1
|
Wang J, Li B, Zhang Y, Luo X, Zhang Y, Li H, Pan Y, Shao L, Zheng S, Yuan C, Li Y, Zheng Q, Sun S, Zhao W, Sun Y. Tislelizumab combined with nab-paclitaxel and cisplatin as the more effective chemoimmunotherapy strategy in the neoadjuvant treatment of locally advanced thoracic esophageal squamous cell carcinoma: A prospective, two-cohort, phase 2 trial. Int J Cancer 2025; 156:1429-1438. [PMID: 39686540 DOI: 10.1002/ijc.35261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/04/2024] [Accepted: 09/17/2024] [Indexed: 12/18/2024]
Abstract
This prospective, two-cohort phase 2 trial with random allocation was conducted to evaluate the safety and efficacy of neoadjuvant tislelizumab combined with nab-paclitaxel/paclitaxel and cisplatin (TP) in patients with esophageal squamous cell carcinoma (ESCC). Patients were enrolled and randomly assigned to the nab-paclitaxel or paclitaxel cohorts at a 1:1 ratio, and received intravenous tislelizumab (200 mg, day 1) combined with cisplatin (25 mg/m2, days 1-3) and either nab-paclitaxel (125 mg/m2, days 1 and 8) or paclitaxel (150 mg/m2, day 1) in a 21-day cycle for two cycles before surgery. The primary endpoint was the major pathological response (MPR) rate. From March 01, 2022 to April 10, 2023, 46 patients were enrolled (n = 23 in each cohort), with 42 patients receiving the full two-cycle treatments and undergoing surgery (n = 22 in the nab-paclitaxel cohort, n = 20 in the paclitaxel cohort). The MPR rate and the pCR rate in the total cohort were 44.2% (19/42) and 19.0% (8/42), respectively, with 59.1% (13/22) and 31.8% (7/22) in the nab-paclitaxel cohort and 30.0% (6/20) and 5.0% (1/20) in paclitaxel cohorts. The most common treatment-related adverse events (TRAEs) were anemia (89.1%) and alopecia (71.7%), and no significant difference in TRAEs was observed between the two cohorts. Up until March 28, 2024, the median follow-up time was 15.5 months (range of 6.0-24.3 months), and the survival analysis revealed that the patients in the nab-paclitaxel cohort had a higher event-free survival (p = .002). In conclusion, neoadjuvant tislelizumab combined with cisplatin and nab-paclitaxel, rather than cisplatin and paclitaxel, is a more effective neoadjuvant strategy for locally advanced thoracic ESCC.
Collapse
Affiliation(s)
- Jie Wang
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bin Li
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yawei Zhang
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoyang Luo
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yiliang Zhang
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hang Li
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yunjian Pan
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Longlong Shao
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shanbo Zheng
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chongze Yuan
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuan Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qiang Zheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Si Sun
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Weixin Zhao
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yihua Sun
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Yang R, Tang H, Xie Y, Cai D, He Y, Zheng Z, Lin Y, Gao H, Tang W, Yan Y, Tan L, Shi H. [ 18F]FDG PET/CT for predicting neoadjuvant PD-L1 blockade monotherapy treatment response in patients with locally advanced esophageal squamous cell carcinoma: a preliminary study. Eur J Nucl Med Mol Imaging 2025; 52:1422-1435. [PMID: 39743618 DOI: 10.1007/s00259-024-07051-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE To investigate the predictive value of 2-[18F]-fluoro-2-deoxy-D-glucose ([18F]FDG) PET/CT for evaluating primary tumor (PT) and lymph node (LN) responses after neoadjuvant programmed death-ligand 1 (PD-L1) blockade monotherapy in patients with locally advanced esophageal squamous cell carcinoma (LA-ESCC). METHODS In the single-arm phase 1b NATION-1907 trial (NCT04215471), 23 patients with LA-ESCC received two cycles of neoadjuvant PD-L1 blockade Adebrelimab followed by surgery. Among these, 18 patients underwent [18F]FDG PET/CT scans both before immunotherapy and prior to surgery. Standardized uptake value corrected for lean body mass (SUL)-derived parameters, including SULmax and SULpeak, were documented for PTs and LNs. Lesions > 1cm3 were segmented using thresholds of 41% and 50% of SULmax, respectively, following European Association of Nuclear Medicine (EANM) guidelines, with metabolic tumor volume (MTV) and total lesion glycolysis (TLG) calculated. Percentage changes of all metabolic parameters were also recorded. Residual viable tumor ≤ 33% were classified as well-responders, whereas residual viable tumor > 33% were classified as poor-responders based on histological evaluation. RESULTS In the PT analysis, 10 patients were classified as PT well-responders and 8 as PT poor-responders. All post-treatment metabolic parameters, except MTV, were significantly lower in well-responders compared to poor-responders. The %ΔMTV, %ΔTLG were significantly higher in the poor-responder group (all P < 0.05). ROC curves indicated %ΔMTV41 exhibited optimum performance in predicting well-responders, with an AUC of 0.875 (cut-off: -31.01). Furthermore, %ΔMTV41 significantly predicted patients' recurrence-free survival (RFS) (P < 0.1). In the LN analysis, 7 LNs were classified as well-responders and 10 as poor-responders. Pre-treatment SULmax, SULpeak were significantly lower in poor-responders compared to well-responders. Post-treatment MTV50 and all percentage changes in parameters were significantly higher in the poor-responder group (all P < 0.05). Receiver operating characteristic curve (ROC) analysis indicated %ΔTLG50 had excellent predictive performance for well-responders, with an AUC of 1.000 (cut-off: -7.5). However, there was no significant correlation between the metabolic response evaluations for PTs and LNs. CONCLUSION The metabolic parameters of [18F]FDG PET/CT, particularly %ΔMTV and %ΔTLG, could effectively predict well-responders among both PTs and LNs to neoadjuvant PD-L1 blockade monotherapy in LA-ESCC, which may facilitate personalized immunotherapy and serve as a stratification tool in future larger-scale studies.
Collapse
Affiliation(s)
- Runjun Yang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Han Tang
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
| | - Yunze Xie
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Danjie Cai
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yibo He
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhe Zheng
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yu Lin
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Huaping Gao
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenxin Tang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yihan Yan
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lijie Tan
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China.
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China.
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China.
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China.
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Sirviö VEJ, Räsänen JV, Helminen O, Helmiö M, Huhta H, Kallio R, Koivukangas V, Kokkola A, Laine S, Lietzen E, Meriläinen S, Pohjanen VM, Rantanen T, Ristimäki A, Saarnio J, Sihvo E, Toikkanen V, Tyrväinen T, Uimonen M, Valtola A, Kauppila JH. Incidence trends of surgical complications after oesophagectomy for oesophageal cancer: a population-based, nationwide cohort study in Finland over 30 years. World J Surg Oncol 2025; 23:59. [PMID: 39966948 PMCID: PMC11834619 DOI: 10.1186/s12957-025-03709-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Knowledge on the incidence of surgical complications after oesophagectomy for oesophageal cancer in nationwide practice is scarce. The aim of this study was to assess complication trends after oesophagectomy in a nationwide, population-based, unselected cohort. METHODS All patients undergoing oesophagectomy for oesophageal cancer in Finland in 1987-2016 were included. All complications defined by the Esophagectomy Complications Consensus Group (ECCG) were reported in three 10-year periods. Chi-square test and Kruskal-Wallis test were used to compare outcomes in these periods. RESULTS A total of 1493 patients were included. From 1987 to 1996 to 2007-2016, improvements were seen in the rate of major complications (49% vs. 43%, p = 0.039), length of hospital stay (19 vs. 14 days, median, p < 0.001), length of ICU-stay (3 vs. 2 days, median, p < 0.001) and 90-day mortality (17.9% vs. 5.4%, p < 0.001), while pneumonia (16% vs. 23%, p = 0.029) and anastomotic leak (8% vs. 12% in total leaks, p = 0.006 for type II leak) increased. The most common complications were pneumonia, pleural effusion requiring drainage (16% vs. 23%, p = 0.080), atrial dysrhythmia (16% vs. 15%, p = 0.464) and anastomotic leak. The most common complication categories defined by ECCG were pulmonary (36% vs. 42%, p = 0.151) and gastrointestinal (21% vs. 23%, p = 0.398) complications. CONCLUSIONS This study reports high postoperative morbidity after oesophagectomy in nationwide practice. Mortality has significantly improved during the years, and it appears that patients who would have died earlier, can now be rescued. While the relative occurrence of complications has remained constant, overall morbidity has decreased as the more severe outcomes have decreased.
Collapse
Affiliation(s)
- Ville E J Sirviö
- Department of Oesophageal and General Thoracic Surgery, Helsinki University Hospital, University of Helsinki, Haartmaninkatu 4 PL 340, Helsinki, 00029 HUS, Finland.
| | - Jari V Räsänen
- Department of Oesophageal and General Thoracic Surgery, Helsinki University Hospital, University of Helsinki, Haartmaninkatu 4 PL 340, Helsinki, 00029 HUS, Finland
| | - Olli Helminen
- Surgery Research Unit, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Mika Helmiö
- The Division of Digestive Surgery and Urology, Turku University Hospital and University of Turku, Turku, Finland
| | - Heikki Huhta
- Surgery Research Unit, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Raija Kallio
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| | - Vesa Koivukangas
- Surgery Research Unit, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Arto Kokkola
- Department of Surgery, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Simo Laine
- The Division of Digestive Surgery and Urology, Turku University Hospital and University of Turku, Turku, Finland
| | - Elina Lietzen
- The Division of Digestive Surgery and Urology, Turku University Hospital and University of Turku, Turku, Finland
| | - Sanna Meriläinen
- Surgery Research Unit, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Vesa-Matti Pohjanen
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Tuomo Rantanen
- Department of Surgery, University of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - Ari Ristimäki
- Department of Pathology and HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Juha Saarnio
- Surgery Research Unit, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Eero Sihvo
- Department of Surgery, Central Finland Central Hospital, Jyväskylä, Finland
| | - Vesa Toikkanen
- Department of Cardiothoracic Surgery, Heart Center, Tampere University Hospital, University of Tampere, Tampere, Tampere, Finland
| | - Tuula Tyrväinen
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
| | - Mikko Uimonen
- Department of Surgery, Central Finland Central Hospital, Jyväskylä, Finland
| | - Antti Valtola
- Department of Surgery, University of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - Joonas H Kauppila
- Surgery Research Unit, Oulu University Hospital, University of Oulu, Oulu, Finland
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Montagut C, Martelli V, Vidal J. FOxTROT: is anti-EGFR a good dancing partner? Ann Oncol 2025:S0923-7534(25)00060-2. [PMID: 39924086 DOI: 10.1016/j.annonc.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 02/01/2025] [Indexed: 02/11/2025] Open
Affiliation(s)
- C Montagut
- Department of Medical Oncology, Hospital del Mar Research Institute, CIBERONC, Universitat Pompeu Fabra, Barcelona, Spain.
| | - V Martelli
- Department of Medical Oncology, Hospital del Mar Research Institute, CIBERONC, Universitat Pompeu Fabra, Barcelona, Spain; Department of Internal Medicine and Medical Specialties (DiMI), Università degli Studi di Genova, Genoa, Italy
| | - J Vidal
- Department of Medical Oncology, Hospital del Mar Research Institute, CIBERONC, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
5
|
Kloft M, Budginaite E, van Kuijk SMJ, Raghuram G, Magee DR, Nankivell MG, Cunningham D, Allum WH, Langley RE, Grabsch HI. Intra-patient comparison of microarchitecture of tumour negative lymph nodes from oesophageal cancer patients - Results from the MRC Oe02 trial. Pathol Res Pract 2025; 266:155818. [PMID: 39842369 DOI: 10.1016/j.prp.2025.155818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Regional lymph node (LN) status is a key prognostic factor in oesophageal cancer (OeC). Tumour-derived antigens can activate immune reactions in LNs, potentially reflecting the host's anti-tumour immune response. It remains unclear whether this response is homogeneous across all tumour negative LNs (LNneg) within individual OeC patients. PURPOSE To investigate the hypotheses: (1) the host anti-tumour immune response is similar in all LNneg from an individual OeC patient reflected in a similar microarchitecture in all LNneg; and (2) immune response measured in the largest LNneg can represent that of all LNnegs. METHODS (y)pN0 patients from the Oe02 trial with at least two LNneg were included. Microarchitectural LN features (germinal centres (GermC), lymphocytes outside GermCs (lymphocytes), histiocytes) were morphometrically quantified. Linear mixed-effects models, intraclass correlation coefficients (ICC) and Bland-Altman plots were used to determine systematic bias, reliability/variability and agreement of LNneg microarchitecture measurements. RESULTS Linear mixed-effects models showed no systematic bias in LNneg microarchitectural features within a patient. The ICC revealed moderate variability for lymphocytes (ICC: 0.39; 95 %CI: 0.01- 0.61, p = 0.02)) and GermC (ICC: 0.50; 95 %CI: 0.22-0.68, p < 0.001), and high variability for histiocytes (ICC: 0.07 (95 %CI: -0.45-0.40, p = 0.38). Bland-Altman plots showed that 5.0 % of GermC, 5.0 % of histiocytes and 8.5 % of lymphocyte measurements were outside the 95 % limits of agreement. CONCLUSIONS This is the first study to systematically assess agreement of microarchitectural features in LNneg within an individual (y)pN0 OeC patient. The absence of systematic bias supports using largest LNneg as surrogate for OeC patient's overall anti-tumour immune response.
Collapse
Affiliation(s)
- Maximilian Kloft
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, the Netherlands; Institute of Clinical Cancer Research, Krankenhaus Nordwest, Frankfurt, Germany
| | - Elzbieta Budginaite
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Precision Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Sander M J van Kuijk
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Gayatri Raghuram
- Department of Pathology, Nottingham University Hospitals, Nottingham, UK
| | | | | | - David Cunningham
- Gastrointestinal and Lymphoma Unit, Royal Marsden Hospital, London, UK
| | | | - Ruth E Langley
- MRC Clinical Trials Unit at UCL, University College London, London, UK
| | - Heike I Grabsch
- Department of Pathology, GROW - Research Institute for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, the Netherlands; Pathology and Data Analytics, Leeds Institute of Medical Research at St. James's, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
6
|
Noronha V, Patil VM, Menon N, Joshi A, Shah MJ, Singh A, Goud S, Shah S, More S, Nawale K, Nakti D, Yadav A, Jogdhankar S, Kaushal RK, Tiwari VK, Niyogi D, Purandare N, Janu A, Chakrabarty N, Mahajan A, Tibdewal A, Agarwal J, Pawar A, Chowdhury OR, Sharma V, Kapu V, Trikha M, Kumar SV, Kolkur M, Bhagyavant P, Peelay Z, Khedkar R, Jain M, Badwe RA, Prabhash K. Phase III randomized trial comparing neoadjuvant paclitaxel plus platinum with 5-fluorouracil plus platinum in esophageal or gastroesophageal junction squamous cell carcinoma. J Natl Cancer Inst 2025; 117:58-75. [PMID: 39222012 DOI: 10.1093/jnci/djae214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/25/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Standard neoadjuvant chemotherapy for locally advanced esophageal or gastroesophageal junction squamous cancer, 5-fluorouracil plus platinum, is toxic and logistically challenging; alternative regimens are needed. METHODS This was a phase III randomized open-label noninferiority trial at Tata Memorial Center, India, in resectable locally advanced esophageal or gastroesophageal junction squamous cancer. Patients were randomly assigned 1:1 to 3 cycles of 3-weekly platinum (cisplatin 75 mg/m2 or carboplatin area under the curve 6) with paclitaxel 175 mg/m2 (day 1) or 5-fluorouracil 1000 mg/m2 continuous infusion (days 1-4), followed by surgery. RESULTS Between August 2014 and June 2022, we enrolled 420 patients; 210 to each arm. Statistically significantly more patients on paclitaxel plus platinum (n =194, 92.3%) received all 3 chemotherapy cycles than on 5-fluorouracil with platinum (n = 170, 85.9%; P = .009). 5-fluorouracil plus platinum caused more grade 3 or higher toxicities (n = 124, 69.7%) than paclitaxel plus platinum (n = 97, 51.9%; P = .001). Surgery was performed in 131 (62.4%) patients on 5-fluorouracil plus platinum vs 139 (66.2%) on paclitaxel plus platinum (P = .415). Paclitaxel plus platinum resulted in higher pathologic primary tumor clearance (n = 33, 25.8%, vs n = 17, 15%; P = .04) and pathologic complete responses in 21.9% compared with 12.4% from 5-fluorouracil plus platinum (P = .053). Median overall survival was 27.5 months (95% confidence interval [CI] = 18.6 to 43.5 months) from paclitaxel plus platinum, which was noninferior to 27.1 months (95% CI = 18.8 to 40.7 months) from 5-fluorouracil plus platinum (hazard ratio [HR] = 0.89, 95% CI = 0.72 to 1.09; P = .346). CONCLUSION Neoadjuvant paclitaxel plus platinum chemotherapy is safer and results in similar R0 resections, higher pathologic tumor clearance and noninferior survival compared with 5-fluorouracil plus platinum. Paclitaxel plus platinum should replace 5-fluorouracil plus platinum as neoadjuvant chemotherapy for resectable locally advanced esophagealor gastroesophageal junction squamous cancer. CLINICAL TRIALS REGISTRY INDIA NUMBER CTRI/2014/04/004516.
Collapse
Affiliation(s)
- Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Vijay Maruti Patil
- Department of Medical Oncology, PD Hinduja Hospital Medical Research Centre, Mumbai, Khar and Mahim, India
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Amit Joshi
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Minit Jalan Shah
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Ajaykumar Singh
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Supriya Goud
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Srushti Shah
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sucheta More
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Kavita Nawale
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Dipti Nakti
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Akanksha Yadav
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Shweta Jogdhankar
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Rajiv Kumar Kaushal
- Department of Pathology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Virendra Kumar Tiwari
- Department of Surgical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Devayani Niyogi
- Department of Surgical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Nilendu Purandare
- Department of Nuclear Medicine, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Amit Janu
- Department of Radiodiagnosis, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Nivedita Chakrabarty
- Department of Radiodiagnosis, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Abhishek Mahajan
- Department of Imaging, Clatterbridge Cancer Centre National Health Service Foundation Trust, Liverpool, United Kingdom and Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - Anil Tibdewal
- Department of Radiation Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Jaiprakash Agarwal
- Department of Radiation Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Akash Pawar
- Department of Biostatistics, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Oindrila Roy Chowdhury
- Department of Biostatistics, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Vibhor Sharma
- Department of Medical Oncology, Yatharth Superspeciality Hospital, Noida Extension, Uttar Pradesh, India
| | - Venkatesh Kapu
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Mehak Trikha
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Srigadha Vivek Kumar
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Manali Kolkur
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Priyanka Bhagyavant
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Zoya Peelay
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Rutvij Khedkar
- Department of Pathology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Medha Jain
- Department of Pathology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Rajendra Achyut Badwe
- Department of Surgical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
7
|
He KJ, Gong G. Prognostic prediction and immune infiltration analysis based on lysosome and senescence state identifies MMP12 as a novel therapy target in gastric cancer. Int Immunopharmacol 2024; 143:113344. [PMID: 39401475 DOI: 10.1016/j.intimp.2024.113344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/21/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND AND AIMS As humans undergo the aging process, they become more vulnerable to various types of cancers, including gastric cancer (GC), which is frequently associated with aging. The senescent phenotype is closely linked to lysosomes, but research on the combined impact of senescence and lysosomes on GC prognosis is scarce. METHODS To construct and validate a prognostic model for gastric cancer (GC), we obtained gene expression and clinical data of GC patients from Cancer Genome Atlas (TCGA) databases. We employed Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression for model construction and ConsensusClusterPlus R package for generating cluster heatmaps. The model's predictive ability was evaluated through Kaplan-Meier survival analysis and ROC curve analysis. Our analysis included an assessment of the senescence and lysosome state using expression profiles and immune infiltration analysis through CIBERSORT methods. Finally, we validated potential gene targets through cellular experiments. RESULTS "In this research, we discovered two subtypes of gastric cancer (GC), Cluster 1 and Cluster 2. These subtypes are characterized by the presence of lysosomes and senescence, and we have identified distinct molecular features unique to each subtype. We observed that Cluster 2 had a lower survival prognosis compared to Cluster 1. Additionally, we have developed a risk prediction model that takes into consideration the presence of lysosomes and senescence. Patients in the high-risk group, as predicted by our model, experienced shorter survival times. Further analysis included immune infiltration, immune checkpoint, and chemotherapy evaluation of GC patients. We have displayed the frequency of mutations and copy number variations (CNVs) in visual formats. Our cellular experiments demonstrated that the MMP12 gene serves as a protective factor in GC cells." CONCLUSIONS In conclusion, we have clarified the extensive relationship between lysosomes and senescence in GC and developed a risk signature to forecast the prognosis of GC patients. MMP12 could be a promising protective factor for GC patients and might present a novel concept for anticipating the efficacy of targeted therapies and immunotherapies in GC patients.
Collapse
Affiliation(s)
- Ke-Jie He
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou City, Zhejiang Province, China.
| | - Guoyu Gong
- School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
8
|
Orabi A, Chillarge G, Di Mauro D, Veeramootoo D, Njere I, Manzelli A, Wajed S. Survival outcomes fifteen years after minimally invasive esophagectomy. Discov Oncol 2024; 15:708. [PMID: 39585588 PMCID: PMC11589040 DOI: 10.1007/s12672-024-01567-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Minimally invasive esophagectomy (MIO) offers a less traumatic resection for cancer patients resulting in improved quality of life. Concerns about the oncological efficacy of the procedure and potential impact on survival may have limited its wider adoption. This study reports survival outcomes fifteen years after patients underwent a total MIO for esophageal cancer. METHODS A single-centre analysis of survival outcomes was conducted on all patients who underwent MIO between 2004 and 2010 and had completed at least 15-years follow-up. Actual overall survival (OS) and disease-free survival (DFS) were evaluated with the Kaplan-Meier method. The pattern of association of patient factors with survival was assessed with the Cox regression analysis. RESULTS A total of 121 patients underwent resection, with 4 (3.3%) in-patient deaths. With a median follow-up time of 15.2 years, the median OS was 41 months. At 15-years there were 20 survivors (17.1%), with numbers at 10 and 5 years being 34 (29.1%) and 45 (38.5%) respectively. Median DFS was 27 months, with 19 (16.2%) patient disease free at 15 years, and 32 (27.4%) and 38 (32.5%) at 10 and 5 years respectively. Predictors of poor survival were the intracorporeal fashioning of the gastric conduit, perioperative blood transfusions, advanced disease stage and recurrence. CONCLUSIONS Long term survival outcomes following MIO support its increasing recognition as the standard of care as for curative resection in esophageal cancer.
Collapse
Affiliation(s)
- Amira Orabi
- Department of Upper GI Surgery, Royal Devon University Healthcare NHS Foundation Trust, Barrack Road, Exeter, EX25DW, UK
| | - Gauri Chillarge
- Department of Upper GI Surgery, Royal Devon University Healthcare NHS Foundation Trust, Barrack Road, Exeter, EX25DW, UK
| | - Davide Di Mauro
- Department of Upper GI Surgery, Royal Devon University Healthcare NHS Foundation Trust, Barrack Road, Exeter, EX25DW, UK.
- College of Medicine and Health, University of Exeter, Exeter, UK.
| | | | - Ikechukwu Njere
- Department of Upper GI Surgery, Royal Devon University Healthcare NHS Foundation Trust, Barrack Road, Exeter, EX25DW, UK
| | - Antonio Manzelli
- Department of Upper GI Surgery, Royal Devon University Healthcare NHS Foundation Trust, Barrack Road, Exeter, EX25DW, UK
| | - Shahjehan Wajed
- Department of Upper GI Surgery, Royal Devon University Healthcare NHS Foundation Trust, Barrack Road, Exeter, EX25DW, UK
- College of Medicine and Health, University of Exeter, Exeter, UK
| |
Collapse
|
9
|
Kahana N, Boaz E, Horesh N, Emile SH, Dourado J, Aeschbacher P, Rogers P, Gefen R, Lo Menzo E, Rosenthal RJ. Evaluation of the robustness of randomized controlled trials for the treatment modalities of esophageal cancer using the fragility index - a systematic review. Surg Endosc 2024:10.1007/s00464-024-11343-3. [PMID: 39443379 DOI: 10.1007/s00464-024-11343-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Esophageal cancer remains a significant global health challenge. Several treatment modalities were explored in randomized controlled trials (RCTs) in recent decades. This study evaluates the robustness of RCTs focusing on esophageal cancer treatment using the fragility index (FI) and reverse fragility index (RFI). METHODS A systematic review of RCTs studying different treatment modalities for esophageal cancer from 2000 to 2023 was conducted. The FI and RFI were utilized to gauge the robustness of statistically significant and non-significant outcomes, respectively. The FI represents the minimal number of patient outcomes that would need to alter to overturn a trial's statistical significance, while RFI indicates the minimal changes required to achieve significance in non-significant results. RESULTS Out of 4028 studies retrieved, 21 RCTs were included for final analysis. The studies spanned 2001 to 2023 with a mean followup of 66 months (range, 29-108 months) and median number of patients of 194 (range, 45-802). The most common treatment modalities examined in these studies were neoadjuvant chemoradiotherapy (n = 7, 33.3%), neoadjuvant chemotherapy (n = 4, 19.0%), and neoadjuvant immunotherapy (n = 2, 9.5%). Only 5 studies (23.8%) had a statistically significant primary outcome result with a median FI of 6 (IQR, 2.5-8.5). Non-significant primary outcomes were seen in 16 studies (76.2%) with a median RFI of 4 (IQR 1-11) and lost to followup of 0 (IQR 0-4). In the study with the highest FI (10), the FI was lower than the number of patients lost to followup (13). CONCLUSION Our findings demonstrate that most RCTs on esophageal cancer treatments did not report significant primary outcomes. The few studies that reported significant results had a low fragility index, suggesting a vulnerability in their findings.
Collapse
Affiliation(s)
- Noam Kahana
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
- Department of General Surgery Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Elad Boaz
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
- Department of General Surgery Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nir Horesh
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
- Department of Surgery and Transplantations, Sheba Medical Center, Ramat Gan, Israel, Tel Aviv University, Tel Aviv, Israel
| | - Sameh Hany Emile
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
- Colorectal Surgery Unit, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Justin Dourado
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
| | - Pauline Aeschbacher
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
| | - Pete Rogers
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
| | - Rachel Gefen
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
- Department of General Surgery, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Emanuele Lo Menzo
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA
| | - Raul J Rosenthal
- Department of General Surgery, Cleveland Clinic Florida, 2950 Cleveland Clinic Blvd., Weston, FL, 33331, USA.
| |
Collapse
|
10
|
Cai Q, Hong Y, Huang X, Chen T, Chen C. Current status and prospects of diagnosis and treatment for esophageal cancer with supraclavicular lymph node metastasis. Front Oncol 2024; 14:1431507. [PMID: 39464710 PMCID: PMC11502295 DOI: 10.3389/fonc.2024.1431507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 09/25/2024] [Indexed: 10/29/2024] Open
Abstract
Patients with supraclavicular lymph node (SLN) metastasis from esophageal cancer encounter significant variations in treatment approaches due to differences in pathological subtypes and the lack of a unified regional staging system between East Asian and Western countries. The Tiger study aims to develop an internationally recognized staging system and to delineate the extent of regional lymph node dissection. In the context of esophageal squamous cell carcinoma (SCC) with SLN metastasis, the treatment paradigms from East Asia offer valuable insights. The Japan Esophageal Society (JES) 12th edition staging system guides a tailored comprehensive treatment strategy, emphasizing either radiotherapy and chemotherapy or surgical intervention. In contrast, esophageal adenocarcinoma (AC) predominates in Western countries, where the 8th edition of the American Joint Committee on Cancer (AJCC) staging system classifies SLN metastasis as a distant metastasis, advocating for systemic therapy as the primary treatment modality. Nonetheless, compelling evidence suggests that a multidisciplinary treatment approach, incorporating either radiotherapy and chemotherapy or surgery as the initial treatment, can yield superior outcomes for these patients compared to chemotherapy alone.
Collapse
Affiliation(s)
- Qingxin Cai
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yingji Hong
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xuehan Huang
- Shantou University Medical College, Shantou, China
| | - Tong Chen
- Shantou University Medical College, Shantou, China
| | - Chuangzhen Chen
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
11
|
Knipper K, Jung JO, Straatman J, Brunner S, Wirsik NM, Lyu SI, Fuchs HF, Gebauer F, Schröder W, Schlößer HA, Quaas A, Bruns CJ, Schmidt T. The role of length of oral resection margin and survival in esophageal cancer surgery after neoadjuvant therapy: A retrospective propensity score-matched study. Surgery 2024; 176:1098-1103. [PMID: 38944588 DOI: 10.1016/j.surg.2024.04.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/05/2024] [Accepted: 04/27/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND Multimodal therapy regimens became the standard of care for patients with esophageal cancer, whereas surgical resection remains at the center of curative treatment modalities. Current guidelines provide no recommendations on the extent of the oral resection margin, especially in the era of neoadjuvant therapy. Therefore, this study aimed to evaluate the relationship between the oral tumor-free resection margin and overall survival. METHODS Retrospective study with 382 1:1 propensity-matched patients out of 660 patients, operated between 2013 and 2019, with an Ivor-Lewis-esophagectomy for adenocarcinoma and squamous cell carcinoma of the esophagus or esophagogastric junction after neoadjuvant therapy. Independent pathologists measured the oral resection margin after formalin fixation. RESULTS The mean oral tumor-free resection margin was 37.2 ± 0.6 mm. The ideal cut-off for survival differences was determined for 33 mm. Patients with an oral resection margin of more than 33 mm had a better median overall survival (≤33 mm: 45.0 months, 95% confidence interval: 22.4-67.6 months, >33 mm: not reached, P = .005). An oral resection margin of more than 33 mm proved to be an independent favorable prognostic factor for patients' overall survival in multivariate Cox regression analyses (P = .049). CONCLUSION This study analyzed a patient cohort retrospectively after curative intended Ivor-Lewis-esophagectomy after neoadjuvant therapy. An oral resection margin of more than 33 mm is a factor for improved overall survival. Therefore, a minimum resection margin of 34 mm after fixation could be suggested.
Collapse
Affiliation(s)
- Karl Knipper
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Jin-On Jung
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Jennifer Straatman
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Stefanie Brunner
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Naita M Wirsik
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Su Ir Lyu
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Hans F Fuchs
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Florian Gebauer
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Wolfgang Schröder
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Hans A Schlößer
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Christiane J Bruns
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Thomas Schmidt
- Faculty of Medicine and University Hospital of Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany.
| |
Collapse
|
12
|
O'Shea R, Withey SJ, Owczarczyk K, Rookyard C, Gossage J, Godfrey E, Jobling C, Parsons SL, Skipworth RJE, Goh V. Multicentre validation of CT grey-level co-occurrence matrix features for overall survival in primary oesophageal adenocarcinoma. Eur Radiol 2024; 34:6919-6928. [PMID: 38526750 PMCID: PMC11399295 DOI: 10.1007/s00330-024-10666-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Personalising management of primary oesophageal adenocarcinoma requires better risk stratification. Lack of independent validation of proposed imaging biomarkers has hampered clinical translation. We aimed to prospectively validate previously identified prognostic grey-level co-occurrence matrix (GLCM) CT features for 3-year overall survival. METHODS Following ethical approval, clinical and contrast-enhanced CT data were acquired from participants from five institutions. Data from three institutions were used for training and two for testing. Survival classifiers were modelled on prespecified variables ('Clinical' model: age, clinical T-stage, clinical N-stage; 'ClinVol' model: clinical features + CT tumour volume; 'ClinRad' model: ClinVol features + GLCM_Correlation and GLCM_Contrast). To reflect current clinical practice, baseline stage was also modelled as a univariate predictor ('Stage'). Discrimination was assessed by area under the receiver operating curve (AUC) analysis; calibration by Brier scores; and clinical relevance by thresholding risk scores to achieve 90% sensitivity for 3-year mortality. RESULTS A total of 162 participants were included (144 male; median 67 years [IQR 59, 72]; training, 95 participants; testing, 67 participants). Median survival was 998 days [IQR 486, 1594]. The ClinRad model yielded the greatest test discrimination (AUC, 0.68 [95% CI 0.54, 0.81]) that outperformed Stage (ΔAUC, 0.12 [95% CI 0.01, 0.23]; p = .04). The Clinical and ClinVol models yielded comparable test discrimination (AUC, 0.66 [95% CI 0.51, 0.80] vs. 0.65 [95% CI 0.50, 0.79]; p > .05). Test sensitivity of 90% was achieved by ClinRad and Stage models only. CONCLUSIONS Compared to Stage, multivariable models of prespecified clinical and radiomic variables yielded improved prediction of 3-year overall survival. CLINICAL RELEVANCE STATEMENT Previously identified radiomic features are prognostic but may not substantially improve risk stratification on their own. KEY POINTS • Better risk stratification is needed in primary oesophageal cancer to personalise management. • Previously identified CT features-GLCM_Correlation and GLCM_Contrast-contain incremental prognostic information to age and clinical stage. • Compared to staging, multivariable clinicoradiomic models improve discrimination of 3-year overall survival.
Collapse
Affiliation(s)
- Robert O'Shea
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Samuel J Withey
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Department of Radiology, Royal Marsden Hospital NHS Trust, Sutton, Surrey, UK
| | - Kasia Owczarczyk
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Department of Clinical Oncology, Guy's & St Thomas' Hospitals NHS Foundation Trust, London, UK
| | - Christopher Rookyard
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - James Gossage
- Department of Surgery, Guy's & St Thomas' Hospitals NHS Foundation Trust, London, UK
| | - Edmund Godfrey
- Department of Radiology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Craig Jobling
- Department of Radiology, Nottingham University Hospitals NHS Foundation Trust, Nottingham, UK
| | - Simon L Parsons
- Department of Surgery, Nottingham University Hospitals NHS Foundation Trust, Nottingham, UK
| | | | - Vicky Goh
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK.
- Department of Radiology, Guy's & St Thomas' Hospitals NHS Foundation Trust, Westminster Bridge Road, London, SE1 7EG, UK.
| |
Collapse
|
13
|
Zheng Y, Liang G, Yuan D, Liu X, Ba Y, Qin Z, Shen S, Li Z, Sun H, Liu B, Gao Q, Li P, Wang Z, Liu S, Zhu J, Wang H, Ma H, Liu Z, Zhao F, Zhang J, Zhang H, Wu D, Qu J, Ma J, Zhang P, Ma W, Yan M, Yu Y, Li Q, Zhang J, Xing W. Perioperative toripalimab plus neoadjuvant chemotherapy might improve outcomes in resectable esophageal cancer: an interim analysis of a phase III randomized clinical trial. Cancer Commun (Lond) 2024; 44:1214-1227. [PMID: 39221992 PMCID: PMC11483553 DOI: 10.1002/cac2.12604] [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: 01/22/2024] [Revised: 08/04/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND In the era of immunotherapy, neoadjuvant immunochemotherapy (NAIC) for the treatment of locally advanced esophageal squamous cell carcinoma (ESCC) is used clinically but lacks of high-level clinical evidence. This study aimed to compare the safety and long-term efficacy of NAIC followed by minimally invasive esophagectomy (MIE) with those of neoadjuvant chemotherapy (NAC) followed by MIE. METHODS A prospective, single-center, open-label, randomized phase III clinical trial was conducted at Henan Cancer Hospital, Zhengzhou, China. Patients were randomly assigned to receive either neoadjuvant toripalimab (240 mg) plus paclitaxel (175 mg/m2) + cisplatin (75 mg/m2) (toripalimab group) or paclitaxel + cisplatin alone (chemotherapy group) every 3 weeks for 2 cycles. After surgery, the toripalimab group received toripalimab (240 mg every 3 weeks for up to 6 months). The primary endpoint was event-free survival (EFS). The pathological complete response (pCR) and overall survival (OS) were key secondary endpoints. Adverse events (AEs) and quality of life were also assessed. RESULTS Between May 15, 2020 and August 13, 2021, 252 ESCC patients ranging from T1N1-3M0 to T2-3N0-3M0 were enrolled for interim analysis, with 127 in the toripalimab group and 125 in the chemotherapy group. The 1-year EFS rate was 77.9% in the toripalimab group compared to 64.3% in the chemotherapy group (hazard ratio [HR] = 0.62; 95% confidence interval [CI] = 0.39 to 1.00; P = 0.05). The 1-year OS rates were 94.1% and 83.0% in the toripalimab and chemotherapy groups, respectively (HR = 0.48; 95% CI = 0.24 to 0.97; P = 0.037). The patients in the toripalimab group had a higher pCR rate (18.6% vs. 4.6%; P = 0.001). The rates of postoperative Clavien-Dindo grade IIIb or higher morbidity were 9.8% in the toripalimab group and 6.8% in the chemotherapy group, with no significant difference observed (P = 0.460). The rates of grade 3 or 4 treatment-related AEs did not differ between the two groups (12.5% versus 12.4%). CONCLUSIONS The interim results of this ongoing trial showed that in resectable ESCC, the addition of perioperative toripalimab to NAC is safe, may improve OS and might change the standard treatment in the future.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Guanghui Liang
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Dongfeng Yuan
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Xianben Liu
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Yufeng Ba
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Zimin Qin
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Sining Shen
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Zhenxuan Li
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Haibo Sun
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Baoxing Liu
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Quanli Gao
- Department of ImmunotherapyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Peng Li
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Zongfei Wang
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Shilei Liu
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Jianping Zhu
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Haoran Wang
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Haibo Ma
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Zhenzhen Liu
- Department of EndoscopyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Fei Zhao
- Department of EndoscopyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Jun Zhang
- Department of EndoscopyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - He Zhang
- Department of PathologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Daoyuan Wu
- Department of PathologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Jinrong Qu
- Department of RadiologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Jie Ma
- Department of BiobankThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Peng Zhang
- Department of Strategic DevelopmentThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Wenjie Ma
- Department of Strategic DevelopmentThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Ming Yan
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Yongkui Yu
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| | - Qing Li
- Department of StatisticsLinkDoc Technology Co., LtdBeijingP. R. China
| | - Jiangong Zhang
- Department of Cancer EpidemiologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouP. R. China
| | - Wenqun Xing
- Department of Thoracic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouHenanP. R. China
| |
Collapse
|
14
|
Patel PH, Patel NM, Doyle JP, Patel HK, Alhasan Y, Luangsomboon A, Petrou N, Bhogal RH, Kumar S, Chaudry MA, Allum WH. Circumferential resection margin rates in esophageal cancer resection: oncological equivalency and comparable clinical outcomes between open versus minimally invasive techniques - a retrospective cohort study. Int J Surg 2024; 110:6257-6267. [PMID: 38526511 PMCID: PMC11486989 DOI: 10.1097/js9.0000000000001296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/22/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Radical surgery for esophageal cancer requires macroscopic and microscopic clearance of all malignant tissue. A critical element of the procedure is achieving a negative circumferential margin (CRM) to minimize local recurrence. The utility of minimally invasive surgery poses challenges in replicating techniques developed in open surgery, particularly for hiatal dissection in esophago-gastrectomy. In this study, the technical approach and clinical and oncological outcomes for open and laparoscopic esophago-gastrectomy are described with particular reference to CRM involvement. MATERIALS AND METHODS This cohort study included all patients undergoing either open or laparoscopic esophago-gastrectomy between January 2004 and June 2022 in a single tertiary center. A standard surgical technique for hiatal dissection of the esophago-gastric junction developed in open surgery was adapted for a laparoscopic approach. Clinical parameters, length of stay (LOS), postoperative complications, and mortality data were collected and analyzed by a Mann-Whitney U or Fisher's exact method. RESULTS Overall 447 patients underwent an esophago-gastrectomy in the study with 219 open and 228 laparoscopic procedures. The CRM involvement was 18.8% in open surgery and 13.6% in laparoscopic surgery. The 90-day-mortality for open surgery was 4.1 compared with 2.2% for laparoscopic procedures. Median Intensive care unit (ITU), inpatient LOS and 30-day readmission rates were shorter for laparoscopic compared with open esophago-gastrectomy (ITU: 5 versus 8 days, P= 0.0004; LOS: 14 versus 20 days, P= 0.022; 30-day re-admission 7.46 versus 10.50%). Postoperative complication rates were comparable across both cohorts. The rates of starting adjuvant chemotherapy were 51.8 after open and 74.4% in laparoscopic esophago-gastrectomy. CONCLUSION This study presents a standardized surgical approach to hiatal dissection for esophageal cancer. The authors present equivalence between open and laparoscopic esophago-gastrectomy in clinical, oncological, and survival outcomes with similar rates of CRM involvement. The authors also observe a significantly shorter hospital length of stay with the minimally invasive approach.
Collapse
Affiliation(s)
- Pranav H. Patel
- Department of Upper GI Academic Surgery, The Royal Marsden Hospital NHS Foundation Trust
| | - Nikhil M. Patel
- Department of Upper GI Academic Surgery, The Royal Marsden Hospital NHS Foundation Trust
| | - Joseph P. Doyle
- Department of Upper GI Academic Surgery, The Royal Marsden Hospital NHS Foundation Trust
| | - Hina K. Patel
- Department of Upper GI Academic Surgery, The Royal Marsden Hospital NHS Foundation Trust
| | - Yousef Alhasan
- Department of Upper GI Academic Surgery, The Royal Marsden Hospital NHS Foundation Trust
| | - Alfa Luangsomboon
- Department of Upper GI Academic Surgery, The Royal Marsden Hospital NHS Foundation Trust
| | - Nikoletta Petrou
- Department of Upper GI Academic Surgery, The Royal Marsden Hospital NHS Foundation Trust
| | - Ricky H. Bhogal
- Department of Upper GI Academic Surgery, The Royal Marsden Hospital NHS Foundation Trust
- Upper Gastrointestinal Surgical Oncology Research Group, Institute of Cancer Research
| | - Sacheen Kumar
- Department of Upper GI Academic Surgery, The Royal Marsden Hospital NHS Foundation Trust
- Upper Gastrointestinal Surgical Oncology Research Group, Institute of Cancer Research
- Department of Upper GI Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic London Hospital, London, SW1X 7HY, United Kingdom
| | - Mohammed A. Chaudry
- Department of Upper GI Academic Surgery, The Royal Marsden Hospital NHS Foundation Trust
| | - William H. Allum
- Department of Upper GI Academic Surgery, The Royal Marsden Hospital NHS Foundation Trust
| |
Collapse
|
15
|
Guo Y, Xu X, Wang T, Liu Y, Gu D, Fang Y, Wang Q, Shi H, Wu D, Zhang Z, Zhou G, Ye J. Efficacy, safety, and survival of neoadjuvant immunotherapy plus chemotherapy in locally advanced esophageal squamous cell carcinoma: A real-world retrospective study. Int Immunopharmacol 2024; 138:112558. [PMID: 38941666 DOI: 10.1016/j.intimp.2024.112558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/23/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND This study aims to analyze the efficacy and safety of neoadjuvant programmed cell death-1 (PD-1) blockade plus chemotherapy in real-world applications. Additionally, we report survival outcomes with a median follow-up of 40.1 months. METHODS From January 2018 to October 2022, we retrospectively recruited patients with esophageal squamous cell carcinoma (ESCC) who underwent surgery after receiving PD-1 blockade (immunotherapy) plus chemotherapy at Jiangsu Cancer Hospital. RESULTS A total of 132 eligible ESCC patients were included, and R0 resection was achieved in 131 cases (99.2 %). A complete pathological response rate (ypT0N0) was observed in 32 patients (24.2 %), and the objective response rate was 59.1 %. The most common grade 3-4 treatment-related adverse events (TRAEs) were leukopenia (18.2 %) and neutropenia (15.9 %). Three cases (2.3 %) of grade 3 immune-related AEs were observed, including increased ALT (0.8 %), rash (0.8 %), and encephalitis (0.8 %). The 1-year disease-free survival (DFS) and overall survival (OS) rates were 68.2 % and 89.4 %, respectively, and the 2-year DFS and OS rates were 55.1 % and 78.6 %, respectively. The pathological responses of 103 cases (94.5 % of 109) of the index lymph node (ILN) were categorized as the worst regression subgroup. In these cases, using the pathological response of the ILN to indicate the status of other lymph nodes would not result to a missed therapeutic lymph node dissection. CONCLUSIONS Neoadjuvant immunotherapy plus chemotherapy is safe and effective for ESCC, with observable survival benefits. The pathological response of the ILN after neoadjuvant therapy may have important value in guiding therapeutic lymph node dissection.
Collapse
Affiliation(s)
- Yiyu Guo
- Department of Radiation Oncology, Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xinyu Xu
- Department of Pathology, Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Tian Wang
- Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ying Liu
- Department of Oncology, Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Dayong Gu
- Department of Radiation Oncology, Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Ying Fang
- Department of Oncology, Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Qiang Wang
- Department of Oncology, Affiliated Jiangyan Hospital of Nanjing University of Chinese Medicine, Taizhou, China
| | - Haifeng Shi
- Department of Oncology, Sheyang People's Hospital, Yancheng, China
| | - Daguang Wu
- Department of Oncology, Funing People's Hospital, Yancheng, China
| | - Zhi Zhang
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Guoren Zhou
- Department of Oncology, Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
| | - Jinjun Ye
- Department of Radiation Oncology, Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
| |
Collapse
|
16
|
Hu H, Zhang J, Li Y, Wang X, Wang Z, Wang H, Kang L, Liu P, Lan P, Wu X, Zhen Y, Pei H, Huang Z, Zhang H, Chen W, Zeng Y, Lai J, Wei H, Huang X, Chen J, Chen J, Tao K, Xu Q, Peng X, Liang J, Cai G, Ding K, Ding Z, Hu M, Zhang W, Tang B, Hong C, Cao J, Huang Z, Cao W, Li F, Wang X, Wang C, Huang Y, Zhao Y, Cai Y, Ling J, Xie X, Wu Z, Shi L, Ling L, Liu H, Wang J, Huang M, Deng Y. Neoadjuvant Chemotherapy With Oxaliplatin and Fluoropyrimidine Versus Upfront Surgery for Locally Advanced Colon Cancer: The Randomized, Phase III OPTICAL Trial. J Clin Oncol 2024; 42:2978-2988. [PMID: 38564700 DOI: 10.1200/jco.23.01889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/29/2023] [Accepted: 02/02/2024] [Indexed: 04/04/2024] Open
Abstract
PURPOSE The role of neoadjuvant chemotherapy (NAC) in colon cancer remains unclear. This trial investigated whether 3 months of modified infusional fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) or capecitabine and oxaliplatin (CAPOX) as NAC could improve outcomes in patients with locally advanced colon cancer versus upfront surgery. PATIENTS AND METHODS OPTICAL was a randomized, phase III trial in patients with clinically staged locally advanced colon cancer (T3 with extramural spread into the mesocolic fat ≥5 mm or T4). Patients were randomly assigned 1:1 to receive six preoperative cycles of mFOLFOX6 or four cycles of CAPOX, followed by surgery and adjuvant chemotherapy (NAC group), or immediate surgery and the physician's choice of adjuvant chemotherapy (upfront surgery group). The primary end point was 3-year disease-free survival (DFS) assessed in the modified intention-to-treat (mITT) population. RESULTS Between January 2016 and April 2021, of the 752 patients enrolled, 744 patients were included in the mITT analysis (371 in the NAC group; 373 in the upfront surgery group). At a median follow-up of 48.0 months (IQR, 46.0-50.1), 3-year DFS rates were 82.1% in the NAC group and 77.5% in the upfront surgery group (stratified hazard ratio [HR], 0.74 [95% CI, 0.54 to 1.03]). The R0 resection was achieved in 98% of patients who underwent surgery in both groups. Compared with upfront surgery, NAC resulted in a 7% pathologic complete response rate (pCR), significantly lower rates of advanced tumor staging (pT3-4: 77% v 94%), lymph node metastasis (pN1-2: 31% v 46%), and potentially improved overall survival (stratified HR, 0.44 [95% CI, 0.25 to 0.77]). CONCLUSION NAC with mFOLFOX6 or CAPOX did not show a significant DFS benefit. However, this neoadjuvant approach was safe, resulted in substantial pathologic downstaging, and appears to be a viable therapeutic option for locally advanced colon cancer.
Collapse
Affiliation(s)
- Huabin Hu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jianwei Zhang
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yunfeng Li
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, People's Republic of China
| | - Xiaozhong Wang
- Department of Gastrointestinal Surgery, Shantou Central Hospital, Shantou, People's Republic of China
| | - Ziqiang Wang
- Department of General Surgery, Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Liang Kang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ping Liu
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, People's Republic of China
| | - Ping Lan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaojian Wu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yunhuan Zhen
- Department of Colorectal Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
| | - Haiping Pei
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhongcheng Huang
- Department of General Surgery, Hunan Provincial People's Hospital, Changsha, People's Republic of China
| | - Hao Zhang
- Department of General Surgery, Dongguan Kanghua Hospital, Dongguan, People's Republic of China
| | - Wenbin Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yongming Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, People's Republic of China
| | - Jiajun Lai
- Department of Gastrointestinal Surgery, Yuebei People's Hospital, Shaoguan, People's Republic of China
| | - Hongbo Wei
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xuefeng Huang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jiansi Chen
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, People's Republic of China
| | - Jigui Chen
- Department of Surgery, The Eighth Hospital of Wuhan, Wuhan, People's Republic of China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qingwen Xu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Xiang Peng
- Department of Gastrointestinal Surgery, The First People's Hospital of Foshan, Foshan, People's Republic of China
| | - Junlin Liang
- Department of Coloproctological Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Guanfu Cai
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangzhou, People's Republic of China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology, Cancer Center, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Zhijie Ding
- Department of Gastrointestinal Surgery, Zhongshan Hospital, Xiamen University, Xiamen, People's Republic of China
| | - Ming Hu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Wei Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Bo Tang
- Department of General Surgery, The First Hospital Affiliated to Army Medical University, Chongqing, People's Republic of China
| | - Chuyuan Hong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jie Cao
- Department of General Surgery, Guangzhou First People's Hospital, Guangzhou, People's Republic of China
| | - Zonghai Huang
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Wuteng Cao
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Fangqian Li
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xinhua Wang
- Department of Radiology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Chao Wang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yandong Zhao
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yue Cai
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiayu Ling
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xiaoyu Xie
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zehua Wu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Lishuo Shi
- Clinical Research Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li Ling
- Department of Medical Statistics, School of Public Health, and Center for Migrant Health Policy, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hao Liu
- Department of Biostatistics and Epidemiology, Biostatistics Shared Resource, Rutgers Cancer Institute of New Jersey, Rutgers School of Public Health, Brunswick, NJ
| | - Jianping Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Meijin Huang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yanhong Deng
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
17
|
Wu YY, Dai L, Yang YB, Yan WP, Cheng H, Fan MY, Gao YM, Chen KN. Long-Term Survival and Recurrence Patterns in Locally Advanced Esophageal Squamous Cell Carcinoma Patients with Pathologic Complete Response After Neoadjuvant Chemotherapy Followed by Surgery. Ann Surg Oncol 2024; 31:5047-5054. [PMID: 38172446 DOI: 10.1245/s10434-023-14809-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND The higher pathologic complete response (pCR) after neoadjuvant chemoradiotherapy compared with neoadjuvant chemotherapy for locally advanced esophageal squamous cell carcinoma (ESCC) has not translated into significant gains in overall survival. Data on the long-term survival of patients who obtained a pCR after neoadjuvant chemotherapy are scarce. Therefore, this study aimed to evaluate the long-term prognosis and recurrence patterns in these patients. METHODS The study enrolled patients with locally advanced ESCC after neoadjuvant chemotherapy followed by surgery in the authors' hospital between January 2007 and December 2020. The factors predictive of pCR were analyzed. Furthermore, propensity score-matching was performed for those who did and those who did not have a pCR using 1:5 ratio for a long-term survival analysis. Finally, the survival and recurrence patterns of patients obtaining pCR after neoadjuvant chemotherapy were analyzed. RESULTS A pCR was achieved for 61 (8.70%) of the 701 patients in the study. Univariate analysis showed that the patients without alcohol drinking had a higher possibility of obtaining a pCR, although multivariate analysis failed to confirm the difference as significant. After propensity score-matching, the 5-year overall survival was 84.50% for the patients who had a pCR and 52.90% for those who did not (p < 0.001). Among the 61 patients with a pCR, 9 patients (14.80%) experienced recurrence, including 6 patients with locoregional recurrence and 3 patients with distant metastasis. CONCLUSION Advanced ESCC patients with pCR after neoadjuvant chemotherapy had a favorable prognosis, yet some still experienced recurrence, particularly locoregional recurrence. Therefore, for this group of patients, regular follow-up evaluation also is needed.
Collapse
Affiliation(s)
- Ya-Ya Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Liang Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Yong-Bo Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Wan-Pu Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Hong Cheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Meng-Ying Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Yi-Mei Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China
| | - Ke-Neng Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), The First Department of Thoracic Surgery, Peking University Cancer Hospital and Institute, Peking University School of Oncology, Beijing, China.
| |
Collapse
|
18
|
Suzumura H, Terauchi T, Yukisawa S, Kimata M, Shinozaki H. Curative Surgery After Neoadjuvant Chemotherapy for Locally Advanced Sigmoid Colon Cancer With Extensive Abdominal Wall Invasion: A Case Report. Cureus 2024; 16:e67444. [PMID: 39310573 PMCID: PMC11415604 DOI: 10.7759/cureus.67444] [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] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
Locally advanced colon cancer (LACC) can be cured under an appropriate treatment strategy, but the decision on the treatment strategy is also important in terms of long-term prognosis. In cases with extensive abdominal wall involvement, it is especially important to secure adequate margins and repair abdominal wall defects. Recently, neoadjuvant chemotherapy (NAC) for LACC has shown promise in improving the chance of cure with tumor shrinkage. Herein, we report a case of curative surgery after NAC for locally advanced sigmoid colon cancer with extensive abdominal wall invasion. A 50-year-old woman visited our hospital with anemia and an abdominal mass. The diagnosis was LACC of the sigmoid colon with abdominal wall invasion (maximum size, 12 cm), and the clinical stage was stage IIIc (T4b[skin]N1bM0). Resection of the involved skin was expected to cause an extensive abdominal wall defect. At first, a colostomy was performed, followed by NAC with leucovorin, 5-fluorouracil, and oxaliplatin (FOLFOX). Ten cycles of chemotherapy were completed without severe adverse events, and the tumor shrank in size by approximately 39%. We performed a curative sigmoidectomy combined with abdominal wall resection with adequate margins. We reconstructed the abdominal wall defect using a left anterolateral thigh skin flap. Pathological examination revealed mucinous carcinoma involving the transverse colon and abdominal wall, with luminal narrowing in the sigmoid colon. The surgical margins were negative, and the tumor was considered to have had a pathological partial response to NAC. Herein, we report a rare case of curative surgery after NAC with FOLFOX for LACC in the sigmoid colon with extensive invasion of the abdominal wall. We reconstructed the extensive abdominal wall defect with a free anterolateral thigh flap. One of the optional treatment strategies for LACC with extensive abdominal wall invasion was reported in our report.
Collapse
Affiliation(s)
| | | | - Seigo Yukisawa
- Department of Medical Oncology, Saiseikai Utsunomiya Hospital, Tochigi, JPN
| | - Masaru Kimata
- Department of Surgery, Saiseikai Utsunomiya Hospital, Tochigi, JPN
| | | |
Collapse
|
19
|
Qiu J, Zhang Z, Liu J, Zhao Y, Li Y, Tang Z, Li L, Tian Y, Tian H. Nomograms to predict tumor regression grade (TRG) and ypTNM staging in patients with locally advanced esophageal cancer receiving neoadjuvant therapy. World J Surg Oncol 2024; 22:198. [PMID: 39068445 PMCID: PMC11282666 DOI: 10.1186/s12957-024-03474-7] [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: 03/09/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Neoadjuvant therapy (NT) has increased survival rates for patients with locally advanced esophageal cancer (EC), but estimating the impact of NT treatment prior to surgery is still very difficult. METHODS A retrospective study of the clinical information of 150 patients with locally advanced EC who got NT at Qilu Hospital of Shandong University between June 2018 and June 2023. Patients were randomized into training and internal validation groups at a 3:1 ratio. Furthermore, an external validation cohort comprised 38 patients who underwent neoadjuvant therapy at Qianfoshan Hospital in the Shandong Province between June 2021 and June 2023. Independent risk factors were identified using univariate and multivariate logistic regression (forward stepwise regression). Predictive models and dynamic web nomograms were developed by integrating these risk factors. RESULTS A total of 188 patients with locally advanced EC were enrolled, of whom 118 achieved stage I of neoadjuvant pathologic TNM (ypTNM) after receiving NT and 129 achieved grades 0-1 in the tumor regression grade (TRG). Logistic regression analysis identified five independent predictors of TRG grades 0-1: pulmonary function tests (PFT), prognostic nutritional index (PNI), triglyceride (TG) levels, squamous cell carcinoma antigen (SCC-Ag) levels, and combination immunotherapy. The areas under the receiver operating characteristic (ROC) curves for the training, internal validation, and external validation groups were 0.87, 0.75, and 0.80, respectively. Meanwhile, two independent predictors of stage I of ypTNM were identified: prealbumin (PA) and SCC antigen. The areas under the ROC curves for the training, internal validation, and external validation groups were 0.78, 0.67, and 0.70, respectively. The Hosmer-Lemeshow test for both predictive models showed excellent calibration, with well-fitted calibration curves. Decision curve analysis (DCA) and clinical impact curves (CIC) have demonstrated that nomograms are of clinical utility. CONCLUSION The nomograms performed well in predicting the likelihood of stage I of ypTNM and TRG grade 0-1 after NT in patients with locally advanced EC. It helps thoracic surgeons to predict the sensitivity of patients to NT before surgery, which enables precise treatment of patients with locally advanced EC.
Collapse
Affiliation(s)
- Jianhao Qiu
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhan Zhang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Junjie Liu
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yue Zhao
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yongmeng Li
- Department of Thoracic Surgery, Qianfoshan Hospital in the Shandong Province, Jinan, Shandong, China
| | - Zhanpeng Tang
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lin Li
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yu Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
20
|
Chen KN. ASO Author Reflections: The Challenging Journey from cCR to pCR After Neoadjuvant Treatment for Esophageal Squamous Cell Carcinoma. Ann Surg Oncol 2024; 31:4317-4320. [PMID: 38366184 DOI: 10.1245/s10434-024-15035-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/18/2024]
Affiliation(s)
- Ke-Neng Chen
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Thoracic Surgery of Department, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
21
|
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
|
22
|
Lv H, Zhang F, Huang C, Xu S, Li J, Sun B, Gai C, Liu Z, Wang M, Li Z, Tian Z. Survival outcomes of neoadjuvant immunochemotherapy versus chemotherapy for locally advanced esophageal squamous cell carcinoma. J Cancer Res Clin Oncol 2024; 150:260. [PMID: 38760614 PMCID: PMC11101546 DOI: 10.1007/s00432-024-05793-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024]
Abstract
PURPOSE Neoadjuvant chemotherapy (NCT) is the standard preoperative treatment for resectable locally advanced esophageal squamous cell carcinoma (ESCC). Some studies reported neoadjuvant immunochemotherapy (NICT) could improve pathological response with manageable safety. However, few studies have compared the efficacy and safety of NICT and NCT, especially survival outcomes. In this study, we compared the efficacy and safety of NICT and NCT after a median follow-up of 36.0 months. METHODS This was a retrospective study with a 1:1 propensity score matching (PSM). Locally advanced ESCC patients treated with neoadjuvant sintilimab plus chemotherapy or chemotherapy followed by esophagectomy were reviewed. The primary outcome was recurrence-free survival (RFS). RESULTS Forty-five patients were identified in each group by PSM. The pathological complete response (pCR) rate in NICT and NCT group were 28.9% and 8.9% (P = 0.02). The hazard ratio (HR) was 0.396 (95% CI 0.171-0.919, p = 0.025) for RFS and 0.377 (95% CI 0.145-0.981, p = 0.038) for overall survival (OS), 3-year RFS was 80.6% and 62.1%, 3-year OS was 86.2% and 68.1%. Patients with pCR, MPR or downstaging had better 3-year RFS and 3-year OS. The incidences of postoperative complications and treatment-related adverse events (TRAEs) were similar. CONCLUSION This trial preliminarily shows that NICT improves pathological and survival outcomes over NCT for resectable locally advanced ESCC, with acceptable and manageable safety.
Collapse
Affiliation(s)
- Huilai Lv
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, NO.12, JianKang Road, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Accurate Diagnosis and Comprehensive Treatment of Esophageal Cancer, Shijiazhuang, Hebei, China
| | - Fan Zhang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, NO.12, JianKang Road, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Accurate Diagnosis and Comprehensive Treatment of Esophageal Cancer, Shijiazhuang, Hebei, China
| | - Chao Huang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, NO.12, JianKang Road, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Accurate Diagnosis and Comprehensive Treatment of Esophageal Cancer, Shijiazhuang, Hebei, China
| | - Shi Xu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, NO.12, JianKang Road, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Accurate Diagnosis and Comprehensive Treatment of Esophageal Cancer, Shijiazhuang, Hebei, China
| | - Jiachen Li
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, NO.12, JianKang Road, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Accurate Diagnosis and Comprehensive Treatment of Esophageal Cancer, Shijiazhuang, Hebei, China
| | - Bokang Sun
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, NO.12, JianKang Road, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Accurate Diagnosis and Comprehensive Treatment of Esophageal Cancer, Shijiazhuang, Hebei, China
| | - Chunyue Gai
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, NO.12, JianKang Road, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Accurate Diagnosis and Comprehensive Treatment of Esophageal Cancer, Shijiazhuang, Hebei, China
| | - Zhao Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, NO.12, JianKang Road, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Accurate Diagnosis and Comprehensive Treatment of Esophageal Cancer, Shijiazhuang, Hebei, China
| | - Mingbo Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, NO.12, JianKang Road, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Accurate Diagnosis and Comprehensive Treatment of Esophageal Cancer, Shijiazhuang, Hebei, China
| | - Zhenhua Li
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, NO.12, JianKang Road, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Accurate Diagnosis and Comprehensive Treatment of Esophageal Cancer, Shijiazhuang, Hebei, China
| | - Ziqiang Tian
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, NO.12, JianKang Road, Shijiazhuang, Hebei, China.
- Hebei Key Laboratory of Accurate Diagnosis and Comprehensive Treatment of Esophageal Cancer, Shijiazhuang, Hebei, China.
| |
Collapse
|
23
|
Egebjerg K, Andersen TS, Bæksgaard L, Garbyal R, Siemsen M, Achiam M, Mau-Sørensen PM. Implementation of perioperative FLOT compared to ECX/EOX chemotherapy regimens in resectable esophagogastric adenocarcinomas: an analysis of real-world data. Acta Oncol 2024; 63:322-329. [PMID: 38745482 PMCID: PMC11332447 DOI: 10.2340/1651-226x.2024.35431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/16/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND AND PURPOSE Perioperative 5-FU, leucovorin, oxaliplatin, and docetaxel (FLOT) is recommended in resectable esophagogastric adenocarcinoma based on randomised trials. However, the effectiveness of FLOT in routine clinical practice remains unknown as randomised trials are subject to selection bias limiting their generalisability. The aim of this study was to evaluate the implementation of FLOT in real-world patients. METHODS Retrospectively collected data were analysed in consecutive patients treated before or after the implementation of FLOT. The primary endpoint was complete pathological response (pCR) and secondary endpoints were margin-free resection (R0), overall survival (OS), relapse-free survival (RFS) tolerability of chemotherapy and surgical complications. RESULTS Mean follow-up time for patients treated with FLOT (n = 205) was 37.7 versus 47.0 months for epirubicin, cis- or oxaliplatin, and capecitabine (ECX/EOX, n = 186). Surgical resection was performed in 88.0% versus 92.0%; pCR were observed in 3.8% versus 2.4%; and R0 resections were achieved in 78.0% versus 86.0% (p = 0.03) in the ECX/EOX and FLOT cohorts, respectively. Survival analysis indicated no significant difference in RFS (p = 0.17) or OS (p = 0.37) between the cohorts with a trend towards increased OS in performance status 0 (hazard ratio [HR] = 0.73, 95% confidence interval [CI]: 0.50-1.04). More patients treated with ECX/EOX completed chemotherapy (39% vs. 28%, p = 0.02). Febrile neutropenia was more common in the FLOT cohort (3.8% vs. 11%, p = 0.0086). 90-days mortality (1.2% vs. 0%) and frequency of anastomotic leakage (8% vs. 6%) were equal and low. INTERPRETATION Patients receiving FLOT did not demonstrate improved pCR, RFS or OS. However, R0 rate was improved and patients in good PS trended towards improved OS.
Collapse
Affiliation(s)
- Kristian Egebjerg
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
| | - Tobias Sørup Andersen
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Lene Bæksgaard
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark. k
| | - Rajendra Garbyal
- Department of Pathology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Mette Siemsen
- Department of Thoracic Surgery, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Michael Achiam
- Department of Surgery and Transplantation, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
24
|
Torii N, Miyata K, Fukaya M, Ebata T. Risk factors for venous thrombosis after esophagectomy. Esophagus 2024; 21:150-156. [PMID: 38214871 DOI: 10.1007/s10388-023-01038-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 12/08/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND Venous thrombosis (VT) after esophagectomy for esophageal cancer is an important complication, potentially leading to pulmonary embolism. However, there are few available information about the risk for the postsurgical VT. METHODS This study included 271 patients who underwent esophagectomy for esophageal cancer between 2006 and 2019. Contrast-enhanced computed tomography (CT) was performed for all patients on the seventh postoperative day to survey complications, including VT. RESULTS VT was radiologically visualized in 48 patients (17.7%), 8 of whom (16.7%) had pulmonary embolism. The thrombus disappeared in 42 patients, the thrombus size was unchanged in 5 patients, and 1 patient died. Multivariate analysis was performed on factors clinically considered to have a significant influence on thrombus formation. The analysis showed that CVC insertion via the femoral vein (odds ratio, 7.67; 95% CI, 2.64-22.27; P < 0.001), retrosternal reconstruction route (odds ratio, 3.94; 95% CI, 1.90-8.17; P < 0.001) and intraoperative fluid balance < 5 ml/kg/hr (odds ratio, 0.38; 95% CI, 0.17-0.85; P = 0.019) were independently related to VT. CONCLUSIONS Intraoperative fluid balance < 5 ml/kg/hr, along with CVC insertion via the femoral vein and retrosternal reconstruction may be potential risk factors for VT after esophagectomy.
Collapse
Affiliation(s)
- Naoya Torii
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kazushi Miyata
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Masahide Fukaya
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomoki Ebata
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
25
|
Kodia K, Alnajar A, Huerta CT, Gupta G, Giri B, Dosch A, Paluvoi N. Nationwide Outcomes After Neoadjuvant Chemotherapy for Locally Advanced Sigmoid Colon Cancer-A Propensity Score-Matched Analysis. Am Surg 2024; 90:866-874. [PMID: 37972411 DOI: 10.1177/00031348231216491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
BACKGROUND The role of neoadjuvant chemotherapy (NAC) in advanced sigmoid colon carcinoma remains to be further characterized. Rationale for NAC includes downstaging on final pathology and optimization of microscopically negative margins (R0 resection). We investigated rates of neoadjuvant chemotherapy use in advanced sigmoid colon cancer at academic cancer centers and assessed factors associated with likelihood of NAC administration. METHODS The National Cancer Database was queried from 2004 to 2017 for patients with clinical T3 or T4, N0-2, M0 sigmoid colon cancer who underwent surgical resection. Those with neoadjuvant radiation or metastatic disease were excluded. The outcomes of patients who did and did not receive neoadjuvant chemotherapy were evaluated for this retrospective cohort study. RESULTS There were 23,597 patients of whom 364 (1.5%) received NAC. More patients received NAC at academic (41%, P < .001) and high-volume centers (27%, P < .001). Patients with Medicare/Medicaid (39%) and private insurance (52%) were more likely to receive NAC (P < .001). There was a significantly higher rate of N2 to N1 downstaging in the NAC group. Propensity-score matching demonstrated comprehensive community cancer programs (CCCP) were less likely to provide NAC (OR 0.4; 95% CI 0.23, 0.70, P < .001). There was no difference in survival (P = .20), R0 resection (P = .090), or 30-day readmission rates (P = .30) in the NAC cohort compared to the non-NAC cohort. CONCLUSIONS Access to centers offering multi-disciplinary care with NAC prior to surgical resection is important. This care was associated with academic and high-volume centers and private or government-sponsored insurance. There was no difference in survival between NAC and non-NAC cohort.
Collapse
Affiliation(s)
- Karishma Kodia
- Division of Colon and Rectal Surgery, Department of Surgery, University of Miami Leonard Miller School of Medicine, Miami, FL, USA
| | - Ahmed Alnajar
- Division of Colon and Rectal Surgery, Department of Surgery, University of Miami Leonard Miller School of Medicine, Miami, FL, USA
| | - Carlos T Huerta
- Division of Colon and Rectal Surgery, Department of Surgery, University of Miami Leonard Miller School of Medicine, Miami, FL, USA
| | - Gaurav Gupta
- Division of Colon and Rectal Surgery, Department of Surgery, University of Miami Leonard Miller School of Medicine, Miami, FL, USA
| | - Bhuwan Giri
- Division of Colon and Rectal Surgery, Department of Surgery, University of Miami Leonard Miller School of Medicine, Miami, FL, USA
| | - Austin Dosch
- Division of Colon and Rectal Surgery, Department of Surgery, University of Miami Leonard Miller School of Medicine, Miami, FL, USA
| | - Nivedh Paluvoi
- Division of Colon and Rectal Surgery, Department of Surgery, University of Miami Leonard Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
26
|
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
|
27
|
Porschen R, Fischbach W, Gockel I, Hollerbach S, Hölscher A, Jansen PL, Miehlke S, Pech O, Stahl M, Vanhoefer U, Ebert MPA. Updated German guideline on diagnosis and treatment of squamous cell carcinoma and adenocarcinoma of the esophagus. United European Gastroenterol J 2024; 12:399-411. [PMID: 38284661 PMCID: PMC11017771 DOI: 10.1002/ueg2.12523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/21/2023] [Indexed: 01/30/2024] Open
Abstract
Diagnosis and therapy of esophageal carcinoma is challenging and requires a multidisciplinary approach. The purpose of the updated German guideline "Diagnosis and Treatment of Squamous Cell Carcinoma and Adenocarcinoma of the Esophagus-version 3.1" is to provide practical and evidence-based advice for the management of patients with esophageal cancer. Recommendations were developed by a multidisciplinary expert panel based on an extensive and systematic evaluation of the published medical literature and the application of well-established methodologies (e.g. Oxford evidence grading scheme, grading of recommendations). Accurate diagnostic evaluation of the primary tumor as well as lymph node and distant metastases is required in order to guide patients to a stage-appropriate therapy after the initial diagnosis of esophageal cancer. In high-grade intraepithelial neoplasia or mucosal carcinoma endoscopic resection shall be performed. Whether endoscopic resection is the definitive therapeutic measure depends on the histopathological evaluation of the resection specimen. Esophagectomy should be performed minimally invasive or in combination with open procedures (hybrid technique). Because the prognosis in locally advanced esophageal carcinoma is poor with surgery alone, multimodality therapy is recommended. In locally advanced adenocarcinomas of the esophagus or esophagogastric junction, perioperative chemotherapy or preoperative radiochemotherapy should be administered. In locally advanced squamous cell carcinomas of the esophagus, preoperative radiochemotherapy followed by complete resection or definitive radiochemotherapy without surgery should be performed. In the case of residual tumor in the resection specimen after neoadjuvant radiochemotherapy and R0 resection of squamous cell carcinoma or adenocarcinoma, adjuvant immunotherapy with nivolumab should be given. Systemic palliative treatment options (chemotherapy, chemotherapy plus immunotherapy, immunotherapy alone) in unresectable or metastastic esophageal cancer depend on histology and are stratified according to PD-L1 and/or Her2 expression.
Collapse
Affiliation(s)
- Rainer Porschen
- Gastroenterologische Praxis am Kreiskrankenhaus OsterholzOsterholz‐ScharmbeckGermany
| | - Wolfgang Fischbach
- Deutsche Gesellschaft zur Bekämpfung der Krankheiten von MagenDarm und Leber sowie von Störungen des Stoffwechsels und der Ernährung (Gastro‐Liga) e. V.GiessenGermany
| | - Ines Gockel
- Klinik für Viszeral‐, Transplantations‐, Thorax‐ und GefäßchirurgieLeipzigGermany
| | | | - Arnulf Hölscher
- Contilia Zentrum für SpeiseröhrenerkrankungenElisabeth Krankenhaus EssenEssenGermany
| | - Petra Lynen Jansen
- Deutsche Gesellschaft für GastroenterologieVerdauungs‐ und StoffwechselkrankheitenBerlinGermany
| | | | - Oliver Pech
- Klinik für Gastroenterologie und Interventionelle EndoskopieKrankenhaus Barmherzige BrüderRegensburgGermany
| | - Michael Stahl
- Klinik für Internistische Onkologie & Onkologische PalliativmedizinEvang. Kliniken Essen‐MitteEssenGermany
| | - Udo Vanhoefer
- Klinik für Hämatologie und OnkologieKath. MarienkrankenhausHamburgGermany
| | - Matthias P. A. Ebert
- Medizinische Fakultät MannheimII. Medizinische KlinikUniversitätsmedizinUniversität HeidelbergMannheimGermany
- DKFZ‐Hector Krebsinstitut an der Universitätsmedizin MannheimMannheimGermany
- Molecular Medicine Partnership UnitEMBLHeidelbergGermany
| |
Collapse
|
28
|
Park S, Lee Y, Lee J, Min YW, Kim HK, Choi JY, Jung HA, Choi YS, Choi YL, Shim YM, Sun JM. Neoadjuvant Nivolumab Therapy for Esophageal Squamous Cell Carcinoma: A Single-Arm, Phase II Study. Cancer Res Treat 2024; 56:567-579. [PMID: 37846467 PMCID: PMC11016664 DOI: 10.4143/crt.2023.897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/12/2023] [Indexed: 10/18/2023] Open
Abstract
PURPOSE Programmed death-1/programmed death-ligand 1 (PD-L1) inhibitors have shown efficacy in metastatic esophageal squamous cell carcinoma (ESCC) therapy. However, data is still limited regarding neoadjuvant immunotherapy for operable ESCC. MATERIALS AND METHODS Patients with clinical stage T2 or T3 and N0 ESCC received three cycles of nivolumab therapy every two weeks before surgical resection. The primary endpoint is major pathologic responses (MPR) rate (≤ 10% of residual viable tumor [RVT]). RESULTS Total 20 patients completed the planned nivolumab therapy. Among them, 17 patients underwent surgery as protocol, showing MPR in two patients (MPR rate, 11.8%), including one pathologic complete response, on conventional pathologic response evaluation. Pathologic response was re-evaluated using the immune-related pathologic response criteria based on immune-related RVT (irRVT). Three patients were classified as immunologic major pathologic response (iMPR; ≤ 10% irRVT, iMPR rate: 17.6%), five as pathologic partial response (> 10% and < 90% irRVT), and nine as pathologic nonresponse (≥ 90% irRVT). The combined positive score (CPS) for PD-L1 in the baseline samples was predictable for iMPR, with the probability as 37.5% in CPS ≥ 10 (3/8) and 0% in CPS < 10 (0/9). CONCLUSION Although the efficacy of neoadjuvant nivolumab therapy was modest in unselected ESCC patients, further researches on neoadjuvant immunotherapy are necessary in patients with PD-L1 expressed ESCC.
Collapse
Affiliation(s)
- Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yurimi Lee
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jiyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yang Won Min
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Kwan Kim
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joon Young Choi
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyun Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yong Soo Choi
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Mog Shim
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
29
|
Tankel J, Ahmed N, Mueller C, Najmeh S, Spicer J, Mulder D, Cool-Lartigue J, Rousseau M, Frechette D, Sud S, Kavan P, Moghrabi A, Champagne M, Lemay F, Dalfen R, Sirhan S, Asselah J, Alcindor T, Ferri L. Docetaxel-Based Neoadjuvant Chemotherapy Followed by En Bloc Resection for Esophageal Adenocarcinoma: A 15-Year Retrospective Analysis from a Regional Upper Gastrointestinal Cancer Network. Ann Surg Oncol 2024; 31:2461-2469. [PMID: 38142255 DOI: 10.1245/s10434-023-14779-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/30/2023] [Indexed: 12/25/2023]
Abstract
BACKROUND Real-world, long-term survival outcomes of neoadjuvant, docetaxel-based therapy for esophageal and junctional adenocarcinoma are lacking. This study describes the long-term survival outcomes of patients with esophageal and junctional adenocarcinoma treated with neoadjuvant docetaxel-based chemotherapy and en bloc transthoracic esophagectomy. METHODS A retrospective cohort analysis of a prospectively maintained database from a regional upper gastrointestinal cancer network in Quebec, Canada, was performed. From January 2007 to December 2021, all patients with locally advanced (cT3 and/or N1) esophageal/Siewert I/II adenocarcinoma treated with neoadjuvant DCFx3 (Docetaxel/Cisplatin/5FU) or FLOTx4 (5FU/Leucovorin/Oxaliplatin/Docetaxel) and transthoracic en bloc esophagectomy were identified. Postoperative, pathological, and survival outcomes were compared. RESULTS Overall, 236 of 420 patients met the inclusion criteria. Tumor location was esophageal/Siewert I/Siewert II (118/33/85), most were cT3-4 (93.6%) and cN+ (61.0%). DCF and FLOT were used in 127 of 236 (53.8%) and 109 of 236 (46.2%). All neoadjuvant cycles were completed in 87.3% with no difference between the regimens. Operative procedures included Ivor Lewis (81.8%), left thoraco-abdominal esophagectomy (10.6%) and McKeown (7.6%) with an R0 resection in 95.3% and pathological complete response in 9.7% (DCF 12.6%/FLOT 6.4%, p = 0.111). The median lymph node yield was 32 (range 4-79), and 60.6% were ypN+. Median follow-up was longer for the DCF group (74.8 months 95% confidence interval [CI] 4-173 vs. 37.8 months 95% CI 2-119, p <0.001. Overall survival was similar between the groups (FLOT 97.3 months, 78.6-115.8 vs. DCF 92.9, 9.2-106.5, p = 0.420). CONCLUSIONS Neoadjuvant DCF and FLOT followed by transthoracic en bloc resection are both highly effective regimens for locally advanced esophageal adenocarcinoma with equivalent survival outcomes despite high disease load.
Collapse
Affiliation(s)
- James Tankel
- Division of Thoracic and Upper Gastrointestinal Surgery, Montreal General Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Nabeel Ahmed
- Division of Thoracic and Upper Gastrointestinal Surgery, Montreal General Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Carmen Mueller
- Division of Thoracic and Upper Gastrointestinal Surgery, Montreal General Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Sarah Najmeh
- Division of Thoracic and Upper Gastrointestinal Surgery, Montreal General Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Jonathan Spicer
- Division of Thoracic and Upper Gastrointestinal Surgery, Montreal General Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - David Mulder
- Division of Thoracic and Upper Gastrointestinal Surgery, Montreal General Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Jonathan Cool-Lartigue
- Division of Thoracic and Upper Gastrointestinal Surgery, Montreal General Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Mathieu Rousseau
- Division of Thoracic and Upper Gastrointestinal Surgery, Montreal General Hospital, McGill University Health Centre, Montreal, QC, Canada
| | | | - Shelly Sud
- Department of Medical Oncology, Hopital de Gatineau, Gatineau, QC, Canada
| | - Petr Kavan
- Deparment of Oncology, Jewish General Hospital, Montreal, QC, Canada
| | - Albert Moghrabi
- Department of Oncology, Hopital de Verdun, Montreal, QC, Canada
| | | | - Frederic Lemay
- Deparmtent of Medicine, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Richard Dalfen
- Division of Medical Oncology, St Mary's Hospital Center, Montreal, QC, Canada
| | - Shireen Sirhan
- Department of Oncology, Brome-Missisquoi-Perkins, Cowansville, QC, Canada
| | - Jamil Asselah
- Division of Medical Oncology, Royal Victoria Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - Thierry Alcindor
- Division of Medical Oncology, Royal Victoria Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - Lorenzo Ferri
- Division of Thoracic and Upper Gastrointestinal Surgery, Montreal General Hospital, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
30
|
Ye YC, Wang YP, Chang TE, Wu PS, Hsin IF, Chen PH, Tai SK, Chu PY, Hou MC, Lu CL. Routine image-enhanced endoscopic surveillance for metachronous esophageal squamous cell neoplasms in head and neck cancer patients. Esophagus 2024; 21:131-140. [PMID: 38194013 DOI: 10.1007/s10388-023-01039-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 12/08/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Esophageal squamous cell neoplasms (ESCNs) are common second primary tumors in patients with head and neck cancer. Image-enhanced endoscopy (IEE) with Lugol chromoendoscopy or magnifying narrow-band imaging both increase the detection of early ESCNs. No evidence-based ESCN surveillance program for head and neck cancer patients without a history of synchronous ESCNs exists. We aimed to evaluate the performance of an IEE surveillance program with magnifying narrow-band imaging endoscopy and Lugol chromoendoscopy. METHODS From April 2016, we routinely used IEE with magnifying narrow-band imaging and Lugol chromoendoscopy to evaluate patients with head and neck cancer history. All patients who were negative for ESCNs at the first surveillance endoscopy and received at least 2 IEEs through December 2019 were included. Demographic profiles, clinical data, cancer characteristics, IEE results and pathology reports were analyzed. RESULTS A total of 178 patients were included. Only 4 patients (2.2%) developed metachronous ESCNs during follow-up, all of whom received curative resection treatment. The interval for the development of metachronous ESCNs was 477 to 717 days. In multivariate Firth logistic regression and Kaplan‒Meier survival curve analysis, Lugol's voiding lesion type C had an increased risk of esophageal cancer development (adjusted odds ratio = 15.71; 95% confidence interval, 1.33-185.87, p = 0.029). Eight patients died during the study period, and none of them had metachronous ESCNs. CONCLUSIONS IEE with magnifying narrow-band imaging and Lugol chromoendoscopy is an effective surveillance program in head and neck cancer patients without a history of ESCNs. Annual surveillance can timely detect early ESCNs with low ESCN-related mortality.
Collapse
Affiliation(s)
- Yong-Cheng Ye
- Endoscopy Center For Diagnosis and Treatment, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou District, Taipei, 11217, Taiwan
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-Po Wang
- Endoscopy Center For Diagnosis and Treatment, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou District, Taipei, 11217, Taiwan.
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
- Institute of Brain Science, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Tien-En Chang
- Endoscopy Center For Diagnosis and Treatment, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou District, Taipei, 11217, Taiwan
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pei-Shan Wu
- Endoscopy Center For Diagnosis and Treatment, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou District, Taipei, 11217, Taiwan
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - I-Fang Hsin
- Endoscopy Center For Diagnosis and Treatment, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou District, Taipei, 11217, Taiwan
| | - Ping-Hsien Chen
- Endoscopy Center For Diagnosis and Treatment, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou District, Taipei, 11217, Taiwan
- Division of Gastroenterology, Department of Medicine, West Garden Hospital, Taipei, Taiwan
| | - Shyh-Kuan Tai
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Otolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Pen-Yuan Chu
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Otolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Chih Hou
- Endoscopy Center For Diagnosis and Treatment, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou District, Taipei, 11217, Taiwan
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ching-Liang Lu
- Endoscopy Center For Diagnosis and Treatment, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou District, Taipei, 11217, Taiwan
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Brain Science, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
31
|
Yang Y, Zhang J, Meng H, Ling X, Wang X, Xin Y, Jiang H, Zhang L, Fang C, Liang H, Ma J, Zhu J. Neoadjuvant camrelizumab combined with paclitaxel and nedaplatin for locally advanced esophageal squamous cell carcinoma: a single-arm phase 2 study (cohort study). Int J Surg 2024; 110:1430-1440. [PMID: 38051925 PMCID: PMC10942145 DOI: 10.1097/js9.0000000000000978] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/20/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND Neoadjuvant administration of immune checkpoint inhibitors (ICIs) combined with chemotherapy demonstrated promising efficacy and manageable safety in locally advanced esophageal squamous cell carcinoma (ESCC). This prospective, single-arm, phase 2 study evaluated the efficacy and safety of neoadjuvant therapy with camrelizumab plus paclitaxel and nedaplatin for 2-4 cycles in ESCC. METHODS Patients with locally advanced stage IIa-IIIb ESCC were enrolled in the study and received camrelizumab (200 mg), paclitaxel (155 mg/m 2 ), and nedaplatin (80 mg/m 2 ) intravenously on day one every 3 weeks. Patients underwent surgery after 2-4 cycles of treatment. The primary endpoint was the pathological complete response (pCR) rate. Secondary endpoints included the major pathological response (MPR) rate, R0 resection rate, tumor regression, objective response rate (ORR), and disease-free survival (DFS). Programmed cell death 1 ligand 1 (PD-L1) expression in tumor tissues was measured and quantified using immunohistochemistry staining and combined positive score (CPS), respectively. RESULTS In total, 75 patients were enrolled and received neoadjuvant treatment. Of them, 45 (60%) received two cycles, 18 (24%) received three cycles, and 10 patients (13.3%) received four cycles of neoadjuvant therapy. Ultimately, 62 patients (82.7%) underwent surgery. The patients achieved a pCR of 27.4% (95% CI: 16.9-40.2), an MPR of 45.2% (95% CI: 33.1-59.2), and an ORR of 48.4% (95% CI: 35.5-61.4); all patients had an R0 resection. T and N downstaging occurred in 39 (62.9%) and 19 (30.6%) patients Moreover, patients with CPS ≥10 tended to have enhanced ORR, pCR, and MPR compared to those with CPS <10. Treatment-related adverse events (TRAEs) of grade 1-2 occurred in 59 (78.7%) patients, grade 3 TRAEs in four (5.3%), and one patient (1.3%) experienced a grade 4 TRAE. CONCLUSIONS Neoadjuvant camrelizumab combined with chemotherapy showed promising efficacy in locally advanced ESCC, with a manageable safety profile, when administered flexibly in two to four cycles.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jinhong Zhu
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| |
Collapse
|
32
|
Liang Y, Maeda O, Miyata K, Kanda M, Sugita S, Shimizu D, Nishida K, Kodera Y, Ando Y. Genetic polymorphisms as predictive biomarkers of adverse events during preoperative chemotherapy in esophageal cancer. Cancer Chemother Pharmacol 2024; 93:121-127. [PMID: 37898586 DOI: 10.1007/s00280-023-04607-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/14/2023] [Indexed: 10/30/2023]
Abstract
PURPOSE This study aimed to explore associations between genetic polymorphisms and adverse effects due to preoperative chemotherapy with docetaxel, cisplatin, and fluorouracil (DCF) for esophageal cancer. METHODS Preoperative DCF (docetaxel, 70 mg/m2/day, day 1; cisplatin, 70 mg/m2/day, day 1; fluorouracil, 750 mg/m2/day, days 1-5) was repeated every 3 weeks for up to three cycles. Genotyping of nine candidate genetic polymorphisms was conducted using blood samples from the enrolled patients. RESULTS According to a multivariable analysis evaluating 50 patients, grade 3 or worse neutropenia was more likely to occur in those with the ABCC2-24C/T or T/T genotype (rs717620) (OR, 5.30, P = 0.013). Additionally, patients with the TYMS 3'-UTR 0 bp/0 bp genotype (rs151264360) showed a trend toward grade 3 or worse hyponatremia (OR, 0.16, P = 0.005). Grade 2 or worse thrombocytopenia was more likely to occur in patients with the TNF-α-1031C/T or T/T genotype (rs1799964) (OR, 6.30, P = 0.016) and IL-6-634C/C genotype (rs1800796) (OR, 0.18, P = 0.034), and grade 2 or worse anemia was more likely to occur in patients with the MCP-1-2518G/G genotype (rs1024611) (OR, 0.19, P = 0.027). CONCLUSIONS ABCC2-24C > T (rs717620), TYMS 3'-UTR 6-bp indel (rs151264360), TNF-α-1031T > C (rs1799964) as well as IL-6-634G > C (rs1800796), and MCP-1-2518A > G (rs1024611) polymorphisms might serve as independent and predictive biomarkers for neutropenia, hyponatremia, thrombocytopenia, and anemia, respectively, during preoperative chemotherapy with docetaxel, cisplatin, and fluorouracil for patients with esophageal cancer.
Collapse
Affiliation(s)
- Yao Liang
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| | - Osamu Maeda
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan.
| | - Kazushi Miyata
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shizuki Sugita
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Dai Shimizu
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuki Nishida
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichi Ando
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
33
|
Tian Y, Shi Z, Wang C, Ke S, Qiu H, Zhao W, Wu Y, Chen J, Zhang Y, Chen Y. A Comparison of Clinicopathologic Outcomes and Patterns of Lymphatic Spread Across Neoadjuvant Chemotherapy, Neoadjuvant Chemoradiotherapy, and Neoadjuvant Immunochemotherapy in Locally Advanced Esophageal Squamous Cell Carcinoma. Ann Surg Oncol 2024; 31:860-871. [PMID: 37947979 DOI: 10.1245/s10434-023-14534-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (NCRT) is recommended as the treatment standard for locally advanced esophageal squamous cell carcinoma (ESCC). The use of immunotherapy in the neoadjuvant setting has gained attention. Multiple, clinical trials have explored the efficacy and safety of neoadjuvant immunochemotherapy (NICT). We evaluated the differences in clinicopathologic outcomes and the patterns of lymphatic spread among patients receiving neoadjuvant chemotherapy (NCT), NCRT, and NICT before esophagectomy for locally advanced ESCC. METHODS A total of 702 patients with ESCC who completed transthoracic esophagectomy followed neoadjuvant therapy were included. Pathological characteristics, including pathologic complete response (pCR), tumor regression grade (TRG) score and patterns of lymphatic spread, were evaluated. RESULTS Compared with the NCT group, the NCRT group and NICT group had an advantage in pathological response (P < 0.05). The pCR rate was 8.1% in the NCT group, 29.9% in the NCRT group, and 23.6% in the NICT group. The TRG score (P < 0.05) and pathologic T stage (P < 0.05) in the NCT group were significantly higher. Compared with NICT, NCRT can significantly reduce the rate of lymph node metastasis rate in station 1R (0 vs. 3.4%, P < 0.05) and 2R (1.1% vs. 6.8%, P < 0.05). Subgroup analysis according to the tumor location distribution showed that NICT group had higher lymph node metastasis rate in station 2R (9.1%) in middle thoracic cases (P < 0.05) and in station 18 (7.5%) (P < 0.05) in lower thoracic cases. CONCLUSIONS NCRT or NICT followed by surgery may result in a promising pCR rate and show a better performance in therapeutic response of primary lesion. For patients with lymph node metastasis in station 1R and 2R, NCRT should be the optimal preoperative treatment strategy.
Collapse
Affiliation(s)
- Yuanyuan Tian
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhenguo Shi
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Oncology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Chenyu Wang
- Department of Thoracic Oncology, Anyang Tumor Hospital, Henan Medical key Laboratory of Precise Prevention and Treatment of Esophageal Cancer, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, China
| | - Shaobo Ke
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hu Qiu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wensi Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yong Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiamei Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yaowen Zhang
- Department of Thoracic Oncology, Anyang Tumor Hospital, Henan Medical key Laboratory of Precise Prevention and Treatment of Esophageal Cancer, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang, China.
| | - Yongshun Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
34
|
Hilton CB, Lander S, Gibson MK. An Ailment with Which I Will Contend: A Narrative Review of 5000 Years of Esophagogastric Cancers and Their Treatments, with Special Emphasis on Recent Advances in Immunotherapeutics. Cancers (Basel) 2024; 16:618. [PMID: 38339368 PMCID: PMC10854527 DOI: 10.3390/cancers16030618] [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: 11/22/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Esophagogastric cancers are among the most common and deadly cancers worldwide. This review traces their chronology from 3000 BCE to the present. The first several thousand years were devoted to palliation, before advances in operative technique and technology led to the first curative surgery in 1913. Systemic therapies were introduced in 1910, and radiotherapy shortly thereafter. Operative technique improved massively over the 20th century, with operative mortality rates reducing from over 50% in 1933 to less than 5% by 1981. In addition to important roles in palliation, endoscopy became a key nonsurgical curative option for patients with limited-stage disease by the 1990s. The first nonrandomized studies on combination therapies (chemotherapy ± radiation ± surgery) were reported in the early 1980s, with survival benefit only for subsets of patients. Randomized trials over the next decades had similar overall results, with increasing nuance. Disparate conclusions led to regional variation in global practice. Starting with the first FDA approval in 2017, multiple immunotherapies now encompass more indications and earlier lines of therapy. As standards of care incorporate these effective yet expensive therapies, care must be given to disparities and methods for increasing access.
Collapse
Affiliation(s)
- C. Beau Hilton
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, 2220 Pierce Ave, Nashville, TN 37232, USA
| | - Steven Lander
- Internal Medicine Residency Program, University of Tennessee Health Sciences Center, 920 Madison Ave, Suite 531, Memphis, TN 38163, USA;
| | - Michael K. Gibson
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, 2220 Pierce Ave, Nashville, TN 37232, USA
| |
Collapse
|
35
|
Tan XZ, Ma R, Liu P, Xiao CH, Zhang HH, Yang F, Liang CH, Liu ZY. Decoding tumor stage by peritumoral and intratumoral radiomics in resectable esophageal squamous cell carcinoma. Abdom Radiol (NY) 2024; 49:301-311. [PMID: 37831168 PMCID: PMC10789665 DOI: 10.1007/s00261-023-04061-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/10/2023] [Accepted: 09/11/2023] [Indexed: 10/14/2023]
Abstract
PURPOSE To evaluate the potential application of radiomics in predicting Tumor-Node-Metastasis (TNM) stage in patients with resectable esophageal squamous cell carcinoma (ESCC). METHODS This retrospective study included 122 consecutive patients (mean age, 57 years; 27 women). Corresponding tumor of interest was identified on axial arterial-phase CT images with manual annotation. Radiomics features were extracted from intra- and peritumoral regions. Features were pruned to train LASSO regression model with 93 patients to construct a radiomics signature, whose performance was validated in a test set of 29 patients. Prognostic value of radiomics-predicted TNM stage was estimated by survival analysis in the entire cohort. RESULTS The radiomics signature incorporating one intratumoral and four peritumoral features was significantly associated with TNM stage. This signature discriminated tumor stage with an area under curve (AUC) of 0.823 in the training set, with similar performance in the test set (AUC 0.813). Recurrence-free survival (RFS) was significantly different between different radiomics-predicted TNM stage groups (Low-risk vs high-risk, log-rank P = 0.004). Univariate and multivariate Cox regression analyses revealed that radiomics-predicted TNM stage was an independent preoperative factor for RFS. CONCLUSIONS The proposed radiomics signature combing intratumoral and peritumoral features was predictive of TNM stage and associated with prognostication in ESCC.
Collapse
Affiliation(s)
- Xian-Zheng Tan
- Department of Radiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China.
| | - Rong Ma
- Department of Radiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Peng Liu
- Department of Radiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Chang-Hui Xiao
- Department of Radiology, The First People's Hospital of Changde City, Changde, 415000, Hunan, China
| | - Hui-Hui Zhang
- Department of Radiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Fan Yang
- Department of Radiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Chang-Hong Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510180, Guangdong, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510180, Guangdong, China.
| | - Zai-Yi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510180, Guangdong, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510180, Guangdong, China.
| |
Collapse
|
36
|
Qian T, Liu D, Cao G, Chen Z, Zhang Q. Neoadjuvant PD-1 Plus Chemotherapy for Locally Advanced Esophageal Squamous Cell Carcinoma. Technol Cancer Res Treat 2024; 23:15330338241231610. [PMID: 38497137 PMCID: PMC10946079 DOI: 10.1177/15330338241231610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/12/2024] [Accepted: 01/10/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND Clinical studies have shown that programmed cell death-1 (PD-1) inhibitors can activate T cells and inhibit cancer growth. Therefore, the use of a PD-1 inhibitor plus chemotherapy as neoadjuvant chemotherapy for locally advanced esophageal cancer is worth further exploration. METHODS Patients with locally advanced esophageal squamous cell carcinoma were enrolled in this study to receive two cycles of a preoperative combination of toripalimab, paclitaxel, and cisplatin. Efficacy was evaluated after two treatment cycles. The patients' postoperative pathological staging was analyzed and compared. Surgery was performed within 42 days of the start date of the last chemotherapy cycle. RESULTS Neoadjuvant immunochemotherapy achieved a high pathologic complete response (pCR) rate (29.0%), major pathological response rate (41.9%), and objective response rate (80.6%) and demonstrated statistically significant downstaging after neoadjuvant therapy (P < .05) with manageable treatment-related adverse effects. No significant association was found between PD-L1 level and pCR (P = .365). In addition, R0 resection was achieved in all 31 (100%) patients during surgery. For all the included patients, the one-year progression-free survival rate was 87.1% (95% CI: 75.3%-98.9%), the one-year overall survival (OS) rate was 96.8% (95% CI: 79.8%-95.9%), and the two-year OS rate was 83.9% (95% CI: 71.6%-92.2%). CONCLUSIONS Our findings indicate that this combination may be a potential neoadjuvant therapy regimen in this setting.
Collapse
Affiliation(s)
- Ting Qian
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Delin Liu
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Guochun Cao
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Zhipeng Chen
- Department of Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Qin Zhang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Cancer Institute of Jiangsu Province, Nanjing, China
| |
Collapse
|
37
|
Okamura A, Watanabe M, Okui J, Matsuda S, Takemura R, Kawakubo H, Kakeji Y, Kono K, Kitagawa Y, Takeuchi H. Outcomes of patients with esophageal squamous cell carcinoma who achieved a pathological complete response in the primary lesion by neoadjuvant treatment: a Japanese nationwide cohort study. Esophagus 2024; 21:2-10. [PMID: 37999900 DOI: 10.1007/s10388-023-01030-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Minimal data was reported regarding the characteristics, risks of lymph node metastasis, and prognostic factors in esophageal cancer patients who achieved remarkable response in the primary lesion to neoadjuvant treatment (NAT). METHODS This study evaluated the nationwide data of esophageal squamous cell carcinoma (ESCC) patients who underwent surgery following NAT in Japan. Of 4484 patients, 300 (6.7%) had ypT0 following NAT and curative esophagectomy. Factors associated with lymph node metastasis and prognosis were analyzed. RESULTS Neoadjuvant chemotherapy (NAC) and neoadjuvant chemoradiotherapy (NACRT) were administered in 260 (86.2%) and 40 (13.8%) patients, respectively. Pathologically, 72 (24.0%) had lymph node metastasis (residual nodal disease; RND), and pretherapeutic lymph node metastasis was the independent risk factor for RND (odd ratio [OR]: 3.21; 95% confidence interval [CI]: 1.44-8.20; P = 0.008). The 5-year overall and relapse-free survivals were significantly longer in patients with pathological complete response (pCR) than in those with RND (both P < 0.001). Pretherapeutic cT3 or T4a tumors (hazard ratio [HR]: 1.71; 95% CI: 1.02-2.88; P = 0.043), RND (HR: 3.30; 95% CI: 1.98-5.50; P < 0.001), and operative blood loss (Liter, HR: 1.53; 95% CI: 1.07-2.19; P = 0.021) were independent risk factors affecting relapse-free survival in multivariable analysis. CONCLUSIONS Of patients with ypT0 after NAT, 24.0% had RND, and pretherapeutic lymph node metastasis was the risk factor. In addition, pretherapeutic cT3, or T4a tumors, RND, and operative blood loss were the poor prognosticators in patients with ypT0 after NAT.
Collapse
Affiliation(s)
- Akihiko Okamura
- Department of Esophageal Surgery, Gastroenterology Center, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Masayuki Watanabe
- Department of Esophageal Surgery, Gastroenterology Center, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| | - Jun Okui
- Department of Preventive Medicine and Public Health, School of Medicine, Keio University, Tokyo, Japan
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Satoru Matsuda
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Ryo Takemura
- Biostatistics Unit, Clinical and Translational Research Center, Keio University Hospital, Tokyo, Japan
| | - Hirofumi Kawakubo
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Yoshihiro Kakeji
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Hyogo, Japan
| | - Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan
| | - Yuko Kitagawa
- Department of Surgery, School of Medicine, Keio University, Tokyo, Japan
| | - Hiroya Takeuchi
- Department of Surgery, Hamamatsu University School of Medicine, Shizuoka, Japan
| |
Collapse
|
38
|
Clements HA, Underwood TJ, Petty RD. Total neoadjuvant therapy in oesophageal and gastro-oesophageal junctional adenocarcinoma. Br J Cancer 2024; 130:9-18. [PMID: 37898721 PMCID: PMC10781745 DOI: 10.1038/s41416-023-02458-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 09/07/2023] [Accepted: 09/28/2023] [Indexed: 10/30/2023] Open
Abstract
Adenocarcinoma of the oesophagus and gastro-oesophageal junction represent a large burden of cancer death in the Western World with an increasing incidence. In the past two decades, the overall survival of patients on a potentially curative treatment pathway has more than doubled due to the addition of perioperative oncological therapies to surgery. However, patients often fail to respond to oncological treatment or struggle to complete their treatment after surgery. In this review, we discuss the current evidence for total neoadjuvant therapy and options for assessment of treatment response.
Collapse
Affiliation(s)
- Hollie A Clements
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK.
| | - Tim J Underwood
- School of Cancer Sciences, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Russell D Petty
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
- Tayside Cancer Centre, Ninewells Hospital and Medical School, NHS Tayside, Dundee, UK
| |
Collapse
|
39
|
Zeng H, Zhang F, Sun Y, Li S, Zhang W. Treatment options for neoadjuvant strategies of esophageal squamous cell carcinoma (Review). Mol Clin Oncol 2024; 20:4. [PMID: 38223404 PMCID: PMC10784769 DOI: 10.3892/mco.2023.2702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/26/2023] [Indexed: 01/16/2024] Open
Abstract
Compared with postoperative adjuvant therapy, neoadjuvant therapy has more potential advantages, such as decreasing tumor stage, killing micrometastatic cells. Because of these advantages, neoadjuvant therapy is recommended for numerous types of tumor, such as breast, lung and rectal cancer. To determine the role of neoadjuvant therapy on overall survival and adverse for patients with resectable esophageal carcinoma. we summarized clinical studies on 7 types of neoadjuvant therapies in this review. Currently, patients with esophageal cancer (EC) in China mainly receive postoperative treatment with <30% of patients receiving neoadjuvant therapy. One reason for the limited use of neoadjuvant therapy in China is inaccurate staging based on imaging and neoadjuvant treatment may increase difficulties in surgery. After neoadjuvant therapy, there may be tissue edema, blurry surgical field of view and unclear tissue gaps, resulting in greater difficulty in surgical procedures. However, oncologists are interested in neoadjuvant treatment, especially neoadjuvant immunotherapy to treat EC. Concurrent chemoradiotherapy for esophageal squamous cell carcinoma (ESCC) is the most common neoadjuvant treatment regimen and increases the pathological complete response (pCR) and 5- and 10-year survival rates. Preoperative induction chemotherapy and sequential concurrent chemoradiotherapy are currently the most widely treatments used in clinical practice in China. However, this treatment strategy does not yield long-term survival. The pCR rate of neoadjuvant immunotherapy is greater than that of concurrent chemoradiotherapy but, to the best of our knowledge, no evidence of long-term survival benefit has been found in phase I and II clinical trials. Neoadjuvant treatment should be considered for patients with locally advanced ESCC.
Collapse
Affiliation(s)
- Hai Zeng
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Fan Zhang
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Yujiao Sun
- Department of Clinical Medicine, Medical School of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Shuang Li
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Weijia Zhang
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
40
|
Wang YJ, Xie XF, He YQ, Bao T, He XD, Li KK, Guo W. Impact of perioperative decreased serum albumin level on anastomotic leakage in esophageal squamous cell carcinoma patients treated with neoadjuvant chemotherapy followed by minimally invasive esophagectomy. BMC Cancer 2023; 23:1212. [PMID: 38066484 PMCID: PMC10704756 DOI: 10.1186/s12885-023-11713-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 12/04/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Anastomotic leakage (AL) is a severe complication following esophagectomy with high mortality. Perioperative decreased serum albumin level is considered a predictive of AL, however, its impact on AL incidence in patients treated with neoadjuvant chemotherapy (NCT) followed by minimally invasive esophagectomy (MIE) is not well defined. METHODS The data of 318 consecutive esophageal cancer patients who underwent MIE were collected retrospectively from January 2021 to December 2021. The perioperative level of albumin was detected and the baseline of altering levels for albumin was established. The incidence of postoperative complications and survival rate were analyzed between groups. RESULTS After exclusion, 137 patients were enrolled and assigned to more decreased albumin (MA) and less decreased albumin (LA) groups. The levels of albumin descended significantly after MIE (p < 0.0001). There was no significant difference in the clinicopathologic characteristics or surgical outcomes between groups. The incidence of postoperative AL was 10.2% in MA group and 1.4% in LA group (p = 0.033). Three patients died due to AL in MA group, while no mortality was observed in LA group (p = 0.120). The rate of other postoperative complications was similar between groups. Progression-free survival (PFS) in LA group was a little higher than that in MA group, but it was no significant difference (p = 0.853). Similarly, no difference was observed in overall survival (OS) between groups (p = 0.277). CONCLUSIONS Severely deficient serum albumin after MIE was an indicator of AL in esophageal cancer patients treated with NCT. TRIAL REGISTRATION Chinese clinical trial registry: ChiCTR2200066694, registered December14th,2022. https://www.chictr.org.cn/edit.aspx?pid=185067&htm=4 .
Collapse
Affiliation(s)
- Ying-Jian Wang
- Department of Thoracic Surgery, Army Medical Center of PLA (Daping Hospital), Changjiang Route #10, Daping, Chongqing, 400042, PR China
| | - Xian-Feng Xie
- Department of Thoracic Surgery, Army Medical Center of PLA (Daping Hospital), Changjiang Route #10, Daping, Chongqing, 400042, PR China
| | - Yi-Qiu He
- Department of pediatrics, Shapingba District Maternity & Infant Health Hospital, Tiancheng Route #2, Shapingba, Choingqing, 401331, PR China
| | - Tao Bao
- Department of Thoracic Surgery, Army Medical Center of PLA (Daping Hospital), Changjiang Route #10, Daping, Chongqing, 400042, PR China
| | - Xian-Dong He
- Department of Thoracic Surgery, Army Medical Center of PLA (Daping Hospital), Changjiang Route #10, Daping, Chongqing, 400042, PR China
| | - Kun-Kun Li
- Department of Thoracic Surgery, Army Medical Center of PLA (Daping Hospital), Changjiang Route #10, Daping, Chongqing, 400042, PR China
| | - Wei Guo
- Department of Thoracic Surgery, Army Medical Center of PLA (Daping Hospital), Changjiang Route #10, Daping, Chongqing, 400042, PR China.
| |
Collapse
|
41
|
Thavanesan N, Bodala I, Walters Z, Ramchurn S, Underwood TJ, Vigneswaran G. Machine learning to predict curative multidisciplinary team treatment decisions in oesophageal cancer. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2023; 49:106986. [PMID: 37463827 DOI: 10.1016/j.ejso.2023.106986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/22/2023] [Accepted: 07/11/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND Rising workflow pressures within the oesophageal cancer (OC) multidisciplinary team (MDT) can lead to variability in decision-making, and health inequality. Machine learning (ML) offers a potential automated data-driven approach to address inconsistency and standardize care. The aim of this experimental pilot study was to develop ML models able to predict curative OC MDT treatment decisions and determine the relative importance of underlying decision-critical variables. METHODS Retrospective complete-case analysis of oesophagectomy patients ± neoadjuvant chemotherapy (NACT) or chemoradiotherapy (NACRT) between 2010 and 2020. Established ML algorithms (Multinomial Logistic regression (MLR), Random Forests (RF), Extreme Gradient Boosting (XGB)) and Decision Tree (DT) were used to train models predicting OC MDT treatment decisions: surgery (S), NACT + S or NACRT + S. Performance metrics included Area Under the Curve (AUC), Accuracy, Kappa, LogLoss, F1 and Precision -Recall AUC. Variable importance was calculated for each model. RESULTS We identified 399 cases with a male-to-female ratio of 3.6:1 and median age of 66.1yrs (range 32-83). MLR outperformed RF, XGB and DT across performance metrics (mean AUC of 0.793 [±0.045] vs 0.757 [±0.068], 0.740 [±0.042], and 0.709 [±0.021] respectively). Variable importance analysis identified age as a major factor in the decision to offer surgery alone or NACT + S across models (p < 0.05). CONCLUSIONS ML techniques can use limited feature-sets to predict curative UGI MDT treatment decisions. Explainable Artificial Intelligence methods provide insight into decision-critical variables, highlighting underlying subconscious biases in cancer care decision-making. Such models may allow prioritization of caseload, improve efficiency, and offer data-driven decision-assistance to MDTs in the future.
Collapse
Affiliation(s)
| | - Indu Bodala
- School of Electronics and Computer Science, University of Southampton, UK
| | - Zoë Walters
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, UK
| | - Sarvapali Ramchurn
- School of Electronics and Computer Science, University of Southampton, UK
| | - Timothy J Underwood
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, UK
| | - Ganesh Vigneswaran
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, UK
| |
Collapse
|
42
|
Yamada M, Tanaka K, Yamamoto K, Matsumoto H, Yamasaki M, Yamashita K, Makino T, Saito T, Yamamoto K, Takahashi T, Kurokawa Y, Nakajima K, Okada Y, Eguchi H, Doki Y. Association between circ_0004365 and cisplatin resistance in esophageal squamous cell carcinoma. Oncol Lett 2023; 26:467. [PMID: 37780544 PMCID: PMC10534278 DOI: 10.3892/ol.2023.14054] [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: 01/03/2023] [Accepted: 07/13/2023] [Indexed: 10/03/2023] Open
Abstract
Cisplatin is one of the most predominant drugs for the chemotherapy of esophageal squamous cell carcinoma (ESCC); however, the underlying resistance mechanisms are still almost unknown. The present study performed RNA sequencing of human circular RNA (circRNA) in TE11 cells and cisplatin-resistant TE11 cells (TE11R). The expression profiles determined using CIRCexplorer2 revealed that the expression of circ_0004365, mapped on the Semaphorin 3C gene, was significantly greater in TE11R compared with in TE11. In reverse transcription-quantitative PCR, circ_0004365 expression was observed in human ESCC and non-tumor tissues and was significantly upregulated in ESCC tumor tissues after chemotherapy. Circ_0004365 expression was significantly upregulated in patients with poor pathological response (P=0.02). Furthermore, patients with advanced pT stage showed an upregulation in circ_0004365 expression after chemotherapy (P=0.02). The MTT assay revealed that knockdown of circ_0003465 in TE11 significantly decreased resistance to cisplatin. In conclusion, the present study suggested that circ_0004365 was associated with cisplatin resistance in ESCC and can be used as both a novel biomarker and a therapeutic target.
Collapse
Affiliation(s)
- Moyuru Yamada
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Koji Tanaka
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Kenichi Yamamoto
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hisatake Matsumoto
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Makoto Yamasaki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Kotaro Yamashita
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Tomoki Makino
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Takuro Saito
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Kazuyoshi Yamamoto
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Kiyokazu Nakajima
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
43
|
Wu L, Gao Y, Xie S, Ye W, Uemura Y, Zhang R, Yu Y, Li J, Chen M, Wu Q, Cui P, Liu H, Mu S, Li Y, Wang L, Liu C, Li J, Zhang L, Jiao S, Zhang G, Liu T. The level of macrophage migration inhibitory factor is negatively correlated with the efficacy of PD-1 blockade immunotherapy combined with chemotherapy as a neoadjuvant therapy for esophageal squamous cell carcinoma. Transl Oncol 2023; 37:101775. [PMID: 37678132 PMCID: PMC10492201 DOI: 10.1016/j.tranon.2023.101775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 08/10/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023] Open
Abstract
PURPOSE This study aimed to screen biomarkers to predict the efficacy of programmed cell death 1 (PD-1) blockade immunotherapy combined with chemotherapy as neoadjuvant therapy for esophageal squamous cell carcinoma (ESCC). METHODS In the first stage of the study, the baseline concentrations of 40 tumor-related chemokines in the serum samples of 50 patients were measured to screen for possible biomarkers. We investigated whether the baseline concentration of the selected chemokine was related to the therapeutic outcomes and tumor microenvironment states of patients treated with the therapy. In the second stage, the reliability of the selected biomarkers was retested in 34 patients. RESULTS The baseline concentration of macrophage migration inhibitory factor (MIF) was negatively correlated with disease-free survival (DFS) and overall survival (OS) in patients treated with the therapy. In addition, a low baseline expression level of MIF is related to a better tumor microenvironment for the treatment of ESCC. A secondary finding was that effective treatment decreased the serum concentration of MIF. CONCLUSION Baseline MIF levels were negatively correlated with neoadjuvant therapy efficacy. Thus, MIF may serve as a predictive biomarker for this therapy. The accuracy of the prediction could be improved if the serum concentration of MIF is measured again after the patient received several weeks of treatment.
Collapse
Affiliation(s)
- Liangliang Wu
- Laboratory of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China; Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yiming Gao
- Department of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shengzhi Xie
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wan Ye
- Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yasushi Uemura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Rong Zhang
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Yanju Yu
- Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jinfeng Li
- Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Man Chen
- Department of Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, Lang Fang, China
| | - Qiyan Wu
- Laboratory of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Pengfei Cui
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongyu Liu
- Department of Neurosurgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan, China
| | - Shuai Mu
- Department of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yilan Li
- Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lingxiong Wang
- Laboratory of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chunxi Liu
- Laboratory of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jiahui Li
- Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lijun Zhang
- Laboratory of Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shunchang Jiao
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Guoqing Zhang
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Tianyi Liu
- Institute of Oncology, Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
44
|
Wakita A, Motoyama S, Sato Y, Nagaki Y, Fujita H, Kemuriyama K, Hayashi K, Imai K, Nanjo H, Minamiya Y. Preoperative neoadjuvant chemoradiotherapy provides borderline resectable thoracic esophageal cancer with equivalent treatment results as clinically T3 thoracic esophageal cancer. Ann Gastroenterol Surg 2023; 7:904-912. [PMID: 37927919 PMCID: PMC10623951 DOI: 10.1002/ags3.12706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/20/2023] [Accepted: 05/28/2023] [Indexed: 11/07/2023] Open
Abstract
Aim Because the optimal treatment strategy for borderline resectable (cT3br) thoracic esophageal cancer patients remains unclear, it is of great interest whether preoperative neoadjuvant therapy for cT3br could achieve results comparable to those seen with resectable T3 cancer (cT3r). We speculated that preoperative neoadjuvant chemoradiotherapy (NACRT) would be particularly effective in cT3br thoracic esophageal cancer patients and compared to cT3br and cT3r. Methods Of 186 cT3 thoracic esophageal cancer patients treated with intended NACRT, 162 received radical esophagectomy. More than 97% were squamous cell carcinomas. Patients were partitioned into two groups according to whether invasion of adjacent organs was suspected (cT3br and cT3r). Treatment outcomes and survival were analyzed. Results Sixty-eight patients (36.6%) were classified as cT3br and 118 (63.4%) as cT3r. The cT3br group had significantly more tumors in the upper and middle mediastinum (p < 0.0001) and more cases with cM1 (lymph node) (p = 0.0104) than the cT3r group. In addition, the cT3br patients receiving esophagectomy exhibited a significantly lower pathological complete response rate than the cT3r patients (p = 0.0374). However, the R0 resection rate did not differ between the cT3br and cT3r patients (p = 0.0978), and the two groups treated with intended NACRT had similar 5-year overall (OS) and disease-specific survival (DSS) (p = 0.3831 and p = 0.9020). In addition, the incidence and patterns of recurrence did not differ between the cT3br and cT3r patients receiving esophagectomy (p = 0.8109 and p = 0.3128). Conclusions Preoperative neoadjuvant chemoradiotherapy appears to be a promising treatment for patients with borderline resectable thoracic esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Akiyuki Wakita
- Division of Esophageal SurgeryAkita University HospitalAkitaJapan
- Department of Thoracic SurgeryAkita University Graduate School of MedicineAkitaJapan
| | - Satoru Motoyama
- Department of Gastroenterological SurgeryJapanese Red Cross Akita HospitalAkitaJapan
| | - Yusuke Sato
- Division of Esophageal SurgeryAkita University HospitalAkitaJapan
- Department of Thoracic SurgeryAkita University Graduate School of MedicineAkitaJapan
| | - Yushi Nagaki
- Division of Esophageal SurgeryAkita University HospitalAkitaJapan
- Department of Thoracic SurgeryAkita University Graduate School of MedicineAkitaJapan
| | - Hiromu Fujita
- Division of Esophageal SurgeryAkita University HospitalAkitaJapan
- Department of Thoracic SurgeryAkita University Graduate School of MedicineAkitaJapan
| | - Kohei Kemuriyama
- Division of Esophageal SurgeryAkita University HospitalAkitaJapan
- Department of Thoracic SurgeryAkita University Graduate School of MedicineAkitaJapan
| | - Kenjiro Hayashi
- Division of Esophageal SurgeryAkita University HospitalAkitaJapan
- Department of Thoracic SurgeryAkita University Graduate School of MedicineAkitaJapan
| | - Kazuhiro Imai
- Department of Thoracic SurgeryAkita University Graduate School of MedicineAkitaJapan
| | - Hiroshi Nanjo
- Division of Clinical PathologyAkita University HospitalAkitaJapan
| | - Yoshihiro Minamiya
- Division of Esophageal SurgeryAkita University HospitalAkitaJapan
- Department of Thoracic SurgeryAkita University Graduate School of MedicineAkitaJapan
| |
Collapse
|
45
|
Tong Y, Chen J, Sun J, Luo T, Duan S, Li K, Zhou K, Zeng J, Lu F. A radiomics nomogram for predicting postoperative recurrence in esophageal squamous cell carcinoma. Front Oncol 2023; 13:1162238. [PMID: 37901318 PMCID: PMC10602760 DOI: 10.3389/fonc.2023.1162238] [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: 02/09/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Purpose To establish and validate a radiomics nomogram for predicting recurrence of esophageal squamous cell carcinoma (ESCC) after esophagectomy with curative intent. Materials and methods The medical records of 155 patients who underwent surgical treatment for pathologically confirmed ESCC were collected. Patients were randomly divided into a training group (n=109) and a validation group (n=46) in a 7:3 ratio. Tumor regions are accurately segmented in computed tomography images of enrolled patients. Radiomic features were then extracted from the segmented tumors. We selected the features by Max-relevance and min-redundancy (mRMR) and least absolute shrinkage and selection operator (LASSO) methods. A radiomics signature was then built by logistic regression analysis. To improve predictive performance, a radiomics nomogram that incorporated the radiomics signature and independent clinical predictors was built. Model performance was evaluated by receiver operating characteristic (ROC) curve, calibration curve, and decision curve analyses (DCA). Results We selected the five most relevant radiomics features to construct the radiomics signature. The radiomics model had general discrimination ability with an area under the ROC curve (AUC) of 0.79 in the training set that was verified by an AUC of 0.76 in the validation set. The radiomics nomogram consisted of the radiomics signature, and N stage showed excellent predictive performance in the training and validation sets with AUCs of 0.85 and 0.83, respectively. Furthermore, calibration curves and the DCA analysis demonstrated good fit and clinical utility of the radiomics nomogram. Conclusion We successfully established and validated a prediction model that combined radiomics features and N stage, which can be used to predict four-year recurrence risk in patients with ESCC who undergo surgery.
Collapse
Affiliation(s)
- Yahan Tong
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Junyi Chen
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
| | - Jingjing Sun
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Taobo Luo
- Department of Thoracic Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shaofeng Duan
- GE Healthcare, Precision Health Institution, Shanghai, China
| | - Kai Li
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Kefeng Zhou
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jian Zeng
- Department of Thoracic Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Fangxiao Lu
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
46
|
Pang T, Nie M, Yin K. The correlation between the margin of resection and prognosis in esophagogastric junction adenocarcinoma. World J Surg Oncol 2023; 21:316. [PMID: 37814242 PMCID: PMC10561513 DOI: 10.1186/s12957-023-03202-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/30/2023] [Indexed: 10/11/2023] Open
Abstract
Adenocarcinoma of the gastroesophageal junction (AEG) has become increasingly common in Western and Asian populations. Surgical resection is the mainstay of treatment for AEG; however, determining the distance from the upper edge of the tumor to the esophageal margin (PM) is essential for accurate prognosis. Despite the relevance of these studies, most have been retrospective and vary widely in their conclusions. The PM is now widely accepted to have an impact on patient outcomes but can be masked by TNM at later stages. Extended PM is associated with improved outcomes, but the optimal PM is uncertain. Academics continue to debate the surgical route, extent of lymphadenectomy, preoperative tumor size assessment, intraoperative cryosection, neoadjuvant therapy, and other aspects to further ensure a negative margin in patients with gastroesophageal adenocarcinoma. This review summarizes and evaluates the findings from these studies and suggests that the choice of approach for patients with adenocarcinoma of the esophagogastric junction should take into account the extent of esophagectomy and lymphadenectomy. Although several guidelines and reviews recommend the routine use of intraoperative cryosections to evaluate surgical margins, its generalizability is limited. Furthermore, neoadjuvant chemotherapy and radiotherapy are more likely to increase the R0 resection rate. In particular, intraoperative cryosections and neoadjuvant chemoradiotherapy were found to be more effective for achieving negative resection margins in signet ring cell carcinoma.
Collapse
Affiliation(s)
- Tao Pang
- Department of Gastrointestinal Tract Surgery, First Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Mingming Nie
- Department of Gastrointestinal Tract Surgery, First Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Kai Yin
- Department of Gastrointestinal Tract Surgery, First Affiliated Hospital of Naval Military Medical University, Shanghai, China.
| |
Collapse
|
47
|
Faron M, Cheugoua-Zanetsie M, Tierney J, Thirion P, Nankivell M, Winter K, Yang H, Shapiro J, Vernerey D, Smithers BM, Walsh T, Piessen G, Nilsson M, Boonstra J, Ychou M, Law S, Cunningham D, de Vathaire F, Stahl M, Urba S, Valmasoni M, Williaume D, Thomas J, Lordick F, Tepper J, Roth J, Gebski V, Burmeister B, Paoletti X, van Sandick J, Fu J, Pignon JP, Ducreux M, Michiels S. Individual Participant Data Network Meta-Analysis of Neoadjuvant Chemotherapy or Chemoradiotherapy in Esophageal or Gastroesophageal Junction Carcinoma. J Clin Oncol 2023; 41:4535-4547. [PMID: 37467395 PMCID: PMC10553121 DOI: 10.1200/jco.22.02279] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/31/2023] [Accepted: 04/24/2023] [Indexed: 07/21/2023] Open
Abstract
PURPOSE The optimal neoadjuvant treatment for resectable carcinoma of the thoracic esophagus (TE) or gastroesophageal junction (GEJ) remains a matter of debate. We performed an individual participant data (IPD) network meta-analysis (NMA) of randomized controlled trials (RCTs) to study the effect of chemotherapy or chemoradiotherapy, with a focus on tumor location and histology subgroups. PATIENTS AND METHODS All, published or unpublished, RCTs closed to accrual before December 31, 2015 and having compared at least two of the following strategies were eligible: upfront surgery (S), chemotherapy followed by surgery (CS), and chemoradiotherapy followed by surgery (CRS). All analyses were conducted on IPD obtained from investigators. The primary end point was overall survival (OS). The IPD-NMA was analyzed by a one-step mixed-effect Cox model adjusted for age, sex, tumor location, and histology. The NMA was registered in PROSPERO (CRD42018107158). RESULTS IPD were obtained for 26 of 35 RCTs (4,985 of 5,807 patients) corresponding to 12 comparisons for CS-S, 12 for CRS-S, and four for CRS-CS. CS and CRS led to increased OS when compared with S with hazard ratio (HR) = 0.86 (0.75 to 0.99), P = .03 and HR = 0.77 (0.68 to 0.87), P < .001 respectively. The NMA comparison of CRS versus CS for OS gave a HR of 0.90 (0.74 to 1.09), P = .27 (consistency P = .26, heterogeneity P = .0038). For CS versus S, a larger effect on OS was observed for GEJ versus TE tumors (P = .036). For the CRS versus S and CRS versus CS, a larger effect on OS was observed for women (P = .003, .012, respectively). CONCLUSION Neoadjuvant chemotherapy and chemoradiotherapy were consistently better than S alone across histology, but with some variation in the magnitude of treatment effect by sex for CRS and tumor location for CS. A strong OS difference between CS and CRS was not identified.
Collapse
Affiliation(s)
- Matthieu Faron
- Oncostat, CESP, Inserm U1018, University Paris-Saclay, labeled Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Maurice Cheugoua-Zanetsie
- Oncostat, CESP, Inserm U1018, University Paris-Saclay, labeled Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Jayne Tierney
- MRC Clinical Trial Unit at UCL, London, United Kingdom
| | | | | | - Kathryn Winter
- NRG Oncology Statistics and Data Management Center, Philadelphia, PA
| | - Hong Yang
- Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Joel Shapiro
- Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - B. Mark Smithers
- University of Queensland, Princess Alexandra Hospital, Brisbane, Australia
| | - Thomas Walsh
- Connolly Hospital Blanchardstown, Dublin, Ireland
| | | | - Magnus Nilsson
- Division of Surgery, Department of Clinical Science, Intervention and Technoglogy, Karolinska Institutet, Stockholm, Sweden
- Department of Upper Abdominal Diseases, Karolinska University Hospital, Stockholm, Sweden
| | | | | | - Simon Law
- Department of Surgery, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - David Cunningham
- National Institute for Health Research, Biomedical Research Centres, Royal Marsden, London, United Kingdom
| | - Florent de Vathaire
- Oncostat, CESP, Inserm U1018, University Paris-Saclay, labeled Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | | | | | - Michele Valmasoni
- Department of Surgery, Oncology and Gastroenterology, Padova University Hospital, Center for Esophageal Diseases, Padova, Italy
| | | | - Janine Thomas
- Princess Alexandra Hospital, Woolloongabba, Australia
| | | | - Joel Tepper
- University of North Carolina School of Medicine, Chapel Hill, NC
| | | | | | | | | | - Johanna van Sandick
- The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Jianhua Fu
- Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jean-Pierre Pignon
- Oncostat, CESP, Inserm U1018, University Paris-Saclay, labeled Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Michel Ducreux
- Departement d’Oncologie Médicale, Gustave Roussy, Villejuif, France
| | - Stefan Michiels
- Oncostat, CESP, Inserm U1018, University Paris-Saclay, labeled Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| |
Collapse
|
48
|
Moore JL, Green M, Santaolalla A, Deere H, Evans RPT, Elshafie M, Lavery A, McManus DT, McGuigan A, Douglas R, Horne J, Walker R, Mir H, Terlizzo M, Kamarajah SK, Van Hemelrijck M, Maisey N, Sita-Lumsden A, Ngan S, Kelly M, Baker CR, Kumar S, Lagergren J, Allum WH, Gossage JA, Griffiths EA, Grabsch HI, Turkington RC, Underwood TJ, Smyth EC, Fitzgerald RC, Cunningham D, Davies AR. Pathologic Lymph Node Regression After Neoadjuvant Chemotherapy Predicts Recurrence and Survival in Esophageal Adenocarcinoma: A Multicenter Study in the United Kingdom. J Clin Oncol 2023; 41:4522-4534. [PMID: 37499209 DOI: 10.1200/jco.23.00139] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/03/2023] [Accepted: 05/24/2023] [Indexed: 07/29/2023] Open
Abstract
PURPOSE There is limited evidence regarding the prognostic effects of pathologic lymph node (LN) regression after neoadjuvant chemotherapy for esophageal adenocarcinoma, and a definition of LN response is lacking. This study aimed to evaluate how LN regression influences survival after surgery for esophageal adenocarcinoma. METHODS Multicenter cohort study of patients with esophageal adenocarcinoma treated with neoadjuvant chemotherapy followed by surgical resection at five high-volume centers in the United Kingdom. LNs retrieved at esophagectomy were examined for chemotherapy response and given a LN regression score (LNRS)-LNRS 1, complete response; 2, <10% residual tumor; 3, 10%-50% residual tumor; 4, >50% residual tumor; and 5, no response. Survival analysis was performed using Cox regression adjusting for confounders including primary tumor regression. The discriminatory ability of different LN response classifications to predict survival was evaluated using Akaike information criterion and Harrell C-index. RESULTS In total, 17,930 LNs from 763 patients were examined. LN response classified as complete LN response (LNRS 1 ≥1 LN, no residual tumor in any LN; n = 62, 8.1%), partial LN response (LNRS 1-3 ≥1 LN, residual tumor ≥1 LN; n = 155, 20.3%), poor/no LN response (LNRS 4-5; n = 303, 39.7%), or LN negative (no tumor/regression; n = 243, 31.8%) demonstrated superior discriminatory ability. Mortality was reduced in patients with complete LN response (hazard ratio [HR], 0.35; 95% CI, 0.22 to 0.56), partial LN response (HR, 0.72; 95% CI, 0.57 to 0.93) or negative LNs (HR, 0.32; 95% CI, 0.25 to 0.42) compared with those with poor/no LN response. Primary tumor regression and LN regression were discordant in 165 patients (21.9%). CONCLUSION Pathologic LN regression after neoadjuvant chemotherapy was a strong prognostic factor and provides important information beyond pathologic TNM staging and primary tumor regression grading. LN regression should be included as standard in the pathologic reporting of esophagectomy specimens.
Collapse
Affiliation(s)
- Jonathan L Moore
- Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom
- School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom
| | - Michael Green
- Department of Histopathology, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Aida Santaolalla
- School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom
| | - Harriet Deere
- Department of Histopathology, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Richard P T Evans
- Department of Upper Gastrointestinal Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Mona Elshafie
- Department of Pathology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Anita Lavery
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Damian T McManus
- Department of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Andrew McGuigan
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Rosalie Douglas
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Joanne Horne
- Department of Histopathology, University Hospitals Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Robert Walker
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Hira Mir
- Department of Histopathology, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Monica Terlizzo
- Department of Histopathology, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Sivesh K Kamarajah
- Department of Pathology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Mieke Van Hemelrijck
- School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom
| | - Nick Maisey
- Department of Medical Oncology, St Thomas' Hospital, London, United Kingdom
| | - Ailsa Sita-Lumsden
- Department of Medical Oncology, St Thomas' Hospital, London, United Kingdom
| | - Sarah Ngan
- Department of Medical Oncology, St Thomas' Hospital, London, United Kingdom
| | - Mark Kelly
- Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom
- School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom
| | - Cara R Baker
- Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom
- School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom
| | - Sacheen Kumar
- Department of Upper Gastrointestinal Surgery, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Jesper Lagergren
- Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom
- School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - William H Allum
- Department of Upper Gastrointestinal Surgery, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - James A Gossage
- Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom
- School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom
| | - Ewen A Griffiths
- Department of Pathology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Heike I Grabsch
- Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, the Netherlands
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Richard C Turkington
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, United Kingdom
| | - Tim J Underwood
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Elizabeth C Smyth
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Rebecca C Fitzgerald
- Early Cancer Institute, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Department of Gastroenterology, Addenbrooke's Hospital, Cambridge University NHS Foundation Trust, Cambridge, United Kingdom
| | - David Cunningham
- Department of Medical Oncology, The Royal Marsden Hospital, London, United Kingdom
| | - Andrew R Davies
- Department of Upper Gastrointestinal and General Surgery, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom
- School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom
| |
Collapse
|
49
|
Han K, Tang JH, Liao LE, Jiang W, Sui QQ, Xiao BY, Li WR, Hong ZG, Li Y, Kong LH, Li DD, Zhang XS, Pan ZZ, Steele SR, Ding PR. Neoadjuvant Immune Checkpoint Inhibition Improves Organ Preservation in T4bM0 Colorectal Cancer With Mismatch Repair Deficiency: A Retrospective Observational Study. Dis Colon Rectum 2023; 66:e996-e1005. [PMID: 35485833 DOI: 10.1097/dcr.0000000000002466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Colorectal cancer with mismatch repair deficiency is usually less aggressive and associated with a lower risk of distant metastasis. Immune checkpoint inhibition, rather than traditional chemoradiotherapy, has shown great advantages in treating such patients. OBJECTIVE This study aimed to verify the hypothesis that locally very advanced (T4b) colorectal cancer without distant metastases might present with higher probability of mismatch repair deficiency and be more sensitive to neoadjuvant immune checkpoint inhibition. DESIGN This study was designed as a single-center retrospective observational study. SETTINGS The study was conducted in a tertiary referral center in China. PATIENTS The study included patients who were clinically diagnosed with T4bM0 colorectal cancer from 2008 to 2019. MAIN OUTCOME MEASURES Clinicopathological characteristics, mismatch repair status, and survival outcomes of patients with mismatch repair deficiency were analyzed. RESULTS A total of 268 patients were included. The incidence of patients with mismatch repair deficiency in the T4bM0 population was 27.6% (75/268), with 84.0% (63/75) in the colon and 16.0% (12/75) in the rectum. For tumors located in the proximal colon, 45.0% (50/111) exhibited mismatch repair deficiency, whereas the incidence of mismatch repair deficiency in sigmoid colon cancer and rectal cancer was only 15.9% (25/157). Neoadjuvant immune checkpoint inhibition significantly reduced the open surgery rate ( p = 0.000) and multivisceral resection rate ( p = 0.025). The pathological complete remission rate in the neoadjuvant immune checkpoint inhibition group was significantly higher than that in neoadjuvant chemoradiotherapy/chemotherapy group (70.0% vs 0%; p = 0.004). No tumor downstaging was observed after neoadjuvant chemotherapy. Neoadjuvant immune checkpoint inhibition provided significantly better disease-free survival ( p = 0.0078) and relatively longer overall survival ( p = 0.15) than other groups. LIMITATIONS This study is limited by the possible selection bias and small sample size. CONCLUSIONS Our data depicted the high incidence of mismatch repair deficiency in T4bM0 mismatch repair deficiency and the effectiveness of the neoadjuvant immune checkpoint inhibition group in organ preservation. Precision oncology requires identification of the protein status of mismatch repair at initial diagnosis to make a rational treatment decision for these patients. See Video Abstract at http://links.lww.com/DCR/B952 . LA INHIBICIN DEL PUNTO DE CONTROL INMUNITARIO NEOADYUVANTE MEJORA LA PRESERVACIN DE RGANOS EN EL CNCER COLORRECTAL TBM CON DEFICIENCIA DE REPARACIN DE ERRORES DE COINCIDENCIA UN ESTUDIO OBSERVACIONAL RETROSPECTIVO ANTECEDENTES:Los pacientes con cáncer colorrectal con deficiencia en la reparación de desajustes suelen (dMMR) ser menos agresivos y se asocian con un menor riesgo de metástasis a distancia. La inhibición del punto de control inmunitario, en lugar de la quimiorradioterapia tradicional, ha mostrado grandes ventajas en el tratamiento de estos pacientes.OBJETIVO:Este estudio tuvo como objetivo verificar nuestra hipótesis de que el CCR localmente muy avanzado (T4b) sin metástasis a distancia podría presentarse con una mayor probabilidad de dMMR y ser más sensible a la inhibición del punto de control inmunitario neoadyuvante.DISEÑO:Este estudio fue diseñado como un estudio observacional retrospectivo de un solo centro.CONFIGURACIÓN:El estudio se realizó en un centro de referencia terciario en China.PACIENTES:Se incluyeron pacientes con diagnóstico clínico de CCR T4bM0 desde 2008 hasta 2019.PRINCIPALES MEDIDAS DE RESULTADO:Se analizaron las características clinicopatológicas, el estado de MMR y los resultados de supervivencia de los pacientes con dMMR.RESULTADOS:Se incluyeron un total de 268 pacientes. La incidencia de dMMR en la población T4bM0 fue del 27,6% (75/268), con un 84,0% (63/75) en colon y un 16,0% (12/75) en recto. Para los tumores ubicados en el colon proximal, el 45,0% (50/111) exhibió dMMR, mientras que la incidencia de dMMR en el cáncer de colon sigmoideo y el cáncer de recto fue solo del 15,9% (25/157). La inhibición del punto de control inmunitario neoadyuvante redujo significativamente la cirugía abierta y la tasa de resección multivisceral ( p = 0,000 y p = 0,025, respectivamente). La tasa de PCR en el grupo de inhibición del punto de control inmunitario neoadyuvante fue significativamente mayor que en el grupo de quimiorradioterapia/quimioterapia neoadyuvante (70,0% frente a 0%, p = 0,004). No se observó reducción del estadio del tumor después de la quimioterapia neoadyuvante. La inhibición del punto de control inmunitario neoadyuvante proporcionó una supervivencia sin enfermedad significativamente mejor ( p = 0,0078) y una supervivencia general relativamente más larga ( p = 0,15) que otros grupos.LIMITACIONES:Este estudio está limitado por el posible sesgo de selección y el pequeño tamaño de la muestra.CONCLUSIONES:Nuestros datos representan la alta incidencia de dMMR en T4bM0 CRC y la eficacia del grupo de inhibición del punto de control inmunitario neoadyuvante en la preservación de órganos. La oncología de precisión requiere la identificación del estado de la proteína MMR en el diagnóstico inicial para tomar una decisión de tratamiento racional para estos pacientes especiales. Consulte el Video Resumen en http://links.lww.com/DCR/B952 . (Traducción-Dr. Yesenia Rojas-Khalil ).
Collapse
Affiliation(s)
- Kai Han
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jing-Hua Tang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Le-En Liao
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wu Jiang
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qiao-Qi Sui
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Bin-Yi Xiao
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wei-Rong Li
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zhi-Gang Hong
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuan Li
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ling-Heng Kong
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Dan-Dan Li
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Biological Therapy Center, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao-Shi Zhang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Biological Therapy Center, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhi-Zhong Pan
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Scott R Steele
- Department of Colorectal Surgery, Digestive Disease & Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Pei-Rong Ding
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
50
|
Dermanis AA, Kamarajah SK, Tan B. The Evolution of Neo-Adjuvant Therapy in the Treatment of Oesophageal and Gastro-Oesophageal Junction Adenocarcinomas. Cancers (Basel) 2023; 15:4741. [PMID: 37835435 PMCID: PMC10571977 DOI: 10.3390/cancers15194741] [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: 08/06/2023] [Revised: 09/11/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Historically, oesophageal and gastro-oesophageal junction adenocarcinomas were associated with a poor prognosis. The advent of neoadjuvant therapy has transformed the management of oesophageal and gastro-oesophageal junction adenocarcinomas further and offers the possibility to reverse disease progression, eliminate micrometastasis, and offer potentially better outcomes for these patients. This review provides an overview of landmark clinical trials in this area, with different treatment regimens considered over the years as well as potential therapeutic agents on the horizon that may transform the management of oesophageal and gastro-oesophageal junction adenocarcinomas further.
Collapse
Affiliation(s)
| | - Sivesh K. Kamarajah
- Queen Elizabeth Hospital, Mindelsohn Way, Birmingham B15 2GW, UK; (A.A.D.)
- Academic Department of Surgery, Institute of Applied Health Research, University of Birmingham, Birmingham B15 2TT, UK
| | - Benjamin Tan
- Queen Elizabeth Hospital, Mindelsohn Way, Birmingham B15 2GW, UK; (A.A.D.)
| |
Collapse
|