched TME, with decreased CD8+ cell infiltration and increased pro-tumour immune cells. Multiplex IHC analysis identified that high intra-tumoural infiltration of CD8+ cells, and low infiltration with CD163+ cells was associated with improved survival. High tumour core CD8+ T cell infiltration, and a low tumour margin infiltration of CD163+ cells was also associated with improved survival. nCRT showed improved survival compared with nCT for patients with low CD8+, or high CD163+ cell infiltration. Poly-functional T cell responses were seen with tumour-antigen specific T cells. Overall, our study supports the development of personalised therapeutic approaches based on the immune microenvironment in OAC. Patients with an immune-inflamed TME show favourable outcomes regardless of treatment modality. However, in those with an immunosuppressed TME with CD163+ cell infiltration, treatment with nCRT can improve outcomes. Our findings support previous studies into the TME of OAC and with more research, immune based biomarker selection of treatment modality may lead in improved outcomes in this deadly disease.
Collapse
Affiliation(s)
- James M. Lonie
- Surgical Oncology Group, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Sandra Brosda
- Surgical Oncology Group, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Vanessa F. Bonazzi
- Surgical Oncology Group, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Lauren G. Aoude
- Surgical Oncology Group, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Kalpana Patel
- Surgical Oncology Group, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Ian Brown
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Envoi Specialist Pathologists, Brisbane, QLD, Australia
- Department of Pathology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Sowmya Sharma
- Medlab Pathology, Sydney, NSW, Australia
- Medical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Guy Lampe
- Department of Anatomical Pathology, Central Laboratory Pathology Queensland, Brisbane, QLD, Australia
| | - Venkateswar Addala
- Medical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | - Scott Wood
- Medical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nicola Waddell
- Medical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Riccardo Dolcetti
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Translational and Clinical Immunotherapy, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Andrew P. Barbour
- Surgical Oncology Group, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
- Department of Surgery, Princess Alexandra Hospital, Brisbane, QLD, Australia
| |
Collapse
|
9
|
Sasaki T, Matsumoto Y, Murakami K, Endo S, Toyozumi T, Otsuka R, Kinoshita K, Hu J, Iida S, Morishita H, Nishioka Y, Nakano A, Uesato M, Matsubara H. Gut microbiome can predict chemoradiotherapy efficacy in patients with esophageal squamous cell carcinoma. Esophagus 2023; 20:691-703. [PMID: 37086309 DOI: 10.1007/s10388-023-01004-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/12/2023] [Indexed: 04/23/2023]
Abstract
PURPOSE The gut microbiome plays an important role in cancer pathogenesis and therapy. Some studies have reported that specific bacteria in tumor tissues may contribute to the prognosis and treatment of esophageal squamous cell carcinoma (ESCC). However, there is limited evidence that the gut microbiome is associated with ESCC. This study assessed the utility of the gut microbiome as a predictive marker of the therapeutic effect in patients with ESCC undergoing chemo-radiotherapy (CRT). PATIENTS AND METHODS Fecal samples were collected from 51 patients with ESCC who had never undergone treatment between April 2021 and May 2022 in the Department of Frontier Surgery, Chiba University. The gut microbiome was analyzed using 16S metagenomics sequencing. The association between the gut microbiome composition and stage according to the TNM classification (American Joint Committee on Cancer 7.0) and CRT response according to the RECIST criteria was evaluated. RESULTS The relative abundance of Fusobacteriaceae was enriched in cStage III-IVb group. Among the 27 patients who received CRT, the relative abundance of Lactobacillaceae was enriched in those with a partial and complete response. Lactobacillaceae also did not correlate with any clinical data, but the high Lactobacillales group had a higher LMR (P = 0.032) and lower PLR (P = 0.045) than in the low Lactobacillales group. CONCLUSIONS In conclusion, we found that the relative abundance of Lactobacillaceae was enriched in patients with a partial or complete response among CRT those with ESCC, thus suggesting that the relative abundance of Lactobacillaceae can predict the effect of CRT.
Collapse
Affiliation(s)
- Takuma Sasaki
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Yasunori Matsumoto
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan.
| | - Kentaro Murakami
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Satoshi Endo
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Takeshi Toyozumi
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Ryota Otsuka
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Kazuya Kinoshita
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Jie Hu
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Shinichiro Iida
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Hiroki Morishita
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Yuri Nishioka
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Akira Nakano
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Masaya Uesato
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, 260-8670, Japan
| |
Collapse
|
10
|
Ho YC, Lai YC, Lin HY, Ko MH, Wang SH, Yang SJ, Chou TW, Hung LC, Huang CC, Chang TH, Lin JB, Lin JC. Cardiac Dose Predicts the Response to Concurrent Chemoradiotherapy in Esophageal Squamous Cell Carcinoma. Cancers (Basel) 2023; 15:4580. [PMID: 37760549 PMCID: PMC10526131 DOI: 10.3390/cancers15184580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Definitive concurrent chemoradiation (CCRT) is the standard treatment for cervical esophageal cancer and non-surgical candidates. Initial treatment response affects survival; however, few validated markers are available for prediction. This study evaluated the clinical variables and chemoradiation parameters associated with treatment response. Between May 2010 and April 2016, 86 completed CCRT patients' clinical, dosimetric, and laboratory data at baseline and during treatment were collected. Cox regression analysis assessed the risk factors for overall survival (OS). A receiver operating characteristic curve with Youden's index was chosen to obtain the optimal cut-off value of each parameter. Treatment response was defined per Response Evaluation Criteria in Solid Tumors v.1.1 at the first post-CCRT computed tomography scan. Responders had complete and partial responses; non-responders had stable and progressive diseases. Logistic regression (LR) was used to evaluate the variables associated with responders. The Cox regression model confirmed the presence of responders (n = 50) vs. non-responders (n = 36) with a significant difference in OS. In multivariate LR, cardiac dose-volume received ≥10 Gy; the baseline hemoglobin level, highest neutrophil to lymphocyte ratio during CCRT, and cumulative cisplatin dose were significantly associated with the responders. The initial clinical treatment response significantly determines disease outcome. Cardiac irradiation may affect the treatment response.
Collapse
Affiliation(s)
- Yu-Chieh Ho
- Department of Radiation Oncology, Changhua Christian Hospital, Changhua 500, Taiwan; (Y.-C.H.); (S.-J.Y.); (T.-W.C.); (L.-C.H.); (C.-C.H.); (T.-H.C.); (J.-C.L.)
| | - Yuan-Chun Lai
- Division of Medical Physics, Department of Radiation Oncology, Changhua Christian Hospital, Changhua 500, Taiwan; (Y.-C.L.); (M.-H.K.)
- Department of Medical Imaging Radiological Science, Central Taiwan University of Science and Technology, Taichung 406, Taiwan
| | - Hsuan-Yu Lin
- Division of Haematology/Oncology, Department of Internal Medicine, Changhua Christian Hospital, Changhua 500, Taiwan;
| | - Ming-Hui Ko
- Division of Medical Physics, Department of Radiation Oncology, Changhua Christian Hospital, Changhua 500, Taiwan; (Y.-C.L.); (M.-H.K.)
| | - Sheng-Hung Wang
- Department of Radiation Oncology, Lukang Christian Hospital, Changhua Christian Medical Foundation, Lukang 505, Taiwan;
| | - Shan-Jun Yang
- Department of Radiation Oncology, Changhua Christian Hospital, Changhua 500, Taiwan; (Y.-C.H.); (S.-J.Y.); (T.-W.C.); (L.-C.H.); (C.-C.H.); (T.-H.C.); (J.-C.L.)
| | - Tsai-Wei Chou
- Department of Radiation Oncology, Changhua Christian Hospital, Changhua 500, Taiwan; (Y.-C.H.); (S.-J.Y.); (T.-W.C.); (L.-C.H.); (C.-C.H.); (T.-H.C.); (J.-C.L.)
| | - Li-Chung Hung
- Department of Radiation Oncology, Changhua Christian Hospital, Changhua 500, Taiwan; (Y.-C.H.); (S.-J.Y.); (T.-W.C.); (L.-C.H.); (C.-C.H.); (T.-H.C.); (J.-C.L.)
| | - Chia-Chun Huang
- Department of Radiation Oncology, Changhua Christian Hospital, Changhua 500, Taiwan; (Y.-C.H.); (S.-J.Y.); (T.-W.C.); (L.-C.H.); (C.-C.H.); (T.-H.C.); (J.-C.L.)
| | - Tung-Hao Chang
- Department of Radiation Oncology, Changhua Christian Hospital, Changhua 500, Taiwan; (Y.-C.H.); (S.-J.Y.); (T.-W.C.); (L.-C.H.); (C.-C.H.); (T.-H.C.); (J.-C.L.)
- Department of Medical Imaging Radiological Science, Central Taiwan University of Science and Technology, Taichung 406, Taiwan
- Department of Medical Imaging and Radiological Technology, Yuanpei University of Science and Technology, Hsinchu 300, Taiwan
| | - Jhen-Bin Lin
- Department of Radiation Oncology, Changhua Christian Hospital, Changhua 500, Taiwan; (Y.-C.H.); (S.-J.Y.); (T.-W.C.); (L.-C.H.); (C.-C.H.); (T.-H.C.); (J.-C.L.)
| | - Jin-Ching Lin
- Department of Radiation Oncology, Changhua Christian Hospital, Changhua 500, Taiwan; (Y.-C.H.); (S.-J.Y.); (T.-W.C.); (L.-C.H.); (C.-C.H.); (T.-H.C.); (J.-C.L.)
- Research Department, Division of Translation Research, Changhua Christian Hospital, Changhua 500, Taiwan
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei 112, Taiwan
| |
Collapse
|
11
|
Mukherjee S, Hurt CN, Adams R, Bateman A, Bradley KM, Bridges S, Falk S, Griffiths G, Gwynne S, Jones CM, Markham PJ, Maughan T, Nixon LS, Radhakrishna G, Roy R, Schoenbuchner S, Sheikh H, Spezi E, Hawkins M, Crosby TD. Efficacy of early PET-CT directed switch to carboplatin and paclitaxel based definitive chemoradiotherapy in patients with oesophageal cancer who have a poor early response to induction cisplatin and capecitabine in the UK: a multi-centre randomised controlled phase II trial. EClinicalMedicine 2023; 61:102059. [PMID: 37409323 PMCID: PMC10318451 DOI: 10.1016/j.eclinm.2023.102059] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 07/07/2023] Open
Abstract
Background The utility of early metabolic response assessment to guide selection of the systemic component of definitive chemoradiotherapy (dCRT) for oesophageal cancer is uncertain. Methods In this multi-centre, randomised, open-label, phase II substudy of the radiotherapy dose-escalation SCOPE2 trial we evaluated the role of 18F-Fluorodeoxyglucose positron emission tomography (PET) at day 14 of cycle 1 of three-weekly induction cis/cap (cisplatin (60 mg/m2)/capecitabine (625 mg/m2 days 1-21)) in patients with oesophageal squamous cell carcinoma (OSCC) or adenocarcinoma (OAC). Non-responders, who had a less than 35% reduction in maximum standardised uptake value (SUVmax) from pre-treatment baseline, were randomly assigned to continue cis/cap or switch to car/pac (carboplatin AUC 5/paclitaxel 175 mg/m2) for a further induction cycle, then concurrently with radiotherapy over 25 fractions. Responders continued cis/cap for the duration of treatment. All patients (including responders) were randomised to standard (50Gy) or high (60Gy) dose radiation as part of the main study. Primary endpoint for the substudy was treatment failure-free survival (TFFS) at week 24. The trial was registered with International Standard Randomized Controlled Trial Number 97125464 and ClinicalTrials.govNCT02741856. Findings This substudy was closed on 1st August 2021 by the Independent Data Monitoring Committee on the grounds of futility and possible harm. To this point from 22nd November 2016, 103 patients from 16 UK centres had participated in the PET-CT substudy; 63 (61.2%; 52/83 OSCC, 11/20 OAC) of whom were non-responders. Of these, 31 were randomised to car/pac and 32 to remain on cis/cap. All patients were followed up until at least 24 weeks, at which point in OSCC both TFFS (25/27 (92.6%) vs 17/25 (68%); p = 0.028) and overall survival (42.5 vs. 20.4 months, adjusted HR 0.36; p = 0.018) favoured cis/cap over car/pac. There was a trend towards worse survival in OSCC + OAC cis/cap responders (33.6 months; 95%CI 23.1-nr) vs. non-responders (42.5 (95%CI 27.0-nr) months; HR = 1.43; 95%CI 0.67-3.08; p = 0.35). Interpretation In OSCC, early metabolic response assessment is not prognostic for TFFS or overall survival and should not be used to personalise systemic therapy in patients receiving dCRT. Funding Cancer Research UK.
Collapse
Affiliation(s)
- Somnath Mukherjee
- Oxford Cancer Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Christopher N. Hurt
- Centre for Trials Research, Cardiff University, Cardiff, UK
- Southampton Clinical Trials Unit, University of Southampton, Southampton, UK
| | - Richard Adams
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Andrew Bateman
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Kevin M. Bradley
- Wales Research and Diagnostic Positron Emission Tomography Centre (PETIC), Cardiff University, Cardiff, UK
| | - Sarah Bridges
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Stephen Falk
- University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Gareth Griffiths
- Southampton Clinical Trials Unit, University of Southampton, Southampton, UK
| | - Sarah Gwynne
- South West Wales Cancer Centre, Swansea Bay University Health Board, Swansea, UK
| | | | | | - Tim Maughan
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | | | - Ganesh Radhakrishna
- The Christie Hospital, The Christie Hospitals NHS Foundation Trust, Manchester, UK
| | - Rajarshi Roy
- Queen's Centre for Oncology, Hull University Teaching Hospitals NHS Trust, UK
| | | | - Hamid Sheikh
- The Christie Hospital, The Christie Hospitals NHS Foundation Trust, Manchester, UK
| | | | - Maria Hawkins
- Department of Medical Physics & Biomedical Engineering, University College London, London, UK
| | | |
Collapse
|
12
|
M Naeini M, Newell F, Aoude LG, Bonazzi VF, Patel K, Lampe G, Koufariotis LT, Lakis V, Addala V, Kondrashova O, Johnston RL, Sharma S, Brosda S, Holmes O, Leonard C, Wood S, Xu Q, Thomas J, Walpole E, Tao Mai G, Ackland SP, Martin J, Burge M, Finch R, Karapetis CS, Shannon J, Nott L, Bohmer R, Wilson K, Barnes E, Zalcberg JR, Mark Smithers B, Simes J, Price T, Gebski V, Nones K, Watson DI, Pearson JV, Barbour AP, Waddell N. Multi-omic features of oesophageal adenocarcinoma in patients treated with preoperative neoadjuvant therapy. Nat Commun 2023; 14:3155. [PMID: 37258531 PMCID: PMC10232490 DOI: 10.1038/s41467-023-38891-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 05/19/2023] [Indexed: 06/02/2023] Open
Abstract
Oesophageal adenocarcinoma is a poor prognosis cancer and the molecular features underpinning response to treatment remain unclear. We investigate whole genome, transcriptomic and methylation data from 115 oesophageal adenocarcinoma patients mostly from the DOCTOR phase II clinical trial (Australian New Zealand Clinical Trials Registry-ACTRN12609000665235), with exploratory analysis pre-specified in the study protocol of the trial. We report genomic features associated with poorer overall survival, such as the APOBEC mutational and RS3-like rearrangement signatures. We also show that positron emission tomography non-responders have more sub-clonal genomic copy number alterations. Transcriptomic analysis categorises patients into four immune clusters correlated with survival. The immune suppressed cluster is associated with worse survival, enriched with myeloid-derived cells, and an epithelial-mesenchymal transition signature. The immune hot cluster is associated with better survival, enriched with lymphocytes, myeloid-derived cells, and an immune signature including CCL5, CD8A, and NKG7. The immune clusters highlight patients who may respond to immunotherapy and thus may guide future clinical trials.
Collapse
Affiliation(s)
- Marjan M Naeini
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Felicity Newell
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Lauren G Aoude
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Vanessa F Bonazzi
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Kalpana Patel
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Guy Lampe
- Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia
| | | | - Vanessa Lakis
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Venkateswar Addala
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Olga Kondrashova
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Rebecca L Johnston
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Sowmya Sharma
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, 4006, Australia
- Anatomical Pathology, Australian Clinical Labs, 2153, Sydney, Australia
| | - Sandra Brosda
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Oliver Holmes
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Conrad Leonard
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Scott Wood
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Qinying Xu
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Janine Thomas
- Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia
- Mater Research Institute, Mater Misericordiae, South Brisbane, QLD, 4101, Australia
| | - Euan Walpole
- Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia
| | - G Tao Mai
- Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia
| | - Stephen P Ackland
- Department of Medical Oncology, Calvary Mater Newcastle, Waratah, NSW, 2298, Australia
| | - Jarad Martin
- Department of Radiation Oncology, Calvary Mater Newcastle, Waratah, NSW, 2298, Australia
| | - Matthew Burge
- Royal Brisbane and Women's Hospital, Herston, QLD, 4029, Australia
| | - Robert Finch
- Royal Brisbane and Women's Hospital, Herston, QLD, 4029, Australia
| | - Christos S Karapetis
- Flinders University Department of Medical Oncology, Flinders Medical Centre, Adelaide, SA, 5042, Australia
| | - Jenny Shannon
- Nepean Cancer Care Centre, Nepean Hospital, Sydney, NSW, 2747, Australia
| | - Louise Nott
- Department of Medical Oncology, Royal Hobart Hospital, Hobart, TAS, Australia
| | - Robert Bohmer
- Department of General Surgery, Royal Hobart Hospital, Hobart, TAS, Australia
| | - Kate Wilson
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - Elizabeth Barnes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - John R Zalcberg
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, 3004, Australia
| | - B Mark Smithers
- Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, 4006, Australia
| | - John Simes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - Timothy Price
- Medical Oncology Unit, The Queen Elizabeth Hospital and University of Adelaide, Adelaide, SA, 5011, Australia
| | - Val Gebski
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - Katia Nones
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - David I Watson
- Flinders University Discipline of Surgery, Flinders Medical Centre, Adelaide, SA, 5042, Australia
| | - John V Pearson
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Andrew P Barbour
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
- Princess Alexandra Hospital, Woolloongabba, QLD, 4102, Australia.
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia.
| |
Collapse
|
13
|
Cowzer D, Keane F, Ku GY. Clinical Utility of 18F-2-Fluoro-deoxy-d-glucose PET Imaging in Locally Advanced Esophageal/Gastroesophageal Junction Adenocarcinoma. Diagnostics (Basel) 2023; 13:1884. [PMID: 37296735 PMCID: PMC10252409 DOI: 10.3390/diagnostics13111884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Esophageal adenocarcinoma, including adenocarcinoma of the gastroesophageal junction, is uncommon in the United States, but is associated with a rising incidence in young adults, and has a traditionally poor prognosis. Despite the incremental benefits that have been made with multimodality approaches to locally advanced disease, most patients will go on to develop metastatic disease, and long-term outcomes remain suboptimal. Over the last decade, PET-CT has emerged as a key tool in the management of this disease, with several prospective and retrospective studies evaluating its role in this disease. Herein, we review the key data pertaining to the use of PET-CT in the management of locally advanced esophageal and GEJ adenocarcinoma, with a focus on staging, prognostication, PET-CT adapted therapy in the neoadjuvant setting, and surveillance.
Collapse
Affiliation(s)
- Darren Cowzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (D.C.); (F.K.)
| | - Fergus Keane
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (D.C.); (F.K.)
| | - Geoffrey Y. Ku
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (D.C.); (F.K.)
- Department of Medicine, Weill Cornell University, New York, NY 10065, USA
| |
Collapse
|
14
|
Yang H, Ai H, Zhang J, Ma J, Liu K, Li Z. UPS: Opportunities and challenges for gastric cancer treatment. Front Oncol 2023; 13:1140452. [PMID: 37077823 PMCID: PMC10106573 DOI: 10.3389/fonc.2023.1140452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
Gastric cancer remains the fourth most frequently diagnosed malignancy and the fifth leading cause of cancer-related mortality worldwide owning to the lack of efficient drugs and targets for therapy. Accumulating evidence indicates that UPS, which consists of E1, E2, and E3 enzymes and proteasome, plays an important role in the GC tumorigenesis. The imbalance of UPS impairs the protein homeostasis network during development of GC. Therefore, modulating these enzymes and proteasome may be a promising strategy for GC target therapy. Besides, PROTAC, a strategy using UPS to degrade the target protein, is an emerging tool for drug development. Thus far, more and more PROTAC drugs enter clinical trials for cancer therapy. Here, we will analyze the abnormal expression enzymes in UPS and summarize the E3 enzymes which can be developed in PROTAC so that it can contribute to the development of UPS modulator and PROTAC technology for GC therapy.
Collapse
Affiliation(s)
- Hang Yang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Huihan Ai
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jialin Zhang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jie Ma
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US Hormel (Henan) Cancer Institute, Zhengzhou, Henan, China
- Research Center of Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Zhi Li, ; Kangdong Liu,
| | - Zhi Li
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- *Correspondence: Zhi Li, ; Kangdong Liu,
| |
Collapse
|
15
|
Barron CC, Wang X, Elimova E. Neoadjuvant Strategies for Esophageal Cancer. Thorac Surg Clin 2023; 33:197-208. [PMID: 37045489 DOI: 10.1016/j.thorsurg.2023.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
Neoadjuvant strategies with multimodal therapy including chemotherapy and radiation are the standard of care in locally advanced esophageal cancer. The role of immunotherapy in the perioperative management of esophageal cancer is expanding, and adjuvant nivolumab for patients with residual disease following trimodality therapy has been shown to improve disease-free survival. Applications of checkpoint blockade and positron emission tomography (PET)-directed therapy in the neoadjuvant setting are under investigation in several clinical trials. We review the perioperative management of locally advanced esophageal cancer and recent evidence exploring the role of immune checkpoint inhibitors and PET in guiding neoadjuvant management.
Collapse
|
16
|
Guo H, Tang HT, Hu WL, Wang JJ, Liu PZ, Yang JJ, Hou SL, Zuo YJ, Deng ZQ, Zheng XY, Yan HJ, Jiang KY, Huang H, Zhou HN, Tian D. The application of radiomics in esophageal cancer: Predicting the response after neoadjuvant therapy. Front Oncol 2023; 13:1082960. [PMID: 37091180 PMCID: PMC10117779 DOI: 10.3389/fonc.2023.1082960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/27/2023] [Indexed: 04/25/2023] Open
Abstract
Esophageal cancer (EC) is one of the fatal malignant neoplasms worldwide. Neoadjuvant therapy (NAT) combined with surgery has become the standard treatment for locally advanced EC. However, the treatment efficacy for patients with EC who received NAT varies from patient to patient. Currently, the evaluation of efficacy after NAT for EC lacks accurate and uniform criteria. Radiomics is a multi-parameter quantitative approach for developing medical imaging in the era of precision medicine and has provided a novel view of medical images. As a non-invasive image analysis method, radiomics is an inevitable trend in NAT efficacy prediction and prognosis classification of EC by analyzing the high-throughput imaging features of lesions extracted from medical images. In this literature review, we discuss the definition and workflow of radiomics, the advances in efficacy prediction after NAT, and the current application of radiomics for predicting efficacy after NAT.
Collapse
Affiliation(s)
- Hai Guo
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Department of Thoracic Surgery, Sichuan Tianfu New Area People’s Hospital, Chengdu, China
| | - Hong-Tao Tang
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Wen-Long Hu
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Jun-Jie Wang
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Pei-Zhi Liu
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Jun-Jie Yang
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Sen-Lin Hou
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Yu-Jie Zuo
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Zhi-Qiang Deng
- College of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Xiang-Yun Zheng
- College of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Hao-Ji Yan
- Department of General Thoracic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kai-Yuan Jiang
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Heng Huang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hai-Ning Zhou
- Department of Thoracic Surgery, Suining Central Hospital, Suining, China
- *Correspondence: Dong Tian, ; Hai-Ning Zhou,
| | - Dong Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Dong Tian, ; Hai-Ning Zhou,
| |
Collapse
|
17
|
Lorenzen S, Quante M, Rauscher I, Slotta-Huspenina J, Weichert W, Feith M, Friess H, Combs SE, Weber WA, Haller B, Angele M, Albertsmeier M, Blankenstein C, Kasper S, Schmid RM, Bassermann F, Schwaiger M, Liffers ST, Siveke JT. PET-directed combined modality therapy for gastroesophageal junction cancer: Results of the multicentre prospective MEMORI trial of the German Cancer Consortium (DKTK). Eur J Cancer 2022; 175:99-106. [PMID: 36099671 DOI: 10.1016/j.ejca.2022.07.027] [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: 05/20/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Positron emission tomography (PET) may differentiate responding and non-responding tumours early in the treatment of locally advanced gastroesophageal junction adenocarcinomas. Early PET non-responders (P-NR) after induction CTX might benefit from changing to chemoradiation (CRT). METHODS Patients underwent baseline 18F-FDG PET followed by 1 cycle of CTX. PET was repeated at day 14-21 and responders (P-R), defined as ≥35% decrease in SUVmean from baseline, continued with CTX. P-NR switched to CRT (CROSS). Patients underwent surgery 4-6 weeks post-CTX/CRT. The primary objective was an improvement in R0 resection rates in P-NR above a proportion of 70%. RESULTS In total, 160 patients with resectable gastroesophageal junction adenocarcinomas were prospectively investigated by PET scanning. Eighty-five patients (53%) were excluded. Seventy-five eligible patients were enrolled in the study. Based on PET criteria, 50 (67.6%)/24 (32.4%) were P-R and P-NR, respectively. Resection was performed on 46 responders, including one patient who withdrew the ICF, and 22 non-responders (per-protocol population). R0 resection rates were 95.6% (43/45) for P-R and 86.4% (19/22) for P-NR. No treatment related deaths occurred. With a median follow-up time of 24.5 months, estimated 18 months DFS was 75.4%/64.2% for P-R/P-NR, respectively. The estimated 18 months OS was 95.5% for P-R and 68.2% for P-NR. CONCLUSION The primary endpoint of the study to increase the R0 resection rate in metabolic NR was not met. PET response after induction CTX is prognostic for outcome with a prolonged OS and DFS in PET responders. TRIAL REGISTRATION NCT00002014-000860-16.
Collapse
Affiliation(s)
- Sylvie Lorenzen
- Technical University of Munich, Klinikum rechts der Isar, III. Medizinische Klinik und Poliklinik, Munich, Germany
| | - Michael Quante
- Technical University Munich, Klinikum rechts der Isar, II. Medizinische Klinik und Poliklinik, Munich, Germany; Department of Internal Medicine II, University of Freiburg, Germany
| | - Isabel Rauscher
- Technical University Munich, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | | | - Wilko Weichert
- Technical University Munich, Institute of Pathology, Munich, Germany
| | - Marcus Feith
- Technical University Munich, Klinikum rechts der Isar, Surgical Clinic and Policlinic, Munich, Germany
| | - Helmut Friess
- Technical University Munich, Klinikum rechts der Isar, Surgical Clinic and Policlinic, Munich, Germany
| | - Stefanie E Combs
- Technical University Munich, Klinikum rechts der Isar, Department of Radiation Oncology, Munich, Germany
| | - Wolfgang A Weber
- Technical University Munich, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | - Bernhard Haller
- Technical University Munich, Klinikum rechts der Isar, Institute of AI and Informatics in Medicine, Munich, Germany
| | - Martin Angele
- Ludwig-Maximilians-Universität (LMU) Munich, LMU University Hospital, Department of General, Visceral and Transplantation Surgery, Munich, Germany
| | - Markus Albertsmeier
- Ludwig-Maximilians-Universität (LMU) Munich, LMU University Hospital, Department of General, Visceral and Transplantation Surgery, Munich, Germany
| | | | - Stefan Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen, Germany
| | - Roland M Schmid
- Technical University Munich, Klinikum rechts der Isar, II. Medizinische Klinik und Poliklinik, Munich, Germany; Department of Internal Medicine II, University of Freiburg, Germany
| | - Florian Bassermann
- Technical University of Munich, Klinikum rechts der Isar, III. Medizinische Klinik und Poliklinik, Munich, Germany
| | - Markus Schwaiger
- Technical University Munich, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | - Sven-Thorsten Liffers
- German Cancer Consortium (DKTK), Partner Site Essen, Germany; Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital, University of Duisburg-Essen, Essen, Germany; Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Jens T Siveke
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen, Germany; Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital, University of Duisburg-Essen, Essen, Germany; Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany.
| |
Collapse
|
18
|
Kumar R, Tchelebi L, Anker CJ, Sharma N, Bianchi NA, Dragovic J, Goodman KA, Herman JM, Jiang Y, Jones WE, Kennedy TJ, Lee P, Kundranda M, Russo S, Small W, Suh WW, Yee N, Jabbour SK. American Radium Society (ARS) Appropriate Use Criteria (AUC) for Locoregional Gastric Adenocarcinoma: Systematic Review and Guidelines. Am J Clin Oncol 2022; 45:391-402. [PMID: 35947781 PMCID: PMC10865426 DOI: 10.1097/coc.0000000000000930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The objective of this study was to systematically evaluate the data regarding the use of neoadjuvant, perioperative, surgical, and adjuvant treatment options in localized gastric cancer patients and to develop Appropriate Use Criteria recommended by a panel of experts convened by the American Radium Society. METHODS Preferred reporting items for systematic reviews and meta-analyses methodology was used to develop an extensive analysis of peer-reviewed phase 2/2R/3 trials, as well as meta-analyses found within the Ovid Medline database between 2010 and 2020. The expert panel then rated the appropriateness of various treatments in 5 representative clinical scenarios through a well-established consensus methodology (modified Delphi). RESULTS For patients with medically operable locally advanced gastric cancer, the strongest recommendation was for perioperative chemotherapy based on high-quality data. Acceptable alternatives included surgery followed by either chemotherapy or concurrent chemoradiotherapy (CRT). For patients with upfront resection of stages I to III gastric cancer (no neoadjuvant therapy), the group strongly recommended adjuvant therapy with either chemotherapy alone or CRT, based on high-quality data. For patients with locally advanced disease who received preoperative chemotherapy without tumor regression, the group strongly recommended postoperative chemotherapy or postoperative CRT. Finally, for medically inoperable gastric cancer patients, there was moderate consensus recommending definitive concurrent CRT. CONCLUSIONS The addition of chemotherapy and/or radiation, either in the neoadjuvant, adjuvant, or perioperative setting, results in improved survival rates for patients compared with surgery alone. For inoperable patients, definitive CRT is a reasonable treatment option, though largely palliative.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yixing Jiang
- UT Health Cancer Center, University of Texas Health Science Center, San Antonio
| | | | | | - Percy Lee
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Suzanne Russo
- School of Medicine, University Hospitals, Case Western Reserve University, Cleveland, OH
| | - William Small
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL
| | - Wonsuk W. Suh
- Ridley-Tree Cancer Center Santa Barbara at Sansum Clinic, Santa Barbara, CA
| | - Nelson Yee
- Northwell Health Cancer Institute, Mount Kisco
| | - Salma K. Jabbour
- Panel Chair, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| |
Collapse
|
19
|
Zhu H, Liu Q, Xu H, Mo M, Wang Z, Lu K, Zhou J, Chen J, Zheng X, Ye J, Ge X, Luo H, Liu Q, Deng J, Ai D, Hao S, Zhang J, Tseng IH, Song S, Chen Y, Zhao K. Dose escalation based on 18F-FDG PET/CT response in definitive chemoradiotherapy of locally advanced esophageal squamous cell carcinoma: a phase III, open-label, randomized, controlled trial (ESO-Shanghai 12). Radiat Oncol 2022; 17:134. [PMID: 35906623 PMCID: PMC9338557 DOI: 10.1186/s13014-022-02099-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 07/07/2022] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION Definitive chemoradiotherapy has established the standard non-surgical treatment for locally advanced esophageal cancer. The standard dose of 50-50.4 Gy has been established decades ago and been confirmed in modern trials. The theorical advantage of better local control and technical advances for less toxicity have encouraged clinicians for dose escalation investigation. 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) have the potential to tailor therapy for esophageal patients not showing response to CRT and pioneers the PET-based dose escalation. METHODS AND ANALYSIS The ESO-Shanghai 12 trial is a prospective multicenter randomized phase 3 study in which patients are randomized to either 61.2 Gy or 50.4 Gy of radiation dose by PET response. Both groups undergo concurrent chemoradiotherapy with paclitaxel/cisplatin regimen for 2 cycles followed by consolidation chemotherapy for 2 cycles. Patients with histologically confirmed ESCC [T1N1-3M0, T2-4NxM0, TxNxM1 (Supraclavicular lymph node metastasis only), (AJCC Cancer Staging Manual, 8th Edition)] and without any prior treatment of chemotherapy, radiotherapy or surgery against esophageal cancer will be eligible. The primary endpoints included overall survival in PET/CT non-responders (SUVmax > 4.0) and overall survival in total population. Patients will be stratified by standardized uptake volume, gross tumor volume and tumor location. The enrollment could be ended, when the number of PET/CT non-responder reached 132 and the total population reached 646 for randomization. ETHICS AND DISSEMINATION This trial has been approved by the Fudan University Shanghai Cancer Center Institutional Review Board. Trial results will be disseminated via peer reviewed scientific journals and conference presentations. Trial registration The trial was initiated in 2018 and is currently recruiting patients. Trial registration number NCT03790553.
Collapse
Affiliation(s)
- Hongcheng Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Qiufang Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hao Xu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Miao Mo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Cancer Prevention and Statistics, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zezhou Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Cancer Prevention and Statistics, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Kui Lu
- Department of Radiation Oncology, Taizhou Second People's Hospital, Taizhou, Jiangsu, China
| | - Jialiang Zhou
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Junqiang Chen
- Department of Radiation Oncology, Fujian Cancer Hospital, Fuzhou, China
| | - Xiangpeng Zheng
- Department of Radiation Oncology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jinjun Ye
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing, China
| | - Xiaolin Ge
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Honglei Luo
- Department of Radiation Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Qi Liu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Jiaying Deng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Dashan Ai
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Shengnan Hao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Junhua Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - I Hsuan Tseng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Shaoli Song
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yun Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. .,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China.
| | - Kuaile Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. .,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China.
| |
Collapse
|
20
|
Disease-free survival as a surrogate endpoint for overall survival in adults with resectable esophageal or gastroesophageal junction cancer: A correlation meta-analysis. Eur J Cancer 2022; 170:119-130. [DOI: 10.1016/j.ejca.2022.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/11/2022] [Accepted: 04/16/2022] [Indexed: 12/24/2022]
|
21
|
Ahmad MU, Javadi C, Poultsides GA. Neoadjuvant Treatment Strategies for Resectable Proximal Gastric, Gastroesophageal Junction and Distal Esophageal Cancer. Cancers (Basel) 2022; 14:1755. [PMID: 35406527 PMCID: PMC8996907 DOI: 10.3390/cancers14071755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 01/27/2023] Open
Abstract
Neoadjuvant treatment strategies for resectable proximal gastric, gastroesophageal junction (GEJ), and distal esophageal cancer have evolved over several decades. Treatment recommendations differ based on histologic type-squamous cell carcinoma (SCC) versus adenocarcinoma (AC)-as well as the exact location of the tumor. Recent and older clinical trials in this area were critically reviewed. Neoadjuvant chemoradiation with concurrent taxane- or fluoropyrimidine-based chemotherapy has an established role for both AC and SCC of the distal esophagus and GEJ. The use of perioperative chemotherapy for gastric AC is based on the FLOT4 and MAGIC trials; however, the utility of neoadjuvant chemoradiation in this setting requires further evaluation. Additional clinical trials evaluating chemotherapy, targeted therapy, immunotherapy, and radiation that are currently in process are highlighted, given the need for further disease control.
Collapse
Affiliation(s)
| | | | - George A. Poultsides
- Section of Surgical Oncology, Department of Surgery, Stanford University, Stanford, CA 94205, USA; (M.U.A.); (C.J.)
| |
Collapse
|
22
|
Mercieca-Bebber R, Barnes EH, Wilson K, Samoon Z, Walpole E, Mai T, Ackland S, Burge M, Dickie G, Watson D, Leung J, Wang T, Bohmer R, Cameron D, Simes J, Gebski V, Smithers M, Thomas J, Zalcberg J, Barbour AP. Patient-reported outcome (PRO) results from the AGITG DOCTOR trial: a randomised phase 2 trial of tailored neoadjuvant therapy for resectable oesophageal adenocarcinoma. BMC Cancer 2022; 22:276. [PMID: 35291965 PMCID: PMC8922838 DOI: 10.1186/s12885-022-09270-4] [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: 07/28/2021] [Accepted: 02/07/2022] [Indexed: 11/12/2022] Open
Abstract
Background AGITG DOCTOR was a randomised phase 2 trial of pre-operative cisplatin, 5 fluorouracil (CF) followed by docetaxel (D) with or without radiotherapy (RT) based on poor early response to CF, detected via PET, for resectable oesophageal adenocarcinoma. This study describes PROs over 2 years. Methods Participants (N = 116) completed the EORTC QLQ-C30 and oesophageal module (QLQ-OES18) before chemotherapy (baseline), before surgery, six and 12 weeks post-surgery and three-monthly until 2 years. We plotted PROs over time and calculated the percentage of participants per treatment group whose post-surgery score was within 10 points (threshold for clinically relevant change) of their baseline score, for each PRO scale. We examined the relationship between Grade 3+ adverse events (AEs) and PROs. This analysis included four groups: CF responders, non-responders randomised to DCF, non-responders randomised to DCF + RT, and “others” who were not randomised. Results Global QOL was clinically similar between groups from 6 weeks post-surgery. All groups had poorer functional and higher symptom scores during active treatment and shortly after surgery, particularly the DCF and DCF + RT groups. DCF + RT reported a clinically significant difference (−13points) in mean overall health/QOL between baseline and pre-surgery. Similar proportions of patients across groups scored +/− 10 points of baseline scores within 2 years for most PRO domains. Instance of grade 3+ AEs were not related to PROs at baseline or 2 years. Conclusions By 2 years, similar proportions of patients scored within 10 points of baseline for most PRO domains, with the exception of pain and insomnia for the DCF + RT group. Non-responders randomised to DCF or DCF + RT experienced additional short-term burden compared to CF responders, reflecting the longer duration of neoadjuvant treatment and additional toxicity. This should be weighed against clinical benefits reported in AGITG DOCTOR. This data will inform communication of the trajectory of treatment options for early CF non-responders. Trial registration Australia New Zealand Clinical Trials Registry (ANZCTR), ACTRN12609000665235. Registered 31 July 2009. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09270-4.
Collapse
Affiliation(s)
- R Mercieca-Bebber
- National Health and Medical Research Council (NHMRC) Clinical Trials Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - E H Barnes
- National Health and Medical Research Council (NHMRC) Clinical Trials Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - K Wilson
- National Health and Medical Research Council (NHMRC) Clinical Trials Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Z Samoon
- National Health and Medical Research Council (NHMRC) Clinical Trials Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - E Walpole
- Division of Cancer Services, Princess Alexandra Hospital, Woolloongabba, Qld, Australia.,School of Clinical Medicine, University of Queensland, Brisbane, Qld, Australia
| | - T Mai
- Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia
| | - S Ackland
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| | - M Burge
- Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia.,Cancer Care Services, Royal Brisbane and Women's Hospital, Brisbane, Qld, Australia
| | - G Dickie
- Cancer Care Services, Royal Brisbane and Women's Hospital, Brisbane, Qld, Australia
| | - D Watson
- Discipline of Surgery, College of Medicine and Public Health, Flinders University, Adelaide, South Australia
| | - J Leung
- GenesisCare St Andrew's Hospital, 352 South Terrace, Adelaide, SA, Australia
| | - T Wang
- Crown Princess Mary Cancer Center, Westmead hospital; Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - R Bohmer
- Hobart Private Hospital, Ground Floor- Suite 6 Corner Argyle & Collins Streets, Hobart, Tasmania, Australia
| | - D Cameron
- Townsville University Hospital, Townsville, Qld, Australia
| | - J Simes
- National Health and Medical Research Council (NHMRC) Clinical Trials Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - V Gebski
- National Health and Medical Research Council (NHMRC) Clinical Trials Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - M Smithers
- Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia.,Divisions of Surgery and Cancer Services, Princess Alexandra Hospital, Woolloongabba, Australia
| | - J Thomas
- GIAST Clinic Mater Medical Centre South Brisbane, Brisbane, Australia
| | - J Zalcberg
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - A P Barbour
- Division of Cancer Services, Princess Alexandra Hospital, Woolloongabba, Qld, Australia. .,Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia.
| |
Collapse
|
23
|
ctDNA as a biomarker of progression in oesophageal adenocarcinoma. ESMO Open 2022; 7:100452. [PMID: 35798469 PMCID: PMC9271467 DOI: 10.1016/j.esmoop.2022.100452] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/09/2022] [Accepted: 02/20/2022] [Indexed: 12/24/2022] Open
Abstract
Background The incidence of oesophageal adenocarcinoma (OAC) is rapidly increasing and despite improvements in treatment, the 5-year survival rate remains poor. Prognostic biomarkers that address the genomic heterogeneity in this highly complex disease will aid the development of precision therapeutics and improve patient survival. The aim of this study was to determine whether circulating tumour DNA (ctDNA) has prognostic significance as a biomarker in OAC patients. Patients and methods We profiled 209 blood and tumour samples from 57 OAC patients. Using a panel of 77 cancer genes, we sequenced ctDNA in plasma samples (n = 127) which were taken at multiple time points before and after therapy. In parallel, we sequenced matched tumour samples from 39 patients using the same gene panel. To assess whether the ctDNA profile reflected the tumour heterogeneity, we sequenced additional multi-region primary tumour samples in 17 patients. In addition, we analysed whole-genome and whole-exome sequencing data from primary tumours for a subset of 18 patients. Results Using a tumour-agnostic approach, we found that detectable ctDNA variants in post-treatment plasma samples were associated with worse disease-specific survival. To evaluate whether the ctDNA originated from the primary tumour, we carried out a tumour-informed analysis which confirmed post-treatment ctDNA variants were associated with worse survival. To determine whether ctDNA could be used as a clinical follow-up test, we assessed blood samples from multiple time points before and after treatment, in a subset of patients. Results showed that the variant allele frequency of ctDNA variants increased with disease recurrence. Conclusion This study demonstrates that ctDNA variants can be detected in patients with OAC and this has potential clinical utility as a prognostic biomarker for survival. Detection of ctDNA variants was associated with worse disease-specific survival in OAC. In a tumour-informed approach, ctDNA variants were confirmed using multiple biopsies from the primary tumour. ctDNA variants reflected the intratumour heterogeneity associated with OAC. ctDNA can be used as a personalised prognostic biomarker for patients with OAC.
Collapse
|
24
|
Lang CCJ, Lloyd M, Alyacoubi S, Rahman S, Pickering O, Underwood T, Breininger SP. The Use of miRNAs in Predicting Response to Neoadjuvant Therapy in Oesophageal Cancer. Cancers (Basel) 2022; 14:1171. [PMID: 35267476 PMCID: PMC8909542 DOI: 10.3390/cancers14051171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Oesophageal cancer (OC) is the ninth most common cancer worldwide. Patients receive neoadjuvant therapy (NAT) as standard of care, but less than 20% of patients with oesophageal adenocarcinoma (OAC) or a third of oesophageal squamous cell carcinoma (OSCC) patients, obtain a clinically meaningful response. Developing a method of determining a patient's response to NAT before treatment will allow rational treatment decisions to be made, thus improving patient outcome and quality of life. (1) Background: To determine the use and accuracy of microRNAs as biomarkers of response to NAT in patients with OAC or OSCC. (2) Methods: MEDLINE, EMBASE, Web of Science and the Cochrane library were searched to identify studies investigating microRNAs in treatment naïve biopsies to predict response to NAT in OC patients. (3) Results: A panel of 20 microRNAs were identified as predictors of good or poor response to NAT, from 15 studies. Specifically, miR-99b, miR-451 and miR-505 showed the strongest ability to predict response in OAC patients along with miR-193b in OSCC patients. (4) Conclusions: MicroRNAs are valuable biomarkers of response to NAT in OC. Research is needed to understand the effects different types of chemotherapy and chemoradiotherapy have on the predictive value of microRNAs; studies also require greater standardization in how response is defined.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Stella P. Breininger
- Cancer Research UK Center, Faculty of Medicine, School of Cancer Science, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK; (C.C.J.L.); (M.L.); (S.A.); (S.R.); (O.P.); (T.U.)
| |
Collapse
|
25
|
Obermannova R, Selingerova I, Rehak Z, Jedlicka V, Slavik M, Fabian P, Novotny I, Zemanova M, Studentova H, Grell P, Zdrazilova Dubska L, Demlova R, Harustiak T, Hejnova R, Kiss I, Vyzula R. PET/CT-tailored treatment of locally advanced oesophago-gastric junction adenocarcinoma: a report on the feasibility of the multicenter GastroPET study. Ther Adv Med Oncol 2022; 13:17588359211065153. [PMID: 35035533 PMCID: PMC8753528 DOI: 10.1177/17588359211065153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Perioperative chemotherapy is a recommended treatment approach for localised oesophago-gastric junction adenocarcinoma, but not all patients respond to neoadjuvant chemotherapy. Early identification of non-responders and treatment adaptation in the preoperative period could improve outcomes. GastroPET is a national, multicentre phase II trial evaluating a 18FDG-PET/CT-guided preoperative treatment strategy with the R0 resection rate as a primary endpoint. Here, we report on the accuracy of the methodology, the feasibility of the study design and patient safety data after enrolment of the first 63 patients. METHODS Patients with locally advanced oesophago-gastric junction adenocarcinoma (Siewert I - III) stage Ib-IIIc underwent baseline 18FDG-PET/CT scanning and re-evaluation after 14 days of oxaliplatinum-5FU-(docetaxel) chemotherapy. Responders were defined by a ⩾ 35% decrease in tumour FDG standardised uptake value (SUV)average from baseline. Responders continued with the same chemotherapy for 2 to 3 months prior to surgery. PET-non-responders switched to preoperative chemoradiotherapy [weekly carboplatin and paclitaxel with concurrent radiotherapy (45 Gy in 25 fractions)]. Here, we aim to confirm the feasibility of FDG-PET-based response assessment in a multicenter setting and to compare local versus central reading. In addition, we report on the feasibility of the study conduct and patient safety data. RESULTS A total of 64 patients received baseline and sequential 14-day 18FDG-PET/CT scanning. And, 63 were allocated to the respective treatment arm according to PET-response [35 (56%) responders and 28 (44%) non-responders]. The concordance of local versus central reading of SUV changes was 100%. Until the date of this analysis, 47 patients (28 responders and 19 non-responders) completed surgery. Postoperative complications of grade ⩾ 3 (Common Terminology Criteria for Adverse Events, CTCAE Version 5.0) were reported in five responders (18%; 95% CI: 7.9-36%) and two non-responders (11%; 95% CI: 2.9-31%), with no statistical difference (p = 0.685). One patient in each arm died after surgery, leading to a postoperative in-hospital mortality rate of 4.3% (2/47 patients; 95% CI: 1.2-14%). CONCLUSION Local and central FDG-SUV quantification and PET-response assessment showed high concordance. This confirms the accuracy of a PET-response-guided treatment algorithm for locally advanced oesophago-gastric junction cancer in a multicenter setting. Preoperative treatment adaptation revealed feasible and safe for patients.
Collapse
Affiliation(s)
- Radka Obermannova
- Department of Comprehensive Cancer Care,
Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53 Brno, Czech
Republic
- Department of Comprehensive Cancer Care,
Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Iveta Selingerova
- Research Centre for Applied Molecular Oncology,
Masaryk Memorial Cancer Institute, Brno, Czech Republic
- Department of Pharmacology, Faculty of
Medicine, Masaryk University, Brno, Czech Republic
| | - Zdenek Rehak
- Department of Nuclear Medicine, Masaryk
Memorial Cancer Institute, Brno, Czech Republic
| | - Vaclav Jedlicka
- Department of Surgery, Masaryk Memorial Cancer
Institute, Brno, Czech Republic
- Department of Surgery, Faculty of Medicine,
Masaryk University, Brno, Czech Republic
| | - Marek Slavik
- Department of Radiation Oncology, Masaryk
Memorial Cancer Institute, Brno, Czech Republic
| | - Pavel Fabian
- Department of Pathology, Masaryk Memorial
Cancer Institute, Brno, Czech Republic
| | - Ivo Novotny
- Department of Gastroenterology, Masaryk
Memorial Cancer Institute, Brno, Czech Republic
| | - Milada Zemanova
- Department of Oncology, First Faculty of
Medicine, Charles University and General University Hospital in Prague,
Prague, Czech Republic
| | - Hana Studentova
- Department of Oncology, University Hospital
Olomouc, Olomouc, Czech Republic
| | - Peter Grell
- Department of Comprehensive Cancer Care,
Masaryk Memorial Cancer Institute, Brno, Czech Republic
- Department of Comprehensive Cancer Care,
Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lenka Zdrazilova Dubska
- Department of Laboratory Medicine – Clinical
Microbiology and Immunology, University Hospital Brno, Brno, Czech
Republic
| | - Regina Demlova
- Department of Pharmacology, Faculty of
Medicine, Masaryk University, Brno, Czech Republic
| | - Tomas Harustiak
- Third Department of Surgery, First Faculty of
Medicine, Charles University, Prague, Czech Republic
| | - Renata Hejnova
- Faculty of Medicine, Masaryk University, Brno,
Czech Republic
| | - Igor Kiss
- Department of Comprehensive Cancer Care,
Masaryk Memorial Cancer Institute, Brno, Czech Republic
- Department of Comprehensive Cancer Care,
Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Rostislav Vyzula
- Department of Comprehensive Cancer Care,
Masaryk Memorial Cancer Institute, Brno, Czech Republic
- Department of Comprehensive Cancer Care,
Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
26
|
Hou S, Pan Z, Hao X, Hang Q, Ding Y. Recent Progress in the Neoadjuvant Treatment Strategy for Locally Advanced Esophageal Cancer. Cancers (Basel) 2021; 13:5162. [PMID: 34680311 PMCID: PMC8533976 DOI: 10.3390/cancers13205162] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 01/03/2023] Open
Abstract
Neoadjuvant therapies, primarily chemotherapy and chemoradiotherapy, are able to improve the overall survival (OS) in patients with locally advanced resectable esophageal cancer (EC) based on the results of several randomized clinical trials. The advantage of neoadjuvant therapy is chiefly attributed to the decreased risk of local-regional recurrence and distant metastasis. Thus, it has been recommended as standard treatment for patients with resectable EC. However, several fundamental problems remain. First, the combination of neoadjuvant chemotherapy (nCT), neoadjuvant chemoradiotherapy (nCRT), and surgery for EC patients with different histological types remain controversial. Furthermore, to reduce the toxicity of preoperative chemotherapy and the risk of complications caused by preoperative radiation therapy, the treatment protocols of nCT and nCRT still need to be investigated and optimized by prospective trials. Moreover, for patients with complete clinical response following neoadjuvant therapy, it is worth ascertaining whether a "watch and wait" surveillance plus surgery-as-needed policy is more favorable, as well as, in addition to preoperative chemoradiotherapy, whether immunotherapy, especially when combined with the traditional neoadjuvant therapy regimens, brings new prospects for EC treatment. In this review, we summarize the recent insights into the research progress and existing problems of neoadjuvant therapy for locally advanced resectable EC.
Collapse
Affiliation(s)
- Sicong Hou
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225000, China;
| | - Ziyin Pan
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.P.); (X.H.)
| | - Xin Hao
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Z.P.); (X.H.)
| | - Qinglei Hang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yanbing Ding
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225000, China;
| |
Collapse
|
27
|
Liao XL, Liang XW, Pang HY, Yang K, Chen XZ, Chen XL, Liu K, Zhao LY, Zhang WH, Hu JK. Safety and Efficacy of Laparoscopic Versus Open Gastrectomy in Patients With Advanced Gastric Cancer Following Neoadjuvant Chemotherapy: A Meta-Analysis. Front Oncol 2021; 11:704244. [PMID: 34422658 PMCID: PMC8377369 DOI: 10.3389/fonc.2021.704244] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/19/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Given the expanding clinical applications of laparoscopic surgery and neoadjuvant chemotherapy in advanced gastric cancer treatment, there is an emerging need to summarize the few evidences that evaluated the safety and efficacy of laparoscopic versus open gastrectomy in patients with advanced gastric cancer (AGC) following neoadjuvant chemotherapy (NAC). METHODS From January 1 to 2, 2021, we searched Ovid Embase, PubMed, Cochrane central register Trials (Ovid), and web of science to find relevant studies published in English, and two authors independently performed literature screening, quality assessment of the included studies, data extraction, and data analysis. This study was registered with PROSPERO (CRD42021228845). RESULTS The initial search retrieved 1567 articles, and 6 studies were finally included in the meta-analysis review, which comprised 2 randomized control trials and 4 observational studies involving 288 laparoscopic gastrectomy (LG) and 416 open gastrectomy (OG) AGC patients treated with NAC. For intraoperative conditions, R0 resection rate, blood transfusion, intraoperative blood loss, number of lymph nodes dissected, proximal margin, and distal margin were comparable between LG group and open OG group. For postoperative short-term clinical outcomes, LG has significantly less postoperative complications (OR = 0.65, 95%CI: 0.42-1.00, p = 0.05) and shorter postoperative time to first aerofluxus (WMD = -0.57d, 95%CI: -0.89-0.25, p = 0.0004) than OG, and anastomotic leakage, pulmonary infection, pleural effusion, surgical site infection, thrombosis, intestinal obstruction, peritoneal effusion or abscess formation, postoperative time to first defecation, postoperative time to first liquid diet, and postoperative length of stay were comparable between the two groups. For postoperative survival outcomes, there were no significant differences in disease-free survival (DFS) and overall survival (OS) between the two groups. CONCLUSION The available evidences indicated that LG is an effective and feasible technology for the treatment of AGC patients treated with NAC, and LG patients have much less postoperative complications and faster bowel function recovery than OG patients. SYSTEMATIC REVIEW REGISTRATION PROSPERO database (identifier, CRD42021228845).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jian-Kun Hu
- Department of Gastrointestinal Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
28
|
Lapa C, Nestle U, Albert NL, Baues C, Beer A, Buck A, Budach V, Bütof R, Combs SE, Derlin T, Eiber M, Fendler WP, Furth C, Gani C, Gkika E, Grosu AL, Henkenberens C, Ilhan H, Löck S, Marnitz-Schulze S, Miederer M, Mix M, Nicolay NH, Niyazi M, Pöttgen C, Rödel CM, Schatka I, Schwarzenboeck SM, Todica AS, Weber W, Wegen S, Wiegel T, Zamboglou C, Zips D, Zöphel K, Zschaeck S, Thorwarth D, Troost EGC. Value of PET imaging for radiation therapy. Strahlenther Onkol 2021; 197:1-23. [PMID: 34259912 DOI: 10.1007/s00066-021-01812-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022]
Abstract
This comprehensive review written by experts in their field gives an overview on the current status of incorporating positron emission tomography (PET) into radiation treatment planning. Moreover, it highlights ongoing studies for treatment individualisation and per-treatment tumour response monitoring for various primary tumours. Novel tracers and image analysis methods are discussed. The authors believe this contribution to be of crucial value for experts in the field as well as for policy makers deciding on the reimbursement of this powerful imaging modality.
Collapse
Affiliation(s)
- Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Ursula Nestle
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- Department of Radiation Oncology, Kliniken Maria Hilf, Mönchengladbach, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christian Baues
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Ambros Beer
- Department of Nuclear Medicine, Ulm University Hospital, Ulm, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Volker Budach
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Rebecca Bütof
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Stephanie E Combs
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
- Department of Radiation Sciences (DRS), Institute of Radiation Medicine (IRM), Neuherberg, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Cihan Gani
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Eleni Gkika
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Anca-L Grosu
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Christoph Henkenberens
- Department of Radiotherapy and Special Oncology, Medical School Hannover, Hannover, Germany
| | - Harun Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Löck
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Simone Marnitz-Schulze
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Matthias Miederer
- Department of Nuclear Medicine, University Hospital Mainz, Mainz, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Maximilian Niyazi
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Christoph Pöttgen
- Department of Radiation Oncology, West German Cancer Centre, University of Duisburg-Essen, Essen, Germany
| | - Claus M Rödel
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Imke Schatka
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | | | - Andrei S Todica
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang Weber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Simone Wegen
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Thomas Wiegel
- Department of Radiation Oncology, Ulm University Hospital, Ulm, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Daniel Zips
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Klaus Zöphel
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Nuclear Medicine, Klinikum Chemnitz gGmbH, Chemnitz, Germany
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Esther G C Troost
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany.
| |
Collapse
|
29
|
Yoshida N, Taniyama Y, Murakami K, Horinouchi T, Takahashi K, Shiraishi S, Eto K, Kamei T, Matsubara H, Baba H. Novel Criterion Using Esophageal Major and Minor Axes is Useful to Evaluate the Therapeutic Effect and Prognosis After Neoadjuvant Chemotherapy Followed by Surgery in Locally Advanced Esophageal Cancer. Ann Surg Oncol 2021; 28:8474-8482. [PMID: 34260005 DOI: 10.1245/s10434-021-10314-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/29/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND An appropriate strategy is needed to determine the therapeutic effect of chemotherapy on primary lesions in esophageal cancer. This multicenter cohort study aimed to examine the usefulness of a novel criterion obtained by multiplying the lengths of the major and minor esophageal axes from helical computed tomography as a tool to evaluate the therapeutic effect of neoadjuvant chemotherapy and to predict prognosis after surgery in locally advanced esophageal cancer. MATERIALS AND METHODS A first investigation evaluated the reproducibility of the new criterion between two independent examiners. In a second investigation, we examined the association of the novel criterion with pathological tumor regression grade and long-term outcomes. Pretreatment primary lesions less than 20 mm on computed tomography were excluded. RESULTS In an initial cohort of 81 patients, the intraclass correlation coefficient for the novel criterion was higher than that for the tumor major axis both before and after neoadjuvant chemotherapy. In the second cohort of 255 patients, the novel criterion significantly correlated with tumor regression grade (p = 0.0003), overall survival (p < 0.0001), and disease-free survival (p < 0.0001). It was also an independent predictor for overall survival (p = 0.0023), along with age, tumor regression grade, and pathological stage. CONCLUSIONS The measurement derived by multiplying the esophageal major and minor axes on computed tomography is easy to obtain and has better objectivity and reproducibility for tumors of any shape. This novel criterion may be clinically useful because it can estimate therapeutic effect, tumor regression grade, and prognosis after neoadjuvant chemotherapy followed by surgery for esophageal cancer.
Collapse
Affiliation(s)
- Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Chuoku, Kumamoto, Japan.,Division of Translational Research and Advanced Treatment against Gastrointestinal Cancer, Kumamoto University, Kumamoto, Japan
| | - Yusuke Taniyama
- Department of Surgery, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Kentaro Murakami
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomo Horinouchi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Chuoku, Kumamoto, Japan
| | - Kozue Takahashi
- Department of Surgery, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Shinya Shiraishi
- Department of Diagnostic Radiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kojiro Eto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Chuoku, Kumamoto, Japan
| | - Takashi Kamei
- Department of Surgery, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Chuoku, Kumamoto, Japan.
| |
Collapse
|
30
|
Lapa C, Nestle U, Albert NL, Baues C, Beer A, Buck A, Budach V, Bütof R, Combs SE, Derlin T, Eiber M, Fendler WP, Furth C, Gani C, Gkika E, Grosu AL, Henkenberens C, Ilhan H, Löck S, Marnitz-Schulze S, Miederer M, Mix M, Nicolay NH, Niyazi M, Pöttgen C, Rödel CM, Schatka I, Schwarzenboeck SM, Todica AS, Weber W, Wegen S, Wiegel T, Zamboglou C, Zips D, Zöphel K, Zschaeck S, Thorwarth D, Troost EGC. Value of PET imaging for radiation therapy. Nuklearmedizin 2021; 60:326-343. [PMID: 34261141 DOI: 10.1055/a-1525-7029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This comprehensive review written by experts in their field gives an overview on the current status of incorporating positron emission tomography (PET) into radiation treatment planning. Moreover, it highlights ongoing studies for treatment individualisation and per-treatment tumour response monitoring for various primary tumours. Novel tracers and image analysis methods are discussed. The authors believe this contribution to be of crucial value for experts in the field as well as for policy makers deciding on the reimbursement of this powerful imaging modality.
Collapse
Affiliation(s)
- Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Ursula Nestle
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,Department of Radiation Oncology, Kliniken Maria Hilf, Mönchengladbach, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christian Baues
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Ambros Beer
- Department of Nuclear Medicine, Ulm University Hospital, Ulm, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Volker Budach
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Rebecca Bütof
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Stephanie E Combs
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany.,Department of Radiation Sciences (DRS), Institute of Radiation Medicine (IRM), Neuherberg, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Cihan Gani
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Eleni Gkika
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Anca L Grosu
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | | | - Harun Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Löck
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Simone Marnitz-Schulze
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Matthias Miederer
- Department of Nuclear Medicine, University Hospital Mainz, Mainz, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Maximilian Niyazi
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Christoph Pöttgen
- Department of Radiation Oncology, West German Cancer Centre, University of Duisburg-Essen, Essen, Germany
| | - Claus M Rödel
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiotherapy and Oncology, Goethe University Frankfurt, Frankfurt, Germany
| | - Imke Schatka
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | | | - Andrei S Todica
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang Weber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Simone Wegen
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Thomas Wiegel
- Department of Radiation Oncology, Ulm University Hospital, Ulm, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Daniel Zips
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Klaus Zöphel
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Nuclear Medicine, Klinikum Chemnitz gGmbH, Chemnitz, Germany
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Esther G C Troost
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | | |
Collapse
|
31
|
Foley KG, Jeffries J, Hannon C, Coles B, Bradley KM, Smyth E. Response rate and diagnostic accuracy of early PET-CT during neo-adjuvant therapies in oesophageal adenocarcinoma: A systematic review and meta-analysis. Int J Clin Pract 2021; 75:e13906. [PMID: 33300222 DOI: 10.1111/ijcp.13906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/03/2020] [Indexed: 11/28/2022] Open
Abstract
PURPOSE Only 25% of oesophageal adenocarcinoma (OAC) patients have a pathological response to neo-adjuvant therapy (NAT) before oesophagectomy. Early response assessment using PET imaging may help guide management of these patients. We performed a systematic review and meta-analysis to synthesise the evidence detailing response rate and diagnostic accuracy of early PET-CT assessment. METHODS We systematically searched several databases including MEDLINE and Embase. Studies with mixed cohorts of histology, tumour location and a repeat PET-CT assessment after more than one cycle of NAT were excluded. Reference standard was pathological response defined by Becker or Mandard classifications. Primary outcome was metabolic response rate after one cycle of NAT defined by a reduction in maximum standardised uptake value (SUVmax) of 35%. Secondary outcome was diagnostic accuracy of treatment response prediction, defined as the sensitivity and specificity of early PET-CT using this threshold. Quality of evidence was also assessed. Random-effects meta-analysis pooled response rates and diagnostic accuracy. This study was registered with PROSPERO (CRD42019147034). RESULTS Overall, 1341 articles were screened, and 6 studies were eligible for analysis. These studies reported data for 518 patients (aged 27-78 years; 452 [87.3%] were men) between 2005 and 2020. Pooled sensitivity of early metabolic response to predict pathological response was 77.2% (95% CI 53.2%-100%). Significant heterogeneity existed between studies (I2 = 80.6% (95% CI 38.9%-93.8%), P = .006). Pooled specificity was 75.0% (95% CI 68.2%-82.5%), however, no significant heterogeneity between studies existed (I2 = 0.0% (95% CI 0.0%-67.4%), P = .73). CONCLUSION High-quality evidence is lacking, and few studies met the inclusion criteria of this systematic review. The sensitivity of PET using a SUVmax reduction threshold of 35% was suboptimal and varied widely. However, specificity was consistent across studies with a pooled value of 75.0%, suggesting early PET assessment is a better predictor of treatment resistance than of pathological response. Further research is required to define optimal PET-guided treatment decisions in OAC.
Collapse
Affiliation(s)
- Kieran G Foley
- Royal Glamorgan Hospital & Velindre Cancer Centre, Cardiff, UK
| | | | - Clare Hannon
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Kevin M Bradley
- Wales Diagnostic and Research Positron Emission Tomography Imaging Centre, Cardiff University, Cardiff, UK
| | - Elizabeth Smyth
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
32
|
Yoon HH, Ou FS, Soori GS, Shi Q, Wigle DA, Sticca RP, Miller RC, Leenstra JL, Peller PJ, Ginos B, Heying E, Wu TT, Drevyanko TF, Ko S, Mattar BI, Nikcevich DA, Behrens RJ, Khalil MF, Kim GP, Alberts SR. Induction versus no induction chemotherapy before neoadjuvant chemoradiotherapy and surgery in oesophageal adenocarcinoma: a multicentre randomised phase II trial (NCCTG N0849 [Alliance]). Eur J Cancer 2021; 150:214-223. [PMID: 33934058 PMCID: PMC8154661 DOI: 10.1016/j.ejca.2021.03.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/05/2021] [Accepted: 03/14/2021] [Indexed: 12/21/2022]
Abstract
AIM report primary results from the first multicentre randomised trial evaluating induction chemotherapy prior to trimodality therapy in patients with oesophageal or gastro-oesophageal junction adenocarcinoma. Notably, recent data from a single-institution randomised trial reported that induction chemotherapy prolonged overall survival (OS) in patients with well/moderately differentiated tumours. METHODS In this phase 2 trial (28 centres in the U.S. NCI-sponsored North Central Cancer Treatment Group [Alliance]), trimodality-eligible patients (T3-4N0, TanyN+) were randomised to receive induction (docetaxel, oxaliplatin, capecitabine; Arm A) or no induction chemotherapy (Arm B) followed by oxaliplatin/5-fluorouracil/radiation and subsequent surgery. The primary endpoint was the rate of pathologic complete response (pathCR). Secondary/exploratory endpoints were OS and disease-free survival (DFS). RESULTS Of 55 patients evaluable for the primary endpoint, the pathCR rate was 28.6% (8/28) in A versus 40.7% (11/27) in B (P = .34). Given interim results indicating futility, accrual was terminated, but patients were followed. After a median follow-up of 60.4 months, a longer median OS in Arm A versus B was unexpectedly observed (3-year rates 57.1% versus 41.7%, respectively) driven by longer DFS after margin-free surgery. In posthoc analysis, induction (versus no induction) chemotherapy was associated with significantly longer OS and DFS among patients with well/moderately differentiated tumours, but not among patients with poorly/undifferentiated tumours (Pinteraction = 0.037). CONCLUSIONS Adding induction chemotherapy prior to trimodality therapy did not improve the primary endpoint, pathCR. However, induction chemotherapy was associated with longer median OS, particularly among patients with well/moderately differentiated tumours. These findings may inform further development of curative-intent trials in this disease.
Collapse
Affiliation(s)
| | - Fang-Shu Ou
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA.
| | | | - Qian Shi
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA.
| | | | | | | | | | | | - Brenda Ginos
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA.
| | - Erica Heying
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA.
| | | | | | | | | | | | | | - Maged F Khalil
- Lehigh Valley Health Network, Allentown, Michigan Cancer Research Consortium, PA, USA.
| | - George P Kim
- 21(st) Century Oncology of Jacksonville, Jacksonville, FL, USA.
| | | |
Collapse
|
33
|
Sugawara K, Kawaguchi Y, Seto Y, Vauthey JN. Multidisciplinary treatment strategy for locally advanced gastric cancer: A systematic review. Surg Oncol 2021; 38:101599. [PMID: 33991939 DOI: 10.1016/j.suronc.2021.101599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/26/2021] [Indexed: 01/17/2023]
Abstract
BACKGROUND Multidisciplinary management of patients with locally advanced gastric cancer (LAGC) remains unstandardized worldwide. We performed a systemic review to summarize the advancements, regional differences, and current recommended multidisciplinary treatment strategies for LAGC. METHODS Eligible studies were identified through a comprehensive search of PubMed, Web of Science, Cochrane Library databases and Embase. Phase 3 randomized controlled trials which investigated survival of patients with LAGC who underwent gastrectomy with pre-/perioperative, postoperative chemotherapy, or chemoradiotherapy were included. RESULTS In total, we identified 11 studies of pre-/perioperative chemotherapy, 38 of postoperative chemotherapy, and 14 of chemoradiotherapy. In Europe and the USA, the current standard of care is perioperative chemotherapy for patients with LAGC using the regimen of 5-FU, folinic acid, oxaliplatin and docetaxel (FLOT). In Eastern Asia, upfront gastrectomy and postoperative chemotherapy is commonly used. The S-1 monotherapy or a regimen of capecitabine and oxaliplatin (CapOx) are used for patients with stage II disease, and the CapOx regimen or the S-1 plus docetaxel regimen are recommended for those with stage III Gastric cancer (GC). The addition of postoperative radiotherapy to peri- or postoperative chemotherapy is currently not recommended. Additionally, clinical trials testing targeted therapy and immunotherapy are increasingly performed worldwide. CONCLUSIONS Recent clinical trials showed a survival benefit of peri-over postoperative chemotherapy and chemoradiotherapy. As such, this strategy may have a potential as a global standard for patients with LAGC. Outcome of the ongoing clinical trials is expected to establish the global standard of multidisciplinary treatment strategy in patients with LAGC.
Collapse
Affiliation(s)
- Kotaro Sugawara
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshikuni Kawaguchi
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jean-Nicolas Vauthey
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
34
|
Lau DK, Athauda A, Chau I. Neoadjuvant and adjuvant multimodality therapies in resectable esophagogastric adenocarcinoma. Expert Opin Pharmacother 2021; 22:1429-1441. [PMID: 33688789 DOI: 10.1080/14656566.2021.1900823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Gastric and esophageal adenocarcinoma is a leading cause of cancer-related death globally. Surgery is the cornerstone modality for cure where feasible. Clinical studies over the past two decades have provided evidence for the use of perioperative chemotherapy and chemoradiotherapy to improve patient outcomes. However, there remains no global consensus in the optimal use of these therapies.Areas covered: In this review, the authors summarize the latest evidence for perioperative multimodality therapy in resectable esophagogastric adenocarcinoma including the use of combination chemotherapy and targeted therapy containing regimens. In addition, the authors discuss some of the clinical and molecular biomarkers, such as PET imaging and microsatellite instability which can inform future practice and further clinical investigation.Expert opinion: A multimodal approach has been proven to improve survival outcomes over surgery alone. Whilst there is no global standard of care for multi-modality therapies in resectable OG cancer, clinical trials are refining the use of chemotherapy and radiotherapy in the neoadjuvant and adjuvant settings. Further investigation is on-going to further optimize therapy and the integration of molecular targeted agents.
Collapse
Affiliation(s)
- David K Lau
- GI and Lymphoma Unit, Royal Marsden NHS Foundation Trust, Sutton and London, UK
| | - Avani Athauda
- GI and Lymphoma Unit, Royal Marsden NHS Foundation Trust, Sutton and London, UK
| | - Ian Chau
- GI and Lymphoma Unit, Royal Marsden NHS Foundation Trust, Sutton and London, UK
| |
Collapse
|
35
|
Veenstra MMK, Smithers BM, Visser E, Edholm D, Brosda S, Thomas JM, Gotley DC, Thomson IG, Wijnhoven BPL, Barbour AP. Complications and survival after hybrid and fully minimally invasive oesophagectomy. BJS Open 2021; 5:6133613. [PMID: 33609389 PMCID: PMC7893474 DOI: 10.1093/bjsopen/zraa033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/29/2020] [Indexed: 12/31/2022] Open
Abstract
Background Minimally invasive oesophagectomy (MIO) is reported to produce fewer respiratory complications than open oesophagectomy. This study assessed differences in postoperative complications between MIO and hybrid MIO (HMIO) employing thoracoscopy and laparotomy, along with the influence of co-morbidities on postoperative outcomes. Methods Patients with oesophageal cancer undergoing three-stage MIO or three-stage HMIO between 1999 and 2018 were identified from a prospectively developed database, which included patient demographics, co-morbidities, preoperative therapies, and cancer stage. The primary outcome was postoperative complications in the two groups. Secondary outcomes included duration of operation, blood transfusion requirement, duration of hospital stay, and overall survival. Results There were 828 patients, of whom 722 had HMIO and 106 MIO, without significant baseline differences. Median duration of operation was longer for MIO (325 versus 289 min; P < 0.001), but with less blood loss (median 250 versus 300 ml; P < 0.001) and a shorter hospital stay (median 12 versus 13 days; P = 0.006). Respiratory complications were not associated with operative approach (31.1 versus 35.2 per cent for MIO and HMIO respectively; P = 0.426). Anastomotic leak rates (10.4 versus 10.2 per cent) and 90-day mortality (1.0 versus 1.7 per cent) did not differ. Cardiac co-morbidity was associated with more medical and surgical complications. Overall survival was associated with AJCC stage and co-morbidities, but not operative approach. Conclusion MIO had a small benefit in terms of blood loss and hospital stay, but not in operating time. Oncological outcomes were similar in the two groups. Postoperative complications were associated with pre-existing cardiorespiratory co-morbidities rather than operative approach.
Collapse
Affiliation(s)
- M M K Veenstra
- Academy of Surgery, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - B M Smithers
- Academy of Surgery, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Upper Gastrointestinal/Soft Tissue Unit, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Mater Research Institute, Mater Health Services, Brisbane, Queensland, Australia
| | - E Visser
- Upper Gastrointestinal/Soft Tissue Unit, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Department of Surgery, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - D Edholm
- Upper Gastrointestinal/Soft Tissue Unit, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - S Brosda
- Diamantina Institute, Translational Research Institute, The University of Queensland, Queensland, Australia
| | - J M Thomas
- Upper Gastrointestinal/Soft Tissue Unit, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Mater Research Institute, Mater Health Services, Brisbane, Queensland, Australia
| | - D C Gotley
- Academy of Surgery, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Upper Gastrointestinal/Soft Tissue Unit, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - I G Thomson
- Academy of Surgery, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Upper Gastrointestinal/Soft Tissue Unit, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - B P L Wijnhoven
- Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, the Netherlands
| | - A P Barbour
- Academy of Surgery, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Upper Gastrointestinal/Soft Tissue Unit, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Diamantina Institute, Translational Research Institute, The University of Queensland, Queensland, Australia
| |
Collapse
|
36
|
Foley KG, Pearson B, Riddell Z, Taylor SA. Opportunities in cancer imaging: a review of oesophageal, gastric and colorectal malignancies. Clin Radiol 2021; 76:748-762. [PMID: 33579518 DOI: 10.1016/j.crad.2021.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/13/2021] [Indexed: 02/07/2023]
Abstract
The incidence of gastrointestinal (GI) malignancy is increasing worldwide. In particular, there is a concerning rise in incidence of GI cancer in younger adults. Direct endoscopic visualisation of luminal tumour sites requires invasive procedures, which are associated with certain risks, but remain necessary because of limitations in current imaging techniques and the continuing need to obtain tissue for diagnosis and genetic analysis; however, management of GI cancer is increasingly reliant on non-invasive, radiological imaging to diagnose, stage, and treat these malignancies. Oesophageal, gastric, and colorectal malignancies require specialist investigation and treatment due to the complex nature of the anatomy, biology, and subsequent treatment strategies. As cancer imaging techniques develop, many opportunities to improve tumour detection, diagnostic accuracy and treatment monitoring present themselves. This review article aims to report current imaging practice, advances in various radiological modalities in relation to GI luminal tumour sites and describes opportunities for GI radiologists to improve patient outcomes.
Collapse
Affiliation(s)
- K G Foley
- Department of Clinical Radiology, Royal Glamorgan Hospital, Llantrisant, UK.
| | - B Pearson
- National Imaging Academy Wales (NIAW), Pencoed, UK
| | - Z Riddell
- National Imaging Academy Wales (NIAW), Pencoed, UK
| | - S A Taylor
- Centre for Medical Imaging, UCL, London, UK
| |
Collapse
|
37
|
Petrillo A, Smyth EC. Biomarkers for Precision Treatment in Gastric Cancer. Visc Med 2020; 36:364-372. [PMID: 33178733 PMCID: PMC7590759 DOI: 10.1159/000510489] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/27/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the most lethal cancers worldwide. Although GC was historically considered a single entity within the organ of origin, nowadays it is acknowledged that GC represents a heterogeneous disease. Nevertheless, in this field there is still a lack of biomarkers able to guide the choice of the best treatment options for each patient. This review aims to summarize the prognostic and predictive biomarkers evaluated in GC and their role as a guide for treatment for precision medicine. SUMMARY Human epidermal growth factor receptor 2 overexpression represents the only predictive molecular biomarker validated in GC, while its prognostic role is still controversial. Microsatellite instability and Epstein-Barr virus status are promising for prediction of the response to immunotherapy. The role of other biomarkers (ctDNA, programmed death ligand 1 [PD-L1], and TMB), as well as the practical application of molecular classifications, requires further evaluation before use in clinical practice. 18-FDG-PET scan could be useful as a predictive tool in non-metastatic GC patients receiving a perioperative approach. Finally, the tumor microenvironment may have an evolving role in the future. KEY MESSAGES GC is a heterogeneous disease and targeted approaches are needed. The finding of prognostic and predictive factors is a hot topic in the field of GC personalized medicine.
Collapse
Affiliation(s)
- Angelica Petrillo
- Medical Oncology Unit, Ospedale del Mare, Naples, Italy
- University of Study of Campania L. Vanvitelli, Naples, Italy
| | - Elizabeth C. Smyth
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
38
|
Braun L, Reinert C, Zips D, Nikolaou K, Pfannenberg C, Gani C. Treatment outcome after radiochemotherapy in anal cancer patients staged with 18F-FDG-PET-CT. Clin Transl Radiat Oncol 2020; 24:83-87. [PMID: 32642564 PMCID: PMC7334798 DOI: 10.1016/j.ctro.2020.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Anal cancer (AC) is a malignancy with increasing incidence and commonly treated with radiochemotherapy. Positron-emission tomography-computed tomography (PET/CT) has been shown to improve treatment outcome in various oncological diseases, however, for AC long-term outcome data is sparse. The aim of the present study is therefore to report outcomes in our cohort of PET/CT staged AC patients treated with radiochemotherapy. METHODS Patients with AC who were treated with radiochemotherapy in curative intent were included in this retrospective study if a PET/CT scan was performed pre-therapeutically. Information from PET/CT was considered for nodal and primary target volume definition. Radiotherapy dose to the primary tumor was 50-66 Gy and concomitant chemotherapy included 5-fluorouracil and mitomycin-C. The uptake of 18F-fluorodeoxyglucose (FDG) was quantified using 50%-isocontour volumes of interests (VOIs) and measuring the standardized uptake value (SUV) and the metabolic tumor volume (MTV).18F-FDG uptake was correlated with baseline clinical parameters and long-term oncological outcome. Survival estimates were determined according to Kaplan-Meier. RESULTS A total of 60 patients were included in this study. Estimates for three-year overall survival (OS) and disease free survival (DFS) were 94.5% and 80%. Five patients developed local (n = 2) or locoregional and local (n = 3) failure. Baseline PET/CT related parameters correlated with primary tumor stage, nodal stage and tumor grading. DFS was independent of T-stage, N-stage and baseline 18F-FDG-uptake. CONCLUSION In this cohort of PET/CT staged AC patients, excellent outcomes for DFS were seen. PET-based markers of tumor burden correlate with local stage of AC, however, are not of prognostic relevance for disease-free survival.
Collapse
Affiliation(s)
- L.H. Braun
- Department of Radiation Oncology, University Hospital and Medical Faculty Tübingen, Eberhard Karls University Tübingen, Germany
- Klinik für Strahlentherapie und Palliativmedizin, Marienhospital Stuttgart, Germany
| | - C.P. Reinert
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University Tubingen, Germany
| | - D. Zips
- Department of Radiation Oncology, University Hospital and Medical Faculty Tübingen, Eberhard Karls University Tübingen, Germany
- German Cancer Research Center (DKFZ) Heidelberg and German Consortium for Translational Cancer Research (DKTK), Partner Site Tübingen, Tübingen, Germany
| | - K. Nikolaou
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University Tubingen, Germany
| | - C. Pfannenberg
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University Tubingen, Germany
| | - C. Gani
- Department of Radiation Oncology, University Hospital and Medical Faculty Tübingen, Eberhard Karls University Tübingen, Germany
- German Cancer Research Center (DKFZ) Heidelberg and German Consortium for Translational Cancer Research (DKTK), Partner Site Tübingen, Tübingen, Germany
| |
Collapse
|
39
|
Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet 2020; 396:635-648. [PMID: 32861308 DOI: 10.1016/s0140-6736(20)31288-5] [Citation(s) in RCA: 2633] [Impact Index Per Article: 526.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
Gastric cancer is the fifth most common cancer and the third most common cause of cancer death globally. Risk factors for the condition include Helicobacter pylori infection, age, high salt intake, and diets low in fruit and vegetables. Gastric cancer is diagnosed histologically after endoscopic biopsy and staged using CT, endoscopic ultrasound, PET, and laparoscopy. It is a molecularly and phenotypically highly heterogeneous disease. The main treatment for early gastric cancer is endoscopic resection. Non-early operable gastric cancer is treated with surgery, which should include D2 lymphadenectomy (including lymph node stations in the perigastric mesentery and along the celiac arterial branches). Perioperative or adjuvant chemotherapy improves survival in patients with stage 1B or higher cancers. Advanced gastric cancer is treated with sequential lines of chemotherapy, starting with a platinum and fluoropyrimidine doublet in the first line; median survival is less than 1 year. Targeted therapies licensed to treat gastric cancer include trastuzumab (HER2-positive patients first line), ramucirumab (anti-angiogenic second line), and nivolumab or pembrolizumab (anti-PD-1 third line).
Collapse
Affiliation(s)
- Elizabeth C Smyth
- Department of Oncology, Cambridge University Hospitals National Health Service Foundation Trust, Hill's Road, Cambridge, UK.
| | - Magnus Nilsson
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden; Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Heike I Grabsch
- Department of Pathology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, Netherlands; Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Nicole Ct van Grieken
- Department of Pathology, Amsterdam University Medical Centre, Cancer Center Amsterdam, VU University, Amsterdam, Netherlands
| | - Florian Lordick
- University Cancer Center Leipzig, Leipzig University Medical Center, Leipzig, Germany
| |
Collapse
|
40
|
Lordick F, Herrmann K. Metabolic response assessment and PET-guided treatment of esophageal cancer. Ann Oncol 2020; 31:163-164. [DOI: 10.1016/j.annonc.2019.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022] Open
|