1
|
Schæbel GH, Johannesen HH, Löfgren J, Gutte H, Bæksgaard L, Achiam MP, Belmouhand M. The prognostic value of positron emission tomography/magnetic resonance imaging in predicting survival in patients with adenocarcinoma of the esophagogastric junction. Ann Nucl Med 2025:10.1007/s12149-025-02058-z. [PMID: 40381134 DOI: 10.1007/s12149-025-02058-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 05/07/2025] [Indexed: 05/19/2025]
Abstract
INTRODUCTION In recent years, the utility of positron emission tomography/magnetic resonance imaging (PET/MRI) has become increasingly significant in diagnostic settings. This study provides a five-year follow-up on a previous pilot study that demonstrated the feasibility of PET/MRI in predicting the resectability of adenocarcinoma of the esophagogastric junction (AEG). We aimed to evaluate whether this imaging modality could further serve as a prognostic tool for survival in AEG patients. METHODS A total of 22 patients were included in the initial pilot study, with 17 of them undergoing surgery. All patients underwent three series of neo-adjuvant chemotherapy (NT). This follow-up study retrospectively analyzed the correlation between the apparent diffusion coefficient (ADC) and standard uptake value (SUV) measurements of the primary tumor from the original study with overall survival and recurrence. ADC and SUV values were measured prior to initiation of NT, and again 17-21 days into the first cycle of NT-administration, and the differences between the scans were calculated as ∆SUVmax, ∆ADCb0, and ∆ADCb50. Early treatment response was assessed using the Response Evaluation Criteria In Solid Tumors (RECIST). Binary logistic regression was employed to evaluate the predictive values of ADC and SUV parameters, and receiver operating characteristic (ROC) curves were generated to determine sensitivity, specificity, and area under the curve (AUC). RESULTS As of January 7, 2022, 8 of the 22 patients were still alive. The AUC was calculated to assess the association of imaging parameters with long-term survival: ∆SUVmax: AUC = 0.74, sensitivity, 87.5%, specificity 62.5% (p = 0.037). ∆ADCb0: AUC = 0.62, sensitivity 85.7%, specificity 57.1% (p = 0.400). ∆ADCb50: AUC = 0.78, sensitivity 78.6%, specificity 85.7% (p = 0.011). Combining all three parameters yielded an AUC of 0.81, with a sensitivity of 78.6% and a specificity of 85.7% (p = 0.002). The results for individual measurements were: SUVmax(pre-NT): AUC = 0.56, sensitivity 78.6%, specificity 50% (p = 0.646). SUVmax(post-NT): AUC = 0.81, sensitivity 85.7%, specificity 87.5% (p = 0.002). ADCb0(pre-NT): AUC = 0.55, sensitivity 71.4%, specificity 62.5% (p = 0.682). ADCb0(post-NT): AUC = 0.63, sensitivity 78.6%, specificity 57.1% (p = 0.339). ADCb50(pre-NT): AUC = 0.51, sensitivity 85.7%, specificity 37.5% (p = 0.952). ADCb50(post-NT): AUC = 0.63, sensitivity 42.9%, specificity 100% (p = 0.279). No significant correlation was found between RECIST group and survival status (p = 0.15). CONCLUSION Our results indicate that PET/MRI is feasible for predicting long-term survival in AEG patients. The highest AUCs were achieved when combining SUV and ADC parameters, and when using post-NT SUVmax alone.
Collapse
Affiliation(s)
- Gustav Holm Schæbel
- Department of Surgery and Transplantation, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Helle Hjorth Johannesen
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Johan Löfgren
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Henrik Gutte
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Lene Bæksgaard
- Department of Oncology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Michael Patrick Achiam
- Department of Surgery and Transplantation, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Mohamed Belmouhand
- Department of Surgery and Transplantation, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Valdemar Hansens Vej 1, 23, 2600, Glostrup, Denmark
| |
Collapse
|
2
|
Alcindor T, Tankel J, Fiset PO, Pal S, Opu T, Strasser M, Dehghani M, Bertos N, Zuo D, Mueller C, Cools-Lartigue J, Hickeson M, Marcus V, Camilleri-Broet S, Spatz A, Evaristo G, Farag M, Artho G, Elkrief A, Saleh R, Bailey S, Park M, Huang S, Sangwan V, Ferri L. Phase 2 trial of perioperative chemo-immunotherapy for gastro-esophageal adenocarcinoma: The role of M2 macrophage landscape in predicting response. Cell Rep Med 2025; 6:102045. [PMID: 40239627 PMCID: PMC12047487 DOI: 10.1016/j.xcrm.2025.102045] [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: 07/18/2024] [Revised: 11/28/2024] [Accepted: 03/06/2025] [Indexed: 04/18/2025]
Abstract
We present the clinical results of a phase 2 trial combining neoadjuvant docetaxel, cisplatin, 5 Flourouracil, and the PD-L1 inhibitor avelumab in locally advanced gastro-esophageal adenocarcinoma (GEA). Fifty-one patients receive neoadjuvant therapy with 50 proceeding to surgery. Grade 3-4 adverse events occur in 40%; complete/major pathological response is found in 7/50 (14%) and 9/50 (18%), with 2-year disease-free survival of 67.5%. There is no correlation between tumor regression and PD-L1 or mismatch repair (MMR) status. Multiplex immunohistochemistry and longitudinal single-cell transcriptomic profiling reveal alterations in certain innate immune cell populations, particularly noting an M2-tumor-associated macrophage (M2-TAM) proliferation in non-responding tumors. These findings describe the effective nature of this treatment regimen for GEA and reveal associated features of the inflammatory milieux associated with response to chemo-immunotherapy. The specific character of the inflammatory environment in non-responders may, in the future, help personalize treatment. This study was registered at ClinicalTrials.gov (NCT03288350).
Collapse
Affiliation(s)
- Thierry Alcindor
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada; Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada.
| | - James Tankel
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Pierre-Olivier Fiset
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Sanjima Pal
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Touhid Opu
- Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | | | - Mehrnoush Dehghani
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Nicholas Bertos
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Dongmei Zuo
- Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Carmen Mueller
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | | | - Marc Hickeson
- Department of Nuclear Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Victoria Marcus
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Sophie Camilleri-Broet
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Alan Spatz
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Gertruda Evaristo
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Mina Farag
- Division of Pathology, Department of Clinical Laboratory Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Giovanni Artho
- Department of Diagnostic Radiology, McGill University Health Centre, Montreal, QC, Canada
| | - Arielle Elkrief
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Ramy Saleh
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada; Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Swneke Bailey
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Morag Park
- Center for Innovative Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Sui Huang
- Institute for Systems Biology, Seattle, WA, USA
| | - Veena Sangwan
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Lorenzo Ferri
- Department of Surgery, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
3
|
Kumar R, Shalaby A, Narra LR, Gokhale S, Deek MP, Jabbour SK. Updates in the Role of Positron Emission Tomography/Computed Tomography in Radiation Oncology in Gastrointestinal Malignancies. PET Clin 2025; 20:219-229. [PMID: 39952884 PMCID: PMC12037145 DOI: 10.1016/j.cpet.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2025]
Abstract
Positron Emission Tomography/Computed Tomography (PET/CT) plays a critical role in managing gastrointestinal (GI) cancers within radiation oncology. It enhances tumor detection, staging, and lymph node involvement assessment, leading to better-targeted radiation treatment. PET/CT also aids in delineating tumor volumes to minimize geographic misses, enabling precise dose escalation to metabolically active regions. Despite its benefits, PET/CT has limitations such as false positives and dependency on complementary imaging. Emerging technologies offer real-time adjustments and personalized treatments, advancing precision medicine in GI radiation oncology. Further research is needed to refine PET/CT integration for improved treatment outcomes and cost-effectiveness.
Collapse
Affiliation(s)
- Ritesh Kumar
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Rutgers University, 195 Little Albany Street, New Brunswick, NJ 08901, USA
| | - Ahmed Shalaby
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Rutgers University, 195 Little Albany Street, New Brunswick, NJ 08901, USA
| | - Lakshmi Rekha Narra
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Rutgers University, 195 Little Albany Street, New Brunswick, NJ 08901, USA
| | - Shivani Gokhale
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Rutgers University, 195 Little Albany Street, New Brunswick, NJ 08901, USA
| | - Matthew P Deek
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Rutgers University, 195 Little Albany Street, New Brunswick, NJ 08901, USA
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute, Rutgers Robert Wood Johnson Medical School, Rutgers University, 195 Little Albany Street, New Brunswick, NJ 08901, USA.
| |
Collapse
|
4
|
Urakawa N, Kanaji S, Sawada R, Koterazawa Y, Ikeda T, Harada H, Goto H, Hasegawa H, Yamashita K, Matsuda T, Kakeji Y. Efficacy of 18F-Fluoro-2-Deoxyglucose Positron Emission Tomography as a Predictor of Treatment Response to Neoadjuvant S-1 + Oxaliplatin Chemotherapy for Gastric Cancer. Cancer Rep (Hoboken) 2025; 8:e70190. [PMID: 40275469 PMCID: PMC12021666 DOI: 10.1002/cnr2.70190] [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: 06/17/2024] [Revised: 02/27/2025] [Accepted: 03/12/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy is widely recognized as the established treatment for advanced gastric cancer. However, predicting its efficacy before surgery remains challenging. AIM The present study aimed to evaluate the effectiveness of 18F-fluoro-2-deoxyglucose positron emission tomography (FDG-PET) as a predictor of treatment response to the S-1+Oxaliplatin regimen (SOX). METHODS AND RESULTS Thirty patients who underwent gastrectomy following neoadjuvant SOX between January 2021 and July 2023 were included. Patients underwent FDG-PET pre- and postsurgery. The maximum standardized uptake value (SUVmax) from FDG-PET was examined in relation to histological tumor response and prognosis. SUVmax decreased significantly after chemotherapy in all patients (p < 0.001), especially in those with Grade 1a, 2, and 3 tumors (p < 0.05). SUV reduction increased stepwise with the histological response grade. Optimal cut-off values for the percentage decrease in SUVmax (ΔSUVmax) predictive of histologic efficacy were identified as 53% (area under curve 0.855, p = 0.0018) for Grade 1b or higher and 75% (area under curve 0.806, p = 0.0044) for Grade 2 or higher. Patients with ΔSUVmax > 50% had improved recurrence-free survival (p = 0.027). CONCLUSION FDG-PET may be useful as a predictor of treatment response in neoadjuvant SOX therapy for gastric cancer. The determination of the optimal ΔSUVmax value may enhance the precision of histological tumor response prediction.
Collapse
Affiliation(s)
- Naoki Urakawa
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Shingo Kanaji
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Ryuichiro Sawada
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Yasufumi Koterazawa
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Taro Ikeda
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Hitoshi Harada
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Hironobu Goto
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Hiroshi Hasegawa
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Kimihiro Yamashita
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Takeru Matsuda
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Yoshihiro Kakeji
- Division of Gastrointestinal Surgery, Department of SurgeryKobe University Graduate School of MedicineKobeJapan
| |
Collapse
|
5
|
Rao V, Singh S, Zade B. Advances in radiotherapy in the treatment of esophageal cancer. World J Clin Oncol 2025; 16:102872. [PMID: 40130058 PMCID: PMC11866087 DOI: 10.5306/wjco.v16.i3.102872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/06/2024] [Accepted: 12/27/2024] [Indexed: 01/21/2025] Open
Abstract
Recent advancements in radiotherapy for esophageal cancer have significantly improved treatment outcomes and patient quality of life. Traditional radiotherapy techniques have been enhanced by the integration of advanced imaging and precision targeting technologies, such as intensity-modulated radiotherapy and proton therapy, which allow for more accurate tumor targeting while minimizing damage to surrounding healthy tissues. Additionally, combining radiotherapy with immunotherapy has shown promising results, leveraging the body's immune response to enhance the effectiveness of cancer treatment. Studies have also highlighted the benefits of neoadjuvant chemoradiation followed by surgical resection, which has been associated with improved overall survival rates compared to radiotherapy alone. These innovations are paving the way for more effective and personalized treatment strategies, offering new hope for patients with esophageal cancer.
Collapse
Affiliation(s)
- Vrushab Rao
- Department of Cyberknife Radiosurgery and Radiation Oncology, Ruby Hall Clinic, Pune 411001, Maharashtra, India
| | - Soumya Singh
- Department of Cyberknife Radiosurgery and Radiation Oncology, Ruby Hall Clinic, Pune 411001, Maharashtra, India
| | - Bhooshan Zade
- Department of Cyberknife Radiosurgery and Radiation Oncology, Ruby Hall Clinic, Pune 411001, Maharashtra, India
| |
Collapse
|
6
|
Liu F, Ma C, Chen M, Chen K, Zhu L, Li L, Zhu X, Qu S, Yan C. A Nomogram Based on Tumor Response to Induction Chemotherapy and Plasma Epstein-Barr Virus DNA Level after Induction Chemotherapy to Explore Individualized Treatment of Patients with Locally Advanced Nasopharyngeal Carcinoma. J Inflamm Res 2025; 18:3677-3693. [PMID: 40099001 PMCID: PMC11913028 DOI: 10.2147/jir.s507926] [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: 11/22/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
Purpose To explore the influence of Epstein-Barr virus (EBV) DNA levels before and after induction chemotherapy (IC), tumor response to IC, and baseline factors on overall survival (OS) in patients with locally advanced nasopharyngeal carcinoma (LA-NPC). A nomogram was subsequently constructed to explore the individualized optimal cumulative cisplatin dose (CCD) in concurrent chemoradiotherapy (CCRT). Methods A total of 581 LA-NPC patients were included, randomly divided into training and validation cohorts in a 7:3 ratio. In the training cohort, a nomogram was subsequently established based on multivariate Cox regression analysis and then validated. Subsequently, patients were classified into different risk groups based on the nomogram, and the impact of different levels of CCD on survival outcomes was evaluated. Results EBV DNA levels after IC, tumor response to IC, age, and LDH were independent prognostic factors of OS. Schoenfeld residual analysis indicated overall satisfaction of the proportional hazards assumption for the Cox regression model. The C-index of the nomogram was 0.758 (95% CI: 0.695-0.821) for the training cohort and 0.701 (95% CI: 0.589-0.813) for the validation cohort. Calibration curves demonstrated good correlation between the nomogram and actual survival outcomes. DCA confirmed the clinical utility enhancement of the nomogram over the TNM staging system. For OS, patients in the medium/high-risk group with a CCD > 200 mg/m² had better outcomes than those with CCD ≤ 200 mg/m², although the difference was not statistically significant (p = 0.097). No significant difference was observed in local relapse-free survival (LRFS), distant metastasis-free survival (DMFS), and progression-free survival (PFS) across various levels of CCD in different risk subgroups (p > 0.05). Conclusion The nomogram based on EBV DNA levels after IC, tumor response, LDH, and age effectively predicts OS in LA-NPC patients, aids in risk stratification, and may guide treatment decisions.
Collapse
Affiliation(s)
- Fushuang Liu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Chengxian Ma
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Meiwen Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Kaihua Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Liru Zhu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Ling Li
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Xiaodong Zhu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
- Department of Oncology, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, 530199, People's Republic of China
| | - Song Qu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Chang Yan
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| |
Collapse
|
7
|
Xu M, Zhang Y, Zhao K, Jiang H, Wang G, Wu Y, Wang Y, Liu N, Su X. Prediction of pathological response to neoadjuvant immunochemotherapy with baseline and post-treatment 18F-FDG PET imaging biomarkers in patients with locally advanced gastric cancer. BMC Cancer 2025; 25:378. [PMID: 40022087 PMCID: PMC11871599 DOI: 10.1186/s12885-025-13765-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Neoadjuvant immunochemotherapy (NICT) has shown promising therapeutic benefits in patients with locally advanced gastric cancer (LAGC). Our study aimed to predict the pathological response to NICT in LAGC before surgery by correlating the metabolic parameters of baseline and post-treatment 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) of the primary lesion with the pathological response following radical surgery. METHODS Thirty-six LAGC patients who received three cycles of NICT (combination of sintilimab and CapeOx), followed by radical surgery, were included in this study. Both baseline 18F-FDG PET/CT (bPET) and post-treatment 18F-FDG PET/CT (pPET) were conducted, the metabolic parameters derived from the PET/CT scans, including the maximum standardized uptake value (SUVmax), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) on bPET and pPET (bSUVmax and pSUVmax, bMTV and pMTV, bTLG and pTLG), as well as their reductions post-treatment (ΔSUVmax, ΔMTV, and ΔTLG), were assessed for their correlation with treatment efficacy and tumor regression grade (TRG) following NICT. RESULTS Out of the 36 patients, 13 patients had a good response (GR), which included 5 cases with TRG 0 and 8 cases with TRG 1. Conversely, 23 patients exhibited a poor response (PR), with 20 patients having TRG 2 and 3 patients having TRG 3. Univariate analysis revealed that pMTV and pTLG in the GR group were significantly lower compared to the PR group (all p < 0.05). The identified cutoff values of pMTV and pTLG were 1.68 cm³ (area under the cure (AUC) = 0.683) and 4.71 cm³ (AUC = 0.683) for the GR and PR groups, respectively. On receiver operating characteristic (ROC) curve analyses, these values corresponded to sensitivity, specificity, and accuracy of 68.8%, 80.0%, and 73.1%, respectively, with no statistically significant differences between them after the DeLong test and McNemar test (all p > 0.05). Furthermore, bSUVmax, bMTV, bTLG, ΔSUVmax, ΔMTV, and ΔTLG in the TRG 0 group were significantly higher than those in the TRG 1 group (all p < 0.05). Upon performing ROC curve analyses for the TRG 0 group, the thresholds for bSUVmax, bMTV, bTLG, ΔSUVmax, ΔMTV, and ΔTLG were determined to be 7.8 (AUC = 0.916), 36.76 (AUC = 0.768), 105.55 (AUC = 0.819), 4.82 (AUC = 0.923), 22.64 (AUC = 0.807), and 104.7 (AUC = 0.845), with no statistically significant differences between them after the DeLong test (all p > 0.05). These thresholds demonstrated high sensitivity (80% for bMTV and 100% for others), specificity (83.9%, 71.0%, 67.7%, 83.9%, 61.3%, and 71.0%), and accuracy (86.1%, 66.7%, 72.2%, 86.1%, 66.7%, and 75.0%) in predicting TRG 0 after NICT, with no statistically significant differences between them after the McNemar test (all p > 0.05). CONCLUSIONS Imaging biomarkers from the combination of baseline and post-treatment 18F-FDG PET/CT showed potential in predicting pathological response to NICT in LAGC patients before surgery.
Collapse
Affiliation(s)
- Mimi Xu
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yafei Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Kui Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Haiping Jiang
- Oncology Department, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Guangfa Wang
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yan Wu
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yu Wang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China
| | - Nian Liu
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Xinhui Su
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
8
|
Dong Y, Wang Z, Hu X, Sun Y, Qin J, Qin Q, Liu S, Yuan S, Yu J, Wei Y. [ 18F]AlF-NOTA-FAPI-04 PET/CT for Predicting Pathologic Response of Resectable Esophageal Squamous Cell Carcinoma to Neoadjuvant Camrelizumab and Chemotherapy: A Phase II Clinical Trial. J Nucl Med 2024; 65:1702-1709. [PMID: 39327020 DOI: 10.2967/jnumed.124.268557] [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: 08/06/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024] Open
Abstract
This single-center, single-arm, phase II trial (ChiCTR2100050057) investigated the ability of 18F-labeled fibroblast activation protein inhibitor ([18F]AlF-NOTA-FAPI-04, denoted as 18F-FAPI) PET/CT to predict the response to neoadjuvant camrelizumab plus chemotherapy (nCC) in locally advanced esophageal squamous cell carcinoma (LA-ESCC). Methods: This study included 32 newly diagnosed LA-ESCC participants who underwent 18F-FAPI PET/CT at baseline, of whom 23 also underwent scanning after 2 cycles of nCC. The participants underwent surgery after 2 cycles of nCC. Recorded PET parameters included maximum, peak, and mean SUVs and tumor-to-background ratios (TBRs), metabolic tumor volume, and total lesion FAP expression. PET parameters were compared between patient groups with good and poor pathologic responses, and the predictive performance for treatment response was analyzed. Results: The good and poor response groups each included 16 participants (16/32, 50.0%). On 18F-FAPI PET/CT, the posttreatment SUVs were significantly lower in good responders than in poor responders, whereas the changes in SUVs with treatment were significantly higher (all P < 0.05). SUVmax (area under the curve [AUC], 0.87; P = 0.0026), SUVpeak (AUC, 0.89; P = 0.0017), SUVmean (AUC, 0.88; P = 0.0021), TBRmax (AUC, 0.86; P = 0.0031), and TBRmean (AUC, 0.88; P = 0.0021) after nCC were significant predictors of pathologic response to nCC, with sensitivities of 63.64%-81.82% and specificities of 83.33%-100%. Changes in SUVmax (AUC, 0.81; P = 0.0116), SUVpeak (AUC, 0.82; P = 0.0097), SUVmean (AUC, 0.81; P = 0.0116), and TBRmean (AUC, 0.74; P = 0.0489) also were significant predictors of the pathologic response to nCC, with sensitivities and specificities in similar ranges. Conclusion: 18F-FAPI PET/CT parameters after treatment and their changes from baseline can predict the pathologic response to nCC in LA-ESCC participants.
Collapse
Affiliation(s)
- Yinjun Dong
- Department of Esophageal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhendan Wang
- Department of Thoracic Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xinying Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yuhong Sun
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; and
| | - Jingjie Qin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Qiming Qin
- Department of Esophageal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shuguang Liu
- Department of Esophageal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shuanghu Yuan
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Department of Radiation Oncology, Division of Life Sciences and Medicine, First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yuchun Wei
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China;
| |
Collapse
|
9
|
Tang Y, Zhu H, Xiao L, Li R, Han H, Tang W, Liu D, Zhou C, Liu D, Yang Z, Zhou L, Xiao B, Rominger A, Shi K, Hu S, Feng L. Individual cerebellar metabolic connectome in patients with MTLE and NTLE associated with surgical prognosis. Eur J Nucl Med Mol Imaging 2024; 51:3600-3616. [PMID: 38805089 DOI: 10.1007/s00259-024-06762-2] [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: 01/31/2024] [Accepted: 05/12/2024] [Indexed: 05/29/2024]
Abstract
PURPOSE This study aimed to comprehensively explore the different metabolic connectivity topological changes in MTLE and NTLE, as well as their association with surgical outcomes. METHODS This study enrolled a cohort of patients with intractable MTLE and NTLE. Each individual's metabolic connectome, as determined by Kullback-Leibler divergence similarity estimation for the [18F]FDG PET image, was employed to conduct a comprehensive analysis of the cerebral metabolic network. Alterations in network connectivity were assessed by extracting and evaluating the strength of edge and weighted connectivity. By utilizing these two connectivity strength metrics with the cerebellum, we explored the network properties of connectivity and its association with prognosis in surgical patients. RESULTS Both MTLE and NTLE patients exhibited substantial alterations in the connectivity of the metabolic network at the edge and nodal levels (p < 0.01, FDR corrected). The key disparity between MTLE and NTLE was observed in the cerebellum. In MTLE, there was a predominance of increased connectivity strength in the cerebellum. Whereas, a decrease in cerebellar connectivity was identified in NTLE. It was found that in MTLE, higher edge connectivity and weighted connectivity strength in the contralateral cerebellar hemisphere correlated with improved surgical outcomes. Conversely, in NTLE, a higher edge metabolic connectivity strength in the ipsilateral cerebellar hemisphere suggested a worse surgical prognosis. CONCLUSION The cerebellum exhibits distinct topological characteristics in the metabolic networks between MTLE and NTLE. The hyper- or hypo-metabolic connectivity in the cerebellum may be a prognostic biomarker of surgical prognosis, which might aid in therapeutic decision-making for TLE individuals.
Collapse
Affiliation(s)
- Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Haoyue Zhu
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, 410008, PR China
| | - Ling Xiao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Rong Li
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Honghao Han
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, China
| | - Weiting Tang
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, 410008, PR China
| | - Ding Liu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Chunyao Zhou
- Department of Neurosurgery, Xiangya Hospital, Central Southern University, Changsha, China
| | - Dingyang Liu
- Department of Neurosurgery, Xiangya Hospital, Central Southern University, Changsha, China
| | - Zhiquan Yang
- Department of Neurosurgery, Xiangya Hospital, Central Southern University, Changsha, China
| | - Luo Zhou
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, 410008, PR China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
- Department of Informatics, Technische Universität München, Munich, Germany
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Li Feng
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Rd, Changsha, 410008, PR China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
10
|
Svrcek M, Voron T, André T, Smyth EC, de la Fouchardière C. Improving individualised therapies in localised gastro-oesophageal adenocarcinoma. Lancet Oncol 2024; 25:e452-e463. [PMID: 39214116 DOI: 10.1016/s1470-2045(24)00180-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/05/2024] [Accepted: 03/20/2024] [Indexed: 09/04/2024]
Abstract
Despite our increased understanding of the biological and molecular aspects of gastro-oesophageal tumourigenesis, the identification of prognostic or predictive factors remains challenging. Patients with resectable gastric and oesophageal adenocarcinoma are often treated similarly after surgical resection, regardless of their tumour biology, clinical characteristics, and histological treatment response. Substantial progress has been made in the past 5 years in managing patients with gastric or oesophageal adenocarcinoma, including the use of immune checkpoint inhibitors and new targeted therapies, leading to substantial improvements in clinical outcomes. These advancements have primarily been established in advanced and metastatic disease, while the management framework for local and locoregional disease is just beginning to shift. We provide an overview of existing data on biomarkers and tumour-related and host-related factors that are relevant to stratify patients into low-risk and high-risk recurrence groups, both before and after surgery, paving the way for more personalised treatment approaches.
Collapse
Affiliation(s)
- Magali Svrcek
- Department of Pathology, APHP, Saint-Antoine Hospital, Sorbonne University, SIRIC CURAMUS, Paris, France
| | - Thibault Voron
- Digestive Surgery Department, APHP, Saint-Antoine Hospital, Sorbonne University, SIRIC CURAMUS, Paris, France
| | - Thierry André
- Department of Medical Oncology, APHP, Saint-Antoine Hospital, Sorbonne University, SIRIC CURAMUS, Paris, France
| | | | | |
Collapse
|
11
|
Hagi T, Shiraishi O, Nakanishi T, Kohda M, Hiraki Y, Kato H, Yasuda A, Shinkai M, Imano M, Yasuda T. Utility of Initial Tumor Reduction as a Prognostic Factor in Esophageal Squamous Cell Cancer Patients Undergoing Neoadjuvant Chemotherapy Followed by Surgery : A Retrospective Cohort Study. Ann Surg Oncol 2024; 31:5064-5074. [PMID: 38664331 DOI: 10.1245/s10434-024-15314-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/01/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND While a neoadjuvant chemotherapy regimen using docetaxel, cisplatin, and 5-fluorouracil (NAC-DCF) is considered the standard treatment for locally advanced esophageal cancer (EC) in Japan, a reliable marker for early prediction of treatment efficacy remains unclear. We investigated the utility of the tumor response after a first course of NAC-DCF as a post-surgery survival predictor in patients with EC. METHODS We enrolled 150 consecutive patients who underwent NAC-DCF followed by surgery for EC between September 2009 and January 2019. The initial tumor reduction (ITR), defined as the percentage decrease in the shorter diameter of the tumor after the first course of NAC-DCF, was evaluated using computed tomography. We analyzed the relationship between ITR, clinicopathological parameters, and survival. RESULTS The median ITR was 21.07% (range -11.45 to 50.13%). The optimal cut-off value for ITR for predicting prognosis was 10% (hazard ratio [HR] 3.30, 95% confidence interval [CI] 1.98-5.51), based on univariate logistic regression analyses for recurrence-free survival (RFS). Compared with patients with ITR <10%, patients with ITR ≥10% showed a significantly higher proportion of ypM0 (80.0% vs. 92.5%) and responders in terms of overall clinical response (50.0% vs. 80.8%). Multivariate analysis for RFS revealed that ypN2-3 (HR 2.78, 95% CI 1.67-4.62), non-response in terms of overall clinical response (HR 1.87, 95% CI 1.10-3.18), and ITR <10% (HR 2.48, 95% CI 1.42-4.32) were independent prognostic factors. CONCLUSIONS Tumor response after the first course of NAC-DCF may be a good predictor of survival in patients with EC who underwent NAC-DCF plus surgery.
Collapse
Affiliation(s)
- Takaomi Hagi
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Osamu Shiraishi
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan.
| | - Tomoya Nakanishi
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Masashi Kohda
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Yoko Hiraki
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Hiroaki Kato
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Atsushi Yasuda
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Masayuki Shinkai
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Motohiro Imano
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| | - Takushi Yasuda
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, Japan
| |
Collapse
|
12
|
Gelzinis TA. The Society of Thoracic Surgeons/American Society for Radiation Oncology/American Society of Clinical Oncology Recommendations on the Care of Patients With Localized Esophageal Cancers. J Cardiothorac Vasc Anesth 2024; 38:1445-1450. [PMID: 38658248 DOI: 10.1053/j.jvca.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 04/26/2024]
|
13
|
Solomon D, Deeb AL, Tarabine K, Xie Y, Mazzola E, Zhao L, Hammer MM, Jaklitsch MT, Swanson SJ, Bueno R, Wee JO. Predicting outcomes in esophageal adenocarcinoma following neoadjuvant chemoradiation: Interactions between tumor response and survival. J Thorac Cardiovasc Surg 2024; 168:278-289.e4. [PMID: 37967764 DOI: 10.1016/j.jtcvs.2023.11.015] [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: 03/30/2023] [Revised: 10/04/2023] [Accepted: 11/05/2023] [Indexed: 11/17/2023]
Abstract
OBJECTIVES The prognostic value of tumor regression scores (TRS) in patients with esophageal adenocarcinoma (EAC) who underwent neoadjuvant chemoradiation remains unclear. We sought to investigate the prognostic value of pathologic and metabolic treatment response among EAC patients undergoing neoadjuvant chemoradiation. METHODS Patients who underwent esophagectomy for EAC after neoadjuvant CROSS protocol between 2016 and 2020 were evaluated. TRS was grouped according to the modified Ryan score; metabolic response, according to the PERCIST criteria. Variables from endoscopic ultrasound, endoscopic biopsies, and positron emission tomography (primary and regional lymph node standardized uptake values [SUVs]) were collected. RESULTS The study population comprised 277 patients. A TRS of 0 (complete response) was identified in 66 patients (23.8%). Seventy-eight patients (28.1%) had TRS 1 (partial response), 97 (35%) had TRS 2 (poor response), and 36 (13%) had TRS 3 (no response). On survival analysis for overall survival (OS), patients with TRS 0 had longer survival compared to those with TRS 1, 2, or 3 (P = .010, P < .001, and P = .005, respectively). On multivariable logistic regression, the presence of signet ring cell features on endoscopic biopsy (odds ratio [OR], 7.54; P = .012) and greater SUV uptake at regional lymph nodes (OR, 1.42; P = .007) were significantly associated with residual tumor at pathology (TRS 1, 2, or 3). On multivariate Cox regression for predictors of OS, higher SUVmax at the most metabolically active nodal station (hazard ratio [HR], 1.08; P = .005) was independently associated with decreased OS, whereas pathologic complete response (HR, 0.61; P = .021) was independently associated with higher OS. CONCLUSIONS Patients with pathologic complete response had prolonged OS, whereas no difference in survival was detected among other TRS categories. At initial staging, the presence of signet ring cells and greater SUV uptake at regional lymph nodes predicted residual disease at pathology and shorter OS, suggesting the need for new treatment strategies for these patients.
Collapse
Affiliation(s)
- Daniel Solomon
- Division of Thoracic and Cardiac Surgery, Brigham and Women's Hospital, Boston, Mass.
| | - Ashley L Deeb
- Division of Thoracic and Cardiac Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Kamal Tarabine
- Department of Radiology, Brigham and Women's Hospital, Boston, Mass
| | - Yue Xie
- Department of Data Sciences, Dana Farber Cancer Institute, Boston, Mass
| | - Emanuele Mazzola
- Department of Data Sciences, Dana Farber Cancer Institute, Boston, Mass
| | - Lei Zhao
- Department of Pathology, Brigham and Women's Hospital, Boston, Mass
| | - Mark M Hammer
- Department of Radiology, Brigham and Women's Hospital, Boston, Mass
| | - Michael T Jaklitsch
- Division of Thoracic and Cardiac Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Scott J Swanson
- Division of Thoracic and Cardiac Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Raphael Bueno
- Division of Thoracic and Cardiac Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Jon O Wee
- Division of Thoracic and Cardiac Surgery, Brigham and Women's Hospital, Boston, Mass
| |
Collapse
|
14
|
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: 35] [Impact Index Per Article: 35.0] [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
|
15
|
Tchelebi LT, Goodman KA. Esophagogastric Cancer: The Current Role of Radiation Therapy. Hematol Oncol Clin North Am 2024; 38:569-583. [PMID: 38485552 DOI: 10.1016/j.hoc.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2024]
Abstract
Radiation therapy is an effective treatment modality in the management of patients with esophageal cancer regardless of tumor location (proximal, middle, or distal esophagus) or histology (squamous cell vs adenocarcinoma). The addition of neoadjuvant CRT to surgery in patients who are surgical candidates has consistently shown a benefit in terms of locoregional recurrence, pathologic downstaging, and overall survival. For patients who are not surgical candidates, CRT has a role as definitive treatment.
Collapse
Affiliation(s)
- Leila T Tchelebi
- Northwell, Lake Success, NY, USA; Department of Radiation Medicine, Northern Westchester Hospital, 400 East Main Street, Mount Kisco, NY 10549, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| | - Karyn A Goodman
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1128, New York, NY 10029-6574, USA. https://twitter.com/KarynAGoodman
| |
Collapse
|
16
|
Zamani-Siahkali N, Mirshahvalad SA, Farbod A, Divband G, Pirich C, Veit-Haibach P, Cook G, Beheshti M. SPECT/CT, PET/CT, and PET/MRI for Response Assessment of Bone Metastases. Semin Nucl Med 2024; 54:356-370. [PMID: 38172001 DOI: 10.1053/j.semnuclmed.2023.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 01/05/2024]
Abstract
Recent developments in hybrid SPECT/CT systems and the use of cadmium-zinc-telluride (CZT) detectors have improved the diagnostic accuracy of bone scintigraphy. These advancements have paved the way for novel quantitative approaches to accurate and reproducible treatment monitoring of bone metastases. PET/CT imaging using [18F]F-FDG and [18F]F-NaF have shown promising clinical utility in bone metastases assessment and monitoring response to therapy and prediction of treatment response in a broad range of malignancies. Additionally, specific tumor-targeting tracers like [99mTc]Tc-PSMA, [68Ga]Ga-PSMA, or [11C]C- or [18F]F-Choline revealed high diagnostic performance for early assessment and prognostication of bone metastases, particularly in prostate cancer. PET/MRI appears highly accurate imaging modality, but has associated limitations notably, limited availability, more complex logistics and high installation costs. Advances in artificial intelligence (Al) seem to improve the accuracy of imaging modalities and provide an assistant role in the evaluation of treatment response of bone metastases.
Collapse
Affiliation(s)
- Nazanin Zamani-Siahkali
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria; Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mirshahvalad
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria; Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, Toronto, Canada
| | - Abolfazl Farbod
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria; Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Christian Pirich
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Patrick Veit-Haibach
- Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, Toronto, Canada
| | - Gary Cook
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
17
|
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
|
18
|
Lee NY, Sherman EJ, Schöder H, Wray R, Boyle JO, Singh B, Grkovski M, Paudyal R, Cunningham L, Zhang Z, Hatzoglou V, Katabi N, Diplas BH, Han J, Imber BS, Pham K, Yu Y, Zakeri K, McBride SM, Kang JJ, Tsai CJ, Chen LC, Gelblum DY, Shah JP, Ganly I, Cohen MA, Cracchiolo JR, Morris LG, Dunn LA, Michel LS, Fetten JV, Kripani A, Pfister DG, Ho AL, Shukla-Dave A, Humm JL, Powell SN, Li BT, Reis-Filho JS, Diaz LA, Wong RJ, Riaz N. Hypoxia-Directed Treatment of Human Papillomavirus-Related Oropharyngeal Carcinoma. J Clin Oncol 2024; 42:940-950. [PMID: 38241600 PMCID: PMC10927322 DOI: 10.1200/jco.23.01308] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/18/2023] [Accepted: 11/08/2023] [Indexed: 01/21/2024] Open
Abstract
PURPOSE Standard curative-intent chemoradiotherapy for human papillomavirus (HPV)-related oropharyngeal carcinoma results in significant toxicity. Since hypoxic tumors are radioresistant, we posited that the aerobic state of a tumor could identify patients eligible for de-escalation of chemoradiotherapy while maintaining treatment efficacy. METHODS We enrolled patients with HPV-related oropharyngeal carcinoma to receive de-escalated definitive chemoradiotherapy in a phase II study (ClinicalTrials.gov identifier: NCT03323463). Patients first underwent surgical removal of disease at their primary site, but not of gross disease in the neck. A baseline 18F-fluoromisonidazole positron emission tomography scan was used to measure tumor hypoxia and was repeated 1-2 weeks intratreatment. Patients with nonhypoxic tumors received 30 Gy (3 weeks) with chemotherapy, whereas those with hypoxic tumors received standard chemoradiotherapy to 70 Gy (7 weeks). The primary objective was achieving a 2-year locoregional control (LRC) of 95% with a 7% noninferiority margin. RESULTS One hundred fifty-eight patients with T0-2/N1-N2c were enrolled, of which 152 patients were eligible for analyses. Of these, 128 patients met criteria for 30 Gy and 24 patients received 70 Gy. The 2-year LRC was 94.7% (95% CI, 89.8 to 97.7), meeting our primary objective. With a median follow-up time of 38.3 (range, 22.1-58.4) months, the 2-year progression-free survival (PFS) and overall survival (OS) rates were 94% and 100%, respectively, for the 30-Gy cohort. The 70-Gy cohort had similar 2-year PFS and OS rates at 96% and 96%, respectively. Acute grade 3-4 adverse events were more common in 70 Gy versus 30 Gy (58.3% v 32%; P = .02). Late grade 3-4 adverse events only occurred in the 70-Gy cohort, in which 4.5% complained of late dysphagia. CONCLUSION Tumor hypoxia is a promising approach to direct dosing of curative-intent chemoradiotherapy for HPV-related carcinomas with preserved efficacy and substantially reduced toxicity that requires further investigation.
Collapse
Affiliation(s)
- Nancy Y. Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Eric J. Sherman
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - HeiKo Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Rick Wray
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jay O. Boyle
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bhuvanesh Singh
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Milan Grkovski
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ramesh Paudyal
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Louise Cunningham
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Zhigang Zhang
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Vaios Hatzoglou
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nora Katabi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bill H. Diplas
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - James Han
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Brandon S. Imber
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Khoi Pham
- Department of Finance, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yao Yu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kaveh Zakeri
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sean M. McBride
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jung J. Kang
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - C. Jillian Tsai
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Linda C. Chen
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daphna Y. Gelblum
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jatin P. Shah
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ian Ganly
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marc A. Cohen
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Luc G.T. Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lara A. Dunn
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Loren S. Michel
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - James V. Fetten
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anuja Kripani
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David G. Pfister
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alan L. Ho
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Amita Shukla-Dave
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - John L. Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Simon N. Powell
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bob T. Li
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jorge S. Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Luis A. Diaz
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Richard J. Wong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
19
|
Sun J, Sheng J, Zhang LJ. Gastrointestinal tract. TRANSPATHOLOGY 2024:281-296. [DOI: 10.1016/b978-0-323-95223-1.00005-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
20
|
Worrell SG, Goodman KA, Altorki NK, Ashman JB, Crabtree TD, Dorth J, Firestone S, Harpole DH, Hofstetter WL, Hong TS, Kissoon K, Ku GY, Molena D, Tepper JE, Watson TJ, Williams T, Willett C. The Society of Thoracic Surgeons/American Society for Radiation Oncology Updated Clinical Practice Guidelines on Multimodality Therapy for Locally Advanced Cancer of the Esophagus or Gastroesophageal Junction. Pract Radiat Oncol 2024; 14:28-46. [PMID: 37921736 DOI: 10.1016/j.prro.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 11/04/2023]
Abstract
Outcomes for patients with esophageal cancer have improved over the last decade with the implementation of multimodality therapy. There are currently no comprehensive guidelines addressing multidisciplinary management of esophageal cancer that have incorporated the input of surgeons, radiation oncologists, and medical oncologists. To address the need for multidisciplinary input in the management of esophageal cancer and to meet current best practices for clinical practice guidelines, the current guidelines were created as a collaboration between The Society of Thoracic Surgeons (STS), American Society for Radiation Oncology (ASTRO), and the American Society of Clinical Oncology (ASCO). Physician representatives chose 8 key clinical questions pertinent to the care of patients with locally advanced, resectable thoracic esophageal cancer (excluding cervical location). A comprehensive literature review was performed identifying 227 articles that met the inclusion criteria covering the use of induction chemotherapy, chemotherapy vs chemoradiotherapy before surgery, optimal radiation dose, the value of esophagectomy, timing of esophagectomy, the approach and extent of lymphadenectomy, the use of minimally invasive esophagectomy, and the value of adjuvant therapy after resection. The relevant data were reviewed and voted on by the panel with 80% of the authors, with 75% agreement on class and level of evidence. These data were then complied into the guidelines document.
Collapse
Affiliation(s)
- Stephanie G Worrell
- Section of Thoracic Surgery, Department of Surgery, University of Arizona College of Medicine, Tucson, Arizona.
| | - Karyn A Goodman
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nasser K Altorki
- Division of Thoracic Surgery, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, New York
| | | | - Traves D Crabtree
- Division of Cardiothoracic Surgery, Department of Surgery, Southern Illinois University School of Medicine, Springfield, Illinois
| | - Jennifer Dorth
- Department of Radiation Oncology, Seidman Cancer Center, University Hospitals, Cleveland, Ohio
| | | | - David H Harpole
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Wayne L Hofstetter
- Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Geoffrey Y Ku
- Gastrointestinal Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniela Molena
- Division of Thoracic Surgery, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joel E Tepper
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, North Carolina
| | - Thomas J Watson
- Thoracic Surgery Group, Beaumont Health, Royal Oak, Michigan
| | - Terence Williams
- Department of Radiation Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California
| | - Christopher Willett
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
21
|
Worrell SG, Goodman KA, Altorki NK, Ashman JB, Crabtree TD, Dorth J, Firestone S, Harpole DH, Hofstetter WL, Hong TS, Kissoon K, Ku GY, Molena D, Tepper JE, Watson TJ, Williams T, Willett C. The Society of Thoracic Surgeons/American Society for Radiation Oncology Updated Clinical Practice Guidelines on Multimodality Therapy for Locally Advanced Cancer of the Esophagus or Gastroesophageal Junction. Ann Thorac Surg 2024; 117:15-32. [PMID: 37921794 DOI: 10.1016/j.athoracsur.2023.09.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/23/2023] [Accepted: 09/05/2023] [Indexed: 11/04/2023]
Abstract
Outcomes for patients with esophageal cancer have improved over the last decade with the implementation of multimodality therapy. There are currently no comprehensive guidelines addressing multidisciplinary management of esophageal cancer that have incorporated the input of surgeons, radiation oncologists, and medical oncologists. To address the need for multidisciplinary input in the management of esophageal cancer and to meet current best practices for clinical practice guidelines, the current guidelines were created as a collaboration between The Society of Thoracic Surgeons (STS), American Society for Radiation Oncology (ASTRO), and the American Society of Clinical Oncology (ASCO). Physician representatives chose 8 key clinical questions pertinent to the care of patients with locally advanced, resectable thoracic esophageal cancer (excluding cervical location). A comprehensive literature review was performed identifying 227 articles that met the inclusion criteria covering the use of induction chemotherapy, chemotherapy vs chemoradiotherapy before surgery, optimal radiation dose, the value of esophagectomy, timing of esophagectomy, the approach and extent of lymphadenectomy, the use of minimally invasive esophagectomy, and the value of adjuvant therapy after resection. The relevant data were reviewed and voted on by the panel with 80% of the authors, with 75% agreement on class and level of evidence. These data were then complied into the guidelines document.
Collapse
Affiliation(s)
- Stephanie G Worrell
- Section of Thoracic Surgery, Department of Surgery, University of Arizona College of Medicine, Tucson, Arizona.
| | - Karyn A Goodman
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nasser K Altorki
- Division of Thoracic Surgery, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, New York
| | | | - Traves D Crabtree
- Division of Cardiothoracic Surgery, Department of Surgery, Southern Illinois University School of Medicine, Springfield, Illinois
| | - Jennifer Dorth
- Department of Radiation Oncology, Seidman Cancer Center, University Hospitals, Cleveland, Ohio
| | | | - David H Harpole
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Wayne L Hofstetter
- Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Geoffrey Y Ku
- Gastrointestinal Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniela Molena
- Division of Thoracic Surgery, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joel E Tepper
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, North Carolina
| | - Thomas J Watson
- Thoracic Surgery Group, Beaumont Health, Royal Oak, Michigan
| | - Terence Williams
- Department of Radiation Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California
| | - Christopher Willett
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
22
|
Veziant J, Bouché O, Aparicio T, Barret M, El Hajbi F, Lepilliez V, Lesueur P, Maingon P, Pannier D, Quero L, Raoul JL, Renaud F, Seitz JF, Serre AA, Vaillant E, Vermersch M, Voron T, Tougeron D, Piessen G. Esophageal cancer - French intergroup clinical practice guidelines for diagnosis, treatments and follow-up (TNCD, SNFGE, FFCD, GERCOR, UNICANCER, SFCD, SFED, SFRO, ACHBT, SFP, RENAPE, SNFCP, AFEF, SFR). Dig Liver Dis 2023; 55:1583-1601. [PMID: 37635055 DOI: 10.1016/j.dld.2023.07.015] [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: 04/19/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION This document is a summary of the French intergroup guidelines regarding the management of esophageal cancer (EC) published in July 2022, available on the website of the French Society of Gastroenterology (SNFGE) (www.tncd.org). METHODS This collaborative work was conducted under the auspices of several French medical and surgical societies involved in the management of EC. Recommendations were graded in three categories (A, B and C), according to the level of evidence found in the literature until April 2022. RESULTS EC diagnosis and staging evaluation are mainly based on patient's general condition assessment, endoscopy plus biopsies, TAP CT-scan and 18F FDG-PET. Surgery alone is recommended for early-stage EC, while locally advanced disease (N+ and/or T3-4) is treated with perioperative chemotherapy (FLOT) or preoperative chemoradiation (CROSS regimen) followed by immunotherapy for adenocarcinoma. Preoperative chemoradiation (CROSS regimen) followed by immunotherapy or definitive chemoradiation with the possibility of organ preservation are the two options for squamous cell carcinoma. Salvage surgery is recommended for incomplete response or recurrence after definitive chemoradiation and should be performed in an expert center. Treatment for metastatic disease is based on systemic therapy including chemotherapy, immunotherapy or combined targeted therapy according to biomarkers testing such as HER2 status, MMR status and PD-L1 expression. CONCLUSION These guidelines are intended to provide a personalised therapeutic strategy for daily clinical practice and are subject to ongoing optimization. Each individual case should be discussed by a multidisciplinary team.
Collapse
Affiliation(s)
- Julie Veziant
- Department of Digestive and Oncological Surgery, Claude Huriez Hospital, CHU Lille, University of Lille, Lille F-59000, France.
| | - Olivier Bouché
- Department of Digestive Oncology, CHU Reims, Reims, France
| | - T Aparicio
- Department of Gastroenterology and Digestive Oncology, AP-HP, Saint-Louis Hospital, Paris, France
| | - M Barret
- Gastroenterology Department, Cochin Hospital, APHP, Paris, France
| | - F El Hajbi
- Department of Oncology, Centre Oscar Lambret, Lille, France
| | - V Lepilliez
- Gastroenterology Department, Jean Mermoz Private Hospital, Ramsay Santé, Lyon, France
| | - P Lesueur
- Department of Radiation Oncology, Centre Guillaume le Conquérant, Le Havre, France
| | - P Maingon
- Department of Radiation Oncology, La Pitié-Salpêtrière, APHP, Sorbonne University, Paris, France
| | - D Pannier
- Department of Oncology, Centre Oscar Lambret, Lille, France
| | - L Quero
- Department of Radiation Oncology, Saint-Louis Hospital, APHP, Paris, France
| | - J L Raoul
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - F Renaud
- Department of Pathology, La Pitié-Salpêtrière, APHP, Sorbonne University, Paris, France
| | - J F Seitz
- Department of Digestive Oncology, La Timone, Aix Marseille Université, Marseille, France
| | - A A Serre
- Department of Radiotherapy, Centre Léon Bérard, Lyon, France
| | | | - M Vermersch
- Medical Imaging Department, Valencienne Hospital Centre, Valencienne 59300, France
| | - T Voron
- Department of General and Digestive Surgery, Sorbonne Université, AP-HP, Hôpital Saint Antoine, 184 rue du faubourg Saint-Antoine, Paris 75012, France
| | - D Tougeron
- Department of Gastro-Enterology and Hepatology, Poitiers University Hospital, Poitiers, France
| | - Guillaume Piessen
- Department of Digestive and Oncological Surgery, Claude Huriez Hospital, CHU Lille, University of Lille, Lille F-59000, France
| |
Collapse
|
23
|
Wada N, Kurokawa Y, Takahashi T, Saito T, Yamamoto K, Yamashita K, Tanaka K, Makino T, Nakajima K, Tatsumi M, Eguchi H, Doki Y. Accurate Preoperative Evaluation of the Location of Esophagogastric Junction Adenocarcinoma Using Positron Emission Tomography-Computed Tomography. Am Surg 2023; 89:6005-6012. [PMID: 37312037 DOI: 10.1177/00031348231183120] [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: 06/15/2023]
Abstract
BACKGROUND The surgical procedure for esophagogastric junction (EGJ) adenocarcinoma usually depends on the location of its epicenter or proximal margin, but accurate evaluation of these positions is often difficult. The usefulness of positron emission tomography-computed tomography (PET-CT) for this purpose is unknown. METHODS Between June 2005 and February 2015, we enrolled 30 patients with cT2-4 EGJ adenocarcinoma (Siewert type I/II) who underwent surgical resection. We ascertained the sensitivity and specificity of preoperative PET-CT for identifying the primary tumor and regional lymph node metastasis, and compared PET-CT and pathological findings in terms of the distance from the EGJ to the tumor epicenter or proximal tumor margin. RESULTS PET-CT detected the primary tumor with a sensitivity of 97% (29/30), and detected lymph node metastasis with a sensitivity and specificity of 22% (4/18) and 100% (8/8), respectively. No significant association was observed between the maximal standardized uptake value and histological type, tumor size, or pT status. Regarding the accuracy of evaluating tumor location, the median differences between PET-CT and pathological measurements were .6 cm for the tumor epicenter and .5 cm for the proximal margin from the EGJ. PET-CT and pathological findings showed agreement regarding Siewert classification type (I or II) and lengths of esophageal involvement exceeding 4 cm or 2 cm in 77% (10/13), 85% (11/13), and 85% (11/13) of cases, respectively. DISCUSSION PET-CT had high sensitivity for primary EGJ adenocarcinoma. It may effectively locate the tumor epicenter and proximal margin and thus help clinicians determine the optimal surgical procedure.
Collapse
Affiliation(s)
- Noriko Wada
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Surgery, Ikeda City Hospital, Osaka, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takuro Saito
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuyoshi Yamamoto
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kotaro Yamashita
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koji Tanaka
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoki Makino
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kiyokazu Nakajima
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Mitsuaki Tatsumi
- Department of Radiology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
24
|
Byun AJ, Grosser RA, Choe JK, Rizk NP, Tang LH, Molena D, Tan KS, Restle D, Cheema W, Zhu A, Gerdes H, Markowitz AJ, Bains MS, Rusch VW, Jones DR, Adusumilli PS. A Prospective Clinical Trial to Evaluate Mesothelin as a Biomarker for the Clinical Management of Patients With Esophageal Adenocarcinoma. Ann Surg 2023; 278:e1003-e1010. [PMID: 37185875 PMCID: PMC10593105 DOI: 10.1097/sla.0000000000005885] [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] [Indexed: 05/17/2023]
Abstract
OBJECTIVE To investigate the utility of serum soluble mesothelin-related peptide (SMRP) and tumor mesothelin expression in the management of esophageal adenocarcinoma (ADC). BACKGROUND Clinical management of esophageal ADC is limited by a lack of accurate evaluation of tumor burden, treatment response, and disease recurrence. Our retrospective data showed that tumor mesothelin and its serum correlate, SMRP, are overexpressed and associated with poor outcomes in patients with esophageal ADC. METHODS Serum SMRP and tumoral mesothelin expression from 101 patients with locally advanced esophageal ADC were analyzed before induction chemoradiation (pretreatment) and at the time of resection (posttreatment), as a biomarker for treatment response, disease recurrence, and overall survival (OS). RESULTS Pre and posttreatment serum SMRP was ≥1 nM in 49% and 53%, and pre and post-treatment tumor mesothelin expression was >25% in 35% and 46% of patients, respectively. Pretreatment serum SMRP was not significantly associated with tumor stage ( P = 0.9), treatment response (radiologic response, P = 0.4; pathologic response, P = 0.7), or recurrence ( P =0.229). Pretreatment tumor mesothelin expression was associated with OS (hazard ratio: 2.08; 95% CI: 1.14-3.79; P = 0.017) but had no statistically significant association with recurrence ( P = 0.9). Three-year OS of patients with pretreatment tumor mesothelin expression of ≤25% was 78% (95% CI: 68%-89%), compared with 49% (95% CI: 35%-70%) among those with >25%. CONCLUSIONS Pretreatment tumor mesothelin expression is prognostic of OS for patients with locally advanced esophageal ADC, whereas serum SMRP is not a reliable biomarker for monitoring treatment response or recurrence.
Collapse
Affiliation(s)
- Alexander J. Byun
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rachel A. Grosser
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jennie K. Choe
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nabil P. Rizk
- Division of Thoracic Surgery, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Laura H. Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniela Molena
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kay See Tan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David Restle
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Waseem Cheema
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amy Zhu
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hans Gerdes
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Arnold J. Markowitz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Manjit S. Bains
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Valerie W. Rusch
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David R. Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Prasad S. Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
25
|
Withey SJ, Owczarczyk K, Grzeda MT, Yip C, Deere H, Green M, Maisey N, Davies AR, Cook GJ, Goh V. Association of dynamic contrast-enhanced MRI and 18F-Fluorodeoxyglucose PET/CT parameters with neoadjuvant therapy response and survival in esophagogastric cancer. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2023; 49:106934. [PMID: 37183047 PMCID: PMC10769883 DOI: 10.1016/j.ejso.2023.05.009] [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: 07/27/2022] [Revised: 04/17/2023] [Accepted: 05/09/2023] [Indexed: 05/16/2023]
Abstract
INTRODUCTION Better predictive markers are needed to deliver individualized care for patients with primary esophagogastric cancer. This exploratory study aimed to assess whether pre-treatment imaging parameters from dynamic contrast-enhanced MRI and 18F-fluorodeoxyglucose (18F-FDG) PET/CT are associated with response to neoadjuvant therapy or outcome. MATERIALS AND METHODS Following ethical approval and informed consent, prospective participants underwent dynamic contrast-enhanced MRI and 18F-FDG PET/CT prior to neoadjuvant chemotherapy/chemoradiotherapy ± surgery. Vascular dynamic contrast-enhanced MRI and metabolic 18F-FDG PET parameters were compared by tumor characteristics using Mann Whitney U test and with pathological response (Mandard tumor regression grade), recurrence-free and overall survival using logistic regression modelling, adjusting for predefined clinical variables. RESULTS 39 of 47 recruited participants (30 males; median age 65 years, IQR: 54, 72 years) were included in the final analysis. The tumor vascular-metabolic ratio was higher in patients remaining node positive following neoadjuvant therapy (median tumor peak enhancement/SUVmax ratio: 0.052 vs. 0.023, p = 0.02). In multivariable analysis adjusted for age, gender, pre-treatment tumor and nodal stage, peak enhancement (highest gadolinium concentration value prior to contrast washout) was associated with pathological tumor regression grade. The odds of response decreased by 5% for each 0.01 unit increase (OR 0.95; 95% CI: 0.90, 1.00, p = 0.04). No 18F-FDG PET/CT parameters were predictive of pathological tumor response. No relationships between pre-treatment imaging and survival were identified. CONCLUSION Pre-treatment esophagogastric tumor vascular and metabolic parameters may provide additional information in assessing response to neoadjuvant therapy.
Collapse
Affiliation(s)
- Samuel J Withey
- Department of Radiology, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, United Kingdom
| | - Kasia Owczarczyk
- Department of Clinical Oncology, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, United Kingdom
| | - Mariusz T Grzeda
- School of Biomedical Engineering & Imaging Sciences, King's College London, United Kingdom
| | - Connie Yip
- Department of Radiation Oncology, National Cancer Centre, Singapore
| | - Harriet Deere
- Department of Pathology, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, United Kingdom
| | - Mike Green
- Department of Pathology, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, United Kingdom
| | - Nick Maisey
- Department of Medical Oncology, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, United Kingdom
| | - Andrew R Davies
- Department of Surgery, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, United Kingdom
| | - Gary J Cook
- School of Biomedical Engineering & Imaging Sciences, King's College London, United Kingdom; The King's College London and Guy's and St Thomas' PET Centre, St Thomas' Hospital, London, United Kingdom
| | - Vicky Goh
- Department of Radiology, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, United Kingdom; School of Biomedical Engineering & Imaging Sciences, King's College London, United Kingdom.
| |
Collapse
|
26
|
Sinnamon AJ, Mehta R, Saeed S, Lauwers GY, Palm RF, Frakes JM, Hoffe SE, Baldonado JJ, Fontaine JP, Pimiento JM. Induction Fluorouracil-Based Chemotherapy and PET-Adapted Consolidation Chemoradiation with Esophagectomy for High-Risk Gastroesophageal Adenocarcinoma. Cancers (Basel) 2023; 15:4375. [PMID: 37686650 PMCID: PMC10486661 DOI: 10.3390/cancers15174375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/11/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Background: Neoadjuvant chemoradiation with esophagectomy is standard management for locally advanced esophageal adenocarcinoma. Induction chemotherapy with a tailored approach to chemoradiation based on metabolic response to therapy on PET was explored as an alternative strategy in the CALGB 80803 trial. We sought to describe real-world institutional experience implementing this approach outside of a clinical trial. Methods: Patients who were treated with induction fluorouracil-leucovorin-oxaliplatin (FOLFOX) or fluorouracil-leucovorin-oxaliplatin-docetaxel (FLOT) with tailored chemoradiation based on PET response and subsequent esophagectomy were identified from a prospectively maintained database. Primary outcomes were pathologic complete response (pCR) and overall survival (OS) following completion of all therapy. Results: There were 35 patients who completed induction chemotherapy, chemoradiation, and esophagectomy. Thirty-three completed restaging PET following induction chemotherapy with metabolic response seen in 76% (n = 25/33). The pCR rate was 31% (n = 11/35) and the ypN0 rate was 71% (n = 25/35). Among the patients who demonstrated metabolic response to induction FOLFOX/FLOT and subsequently continued fluorouracil-based chemoradiation, the pCR rate was 39% (n = 9/23). The rate of pathologically negative lymph nodes in this group was high (n = 19/23, 83%) with 100% R0 resection rate (n = 23/23). With the median follow-up of 43 months, the median OS was not reached for this group and was significantly longer than the OS for the remainder of the cohort (p = 0.027, p = 0.046 adjusted for clinical stage). Conclusions: Induction FOLFOX/FLOT chemotherapy with evaluation of sensitivity via metabolic response and tailored chemoradiation seems to lead to high pCR and ypN0 rates in high-risk patients with adenocarcinoma of the esophagus and GE junction. This approach in clinical practice seems to recapitulate encouraging results in clinical trials.
Collapse
Affiliation(s)
- Andrew J Sinnamon
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Rutika Mehta
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Samir Saeed
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Gregory Y Lauwers
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Russell F Palm
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Jessica M Frakes
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Sarah E Hoffe
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Jobelle J Baldonado
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jacques P Fontaine
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jose M Pimiento
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
27
|
Hörst F, Ting S, Liffers ST, Pomykala KL, Steiger K, Albertsmeier M, Angele MK, Lorenzen S, Quante M, Weichert W, Egger J, Siveke JT, Kleesiek J. Histology-Based Prediction of Therapy Response to Neoadjuvant Chemotherapy for Esophageal and Esophagogastric Junction Adenocarcinomas Using Deep Learning. JCO Clin Cancer Inform 2023; 7:e2300038. [PMID: 37527475 DOI: 10.1200/cci.23.00038] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/27/2023] [Accepted: 06/07/2023] [Indexed: 08/03/2023] Open
Abstract
PURPOSE Quantifying treatment response to gastroesophageal junction (GEJ) adenocarcinomas is crucial to provide an optimal therapeutic strategy. Routinely taken tissue samples provide an opportunity to enhance existing positron emission tomography-computed tomography (PET/CT)-based therapy response evaluation. Our objective was to investigate if deep learning (DL) algorithms are capable of predicting the therapy response of patients with GEJ adenocarcinoma to neoadjuvant chemotherapy on the basis of histologic tissue samples. METHODS This diagnostic study recruited 67 patients with I-III GEJ adenocarcinoma from the multicentric nonrandomized MEMORI trial including three German university hospitals TUM (University Hospital Rechts der Isar, Munich), LMU (Hospital of the Ludwig-Maximilians-University, Munich), and UME (University Hospital Essen, Essen). All patients underwent baseline PET/CT scans and esophageal biopsy before and 14-21 days after treatment initiation. Treatment response was defined as a ≥35% decrease in SUVmax from baseline. Several DL algorithms were developed to predict PET/CT-based responders and nonresponders to neoadjuvant chemotherapy using digitized histopathologic whole slide images (WSIs). RESULTS The resulting models were trained on TUM (n = 25 pretherapy, n = 47 on-therapy) patients and evaluated on our internal validation cohort from LMU and UME (n = 17 pretherapy, n = 15 on-therapy). Compared with multiple architectures, the best pretherapy network achieves an area under the receiver operating characteristic curve (AUROC) of 0.81 (95% CI, 0.61 to 1.00), an area under the precision-recall curve (AUPRC) of 0.82 (95% CI, 0.61 to 1.00), a balanced accuracy of 0.78 (95% CI, 0.60 to 0.94), and a Matthews correlation coefficient (MCC) of 0.55 (95% CI, 0.18 to 0.88). The best on-therapy network achieves an AUROC of 0.84 (95% CI, 0.64 to 1.00), an AUPRC of 0.82 (95% CI, 0.56 to 1.00), a balanced accuracy of 0.80 (95% CI, 0.65 to 1.00), and a MCC of 0.71 (95% CI, 0.38 to 1.00). CONCLUSION Our results show that DL algorithms can predict treatment response to neoadjuvant chemotherapy using WSI with high accuracy even before therapy initiation, suggesting the presence of predictive morphologic tissue biomarkers.
Collapse
Affiliation(s)
- Fabian Hörst
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany
- Cancer Research Center Cologne Essen (CCCE), West German Cancer Center Essen, University Hospital Essen (AöR), Essen, Germany
| | - Saskia Ting
- Institute of Pathology, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany
- Current address: Institute of Pathology Nordhessen, Kassel, Germany
| | - Sven-Thorsten Liffers
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center Essen, University Hospital Essen (AöR), Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner site Essen) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kelsey L Pomykala
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany
| | - Katja Steiger
- Institute of Pathology, Technical University of Munich (TUM), Munich, Germany
| | - Markus Albertsmeier
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Martin K Angele
- Department of General, Visceral and Transplantation Surgery, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Sylvie Lorenzen
- Clinic for Internal Medicine III, University Hospital rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Michael Quante
- Clinic for Internal Medicine II, Gastrointestinal Oncology, University Medical Center of Freiburg, Freiburg, Germany
- Department of Internal Medicine II, University Hospital rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich (TUM), Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Egger
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany
- Cancer Research Center Cologne Essen (CCCE), West German Cancer Center Essen, University Hospital Essen (AöR), Essen, Germany
| | - Jens T Siveke
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center Essen, University Hospital Essen (AöR), Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner site Essen) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen (AöR), Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - Jens Kleesiek
- Institute for Artificial Intelligence in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany
- Cancer Research Center Cologne Essen (CCCE), West German Cancer Center Essen, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK, Partner site Essen), Heidelberg, Germany
| |
Collapse
|
28
|
Tsagkalidis V, Blaszczyk MB, In H. Interpretation of Tumor Response Grade following Preoperative Therapy for Gastric Cancer: An Overview. Cancers (Basel) 2023; 15:3662. [PMID: 37509323 PMCID: PMC10377503 DOI: 10.3390/cancers15143662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Gastric cancer is among the top five causes of cancer-related death worldwide. Preoperative chemotherapy has been established as an option in patients with locally advanced gastric cancer. However, chemotherapy yields variable results, owing to the cellular and molecular heterogeneity of this disease. Identifying patients who did or did not respond to preoperative therapy can allow clinicians to alter treatment modalities and provide important information related to prognostication. A pathologic response to preoperative therapies, called the Tumor Response Grade (TRG), has been evaluated to quantify treatment response. Multiple systems for TRG have been established. However, the literature has demonstrated inconsistent results for TRG systems and prognosis, possibly due to variability in interpretation of tumor response between systems and interobserver variability. Radiographic responses to preoperative therapies using RECIST 1.1 criteria and endoscopically assessed tumor response have demonstrated association with survival; however, their use in gastric cancer remains challenging given the inability to accurately and consistently identify and measure the tumor, especially in the setting of neoadjuvant therapy, where treatment-related changes can obscure the gastric wall layers. While the response to preoperative therapies with positron emission tomography (PET) has shown promising results in esophageal and esophagogastric junction (EGJ) malignancies, its role in gastric cancer is still under investigation. This review is focused on summarizing the available literature related to evaluating TRG in gastric cancer, as well as providing a brief overview of the use of radiographic and endoscopic methods to assess response to preoperative therapies. Lastly, we outline future directions regarding the use of a universal TRG system to guide care and assist with prognosis.
Collapse
Affiliation(s)
- Vasileios Tsagkalidis
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Maryjka B Blaszczyk
- Department of Pathology and Laboratory Medicine, Rutgers Biomedical and Health Sciences, New Brunswick, NJ 08901, USA
| | - Haejin In
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Department of Health Behavior, Society and Policy, Rutgers School of Public Health, Piscataway, NJ 08854, USA
| |
Collapse
|
29
|
Mukherjee S, Hurt CN, Adams R, Bateman A, Bradley KM, Bridges S, Falk S, Griffiths G, Gwynne S, Jones CM, Markham PJ, Maughan T, Nixon LS, Radhakrishna G, Roy R, Schoenbuchner S, Sheikh H, Spezi E, Hawkins M, Crosby TD. Efficacy of early PET-CT directed switch to carboplatin and paclitaxel based definitive chemoradiotherapy in patients with oesophageal cancer who have a poor early response to induction cisplatin and capecitabine in the UK: a multi-centre randomised controlled phase II trial. EClinicalMedicine 2023; 61:102059. [PMID: 37409323 PMCID: PMC10318451 DOI: 10.1016/j.eclinm.2023.102059] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 07/07/2023] Open
Abstract
Background The utility of early metabolic response assessment to guide selection of the systemic component of definitive chemoradiotherapy (dCRT) for oesophageal cancer is uncertain. Methods In this multi-centre, randomised, open-label, phase II substudy of the radiotherapy dose-escalation SCOPE2 trial we evaluated the role of 18F-Fluorodeoxyglucose positron emission tomography (PET) at day 14 of cycle 1 of three-weekly induction cis/cap (cisplatin (60 mg/m2)/capecitabine (625 mg/m2 days 1-21)) in patients with oesophageal squamous cell carcinoma (OSCC) or adenocarcinoma (OAC). Non-responders, who had a less than 35% reduction in maximum standardised uptake value (SUVmax) from pre-treatment baseline, were randomly assigned to continue cis/cap or switch to car/pac (carboplatin AUC 5/paclitaxel 175 mg/m2) for a further induction cycle, then concurrently with radiotherapy over 25 fractions. Responders continued cis/cap for the duration of treatment. All patients (including responders) were randomised to standard (50Gy) or high (60Gy) dose radiation as part of the main study. Primary endpoint for the substudy was treatment failure-free survival (TFFS) at week 24. The trial was registered with International Standard Randomized Controlled Trial Number 97125464 and ClinicalTrials.govNCT02741856. Findings This substudy was closed on 1st August 2021 by the Independent Data Monitoring Committee on the grounds of futility and possible harm. To this point from 22nd November 2016, 103 patients from 16 UK centres had participated in the PET-CT substudy; 63 (61.2%; 52/83 OSCC, 11/20 OAC) of whom were non-responders. Of these, 31 were randomised to car/pac and 32 to remain on cis/cap. All patients were followed up until at least 24 weeks, at which point in OSCC both TFFS (25/27 (92.6%) vs 17/25 (68%); p = 0.028) and overall survival (42.5 vs. 20.4 months, adjusted HR 0.36; p = 0.018) favoured cis/cap over car/pac. There was a trend towards worse survival in OSCC + OAC cis/cap responders (33.6 months; 95%CI 23.1-nr) vs. non-responders (42.5 (95%CI 27.0-nr) months; HR = 1.43; 95%CI 0.67-3.08; p = 0.35). Interpretation In OSCC, early metabolic response assessment is not prognostic for TFFS or overall survival and should not be used to personalise systemic therapy in patients receiving dCRT. Funding Cancer Research UK.
Collapse
Affiliation(s)
- Somnath Mukherjee
- Oxford Cancer Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Christopher N. Hurt
- Centre for Trials Research, Cardiff University, Cardiff, UK
- Southampton Clinical Trials Unit, University of Southampton, Southampton, UK
| | - Richard Adams
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Andrew Bateman
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Kevin M. Bradley
- Wales Research and Diagnostic Positron Emission Tomography Centre (PETIC), Cardiff University, Cardiff, UK
| | - Sarah Bridges
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Stephen Falk
- University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Gareth Griffiths
- Southampton Clinical Trials Unit, University of Southampton, Southampton, UK
| | - Sarah Gwynne
- South West Wales Cancer Centre, Swansea Bay University Health Board, Swansea, UK
| | | | | | - Tim Maughan
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | | | - Ganesh Radhakrishna
- The Christie Hospital, The Christie Hospitals NHS Foundation Trust, Manchester, UK
| | - Rajarshi Roy
- Queen's Centre for Oncology, Hull University Teaching Hospitals NHS Trust, UK
| | | | - Hamid Sheikh
- The Christie Hospital, The Christie Hospitals NHS Foundation Trust, Manchester, UK
| | | | - Maria Hawkins
- Department of Medical Physics & Biomedical Engineering, University College London, London, UK
| | | |
Collapse
|
30
|
Cinicola J, Mamidanna S, Yegya-Raman N, Spencer K, Deek MP, Jabbour SK. A Review of Advances in Radiotherapy in the Setting of Esophageal Cancers. Surg Oncol Clin N Am 2023; 32:433-459. [PMID: 37182986 DOI: 10.1016/j.soc.2023.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Esophageal cancer is the eighth most common cancer worldwide and is the sixth most common cause of cancer-related mortality. The paradigm has shifted to include a multimodality approach with surgery, chemotherapy, targeted therapy (including immunotherapy), and radiation therapy. Advances in radiotherapy through techniques such as intensity modulated radiotherapy and proton beam therapy have allowed for the more dose homogeneity and improved organ sparing. In addition, recent studies of targeted therapies and predictive approaches in patients with locally advanced disease provide clinicians with new approaches to modify multimodality treatment to improve clinical outcomes.
Collapse
Affiliation(s)
- Joshua Cinicola
- Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Swati Mamidanna
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson School of Medicine, Rutgers University, New Brunswick, NJ, USA
| | - Nikhil Yegya-Raman
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristen Spencer
- New York Langone Perlmutter Cancer Center, New York, NY, USA
| | - Matthew P Deek
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson School of Medicine, Rutgers University, New Brunswick, NJ, USA
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson School of Medicine, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
31
|
Authors, und die Mitarbeiter der Leitlinienkommission, Collaborators:. S3-Leitlinie Diagnostik und Therapie der Plattenepithelkarzinome und Adenokarzinome des Ösophagus. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2023; 61:e209-e307. [PMID: 37285869 DOI: 10.1055/a-1771-6953] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
|
32
|
Cowzer D, Keane F, Ku GY. Clinical Utility of 18F-2-Fluoro-deoxy-d-glucose PET Imaging in Locally Advanced Esophageal/Gastroesophageal Junction Adenocarcinoma. Diagnostics (Basel) 2023; 13:1884. [PMID: 37296735 PMCID: PMC10252409 DOI: 10.3390/diagnostics13111884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Esophageal adenocarcinoma, including adenocarcinoma of the gastroesophageal junction, is uncommon in the United States, but is associated with a rising incidence in young adults, and has a traditionally poor prognosis. Despite the incremental benefits that have been made with multimodality approaches to locally advanced disease, most patients will go on to develop metastatic disease, and long-term outcomes remain suboptimal. Over the last decade, PET-CT has emerged as a key tool in the management of this disease, with several prospective and retrospective studies evaluating its role in this disease. Herein, we review the key data pertaining to the use of PET-CT in the management of locally advanced esophageal and GEJ adenocarcinoma, with a focus on staging, prognostication, PET-CT adapted therapy in the neoadjuvant setting, and surveillance.
Collapse
Affiliation(s)
- Darren Cowzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (D.C.); (F.K.)
| | - Fergus Keane
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (D.C.); (F.K.)
| | - Geoffrey Y. Ku
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (D.C.); (F.K.)
- Department of Medicine, Weill Cornell University, New York, NY 10065, USA
| |
Collapse
|
33
|
Shah MA, Altorki N, Patel P, Harrison S, Bass A, Abrams JA. Improving outcomes in patients with oesophageal cancer. Nat Rev Clin Oncol 2023; 20:390-407. [PMID: 37085570 DOI: 10.1038/s41571-023-00757-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 04/23/2023]
Abstract
The care of patients with oesophageal cancer or of individuals who have an elevated risk of oesophageal cancer has changed dramatically. The epidemiology of squamous cell and adenocarcinoma of the oesophagus has diverged over the past several decades, with a marked increase in incidence only for oesophageal adenocarcinoma. Only in the past decade, however, have molecular features that distinguish these two forms of the disease been identified. This advance has the potential to improve screening for oesophageal cancers through the development of novel minimally invasive diagnostic technologies predicated on cancer-specific genomic or epigenetic alterations. Surgical techniques have also evolved towards less invasive approaches associated with less morbidity, without compromising oncological outcomes. With improvements in multidisciplinary care, advances in radiotherapy and new tools to detect minimal residual disease, certain patients may no longer even require surgical tumour resection. However, perhaps the most anticipated advance in the treatment of patients with oesophageal cancer is the advent of immune-checkpoint inhibitors, which harness and enhance the host immune response against cancer. In this Review, we discuss all these advances in the management of oesophageal cancer, representing only the beginning of a transformation in our quest to improve patient outcomes.
Collapse
Affiliation(s)
- Manish A Shah
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Nasser Altorki
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Pretish Patel
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sebron Harrison
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Adam Bass
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Julian A Abrams
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
34
|
Mirshahvalad SA, Seyedinia SS, Huemer F, Schweighofer-Zwink G, Koch O, Hitzl W, Weiss L, Emannuel K, Greil R, Pirich C, Beheshti M. Prognostic value of [ 18F]FDG PET/CT on treatment response and progression-free survival of gastroesophageal cancer patients undergoing perioperative FLOT chemotherapy. Eur J Radiol 2023; 163:110843. [PMID: 37119707 DOI: 10.1016/j.ejrad.2023.110843] [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: 11/25/2022] [Revised: 03/17/2023] [Accepted: 04/17/2023] [Indexed: 05/01/2023]
Abstract
PURPOSE To evaluate the prognostic role of [18F]FDG PET/CT metabolic parameters in gastric cancer (GC) and gastroesophageal adenocarcinoma (GEJAC) patients receiving neoadjuvant chemotherapy. METHOD In this retrospective study, 31 patients with biopsy-proven GC or GEJAC were included between August 2016 and March 2020. [18F]FDG PET/CT was performed before the neoadjuvant chemotherapy. Primary tumours' semi-quantitative metabolic parameters were extracted. All patients received a perioperative FLOT regimen thereafter. Post-chemotherapy [18F]FDG PET/CT was performed in most patients (17/31). All patients underwent surgical resection. Histopathology response to treatment and progression-free survival (PFS) were evaluated. Two-sided p-values < 0.05 were considered statistically significant. RESULTS Thirty-one patients (mean age = 62 ± 8), including 21 GC and 10 GEJAC patients, were evaluated. 20/31(65%) patients were histopathology responders to neoadjuvant chemotherapy, including twelve complete and eight partial responders. During the median follow-up of 42.0 months, nine patients experienced recurrence. The median PFS was 60(95% CI:32.9-87.1) months. Pre-neoadjuvant chemotherapy SULpeak was significantly correlated with pathological response to treatment (p-value = 0.03;odds ratio = 16.75). In survival analysis, SUVmax (p-value = 0.01;hazard ratio[HR] = 1.55), SUVmean (p-value = 0.04;HR = 2.73), SULpeak (p-value < 0.001;HR = 1.91) and SULmean (p-value = 0.04;HR = 4.22) in the post-neoadjuvant chemotherapy pre-operative [18F]FDG PET/CT showed significant correlation with PFS. Additionally, aspects of staging were significantly correlated with PFS (p-value = 0.01;HR = 2.21). CONCLUSIONS Pre-neoadjuvant chemotherapy [18F]FDG PET/CT parameters, especially SULpeak, could predict the pathological response to treatment in GC and GEJAC patients. Additionally, in survival analysis, post-chemotherapy metabolic parameters significantly correlated with PFS. Thus, performing [18F]FDG PET/CT before chemotherapy may help to identify patients at risk for inadequate response to perioperative FLOT and, after chemotherapy, may predict clinical outcomes.
Collapse
Affiliation(s)
- Seyed Ali Mirshahvalad
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria; Joint Department of Medical Imaging, Toronto General Hospital, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Seyedeh Sara Seyedinia
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Florian Huemer
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology, and Rheumatology, Oncologic Center, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Gregor Schweighofer-Zwink
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Oliver Koch
- Department of Surgery, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Wolfgang Hitzl
- Biostatistics and Publication of Clinical Trial Studies, Research and Innovation Management (RIM), Paracelsus Medical University, 5020 Salzburg, Austria; Department of Ophthalmology and Optometry, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; Research Program Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Lukas Weiss
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology, and Rheumatology, Oncologic Center, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Klaus Emannuel
- Department of Surgery, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Richard Greil
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology, and Rheumatology, Oncologic Center, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Christian Pirich
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Mohsen Beheshti
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria.
| |
Collapse
|
35
|
Metabolic tumour and nodal response to neoadjuvant chemotherapy on FDG PET-CT as a predictor of pathological response and survival in patients with oesophageal adenocarcinoma. Eur Radiol 2023; 33:3647-3659. [PMID: 36920518 PMCID: PMC10121512 DOI: 10.1007/s00330-023-09482-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/23/2022] [Accepted: 01/27/2023] [Indexed: 03/16/2023]
Abstract
OBJECTIVES 2-deoxy-2[18F]Fluoro-D-glucose (FDG) PET-CT has an emerging role in assessing response to neoadjuvant therapy in oesophageal cancer. This study evaluated FDG PET-CT in predicting pathological tumour response (pTR), pathological nodal response (pNR) and survival. METHODS Cohort study of 75 patients with oesophageal or oesophago-gastric junction (GOJ) adenocarcinoma treated with neoadjuvant chemotherapy then surgery at Guy's and St Thomas' NHS Foundation Trust, London (2017-2020). Standardised uptake value (SUV) metrics on pre- and post-treatment FDG PET-CT in the primary tumour (mTR) and loco-regional lymph nodes (mNR) were derived. Optimum SUVmax thresholds for predicting pathological response were identified using receiver operating characteristic analysis. Predictive accuracy was compared to PERCIST (30% SUVmax reduction) and MUNICON (35%) criteria. Survival was assessed using Cox regression. RESULTS Optimum tumour SUVmax decrease for predicting pTR was 51.2%. A 50% cut-off predicted pTR with 73.5% sensitivity, 69.2% specificity and greater accuracy than PERCIST or MUNICON (area under the curve [AUC] 0.714, PERCIST 0.631, MUNICON 0.659). Using a 30% SUVmax threshold, mNR predicted pNR with high sensitivity but low specificity (AUC 0.749, sensitivity 92.6%, specificity 57.1%, p = 0.010). pTR, mTR, pNR and mNR were independent predictive factors for survival (pTR hazard ratio [HR] 0.10 95% confidence interval [CI] 0.03-0.34; mTR HR 0.17 95% CI 0.06-0.48; pNR HR 0.17 95% CI 0.06-0.54; mNR HR 0.13 95% CI 0.02-0.66). CONCLUSIONS Metabolic tumour and nodal response predicted pTR and pNR, respectively, in patients with oesophageal or GOJ adenocarcinoma. However, currently utilised response criteria may not be optimal. pTR, mTR, pNR and mNR were independent predictors of survival. KEY POINTS • FDG PET-CT has an emerging role in evaluating response to neoadjuvant therapy in patients with oesophageal cancer. • Prospective cohort study demonstrated that metabolic response in the primary tumour and lymph nodes was predictive of pathological response in a cohort of patients with adenocarcinoma of the oesophagus or oesophago-gastric junction treated with neoadjuvant chemotherapy followed by surgical resection. • Patients who demonstrated a response to neoadjuvant chemotherapy in the primary tumour or lymph nodes on FDG PET-CT demonstrated better survival and reduced rates of tumour recurrence.
Collapse
|
36
|
Carr RA, Hsu M, Harrington CA, Tan KS, Bains MS, Bott MJ, Ilson DH, Isbell JM, Janjigian YY, Maron SB, Park BJ, Rusch VW, Sihag S, Wu AJ, Jones DR, Ku GY, Molena D. Induction FOLFOX and PET-Directed Chemoradiation for Locally Advanced Esophageal Adenocarcinoma. Ann Surg 2023; 277:e538-e544. [PMID: 34387205 PMCID: PMC8840992 DOI: 10.1097/sla.0000000000005163] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To compare the efficacy and safety of induction FOLFOX followed by PET-directed nCRT, induction CP followed by PET-directed nCRT, and nCRT with CP alone in patients with EAC. SUMMARY OF BACKGROUND DATA nCRT with CP is a standard treatment for locally advanced EAC. The results of cancer and leukemia group B 80803 support the use of induction chemotherapy followed by PET-directed chemo-radiation therapy. METHODS We retrospectively identified all patients with EAC who underwent the treatments above followed by esophagectomy. We assessed incidences of pathologic complete response (pCR), near-pCR (ypN0 with ≥90% response), and surgical complications between treatment groups using Fisher exact test and logistic regression; disease-free survival (DFS) and overall survival (OS) were estimated by the Kaplan-Meier method and evaluated using the log-rank test and extended Cox regression. RESULTS In total, 451 patients were included: 309 (69%) received induction chemotherapy before nCRT (FOLFOX, n = 70; CP, n = 239); 142 (31%) received nCRT with CP. Rates of pCR (33% vs. 16%, P = 0.004), near-pCR (57% vs. 33%, P < 0.001), and 2-year DFS (68% vs. 50%, P = 0.01) were higher in the induction FOLFOX group than in the induction CP group. Similarly, the rate of near-pCR (57% vs. 42%, P = 0.04) and 2-year DFS (68% vs. 44%, P < 0.001) were significantly higher in the FOLFOX group than in the no-induction group. CONCLUSIONS Induction FOLFOX followed by PET-directed nCRT may result in better histopathologic response rates and DFS than either induction CP plus PET-directed nCRT or nCRT with CP alone.
Collapse
Affiliation(s)
- Rebecca A. Carr
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Meier Hsu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Caitlin A. Harrington
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kay See Tan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Manjit S. Bains
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matthew J. Bott
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David H. Ilson
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James M. Isbell
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yelena Y. Janjigian
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steven B. Maron
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bernard J. Park
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Valerie W. Rusch
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Smita Sihag
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Abraham J. Wu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David R. Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Geoffrey Y. Ku
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniela Molena
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
37
|
Barron CC, Wang X, Elimova E. Neoadjuvant Strategies for Esophageal Cancer. Thorac Surg Clin 2023; 33:197-208. [PMID: 37045489 DOI: 10.1016/j.thorsurg.2023.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
Neoadjuvant strategies with multimodal therapy including chemotherapy and radiation are the standard of care in locally advanced esophageal cancer. The role of immunotherapy in the perioperative management of esophageal cancer is expanding, and adjuvant nivolumab for patients with residual disease following trimodality therapy has been shown to improve disease-free survival. Applications of checkpoint blockade and positron emission tomography (PET)-directed therapy in the neoadjuvant setting are under investigation in several clinical trials. We review the perioperative management of locally advanced esophageal cancer and recent evidence exploring the role of immune checkpoint inhibitors and PET in guiding neoadjuvant management.
Collapse
|
38
|
Shi Y, Wang M, Zhang J, Xiang Z, Li C, Zhang J, Ma X. Tailoring the clinical management of colorectal cancer by 18F-FDG PET/CT. Front Oncol 2022; 12:1062704. [PMID: 36620584 PMCID: PMC9814158 DOI: 10.3389/fonc.2022.1062704] [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: 10/06/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is among the most commonly diagnosed gastrointestinal malignancies worldwide. It is inadequate to handle in terms of staging and restaging only based on morphological imaging modalities and serum surrogate markers. And the correct and timely staging of CRC is imperative to prognosis and management. When compared to established sequential, multimodal conventional diagnostic methods, the molecular and functional imaging 18F-FDG PET/CT shows superiorities for tailoring appropriate treatment maneuvers to each patient. This review aims to summarize the utilities of 18F-FDG PET/CT in CRC, focusing on primary staging, follow-up assessment of tumor responses and diagnostic of recurrence. In addition, we also summarize the technical considerations of PET/CT and the conventional imaging modalities in those patients who are either newly diagnosed with CRC or has already been treated from this cancer.
Collapse
Affiliation(s)
- Yang Shi
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,State Key Laboratory for the Prevention and Treatment of Esophageal Cancer, Zhengzhou University, Zhengzhou, China,*Correspondence: Yang Shi, ; ; Jingjing Zhang, ; Xing Ma,
| | - Meiqi Wang
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jiyu Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,State Key Laboratory for the Prevention and Treatment of Esophageal Cancer, Zhengzhou University, Zhengzhou, China
| | - Zheng Xiang
- Department of Pathology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Can Li
- Department of Administration, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jingjing Zhang
- Department of Nuclear Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China,*Correspondence: Yang Shi, ; ; Jingjing Zhang, ; Xing Ma,
| | - Xing Ma
- Department of Nuclear Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China,*Correspondence: Yang Shi, ; ; Jingjing Zhang, ; Xing Ma,
| |
Collapse
|
39
|
The prognostic value of positron emission tomography/computed tomography-based parameters in locally advanced esophageal squamous cell carcinoma treated with chemoradiation therapy. Nucl Med Commun 2022; 43:1239-1246. [DOI: 10.1097/mnm.0000000000001619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
40
|
Seifert R, Kersting D, Rischpler C, Sandach P, Ferdinandus J, Fendler WP, Rahbar K, Weckesser M, Umutlu L, Hanoun C, Hüttmann A, Reinhardt HC, von Tresckow B, Herrmann K, Dührsen U, Schäfers M. Interim FDG-PET analysis to identify patients with aggressive non-Hodgkin lymphoma who benefit from treatment intensification: a post-hoc analysis of the PETAL trial. Leukemia 2022; 36:2845-2852. [PMID: 36241697 PMCID: PMC9712103 DOI: 10.1038/s41375-022-01713-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/13/2022] [Accepted: 09/16/2022] [Indexed: 11/08/2022]
Abstract
The randomized PETAL trial failed to demonstrate a benefit of interim FDG-PET (iPET)-based treatment intensification over continued standard therapy with CHOP (plus rituximab (R) in CD20-positive lymphomas). We hypothesized that PET analysis of all lymphoma manifestations may identify patients who benefitted from treatment intensification. A previously developed neural network was employed for iPET analysis to identify the highest pathological FDG uptake (max-SUVAI) and the mean FDG uptake of all lymphoma manifestations (mean-SUVAI). High mean-SUVAI uptake was determined separately for iPET-positive and iPET-negative patients. The endpoint was time-to-progression (TTP). There was a significant interaction of additional rituximab and mean-SUVAI in the iPET-negative group (HR = 0.6, p < 0.05). Patients with high mean-SUVAI had significantly prolonged TTP when treated with 6xR-CHOP + 2 R (not reached versus 52 months, p < 0.05), whereas max-SUVmanual failed to show an impact of additional rituximab. In the iPET-positive group, patients with high mean-SUVAI had a significantly longer TTP with (R-)CHOP than with the Burkitt protocol (14 versus 4 months, p < 0.01). Comprehensive iPET evaluation may provide new prognosticators in aggressive lymphoma. Additional application of rituximab was associated with prolonged TTP in iPET-negative patients with high mean-SUVAI. Comprehensive iPET interpretation could identify high-risk patients who benefit from study-specific interventions.
Collapse
Affiliation(s)
- Robert Seifert
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany.
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany.
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany.
- West German Cancer Center, University Hospital Essen, Essen, Germany.
| | - David Kersting
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Christoph Rischpler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Patrick Sandach
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Justin Ferdinandus
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Kambiz Rahbar
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Matthias Weckesser
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Lale Umutlu
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Christine Hanoun
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Andreas Hüttmann
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hans Christian Reinhardt
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bastian von Tresckow
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Ulrich Dührsen
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael Schäfers
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| |
Collapse
|
41
|
Lewis S, Lukovic J. Neoadjuvant Therapy in Esophageal Cancer. Thorac Surg Clin 2022; 32:447-456. [DOI: 10.1016/j.thorsurg.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
42
|
Prognostic value of PERCIST and PET/CT metabolic parameters after neoadjuvant treatment in patients with esophageal cancer. Rev Esp Med Nucl Imagen Mol 2022; 41:360-367. [DOI: 10.1016/j.remnie.2021.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/21/2021] [Accepted: 09/21/2021] [Indexed: 11/08/2022]
|
43
|
Coit DG, Strong VE. Fifty years of progress in gastric cancer. J Surg Oncol 2022; 126:865-871. [PMID: 36087088 PMCID: PMC9469502 DOI: 10.1002/jso.27060] [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: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/06/2022]
Abstract
As with every human malignancy, the diagnosis, staging, and treatment of patients with gastric cancer have undergone enormous evidence-based change over the last 50 years, largely as a result of increasingly rapid developments in technology and science. Some of the changes in clinical practice have derived from prospective randomized controlled trials (RCTs), whereas others have come from study of meticulously maintained prospective databases, which define the disease's natural history over time, and occasionally from in-depth analysis of a single patient with an unexpectedly good or poor outcome. Herein we summarize the more important changes in gastric cancer management and the data supporting those changes.
Collapse
Affiliation(s)
- Daniel G Coit
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Vivian E Strong
- Department of Surgery, Gastric and Mixed Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
44
|
Differences and Similarities in the Pattern of Early Metabolic and Morphologic Response after Induction Chemo-Immunotherapy versus Induction Chemotherapy Alone in Locally Advanced Squamous Cell Head and Neck Cancer. Cancers (Basel) 2022; 14:cancers14194811. [PMID: 36230733 PMCID: PMC9563870 DOI: 10.3390/cancers14194811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/30/2022] Open
Abstract
Background: In head and neck cancer patients, parameters of metabolic and morphologic response of the tumor to single-cycle induction chemotherapy (IC) with docetaxel, cis- or carboplatin are used to decide the further course of treatment. This study investigated the effect of adding a double immune checkpoint blockade (DICB) of tremelimumab and durvalumab to IC on imaging parameters and their significance with regard to tumor cell remission. Methods: Response variables of 53 patients treated with IC+DICB (ICIT) were compared with those of 104 who received IC alone. Three weeks after one cycle, pathologic and, in some cases, clinical and endoscopic primary tumor responses were evaluated and correlated with a change in 18F-FDG PET and CT/MRI-based maximum-standardized uptake values (SUVmax) before (SUVmaxpre), after treatment (SUVmaxpost) and residually (resSUVmax in % of SUVmaxpre), and in maximum tumor diameter (Dmax) before (Dmaxpre) and after treatment (Dmaxpost) and residually (resD). Results: Reduction of SUVmax and Dmax occurred in both groups; values were SUVmaxpre: 14.4, SUVmaxpost: 6.6, Dmaxpre: 30 mm and Dmaxpost: 23 mm for ICIT versus SUVmaxpre: 16.5, SUVmaxpost: 6.4, Dmaxpre: 21 mm, and Dmaxpost: 16 mm for IC alone (all p < 0.05). ResSUVmax was the best predictor of complete response (IC: AUC: 0.77; ICIT: AUC: 0.76). Metabolic responders with resSUVmax ≤ 40% tended to have a higher rate of CR to ICIT (88%; n = 15/17) than to IC (65%; n = 30/46; p = 0.11). Of the metabolic nonresponders (resSUVmax > 80%), 33% (n = 5/15) achieved a clinical CR to ICIT versus 6% (n = 1/15) to IC (p = 0.01). Conclusions: ICIT and IC quickly induce a response and 18F-FDG PET is the more accurate modality for identifying complete remission. The rate of discrepant response, i.e., pCR with metabolic nonresponse after ICIT was >30%.
Collapse
|
45
|
Lavertu S, Barkati M, Beaulieu S, Martin J, Campeau MP, Donath D, Roberge D. Prognostic Assessment of Interim F18-Fluorodeoxyglucose Positron Emission Tomography in Esophageal Cancer Treated With Chemoradiation With or Without Surgery. Cureus 2022; 14:e29086. [PMID: 36259030 PMCID: PMC9558932 DOI: 10.7759/cureus.29086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 09/11/2022] [Indexed: 11/22/2022] Open
Abstract
Purpose This study aimed to evaluate if the F18-fluorodeoxyglucose positron emission tomography (F18-FDG PET) response after two weeks of chemoradiation for locoregionally advanced esophageal cancer (staged Tumor (T) 3 and/or Nodes (N)+ Metastases (M) 0) was linked to the pathologic response for patients undergoing surgery, to disease-free survival (DFS) or overall survival (OS). Materials and Methods Between March 2006 and September 2017, 40 patients were prospectively enrolled in our study, gave written consent, and had PET scans performed before treatment and after two weeks of chemoradiation. One patient did not undergo his two-week PET without informing study coordinators and was excluded from analyses. Results The median age at diagnosis was 62 years. Seventy-two percent of patients had N+ disease. Median OS for the entire group was 24 months. Five-year overall survival was 17%. Survival curves for patients with no PET response, minor PET response, or good PET response overlapped and were not statistically different. For the 25 patients who underwent surgery, the positive predictive value (PPV) of the PET response relative to the pathologic response was 75% and the negative predictive value (NPV) was 62%. In study patients, the crude recurrence rate was 68% and there was no correlation between PET response and DFS. Conclusion In our study, interim PET response after two weeks of chemoradiation for locoregionally advanced esophageal cancer was not predictive of outcome or pathologic response. Based on our data and current literature, interim PET should not be used to alter treatment (whether to escalate neo-adjuvant treatment or omit surgery).
Collapse
|
46
|
Lorenzen S, Quante M, Rauscher I, Slotta-Huspenina J, Weichert W, Feith M, Friess H, Combs SE, Weber WA, Haller B, Angele M, Albertsmeier M, Blankenstein C, Kasper S, Schmid RM, Bassermann F, Schwaiger M, Liffers ST, Siveke JT. PET-directed combined modality therapy for gastroesophageal junction cancer: Results of the multicentre prospective MEMORI trial of the German Cancer Consortium (DKTK). Eur J Cancer 2022; 175:99-106. [PMID: 36099671 DOI: 10.1016/j.ejca.2022.07.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Positron emission tomography (PET) may differentiate responding and non-responding tumours early in the treatment of locally advanced gastroesophageal junction adenocarcinomas. Early PET non-responders (P-NR) after induction CTX might benefit from changing to chemoradiation (CRT). METHODS Patients underwent baseline 18F-FDG PET followed by 1 cycle of CTX. PET was repeated at day 14-21 and responders (P-R), defined as ≥35% decrease in SUVmean from baseline, continued with CTX. P-NR switched to CRT (CROSS). Patients underwent surgery 4-6 weeks post-CTX/CRT. The primary objective was an improvement in R0 resection rates in P-NR above a proportion of 70%. RESULTS In total, 160 patients with resectable gastroesophageal junction adenocarcinomas were prospectively investigated by PET scanning. Eighty-five patients (53%) were excluded. Seventy-five eligible patients were enrolled in the study. Based on PET criteria, 50 (67.6%)/24 (32.4%) were P-R and P-NR, respectively. Resection was performed on 46 responders, including one patient who withdrew the ICF, and 22 non-responders (per-protocol population). R0 resection rates were 95.6% (43/45) for P-R and 86.4% (19/22) for P-NR. No treatment related deaths occurred. With a median follow-up time of 24.5 months, estimated 18 months DFS was 75.4%/64.2% for P-R/P-NR, respectively. The estimated 18 months OS was 95.5% for P-R and 68.2% for P-NR. CONCLUSION The primary endpoint of the study to increase the R0 resection rate in metabolic NR was not met. PET response after induction CTX is prognostic for outcome with a prolonged OS and DFS in PET responders. TRIAL REGISTRATION NCT00002014-000860-16.
Collapse
Affiliation(s)
- Sylvie Lorenzen
- Technical University of Munich, Klinikum rechts der Isar, III. Medizinische Klinik und Poliklinik, Munich, Germany
| | - Michael Quante
- Technical University Munich, Klinikum rechts der Isar, II. Medizinische Klinik und Poliklinik, Munich, Germany; Department of Internal Medicine II, University of Freiburg, Germany
| | - Isabel Rauscher
- Technical University Munich, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | | | - Wilko Weichert
- Technical University Munich, Institute of Pathology, Munich, Germany
| | - Marcus Feith
- Technical University Munich, Klinikum rechts der Isar, Surgical Clinic and Policlinic, Munich, Germany
| | - Helmut Friess
- Technical University Munich, Klinikum rechts der Isar, Surgical Clinic and Policlinic, Munich, Germany
| | - Stefanie E Combs
- Technical University Munich, Klinikum rechts der Isar, Department of Radiation Oncology, Munich, Germany
| | - Wolfgang A Weber
- Technical University Munich, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | - Bernhard Haller
- Technical University Munich, Klinikum rechts der Isar, Institute of AI and Informatics in Medicine, Munich, Germany
| | - Martin Angele
- Ludwig-Maximilians-Universität (LMU) Munich, LMU University Hospital, Department of General, Visceral and Transplantation Surgery, Munich, Germany
| | - Markus Albertsmeier
- Ludwig-Maximilians-Universität (LMU) Munich, LMU University Hospital, Department of General, Visceral and Transplantation Surgery, Munich, Germany
| | | | - Stefan Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen, Germany
| | - Roland M Schmid
- Technical University Munich, Klinikum rechts der Isar, II. Medizinische Klinik und Poliklinik, Munich, Germany; Department of Internal Medicine II, University of Freiburg, Germany
| | - Florian Bassermann
- Technical University of Munich, Klinikum rechts der Isar, III. Medizinische Klinik und Poliklinik, Munich, Germany
| | - Markus Schwaiger
- Technical University Munich, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | - Sven-Thorsten Liffers
- German Cancer Consortium (DKTK), Partner Site Essen, Germany; Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital, University of Duisburg-Essen, Essen, Germany; Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Jens T Siveke
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen, Germany; Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital, University of Duisburg-Essen, Essen, Germany; Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany.
| |
Collapse
|
47
|
Withey SJ, Goh V, Foley KG. State-of-the-art imaging in oesophago-gastric cancer. Br J Radiol 2022; 95:20220410. [PMID: 35671095 PMCID: PMC10996959 DOI: 10.1259/bjr.20220410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 11/05/2022] Open
Abstract
Radiological investigations are essential in the management of oesophageal and gastro-oesophageal junction cancers. The current multimodal combination of CT, 18F-fluorodeoxyglucose positron emission tomography combined with CT (PET/CT) and endoscopic ultrasound (EUS) has limitations, which hinders the prognostic and predictive information that can be used to guide optimum treatment decisions. Therefore, the development of improved imaging techniques is vital to improve patient management. This review describes the current evidence for state-of-the-art imaging techniques in oesophago-gastric cancer including high resolution MRI, diffusion-weighted MRI, dynamic contrast-enhanced MRI, whole-body MRI, perfusion CT, novel PET tracers, and integrated PET/MRI. These novel imaging techniques may help clinicians improve the diagnosis, staging, treatment planning, and response assessment of oesophago-gastric cancer.
Collapse
Affiliation(s)
- Samuel J Withey
- Department of Radiology, The Royal Marsden NHS Foundation
Trust, London,
UK
| | - Vicky Goh
- Cancer Imaging, School of Biomedical Engineering & Imaging
Sciences, King’s College London,
London, UK
- Department of Radiology, Guy’s and St Thomas’ NHS
Foundation Trust, London,
UK
| | - Kieran G Foley
- Division of Cancer & Genetics, School of Medicine, Cardiff
University, Wales,
UK
- Department of Radiology, Velindre Cancer Centre,
Cardiff, UK
| |
Collapse
|
48
|
Fooladi M, Shirazi A, Sheikhzadeh P, Amirrashedi M, Ghahramani F, Cheki M, Khoobi M. Investigating the attenuating effect of telmisartan against radiation-induced intestinal injury using 18F-FDG micro-PET imaging. Int J Radiat Biol 2022; 99:446-458. [PMID: 35930426 DOI: 10.1080/09553002.2022.2110295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
BACKGROUND AND OBJECTIVE This study was aimed to investigate the ability of 18F-Fluro-deoxy-glucose (18F-FDG)-based micro-positron emission tomography (microPET) imaging to evaluate the efficacy of telmisartan, a highly selective angiotensin II receptor antagonist (ARA), in intestinal tissue recovery process after in vivo irradiation. METHODS Male Balb/c mice were randomly divided into four groups of control, telmisartan, irradiation, and telmisartan + irradiation. A solution of telmisartan in phosphate-buffered saline (PBS) was administered orally at 12 mg/kg body weight for seven consecutive days prior to whole body exposing to a single sub-lethal dose of 5 Gy X-rays. The mice were imaged using 18F-FDG microPET at 9 and 30 days post-irradiation. The 18F-FDG uptake in jejunum was determined according to the mean standardized uptake value (SUVmean) index. Tissues were also processed in similar time points for histological analysis. RESULTS The 18F-FDG microPET imaging confirmed the efficacy of telmisartan as a potent attenuating agent for ionizing radiation-induced injury of intestine in mice model. The results were also in line with the histological analysis indicating that pretreatment with telmisartan reduced damage to the villi, crypts, and intestinal mucosa compared with irradiated and non-treated group from day 9 to 30 after irradiation. CONCLUSION The results revealed that 18F-FDG microPET imaging could be a good candidate to replace time-consuming and invasive biological techniques for screening of radioprotective agents. These findings were also confirmed by histological examinations which indicated that telmisartan can effectively attenuates radiation injury caused by ionizing-irradiation.
Collapse
Affiliation(s)
- Masoomeh Fooladi
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Shirazi
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Peyman Sheikhzadeh
- Department of Nuclear Medicine, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Amirrashedi
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ghahramani
- Radiotherapy-Oncology Center, Yas Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Cheki
- Department of Medical Imaging and Radiation Sciences, Faculty of Paramedicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mehdi Khoobi
- Biomaterials Group, Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Sakin A, Ozcelik M, Sahin S, Aydemir O, Aldemir MN, Iliklerden UH, Kotan MC. The prognostic effect of pretreatment 18F-FDG PET/CT metabolic parameters in locally advanced Esophageal Squamous Cell Carcinoma treated with definitive chemoradiotherapy. Surg Oncol 2022; 43:101809. [DOI: 10.1016/j.suronc.2022.101809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/01/2022] [Accepted: 07/03/2022] [Indexed: 12/27/2022]
|
50
|
Zhu H, Liu Q, Xu H, Mo M, Wang Z, Lu K, Zhou J, Chen J, Zheng X, Ye J, Ge X, Luo H, Liu Q, Deng J, Ai D, Hao S, Zhang J, Tseng IH, Song S, Chen Y, Zhao K. Dose escalation based on 18F-FDG PET/CT response in definitive chemoradiotherapy of locally advanced esophageal squamous cell carcinoma: a phase III, open-label, randomized, controlled trial (ESO-Shanghai 12). Radiat Oncol 2022; 17:134. [PMID: 35906623 PMCID: PMC9338557 DOI: 10.1186/s13014-022-02099-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 07/07/2022] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION Definitive chemoradiotherapy has established the standard non-surgical treatment for locally advanced esophageal cancer. The standard dose of 50-50.4 Gy has been established decades ago and been confirmed in modern trials. The theorical advantage of better local control and technical advances for less toxicity have encouraged clinicians for dose escalation investigation. 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) have the potential to tailor therapy for esophageal patients not showing response to CRT and pioneers the PET-based dose escalation. METHODS AND ANALYSIS The ESO-Shanghai 12 trial is a prospective multicenter randomized phase 3 study in which patients are randomized to either 61.2 Gy or 50.4 Gy of radiation dose by PET response. Both groups undergo concurrent chemoradiotherapy with paclitaxel/cisplatin regimen for 2 cycles followed by consolidation chemotherapy for 2 cycles. Patients with histologically confirmed ESCC [T1N1-3M0, T2-4NxM0, TxNxM1 (Supraclavicular lymph node metastasis only), (AJCC Cancer Staging Manual, 8th Edition)] and without any prior treatment of chemotherapy, radiotherapy or surgery against esophageal cancer will be eligible. The primary endpoints included overall survival in PET/CT non-responders (SUVmax > 4.0) and overall survival in total population. Patients will be stratified by standardized uptake volume, gross tumor volume and tumor location. The enrollment could be ended, when the number of PET/CT non-responder reached 132 and the total population reached 646 for randomization. ETHICS AND DISSEMINATION This trial has been approved by the Fudan University Shanghai Cancer Center Institutional Review Board. Trial results will be disseminated via peer reviewed scientific journals and conference presentations. Trial registration The trial was initiated in 2018 and is currently recruiting patients. Trial registration number NCT03790553.
Collapse
Affiliation(s)
- Hongcheng Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Qiufang Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hao Xu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Miao Mo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Cancer Prevention and Statistics, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zezhou Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Cancer Prevention and Statistics, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Kui Lu
- Department of Radiation Oncology, Taizhou Second People's Hospital, Taizhou, Jiangsu, China
| | - Jialiang Zhou
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Junqiang Chen
- Department of Radiation Oncology, Fujian Cancer Hospital, Fuzhou, China
| | - Xiangpeng Zheng
- Department of Radiation Oncology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jinjun Ye
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing, China
| | - Xiaolin Ge
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Honglei Luo
- Department of Radiation Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Qi Liu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Jiaying Deng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Dashan Ai
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Shengnan Hao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Junhua Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - I Hsuan Tseng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Shaoli Song
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yun Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. .,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China.
| | - Kuaile Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China. .,Shanghai Key Laboratory of Radiation Oncology, Shanghai, China.
| |
Collapse
|