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
|
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
|
3
|
Cytryn SL, Pandit-Taskar N, Lumish MA, Maron SB, Gu P, Ku GY, Chou JF, Capanu M, Antoine A, Loegel D, Feder L, Philemond S, Lyashchenko SK, Lewis JS, Paroder V, Srivastava A, Tang LH, Schoder H, Janjigian YY. 18F-BMS-986229 PET to Assess Programmed-Death Ligand 1 Status in Gastroesophageal Cancer. J Nucl Med 2024; 65:722-727. [PMID: 38514081 PMCID: PMC11064823 DOI: 10.2967/jnumed.123.267186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/13/2024] [Indexed: 03/23/2024] Open
Abstract
Anti-programmed death 1 (PD-1) inhibitors are the standard of care for advanced gastroesophageal cancer. Although recommendations and approval by regulatory agencies are often based on programmed death ligand 1 (PD-L1) expression, pathologic assessments of PD-L1 status have several limitations. Single-site biopsies do not adequately capture disease heterogeneity within individual tumor lesions or among several lesions within the same patient, the PD-L1 combined positive score is a dynamic biomarker subject to evolution throughout a patient's disease course, and repeated biopsies are invasive and not always feasible. Methods: This was a prospective pilot study of the PD-L1-targeting radiotracer, 18F-BMS-986229, with PET imaging (PD-L1 PET) in patients with gastroesophageal cancer. Patients were administered the 18F-BMS-986229 radiotracer intravenously at a dose of 370 MBq over 1-2 min and underwent whole-body PET/CT imaging 60 min later. The primary objective of this study was to evaluate the safety and feasibility of 18F-BMS-986229. The trial is registered with ClinicalTrials.gov (NCT04161781). Results: Between February 3, 2020, and February 2, 2022, 10 patients with gastroesophageal adenocarcinoma underwent PD-L1 PET. There were no adverse events associated with the 18F-BMS-986229 tracer, and imaging did not result in treatment delays; the primary endpoint was achieved. Radiographic evaluation of PD-L1 expression was concordant with pathologic assessment in 88% of biopsied lesions, and 18F-BMS-986229 uptake on PET imaging correlated with pathologic evaluation by the combined positive score (Spearman rank correlation coefficient, 0.64). Seventy-one percent of patients with 18F-BMS-986229 accumulation on PET imaging also had lesions without 18F-BMS-986229 uptake, highlighting the intrapatient heterogeneity of PD-L1 expression. Patients treated with frontline programmed death 1 inhibitors who had 18F-BMS-986229 accumulation in any lesions on PET imaging had longer progression-free survival than patients without tracer accumulation in any lesions (median progression-free survival, 28.4 vs. 9.9 mo), though the small sample size prevents any definitive conclusions. Conclusion: PD-L1 PET imaging was safe, feasible, and concordant with pathologic evaluation and offers a potential noninvasive tool to assess PD-L1 expression.
Collapse
Affiliation(s)
- Samuel L Cytryn
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Melissa A Lumish
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Steven B Maron
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Ping Gu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Geoffrey Y Ku
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Joanne F Chou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Marinela Capanu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Ariel Antoine
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Diane Loegel
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Lara Feder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Steven Philemond
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Serge K Lyashchenko
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Viktoriya Paroder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Amitabh Srivastava
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laura H Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Heiko Schoder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Yelena Y Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York;
- Department of Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
4
|
Mineur L, Plat F, Desseigne F, Deplanque G, Belkacemi M, Moureau-Zabotto L, Beyrne CD, Jalali K, Obled S, Smith D, Vazquez L, Boustany R. NESC Multicenter Phase II Trial in the Preoperative Treatment of Gastric Adenocarcinoma with Chemotherapy (Docetaxel-Cisplatin-5FU+Lenograstim) Followed by Chemoradiation Based 5FU and Oxaliplatin and Surgery. Cancer Res Treat 2024; 56:580-589. [PMID: 37817565 PMCID: PMC11016650 DOI: 10.4143/crt.2023.812] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/04/2023] [Indexed: 10/12/2023] Open
Abstract
PURPOSE Preoperative chemoradiation (CRT) is expected to increase the rate of curative resection and complete histological response. In this trial, we investigated the efficacy of a neoadjuvant CRT regimen in gastric adenocarcinoma (NCT01565109 trial). MATERIALS AND METHODS Patients with stage IB to IIIC gastric adenocarcinoma, endoscopy ultrasound and computed tomography-scan diagnosed, were eligible for this phase II trial. Neoadjuvant treatment consisted of 2 cycles of chemotherapy with DCF (docetaxel, cisplatin, and 5-fluorouracil [5FU]) followed by preoperative CRT with oxaliplatin, continuous 5FU and radiotherapy (45 Gy in 25 fractions of 1.8 Gy, 5 fractions per week for 5 weeks) administered before surgery. R0-resection rate, pathological complete response (pathCR) rate, and survival (progression-free survival [PFS] and overall survival [OS]) were evaluated as primary endpoints. RESULTS Among 33 patients included, 32 patients (97%) received CRT and 26 (78.8%) were resected (R0 resection for all patients resected). Among resected patients, we report pathCR in 23,1% and pathologic major response (tumor regression grade 2 according to Mandard's classification) in 26,9%. With a median follow-up duration of 5.82 years (range, 0.4 to 9.24 years), the estimated median OS for all 33 patients was not reached; 1-, 3-, and 5-year OS rates were 85%, 61%, and 52%, respectively. Among resected patients, those whose histological response was tumor grade regression (TRG) 1-2 had significantly better OS and PFS rates than those with a TRG 3-4-5 response (p=0.019 and p=0.016, respectively). CONCLUSION Promising results from trials involving preoperative chemoradiation followed by surgery in gastric cancer need to be further evaluated in a phase III trial.
Collapse
|
5
|
Ebert MP, Fischbach W, Hollerbach S, Höppner J, Lorenz D, Stahl M, Stuschke M, Pech O, Vanhoefer U, Porschen R. S3-Leitlinie Diagnostik und Therapie der Plattenepithelkarzinome und Adenokarzinome des Ösophagus. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:535-642. [PMID: 38599580 DOI: 10.1055/a-2239-9802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Affiliation(s)
- Matthias P Ebert
- II. Medizinische Klinik, Medizinische Fakultät Mannheim, Universitätsmedizin, Universität Heidelberg, Mannheim
- DKFZ-Hector Krebsinstitut an der Universitätsmedizin Mannheim, Mannheim
- Molecular Medicine Partnership Unit, EMBL, Heidelberg
| | - Wolfgang Fischbach
- Deutsche Gesellschaft zur Bekämpfung der Krankheiten von Magen, Darm und Leber sowie von Störungen des Stoffwechsels und der Ernährung (Gastro-Liga) e. V., Giessen
| | | | - Jens Höppner
- Klinik für Allgemeine Chirurgie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Lübeck
| | - Dietmar Lorenz
- Chirurgische Klinik I, Allgemein-, Viszeral- und Thoraxchirurgie, Klinikum Darmstadt, Darmstadt
| | - Michael Stahl
- Klinik für Internistische Onkologie und onkologische Palliativmedizin, Evang. Huyssensstiftung, Evang. Kliniken Essen-Mitte, Essen
| | - Martin Stuschke
- Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Essen, Essen
| | - Oliver Pech
- Klinik für Gastroenterologie und Interventionelle Endoskopie, Krankenhaus Barmherzige Brüder, Regensburg
| | - Udo Vanhoefer
- Klinik für Hämatologie und Onkologie, Katholisches Marienkrankenhaus, Hamburg
| | - Rainer Porschen
- Gastroenterologische Praxis am Kreiskrankenhaus Osterholz, Osterholz-Scharmbeck
| |
Collapse
|
6
|
Okazumi S, Ohira G, Hayano K, Aoyagi T, Imanishi S, Matsubara H. Novel Advances in Qualitative Diagnostic Imaging for Decision Making in Multidisciplinary Treatment for Advanced Esophageal Cancer. J Clin Med 2024; 13:632. [PMID: 38276137 PMCID: PMC10816440 DOI: 10.3390/jcm13020632] [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: 11/06/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Background: Recently, neoadjuvant therapy and the succeeding surgery for advanced esophageal cancer have been evaluated. In particular, the response to the therapy has been found to affect surgical outcomes, and thus a precise evaluation of treatment effect is important for this strategy. In this study, articles on qualitative diagnostic modalities to evaluate tumor activities were reviewed, and the diagnostic indices were examined. Methods: For prediction of the effect, perfusion CT and diffusion MRI were estimated. For the histological response evaluation, perfusion CT, diffusion-MRI, and FDG-PET were estimated. For downstaging evaluation of T4, tissue-selective image reconstruction using enhanced CT was estimated and diagnostic indices were reviewed. Results: The prediction of the effect using perfusion CT with 'pre CRT blood flow' and diffusion MRI with 'pre CRT ADC value'; the estimation of the histological response using perfusion CT with 'post CRT blood flow reduction, using diffusion MRI with 'post CRT ADC increasing', and using FDG-PET with 'post CRT SUV reduction'; and the downstaging evaluation of T4 using CT image reconstruction with 'fibrous changed layer' were performed well, respectively. Conclusions: Qualitative imaging modalities for prediction or response evaluation of neoadjuvant therapy for progressive esophageal cancer were useful for the decision making of the treatment strategy of the multidisciplinary treatment.
Collapse
Affiliation(s)
- Shinichi Okazumi
- Department of Surgery, Toho University Sakura Medical Center, Chiba 285-8741, Japan;
| | - Gaku Ohira
- Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (K.H.); (H.M.)
| | - Koichi Hayano
- Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (K.H.); (H.M.)
| | - Tomoyoshi Aoyagi
- Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (K.H.); (H.M.)
| | - Shunsuke Imanishi
- Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (K.H.); (H.M.)
| | - Hisahiro Matsubara
- Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (K.H.); (H.M.)
| |
Collapse
|
7
|
Linders DGJ, Deken MM, van Dam MA, Wasser MNJM, Voormolen EMC, Kroep JR, van Dongen GAMS, Vugts D, Oosterkamp HM, Straver ME, van de Velde CJH, Cohen D, Dibbets-Schneider P, van Velden FHP, Pereira Arias-Bouda LM, Vahrmeijer AL, Liefers GJ, de Geus-Oei LF, Hilling DE. 89Zr-Trastuzumab PET/CT Imaging of HER2-Positive Breast Cancer for Predicting Pathological Complete Response after Neoadjuvant Systemic Therapy: A Feasibility Study. Cancers (Basel) 2023; 15:4980. [PMID: 37894346 PMCID: PMC10605041 DOI: 10.3390/cancers15204980] [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: 09/07/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Approximately 20% of invasive ductal breast malignancies are human epidermal growth factor receptor 2 (HER2)-positive. These patients receive neoadjuvant systemic therapy (NAT) including HER2-targeting therapies. Up to 65% of patients achieve a pathological complete response (pCR). These patients might not have needed surgery. However, accurate preoperative identification of a pCR remains challenging. A radiologic complete response (rCR) on MRI corresponds to a pCR in only 73% of patients. The current feasibility study investigates if HER2-targeted PET/CT-imaging using Zirconium-89 (89Zr)-radiolabeled trastuzumab can be used for more accurate NAT response evaluation. METHODS HER2-positive breast cancer patients scheduled to undergo NAT and subsequent surgery received a 89Zr-trastuzumab PET/CT both before (PET/CT-1) and after (PET/CT-2) NAT. Qualitative and quantitative response evaluation was performed. RESULTS Six patients were enrolled. All primary tumors could be identified on PET/CT-1. Four patients had a pCR and two a pathological partial response (pPR) in the primary tumor. Qualitative assessment of PET/CT resulted in an accuracy of 66.7%, compared to 83.3% of the standard-of-care MRI. Quantitative assessment showed a difference between the SUVR on PET/CT-1 and PET/CT-2 (ΔSUVR) in patients with a pPR and pCR of -48% and -90% (p = 0.133), respectively. The difference in tumor-to-blood ratio on PET/CT-1 and PET/CT-2 (ΔTBR) in patients with pPR and pCR was -79% and -94% (p = 0.133), respectively. Three patients had metastatic lymph nodes at diagnosis that were all identified on PET/CT-1. All three patients achieved a nodal pCR. Qualitative assessment of the lymph nodes with PET/CT resulted in an accuracy of 66.7%, compared to 50% of the MRI. CONCLUSIONS NAT response evaluation using 89Zr-trastuzumab PET/CT is feasible. In the current study, qualitative assessment of the PET/CT images is not superior to standard-of-care MRI. Our results suggest that quantitative assessment of 89Zr-trastuzumab PET/CT has potential for a more accurate response evaluation of the primary tumor after NAT in HER2-positive breast cancer.
Collapse
Affiliation(s)
- D. G. J. Linders
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands (D.E.H.)
| | - M. M. Deken
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands (D.E.H.)
| | - M. A. van Dam
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands (D.E.H.)
| | - M. N. J. M. Wasser
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - E. M. C. Voormolen
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - J. R. Kroep
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - G. A. M. S. van Dongen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, 1081 HV Amsterdam, The Netherlands
| | - D. Vugts
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, 1081 HV Amsterdam, The Netherlands
| | - H. M. Oosterkamp
- Department of Internal Medicine, Haaglanden Medical Center, 2512 VA The Hague, The Netherlands
| | - M. E. Straver
- Department of Surgery, Haaglanden Medical Center, 2512 VA The Hague, The Netherlands
| | - C. J. H. van de Velde
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands (D.E.H.)
| | - D. Cohen
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - P. Dibbets-Schneider
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - F. H. P. van Velden
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - L. M. Pereira Arias-Bouda
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Nuclear Medicine, Alrijne Hospital, 2353 GA Leiderdorp, The Netherlands
| | - A. L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands (D.E.H.)
| | - G. J. Liefers
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands (D.E.H.)
| | - L. F. de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
- Department of Radiation Science and Technology, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - D. E. Hilling
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands (D.E.H.)
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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]
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
Barron CC, Wang X, Elimova E. Neoadjuvant Strategies for Esophageal Cancer. Thorac Surg Clin 2023; 33:197-208. [PMID: 37045489 DOI: 10.1016/j.thorsurg.2023.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
Neoadjuvant strategies with multimodal therapy including chemotherapy and radiation are the standard of care in locally advanced esophageal cancer. The role of immunotherapy in the perioperative management of esophageal cancer is expanding, and adjuvant nivolumab for patients with residual disease following trimodality therapy has been shown to improve disease-free survival. Applications of checkpoint blockade and positron emission tomography (PET)-directed therapy in the neoadjuvant setting are under investigation in several clinical trials. We review the perioperative management of locally advanced esophageal cancer and recent evidence exploring the role of immune checkpoint inhibitors and PET in guiding neoadjuvant management.
Collapse
|
16
|
Zhang C, Wu B, Yang H, Yao Z, Zhang N, Tan F, Liu M, Xu K, Chen L, Xing J, Cui M, Su X. The validity of neoadjuvant chemotherapy with paclitaxel plus S-1 is not inferior to that of SOX regimen for locally advanced gastric cancer: an observational study. BMC Cancer 2022; 22:1223. [PMID: 36443694 PMCID: PMC9703660 DOI: 10.1186/s12885-022-10230-1] [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: 06/03/2022] [Accepted: 10/25/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Paclitaxel plus S-1(PTXS) has shown definite efficacy for advanced gastric cancer. However, the efficacy and safety of this regimen in neoadjuvant setting for locally advanced gastric cancer (LAGC) are unclear. This study aimed to compare the efficacy of neoadjuvant chemotherapy (NAC) PTXS and oxaliplatin plus S-1 (SOX) regime for patients with LAGC. METHODS A total of 103 patients with LAGC (cT3/4NanyM0/x) who were treated with three cycles of neoadjuvant SOX regimen (n = 77) or PTXS regimen (n = 26) between 2011 and 2017 were enrolled in this study. NAC-related clinical response, pathological response, postoperative complication, and overall survival were analyzed between the groups. RESULTS The baseline data did not differ significantly between both groups. After NAC, the disease control rate of the SOX group (94.8%) was comparable with that of the PTXS group (92.3%) (p = 0.641). Twenty-three cases (29.9%) in the SOX group and 10 cases (38.5%) in the PTX group got the descending stage with no statistical difference (p = 0.417). No significant differences were observed in the overall pathological response rate and the overall postoperative complication rate between the two groups (p > 0.05). There were also no differences between groups in terms of 5-year overall and disease-free survival (p > 0.05). CONCLUSIONS The validity of NAC PTXS was not inferior to that of SOX regimen for locally advanced gastric cancer in terms of treatment response and overall survival. PTXS regimen could be expected to be ideal neoadjuvant chemotherapy for patients with LAGC and should be adopted for the test arm of a large randomized controlled trial.
Collapse
Affiliation(s)
- Chenghai Zhang
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fu-Cheng Road, Hai-Dian District, Beijing, 100142 China
| | - Binghong Wu
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fu-Cheng Road, Hai-Dian District, Beijing, 100142 China
| | - Hong Yang
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fu-Cheng Road, Hai-Dian District, Beijing, 100142 China
| | - Zhendan Yao
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fu-Cheng Road, Hai-Dian District, Beijing, 100142 China
| | - Nan Zhang
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fu-Cheng Road, Hai-Dian District, Beijing, 100142 China
| | - Fei Tan
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fu-Cheng Road, Hai-Dian District, Beijing, 100142 China
| | - Maoxing Liu
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fu-Cheng Road, Hai-Dian District, Beijing, 100142 China
| | - Kai Xu
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fu-Cheng Road, Hai-Dian District, Beijing, 100142 China
| | - Lei Chen
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fu-Cheng Road, Hai-Dian District, Beijing, 100142 China
| | - Jiadi Xing
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fu-Cheng Road, Hai-Dian District, Beijing, 100142 China
| | - Ming Cui
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fu-Cheng Road, Hai-Dian District, Beijing, 100142 China
| | - Xiangqian Su
- grid.412474.00000 0001 0027 0586Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fu-Cheng Road, Hai-Dian District, Beijing, 100142 China
| |
Collapse
|
17
|
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]
|
18
|
Liu XY, Gao Y, Kui XY, Liu XS, Zhang YH, Zhang Y, Ke CB, Pei ZJ. High expression of HNRNPR in ESCA combined with 18F-FDG PET/CT metabolic parameters are novel biomarkers for preoperative diagnosis of ESCA. Lab Invest 2022; 20:450. [PMID: 36195940 PMCID: PMC9533615 DOI: 10.1186/s12967-022-03665-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/25/2022] [Indexed: 01/17/2023]
Abstract
Background The aim of this study was to determine the expression and function of heterogeneous nuclear ribonucleoprotein R (HNRNPR) in esophageal carcinoma (ESCA), the correlation between its expression and 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computerized tomography scan (PET/CT)-related parameters. We also investigated whether 18F-FDG PET/CT can be used to predict the expression of HNRNPR in ESCA. Methods We analyzed patients with ESCA who underwent 18F-FDG PET/CT before surgery, and their tissues were stained with HNRNPR IHC. The associated parameters were derived using the 18F-FDG PET imaging data, and the correlation with the IHC score was evaluated. The Oncomine, TCGA, and GEO datasets were used to investigate HNRNPR expression in the pan- and esophageal cancers, as well as its relationship with N6-methyladenosine (m6A) modification and glycolysis. The R software, LinkedOmics, GeneMANIA, and StringOnline tools were used to perform GO/KEGG, GGI, and PPI analyses on the HNRNPR. Results HNRNPR is highly expressed in the majority of pan-cancers, including ESCA, and is associated with BMI, weight, and history of reflux in patients with ESCA. HNRNPR is somewhat accurate in predicting the clinical prognosis of ESCA. HNRNPR expression was positively correlated with SUVmax, SUVmean, and TLG in ESCA (p < 0.05). The combination of these three variables provides a strong predictive value for HNRNPR expression in ESCA. GO/KEGG analysis showed that HNRNPR played a role in the regulation of cell cycle, DNA replication, and the Fannie anemia pathway. The analysis of the TCGA and GEO data sets revealed a significant correlation between HNRNPR expression and m6A and glycolysis-related genes. GSEA analysis revealed that HNRNPR was involved in various m6A and glycolysis related-pathways. Conclusion HNRNPR overexpression correlates with 18F-FDG uptake in ESCA and may be involved in the regulation of the cell cycle, m6A modification, and cell glycolysis. 18F-FDG PET/CT-related parameters can predict the diagnostic accuracy of HNRNPR expression in ESCA.
Collapse
Affiliation(s)
- Xiao-Yu Liu
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yan Gao
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xue-Yan Kui
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.,Taihe Hospital, Postgraduate Training Basement of Jinzhou Medical University, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xu-Sheng Liu
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yao-Hua Zhang
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yu Zhang
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Chang-Bin Ke
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
| | - Zhi-Jun Pei
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China. .,Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan, Hubei, China.
| |
Collapse
|
19
|
Lorenzen S, Quante M, Rauscher I, Slotta-Huspenina J, Weichert W, Feith M, Friess H, Combs SE, Weber WA, Haller B, Angele M, Albertsmeier M, Blankenstein C, Kasper S, Schmid RM, Bassermann F, Schwaiger M, Liffers ST, Siveke JT. PET-directed combined modality therapy for gastroesophageal junction cancer: Results of the multicentre prospective MEMORI trial of the German Cancer Consortium (DKTK). Eur J Cancer 2022; 175:99-106. [PMID: 36099671 DOI: 10.1016/j.ejca.2022.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Positron emission tomography (PET) may differentiate responding and non-responding tumours early in the treatment of locally advanced gastroesophageal junction adenocarcinomas. Early PET non-responders (P-NR) after induction CTX might benefit from changing to chemoradiation (CRT). METHODS Patients underwent baseline 18F-FDG PET followed by 1 cycle of CTX. PET was repeated at day 14-21 and responders (P-R), defined as ≥35% decrease in SUVmean from baseline, continued with CTX. P-NR switched to CRT (CROSS). Patients underwent surgery 4-6 weeks post-CTX/CRT. The primary objective was an improvement in R0 resection rates in P-NR above a proportion of 70%. RESULTS In total, 160 patients with resectable gastroesophageal junction adenocarcinomas were prospectively investigated by PET scanning. Eighty-five patients (53%) were excluded. Seventy-five eligible patients were enrolled in the study. Based on PET criteria, 50 (67.6%)/24 (32.4%) were P-R and P-NR, respectively. Resection was performed on 46 responders, including one patient who withdrew the ICF, and 22 non-responders (per-protocol population). R0 resection rates were 95.6% (43/45) for P-R and 86.4% (19/22) for P-NR. No treatment related deaths occurred. With a median follow-up time of 24.5 months, estimated 18 months DFS was 75.4%/64.2% for P-R/P-NR, respectively. The estimated 18 months OS was 95.5% for P-R and 68.2% for P-NR. CONCLUSION The primary endpoint of the study to increase the R0 resection rate in metabolic NR was not met. PET response after induction CTX is prognostic for outcome with a prolonged OS and DFS in PET responders. TRIAL REGISTRATION NCT00002014-000860-16.
Collapse
Affiliation(s)
- Sylvie Lorenzen
- Technical University of Munich, Klinikum rechts der Isar, III. Medizinische Klinik und Poliklinik, Munich, Germany
| | - Michael Quante
- Technical University Munich, Klinikum rechts der Isar, II. Medizinische Klinik und Poliklinik, Munich, Germany; Department of Internal Medicine II, University of Freiburg, Germany
| | - Isabel Rauscher
- Technical University Munich, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | | | - Wilko Weichert
- Technical University Munich, Institute of Pathology, Munich, Germany
| | - Marcus Feith
- Technical University Munich, Klinikum rechts der Isar, Surgical Clinic and Policlinic, Munich, Germany
| | - Helmut Friess
- Technical University Munich, Klinikum rechts der Isar, Surgical Clinic and Policlinic, Munich, Germany
| | - Stefanie E Combs
- Technical University Munich, Klinikum rechts der Isar, Department of Radiation Oncology, Munich, Germany
| | - Wolfgang A Weber
- Technical University Munich, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | - Bernhard Haller
- Technical University Munich, Klinikum rechts der Isar, Institute of AI and Informatics in Medicine, Munich, Germany
| | - Martin Angele
- Ludwig-Maximilians-Universität (LMU) Munich, LMU University Hospital, Department of General, Visceral and Transplantation Surgery, Munich, Germany
| | - Markus Albertsmeier
- Ludwig-Maximilians-Universität (LMU) Munich, LMU University Hospital, Department of General, Visceral and Transplantation Surgery, Munich, Germany
| | | | - Stefan Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen, Germany
| | - Roland M Schmid
- Technical University Munich, Klinikum rechts der Isar, II. Medizinische Klinik und Poliklinik, Munich, Germany; Department of Internal Medicine II, University of Freiburg, Germany
| | - Florian Bassermann
- Technical University of Munich, Klinikum rechts der Isar, III. Medizinische Klinik und Poliklinik, Munich, Germany
| | - Markus Schwaiger
- Technical University Munich, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | - Sven-Thorsten Liffers
- German Cancer Consortium (DKTK), Partner Site Essen, Germany; Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital, University of Duisburg-Essen, Essen, Germany; Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Jens T Siveke
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner Site Essen, Germany; Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital, University of Duisburg-Essen, Essen, Germany; Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany.
| |
Collapse
|
20
|
Lumish MA, Ku GY. Approach to Resectable Gastric Cancer: Evolving Paradigm of Neoadjuvant and Adjuvant Treatment. Curr Treat Options Oncol 2022; 23:1044-1058. [PMID: 35524838 DOI: 10.1007/s11864-021-00917-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 12/22/2022]
Abstract
OPINION STATEMENT Recent therapeutic advances have prolonged survival in patients with metastatic gastric cancer, though the prognosis for patients with locally advanced resectable gastric cancer remains poor. Long-term survival after resection of locally advanced gastric adenocarcinoma is dependent on early eradication of micrometastatic disease and optimal surgical resection. Preoperative therapy with a docetaxel-containing three-drug regimen has recently been shown to be superior to an anthracycline-containing three-drug regimen or two-drug therapy with a fluoropyrimidine and platinum. Chemoradiation is not essential and is reserved for patients with suboptimal resection. Emerging research strategies include introduction of pre- and postoperative checkpoint blockade and biomarker-directed therapy.
Collapse
Affiliation(s)
- Melissa A Lumish
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Geoffrey Y Ku
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 300 E. 66th Street, Rm 1035, New York, NY, 10065, USA.
| |
Collapse
|
21
|
Jayaprakasam VS, Gibbs P, Gangai N, Bajwa R, Sosa RE, Yeh R, Greally M, Ku GY, Gollub MJ, Paroder V. Can 18F-FDG PET/CT Radiomics Features Predict Clinical Outcomes in Patients with Locally Advanced Esophageal Squamous Cell Carcinoma? Cancers (Basel) 2022; 14:cancers14123035. [PMID: 35740700 PMCID: PMC9221147 DOI: 10.3390/cancers14123035] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary PET/CT is an important staging modality in the baseline assessment of locally advanced esophageal squamous cell carcinoma. Accurate staging and response prediction in these patients is essential for management. The aim of this retrospective study was to assess the usefulness of 18F-FDG PET/CT radiomics features in predicting outcomes such as tumor and nodal categories, PET-based response to induction chemotherapy, progression-free survival, and overall survival. In a final cohort of 74 patients, we found that the developed radiomics models can predict these clinical and prognostic outcomes with reasonable accuracy, similar or better than those derived from conventional imaging. Future studies with a larger cohort would be helpful in establishing the significance of these models. Abstract This study aimed to assess the usefulness of radiomics features of 18F-FDG PET/CT in patients with locally advanced esophageal cancers (ESCC) in predicting outcomes such as clinical tumor (cT) and nodal (cN) categories, PET response to induction chemotherapy (PET response), progression-free survival (PFS), and overall survival (OS). Pretreatment PET/CT images from patients who underwent concurrent chemoradiotherapy from July 2002 to February 2017 were segmented, and data were split into training and test sets. Model development was performed on the training datasets and a maximum of five features were selected. Final diagnostic accuracies were determined using the test dataset. A total of 86 PET/CTs (58 men and 28 women, mean age 65 years) were segmented. Due to small lesion size, 12 patients were excluded. The diagnostic accuracies as derived from the CT, PET, and combined PET/CT test datasets were as follows: cT category—70.4%, 70.4%, and 81.5%, respectively; cN category—69.0%, 86.2%, and 86.2%, respectively; PET response—60.0%, 66.7%, and 70.0%, respectively; PFS—60.7%, 75.0%, and 75.0%, respectively; and OS—51.7%, 55.2%, and 62.1%, respectively. A radiomics assessment of locally advanced ESCC has the potential to predict various clinical outcomes. External validation of these models would be further helpful.
Collapse
Affiliation(s)
- Vetri Sudar Jayaprakasam
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (V.S.J.); (R.Y.)
| | - Peter Gibbs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.G.); (N.G.); (R.B.); (R.E.S.); (M.J.G.)
| | - Natalie Gangai
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.G.); (N.G.); (R.B.); (R.E.S.); (M.J.G.)
| | - Raazi Bajwa
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.G.); (N.G.); (R.B.); (R.E.S.); (M.J.G.)
| | - Ramon E. Sosa
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.G.); (N.G.); (R.B.); (R.E.S.); (M.J.G.)
| | - Randy Yeh
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (V.S.J.); (R.Y.)
| | | | - Geoffrey Y. Ku
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Marc J. Gollub
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.G.); (N.G.); (R.B.); (R.E.S.); (M.J.G.)
| | - Viktoriya Paroder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.G.); (N.G.); (R.B.); (R.E.S.); (M.J.G.)
- Correspondence:
| |
Collapse
|
22
|
Jayaprakasam VS, Gibbs P, Gangai N, Bajwa R, Sosa RE, Yeh R, Greally M, Ku GY, Gollub MJ, Paroder V. Can 18F-FDG PET/CT Radiomics Features Predict Clinical Outcomes in Patients with Locally Advanced Esophageal Squamous Cell Carcinoma? Cancers (Basel) 2022; 14:3035. [PMID: 35740700 PMCID: PMC9221147 DOI: 10.3390/cancers14123035&n999822=v982537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
This study aimed to assess the usefulness of radiomics features of 18F-FDG PET/CT in patients with locally advanced esophageal cancers (ESCC) in predicting outcomes such as clinical tumor (cT) and nodal (cN) categories, PET response to induction chemotherapy (PET response), progression-free survival (PFS), and overall survival (OS). Pretreatment PET/CT images from patients who underwent concurrent chemoradiotherapy from July 2002 to February 2017 were segmented, and data were split into training and test sets. Model development was performed on the training datasets and a maximum of five features were selected. Final diagnostic accuracies were determined using the test dataset. A total of 86 PET/CTs (58 men and 28 women, mean age 65 years) were segmented. Due to small lesion size, 12 patients were excluded. The diagnostic accuracies as derived from the CT, PET, and combined PET/CT test datasets were as follows: cT category-70.4%, 70.4%, and 81.5%, respectively; cN category-69.0%, 86.2%, and 86.2%, respectively; PET response-60.0%, 66.7%, and 70.0%, respectively; PFS-60.7%, 75.0%, and 75.0%, respectively; and OS-51.7%, 55.2%, and 62.1%, respectively. A radiomics assessment of locally advanced ESCC has the potential to predict various clinical outcomes. External validation of these models would be further helpful.
Collapse
Affiliation(s)
- Vetri Sudar Jayaprakasam
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (V.S.J.); (R.Y.)
| | - Peter Gibbs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.G.); (N.G.); (R.B.); (R.E.S.); (M.J.G.)
| | - Natalie Gangai
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.G.); (N.G.); (R.B.); (R.E.S.); (M.J.G.)
| | - Raazi Bajwa
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.G.); (N.G.); (R.B.); (R.E.S.); (M.J.G.)
| | - Ramon E. Sosa
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.G.); (N.G.); (R.B.); (R.E.S.); (M.J.G.)
| | - Randy Yeh
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (V.S.J.); (R.Y.)
| | | | - Geoffrey Y. Ku
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Marc J. Gollub
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.G.); (N.G.); (R.B.); (R.E.S.); (M.J.G.)
| | - Viktoriya Paroder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.G.); (N.G.); (R.B.); (R.E.S.); (M.J.G.)
- Correspondence:
| |
Collapse
|
23
|
Tustumi F, Albenda DG, Sallum RAA, Nahas SC, Ribeiro Junior U, Buchpiguel CA, Cecconello I, Duarte PS. 18F-FDG-PET/CT-measured parameters as potential predictors of residual disease after neoadjuvant chemoradiotherapy in patients with esophageal carcinoma. Radiol Bras 2022; 55:286-292. [PMID: 36320366 PMCID: PMC9620845 DOI: 10.1590/0100-3984.2021.0135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/27/2021] [Indexed: 12/22/2022] Open
Abstract
Objective To evaluate the maximum and mean standardized uptake values, together with
the metabolic tumor value and the total lesion glycolysis, at the primary
tumor site, as determined by 18F-fluorodeoxyglucose
positron-emission tomography/computed tomography
(18F-FDG-PET/CT), performed before and after neoadjuvant
chemoradiotherapy (nCRT), as predictors of residual disease (RD) in patients
with esophageal cancer. Materials and Methods The standardized uptake values and the volumetric parameters (metabolic tumor
value and total lesion glycolysis) were determined by
18F-FDG-PET/CT to identify RD in 39 patients before and after
nCRT for esophageal carcinoma. We used receiver operating characteristic
curves to analyze the diagnostic performance of 18F-FDG-PET/CT
parameters in the definition of RD. The standard of reference was
histopathological analysis of the surgical specimen. Results Eighteen patients (46%) presented RD after nCRT. Statistically significant
areas under the curve (approximately 0.72) for predicting RD were obtained
for all four of the variables evaluated after nCRT. Considering the presence
of visually detectable uptake (higher than the background level) at the
primary tumor site after nCRT as a positive result, we achieved a
sensitivity of 94% and a specificity of 48% for the detection of RD. Conclusion The use of 18F-FDG-PET/CT can facilitate the detection of RD after
nCRT in patients with esophageal cancer.
Collapse
|
24
|
Lee BM, Lee CG. Significance of mid-radiotherapy 18F-fluorodeoxyglucose positron emission tomography/computed tomography in esophageal cancer. Radiother Oncol 2022; 171:114-120. [PMID: 35447284 DOI: 10.1016/j.radonc.2022.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/27/2022] [Accepted: 04/11/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND AND PURPOSE Metabolic parameters evaluated by 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET)/computed tomography (CT) are known as prognostic markers in various cancers. We aimed to validate the predictive value of mid-radiotherapy (RT) FDG PET/CT parameters in esophageal cancer. MATERIALS AND METHODS Eighty-three patients treated with RT with or without chemotherapy between 2015 and 2020 were included. PET parameters including metabolic tumor volume (MTV), total lesion glycolysis, and mean (SUVmean) and maximum standardized uptake value (SUVmax) were analyzed. Locoregional recurrence-free rate (LRFR) and distant metastasis-free rate (DMFR) were analyzed. RESULTS The median follow-up period was 10.5 months. Mid-RT SUVmax was significantly associated with LRFR (HR 1.07, p = 0.009) and DMFR (HR 1.13, p=0.047) while mid-RT MTV was associated with DMFR (HR 1.06, p=0.007). Treatment response after RT was associated with overall survival (HR, 1.52, p=0.025). Further, treatment response was significantly associated with mid-RT SUVmax. The optimal cutoff value for mid-RT SUVmax in predicting LRFR and DMFR was 11 while cutoff value for mid-RT MTV was 15. The patients with mid-RT SUVmax≤11 showed superior LRFR and DMFR compared to SUVmax>11 (1-year LRFR; 73.4% vs 48.4%, p=0.028, 1-year DMFR; 74.6% vs 40.7%, p=0.007). The 1-year DMFR was significantly different between patients with mid-RT MTV≤15 and >15 (1-year DMFR; 78.2% vs 31.9%, p=0.002). CONCLUSION Tumor metabolism changes during RT can be a useful predictive tool for treatment response and recurrence in patients with esophageal cancer. Clinicians may consider early response evaluation with PET during RT for information about prognosis.
Collapse
Affiliation(s)
- Byung Min Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Radiation Oncology, Uijeongbu St. Mary's Hospital, The Catholic University of Korea, Uijeongbu, Gyeonggi-do, Republic of Korea
| | - Chang Geol Lee
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
25
|
Advances in the curative management of oesophageal cancer. Br J Cancer 2022; 126:706-717. [PMID: 34675397 PMCID: PMC8528946 DOI: 10.1038/s41416-021-01485-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/01/2021] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
The incidence of oesophageal cancer, in particular adenocarcinoma, has markedly increased over the last four decades with adenocarcinoma becoming the dominant subtype in the West, and mortality rates are high. Nevertheless, overall survival of patients with oesophageal cancer has doubled in the past 20 years, with earlier diagnosis and improved treatments benefiting those patients who can be treated with curative intent. Advances in endotherapy, surgical approaches, and multimodal and other combination therapies have been reported. New vistas have emerged in targeted therapies and immunotherapy, informed by new knowledge in genomics and molecular biology, which present opportunities for personalised cancer therapy and novel clinical trials. This review focuses exclusively on the curative intent treatment pathway, and highlights emerging advances.
Collapse
|
26
|
Bauer L, Lang K. [Randomized phase II study of PET response-adapted combined multimodal therapy for esophageal cancer: results of the CALGB 80803 (Alliance) trial]. Strahlenther Onkol 2022; 198:322-324. [PMID: 35015087 PMCID: PMC8863770 DOI: 10.1007/s00066-021-01898-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Lukas Bauer
- Universitätsklinikum Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Deutschland
| | - Kristin Lang
- Universitätsklinikum Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Deutschland.
| |
Collapse
|
27
|
Obermannova R, Selingerova I, Rehak Z, Jedlicka V, Slavik M, Fabian P, Novotny I, Zemanova M, Studentova H, Grell P, Zdrazilova Dubska L, Demlova R, Harustiak T, Hejnova R, Kiss I, Vyzula R. PET/CT-tailored treatment of locally advanced oesophago-gastric junction adenocarcinoma: a report on the feasibility of the multicenter GastroPET study. Ther Adv Med Oncol 2022; 13:17588359211065153. [PMID: 35035533 PMCID: PMC8753528 DOI: 10.1177/17588359211065153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Perioperative chemotherapy is a recommended treatment approach for localised oesophago-gastric junction adenocarcinoma, but not all patients respond to neoadjuvant chemotherapy. Early identification of non-responders and treatment adaptation in the preoperative period could improve outcomes. GastroPET is a national, multicentre phase II trial evaluating a 18FDG-PET/CT-guided preoperative treatment strategy with the R0 resection rate as a primary endpoint. Here, we report on the accuracy of the methodology, the feasibility of the study design and patient safety data after enrolment of the first 63 patients. METHODS Patients with locally advanced oesophago-gastric junction adenocarcinoma (Siewert I - III) stage Ib-IIIc underwent baseline 18FDG-PET/CT scanning and re-evaluation after 14 days of oxaliplatinum-5FU-(docetaxel) chemotherapy. Responders were defined by a ⩾ 35% decrease in tumour FDG standardised uptake value (SUV)average from baseline. Responders continued with the same chemotherapy for 2 to 3 months prior to surgery. PET-non-responders switched to preoperative chemoradiotherapy [weekly carboplatin and paclitaxel with concurrent radiotherapy (45 Gy in 25 fractions)]. Here, we aim to confirm the feasibility of FDG-PET-based response assessment in a multicenter setting and to compare local versus central reading. In addition, we report on the feasibility of the study conduct and patient safety data. RESULTS A total of 64 patients received baseline and sequential 14-day 18FDG-PET/CT scanning. And, 63 were allocated to the respective treatment arm according to PET-response [35 (56%) responders and 28 (44%) non-responders]. The concordance of local versus central reading of SUV changes was 100%. Until the date of this analysis, 47 patients (28 responders and 19 non-responders) completed surgery. Postoperative complications of grade ⩾ 3 (Common Terminology Criteria for Adverse Events, CTCAE Version 5.0) were reported in five responders (18%; 95% CI: 7.9-36%) and two non-responders (11%; 95% CI: 2.9-31%), with no statistical difference (p = 0.685). One patient in each arm died after surgery, leading to a postoperative in-hospital mortality rate of 4.3% (2/47 patients; 95% CI: 1.2-14%). CONCLUSION Local and central FDG-SUV quantification and PET-response assessment showed high concordance. This confirms the accuracy of a PET-response-guided treatment algorithm for locally advanced oesophago-gastric junction cancer in a multicenter setting. Preoperative treatment adaptation revealed feasible and safe for patients.
Collapse
Affiliation(s)
- Radka Obermannova
- Department of Comprehensive Cancer Care,
Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53 Brno, Czech
Republic
- Department of Comprehensive Cancer Care,
Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Iveta Selingerova
- Research Centre for Applied Molecular Oncology,
Masaryk Memorial Cancer Institute, Brno, Czech Republic
- Department of Pharmacology, Faculty of
Medicine, Masaryk University, Brno, Czech Republic
| | - Zdenek Rehak
- Department of Nuclear Medicine, Masaryk
Memorial Cancer Institute, Brno, Czech Republic
| | - Vaclav Jedlicka
- Department of Surgery, Masaryk Memorial Cancer
Institute, Brno, Czech Republic
- Department of Surgery, Faculty of Medicine,
Masaryk University, Brno, Czech Republic
| | - Marek Slavik
- Department of Radiation Oncology, Masaryk
Memorial Cancer Institute, Brno, Czech Republic
| | - Pavel Fabian
- Department of Pathology, Masaryk Memorial
Cancer Institute, Brno, Czech Republic
| | - Ivo Novotny
- Department of Gastroenterology, Masaryk
Memorial Cancer Institute, Brno, Czech Republic
| | - Milada Zemanova
- Department of Oncology, First Faculty of
Medicine, Charles University and General University Hospital in Prague,
Prague, Czech Republic
| | - Hana Studentova
- Department of Oncology, University Hospital
Olomouc, Olomouc, Czech Republic
| | - Peter Grell
- Department of Comprehensive Cancer Care,
Masaryk Memorial Cancer Institute, Brno, Czech Republic
- Department of Comprehensive Cancer Care,
Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lenka Zdrazilova Dubska
- Department of Laboratory Medicine – Clinical
Microbiology and Immunology, University Hospital Brno, Brno, Czech
Republic
| | - Regina Demlova
- Department of Pharmacology, Faculty of
Medicine, Masaryk University, Brno, Czech Republic
| | - Tomas Harustiak
- Third Department of Surgery, First Faculty of
Medicine, Charles University, Prague, Czech Republic
| | - Renata Hejnova
- Faculty of Medicine, Masaryk University, Brno,
Czech Republic
| | - Igor Kiss
- Department of Comprehensive Cancer Care,
Masaryk Memorial Cancer Institute, Brno, Czech Republic
- Department of Comprehensive Cancer Care,
Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Rostislav Vyzula
- Department of Comprehensive Cancer Care,
Masaryk Memorial Cancer Institute, Brno, Czech Republic
- Department of Comprehensive Cancer Care,
Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
28
|
Bott RK, George G, McEwen R, Zylstra J, Knight WRC, Baker CR, Kelly M, Griffin N, McAddy N, Maisey N, Van Hemelrijck M, Gossage JA, Lagergren J, Davies AR. Predicting response to neoadjuvant chemotherapy in patients with oesophageal adenocarcinoma. Acta Oncol 2021; 60:1629-1636. [PMID: 34613874 DOI: 10.1080/0284186x.2021.1986228] [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: 12/24/2022]
Abstract
BACKGROUND Neoadjuvant chemotherapy is often used prior to surgical resection for oesophageal adenocarcinoma but remains ineffective in a high proportion of patients. The histological Mandard tumour regression grade is used to determine chemoresponse but is not available at the time of treatment decision-making. The aim of this cohort study was to identify factors that predict chemotherapy response prior to surgery. METHODS A prospectively collected database of patients undergoing surgical resection for oesophageal adenocarcinoma from a high-volume UK institution was used. Patients were subcategorised using pathological tumour response into 'responders' (Mandard grade 1-3) and 'non-responders' (Mandard grade 4 and 5). Multivariable logistic regression analysis was performed to calculate crude and adjusted odds ratios (OR) with 95% confidence intervals (CI) for responder status adjusting for a variety of parameters. Receiver operating characteristic (ROC) curves were calculated. RESULTS Among 315 patients included, 102 (32%) were responders and 213 (68%) non-responders. A decrease in radiological tumour volume (OR 1.92 95%CI 1.02-3.62; p = 0.05), a 'partial response' RECIST score (OR 7.16 95%CI 1.49-34.36; p = 0.01), a clinically improved dysphagia score (OR 2.79 95%CI 1.05-7.04; p = 0.04) and lymphovascular invasion (OR 0.06 95%CI 0.02-0.13; p = 0.000) influenced responder status. ROC curve analysis for responder status utilising all available parameters had an area under the curve (AUC) of 0.86. CONCLUSION This study has highlighted the potential for using pre-defined factors to identify those patients who have responded to neoadjuvant chemotherapy, prior to surgical resection, potentially facilitating a more individualised therapeutic approach.
Collapse
Affiliation(s)
- Rebecca K. Bott
- Department of Upper Gastrointestinal and General Surgery, St Thomas’ Hospital, London, UK
- School of Cancer and Pharmaceutical Sciences, King’s College London, London, UK
| | - Gincy George
- School of Cancer and Pharmaceutical Sciences, Translational Oncology and Urology Research (TOUR), King’s College London, London, UK
| | - Ricardo McEwen
- Department of Upper Gastrointestinal and General Surgery, St Thomas’ Hospital, London, UK
| | - Janine Zylstra
- Department of Upper Gastrointestinal and General Surgery, St Thomas’ Hospital, London, UK
| | - William R. C. Knight
- Department of Upper Gastrointestinal and General Surgery, St Thomas’ Hospital, London, UK
- School of Cancer and Pharmaceutical Sciences, King’s College London, London, UK
| | - Cara R. Baker
- Department of Upper Gastrointestinal and General Surgery, St Thomas’ Hospital, London, UK
- School of Cancer and Pharmaceutical Sciences, King’s College London, London, UK
| | - Mark Kelly
- Department of Upper Gastrointestinal and General Surgery, St Thomas’ Hospital, London, UK
- School of Cancer and Pharmaceutical Sciences, King’s College London, London, UK
| | - Nyree Griffin
- Department of Radiology, St Thomas’ Hospital, London, UK
| | - Naami McAddy
- Department of Radiology, St Thomas’ Hospital, London, UK
| | - Nick Maisey
- Department of Medical Oncology, Guy’s Hospital London, London, UK
| | - Mieke Van Hemelrijck
- School of Cancer and Pharmaceutical Sciences, Translational Oncology and Urology Research (TOUR), King’s College London, London, UK
| | - James A. Gossage
- Department of Upper Gastrointestinal and General Surgery, St Thomas’ Hospital, London, UK
- School of Cancer and Pharmaceutical Sciences, King’s College London, London, UK
| | - Jesper Lagergren
- School of Cancer and Pharmaceutical Sciences, King’s College London, London, UK
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Andrew R. Davies
- Department of Upper Gastrointestinal and General Surgery, St Thomas’ Hospital, London, UK
- School of Cancer and Pharmaceutical Sciences, King’s College London, London, UK
| |
Collapse
|
29
|
Goodman KA, Ou FS, Hall NC, Bekaii-Saab T, Fruth B, Twohy E, Meyers MO, Boffa DJ, Mitchell K, Frankel WL, Niedzwiecki D, Noonan A, Janjigian YY, Thurmes PJ, Venook AP, Meyerhardt JA, O'Reilly EM, Ilson DH. Randomized Phase II Study of PET Response-Adapted Combined Modality Therapy for Esophageal Cancer: Mature Results of the CALGB 80803 (Alliance) Trial. J Clin Oncol 2021; 39:2803-2815. [PMID: 34077237 PMCID: PMC8407649 DOI: 10.1200/jco.20.03611] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/01/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022] Open
Abstract
PURPOSE To evaluate the use of early assessment of chemotherapy responsiveness by positron emission tomography (PET) imaging to tailor therapy in patients with esophageal and esophagogastric junction adenocarcinoma. METHODS After baseline PET, patients were randomly assigned to an induction chemotherapy regimen: modified oxaliplatin, leucovorin, and fluorouracil (FOLFOX) or carboplatin-paclitaxel (CP). Repeat PET was performed after induction; change in maximum standardized uptake value (SUV) from baseline was assessed. PET nonresponders (< 35% decrease in SUV) crossed over to the alternative chemotherapy during chemoradiation (50.4 Gy/28 fractions). PET responders (≥ 35% decrease in SUV) continued on the same chemotherapy during chemoradiation. Patients underwent surgery at 6 weeks postchemoradiation. Primary end point was pathologic complete response (pCR) rate in nonresponders after switching chemotherapy. RESULTS Two hundred forty-one eligible patients received Protocol treatment, of whom 225 had an evaluable repeat PET. The pCR rates for PET nonresponders after induction FOLFOX who crossed over to CP (n = 39) or after induction CP who changed to FOLFOX (n = 50) was 18.0% (95% CI, 7.5 to 33.5) and 20% (95% CI, 10 to 33.7), respectively. The pCR rate in responders who received induction FOLFOX was 40.3% (95% CI, 28.9 to 52.5) and 14.1% (95% CI, 6.6 to 25.0) in responders to CP. With a median follow-up of 5.2 years, median overall survival was 48.8 months (95% CI, 33.2 months to not estimable) for PET responders and 27.4 months (95% CI, 19.4 months to not estimable) for nonresponders. For induction FOLFOX patients who were PET responders, median survival was not reached. CONCLUSION Early response assessment using PET imaging as a biomarker to individualize therapy for patients with esophageal and esophagogastric junction adenocarcinoma was effective, improving pCR rates in PET nonresponders. PET responders to induction FOLFOX who continued on FOLFOX during chemoradiation achieved a promising 5-year overall survival of 53%.
Collapse
Affiliation(s)
| | - Fang-Shu Ou
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN
| | - Nathan C. Hall
- Hospital of the University of Pennsylvania, Philadelphia, PA
| | | | - Briant Fruth
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN
| | - Erin Twohy
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | - Anne Noonan
- The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - Paul J. Thurmes
- Metro Minnesota Community Oncology Research Consortium, Minneapolis, MN
| | - Alan P. Venook
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | | | | | | |
Collapse
|
30
|
Abstract
Gastrointestinal malignancies encompass a variety of primary tumor sites, each with different staging criteria and treatment approaches. In this review we discuss technical aspects of 18F-FDG-PET/CT scanning to optimize information from both the PET and computed tomography components. Specific applications for 18F-FDG-PET/CT are summarized for initial staging and follow-up of the major disease sites, including esophagus, stomach, hepatobiliary system, pancreas, colon, rectum, and anus.
Collapse
Affiliation(s)
- Brandon A Howard
- Division of Nuclear Medicine and Radiotheranostics, Department of Radiology, Duke University Medical Center, DUMC Box 3949, 2301 Erwin Road, Durham, NC 27710, USA.
| | - Terence Z Wong
- Division of Nuclear Medicine and Radiotheranostics, Department of Radiology, Duke University Medical Center, DUMC Box 3949, 2301 Erwin Road, Durham, NC 27710, USA
| |
Collapse
|
31
|
Gastric cancer with positive peritoneal cytology: survival benefit after induction chemotherapy and conversion to negative peritoneal cytology. World J Surg Oncol 2021; 19:245. [PMID: 34404403 PMCID: PMC8371873 DOI: 10.1186/s12957-021-02351-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022] Open
Abstract
Background The optimal treatment in patients with gastric cancer and peritoneal disease remains controversial. Some guidelines indicate palliative treatment only, while others consider surgical treatment in case of positive lavage cytology (CY+) or limited peritoneal disease. Here, we analyzed the role of peritoneal disease in patients with gastric cancer, and the prognostic relevance of response to neoadjuvant therapy. Methods In this retrospective cohort analysis, we analyzed patients with adenocarcinoma of the stomach or esophago-gastric junction from a single center operated between 2011 and 2019. According to histology and lavage cytology, patients were classified into four risk groups: (A) no peritoneal disease, (B) CY+ who converted to negative lavage cytology (CY−) after neoadjuvant chemotherapy, (C) CY+ without conversion after chemotherapy, and (D) patients with visible peritoneal metastasis. Results Overall, n = 172 patients were included. At initial presentation, n = 125 (73%) had no peritoneal disease, and about a third of patients (n = 47, 27%) had microscopic or macroscopic peritoneal disease. Among them, n = 14 (8%) were CY+ without visible peritoneal metastasis, n = 9 converted to CY− after chemotherapy, and in n = 5 no conversion was observed. Median overall survival was not reached in patients who had initially no peritoneal disease and in patients who converted after chemotherapy, resulting in 3-year survival rates of 65% and 53%. In contrast, median overall survival was reduced to 13 months (95% CI 8.7–16.7) in patients without conversion and was 16 months (95% CI 12–20.5) in patients with peritoneal metastasis without difference between the two groups (p = .364). The conversion rate from CY+ to CY− was significantly higher after neoadjuvant treatment with FLOT (5-fluorouracil plus leucovorin, oxaliplatin, and docetaxel) compared to ECF (epirubicin, cisplatin, and 5-fluorouracil) (p = 0.027). Conclusion Conversion of CY+ to CY− after neoadjuvant chemotherapy with FLOT is a significant prognostic factor for a better overall survival. Surgical treatment in well-selected patients should therefore be considered. However, peritoneal recurrence remains frequent despite conversion, urging for a better local control.
Collapse
|
32
|
Benz MR, Armstrong WR, Ceci F, Polverari G, Donahue TR, Wainberg ZA, Quon A, Auerbach M, Girgis MD, Herrmann K, Czernin J, Calais J. 18F-FDG PET/CT imaging biomarkers for early and late evaluation of response to first-line chemotherapy in patients with pancreatic ductal adenocarcinoma. J Nucl Med 2021; 63:199-204. [PMID: 34272317 DOI: 10.2967/jnumed.121.261952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/05/2021] [Indexed: 11/16/2022] Open
Abstract
Purpose: The purpose of this study was to evaluate 18F-FDG-PET/CT as an early and late interim imaging biomarker in patients with pancreatic ductal adenocarcinoma (PDAC) who undergo first-line systemic therapy. Methods: This was a prospective, single-center, single-arm, open-label study (IRB12-000770). Patient receiving first line chemotherapy were planned to undergo a baseline 18F-FDG-PET/CT (PET1), early interim 18F-FDG PET/CT (PET2) and late interim 18F-FDG-PET/CT (PET3). ROC selected and established (mPERCIST / RECIST1.1) cut-offs for metabolic and radiographic tumor response assessment were applied. Patients were followed to collect data on further treatments and overall survival (OS). Results: The study population consisted of 28 patients who underwent PET1. Twenty-three of these (82%) underwent PET2 and 21 (75%) PET3, respectively. Twenty-three deaths occurred during a median follow up period of 14 months (maximum follow up, 58.3 months). The median OS was 36.2 months (95%CI, 28-NYR) in early metabolic responders (6/23 (26%), P = 0.016) and 25.4 months (95%CI, 19.6-NYR) in early radiographic responders (7/23 (30%), P = 0.16). The median overall survival was 27.4 months (95%CI, 21.4-NYR) in late metabolic responders (10/21 (48%), P = 0.058) and 58.2 months (95%CI, 21.4-NYR) in late radiographic responders (7/21 (33%), P = 0.008). Conclusion: 18F-FDG PET may serve as early interim imaging biomarker (~ at 4 weeks) for evaluation of response to first-line chemotherapy in patients with PDAC. Radiographic changes might be sufficient for response evaluation after the completion of first line chemotherapy.
Collapse
Affiliation(s)
- Matthias R Benz
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, United States
| | - Wesley R Armstrong
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, United States
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Milan, Italy
| | | | | | - Zev A Wainberg
- Department of Medical Oncology, University of California, Los Angeles, CA
| | - Andrew Quon
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, United States
| | - Martin Auerbach
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, United States
| | - Mark D Girgis
- Department of Surgery, University of California, Los Angeles, CA
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Johannes Czernin
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, United States
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, United States
| |
Collapse
|
33
|
Pucher PH, Wijnhoven BPL, Underwood TJ, Reynolds JV, Davies AR. Thinking through the multimodal treatment of localized oesophageal cancer: the point of view of the surgeon. Curr Opin Oncol 2021; 33:353-361. [PMID: 33966001 DOI: 10.1097/cco.0000000000000751] [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: 10/21/2022]
Abstract
PURPOSE OF REVIEW This review examines current developments and controversies in the multimodal management of oesophageal cancer, with an emphasis on surgical dilemmas and outcomes from the surgeon's perspective. RECENT FINDINGS Despite the advancement of oncological neoadjuvant treatments, there is still no consensus on what regimen is superior. The majority of patients may still fail to respond to neoadjuvant therapy and suffer potential harm without any survival advantage as a result. In patients who do not respond, adjuvant therapy is still often recommended after surgery despite any evidence for its benefit. We examine the implications of different regimens and treatment approaches for both squamous cell cancer and adenocarcinoma of the oesophagus. SUMMARY The efficacy of neoadjuvant treatment is highly variable and likely relates to variability of tumour biology. Ongoing work to identify responders, or optimize treatment on an individual patient, should increase the efficacy of multimodal therapy and improve patient outcomes.
Collapse
Affiliation(s)
- Philip H Pucher
- Department of Surgery, Guy's and St Thomas' NHS Foundation Trust, London
- Department of Surgery, Portsmouth University Hospitals NHS Trust, Portsmouth, UK
| | - Bas P L Wijnhoven
- Department of Surgery, Erasmus MC-Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Timothy J Underwood
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, UK
| | - John V Reynolds
- Department of Surgery, National Oesophageal and Gastric Center, St. James's Hospital and Trinity College, Dublin, Ireland
| | - Andrew R Davies
- Department of Surgery, Guy's and St Thomas' NHS Foundation Trust, London
- King's College London, London, UK
| |
Collapse
|
34
|
Foley KG, Lavery A, Napier E, Campbell D, Eatock MM, Kennedy RD, Bradley KM, Turkington RC. A DNA-damage immune response assay combined with PET biomarkers predicts response to neo-adjuvant chemotherapy and survival in oesophageal adenocarcinoma. Sci Rep 2021; 11:13061. [PMID: 34158588 PMCID: PMC8219719 DOI: 10.1038/s41598-021-92545-w] [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: 12/08/2020] [Accepted: 06/07/2021] [Indexed: 11/24/2022] Open
Abstract
18F-fluorodeoxyglucose PET-CT may guide treatment decisions in patients with oesophageal adenocarcinoma (OAC). This study evaluated the added value of maximum standardised uptake value (SUVmax) to a novel DNA-damage immune response (DDIR) assay to improve pathological response prediction. The diagnostic accuracy of PET response and the prognostic significance of PET metrics for recurrence-free survival (RFS) and overall survival (OS) were assessed. This was a retrospective, single-centre study of OAC patients treated with neo-adjuvant chemotherapy from 2003 to 2014. SUVmax was recorded from baseline and repeat PET-CT after completion of pre-operative chemotherapy. Logistic regression models tested the additional predictive value of PET metrics combined with the DDIR assay for pathological response. Cox regression models tested the prognostic significance of PET metrics for RFS and OS. In total, 113 patients were included; 25 (22.1%) were DDIR positive and 88 (77.9%) were DDIR negative. 69 (61.1%) were PET responders (SUVmax reduction of 35%) and 44 (38.9%) were PET non-responders. After adding PET metrics to DDIR status, post-chemotherapy SUVmax (hazard ratio (HR) 0.75, p = 0.02), SUVmax change (HR 1.04, p = 0.003) and an optimum SUVmax reduction of 46.5% (HR 4.36, p = 0.021) showed additional value for predicting pathological response. The optimised SUVmax threshold was independently significant for RFS (HR 0.47, 95% CI 0.26–0.85, p = 0.012) and OS (HR 0.51, 95% CI 0.26–0.99, p = 0.047). This study demonstrated the additional value of PET metrics, when combined with a novel DDIR assay, to predict pathological response in OAC patients treated with neo-adjuvant chemotherapy. Furthermore, an optimised SUVmax reduction threshold for pathological response was calculated and was independently significant for RFS and OS.
Collapse
Affiliation(s)
| | | | - Eoin Napier
- Belfast Health and Social Care Trust, Belfast, UK
| | | | - Martin M Eatock
- Queen's University Belfast, Belfast, UK.,Belfast Health and Social Care Trust, Belfast, UK
| | | | - Kevin M Bradley
- Wales Research & Diagnostic Positron Emission Tomography Imaging Centre (PETIC), Cardiff University, Cardiff, UK
| | | |
Collapse
|
35
|
Foley KG, Jeffries J, Hannon C, Coles B, Bradley KM, Smyth E. Response rate and diagnostic accuracy of early PET-CT during neo-adjuvant therapies in oesophageal adenocarcinoma: A systematic review and meta-analysis. Int J Clin Pract 2021; 75:e13906. [PMID: 33300222 DOI: 10.1111/ijcp.13906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/03/2020] [Indexed: 11/28/2022] Open
Abstract
PURPOSE Only 25% of oesophageal adenocarcinoma (OAC) patients have a pathological response to neo-adjuvant therapy (NAT) before oesophagectomy. Early response assessment using PET imaging may help guide management of these patients. We performed a systematic review and meta-analysis to synthesise the evidence detailing response rate and diagnostic accuracy of early PET-CT assessment. METHODS We systematically searched several databases including MEDLINE and Embase. Studies with mixed cohorts of histology, tumour location and a repeat PET-CT assessment after more than one cycle of NAT were excluded. Reference standard was pathological response defined by Becker or Mandard classifications. Primary outcome was metabolic response rate after one cycle of NAT defined by a reduction in maximum standardised uptake value (SUVmax) of 35%. Secondary outcome was diagnostic accuracy of treatment response prediction, defined as the sensitivity and specificity of early PET-CT using this threshold. Quality of evidence was also assessed. Random-effects meta-analysis pooled response rates and diagnostic accuracy. This study was registered with PROSPERO (CRD42019147034). RESULTS Overall, 1341 articles were screened, and 6 studies were eligible for analysis. These studies reported data for 518 patients (aged 27-78 years; 452 [87.3%] were men) between 2005 and 2020. Pooled sensitivity of early metabolic response to predict pathological response was 77.2% (95% CI 53.2%-100%). Significant heterogeneity existed between studies (I2 = 80.6% (95% CI 38.9%-93.8%), P = .006). Pooled specificity was 75.0% (95% CI 68.2%-82.5%), however, no significant heterogeneity between studies existed (I2 = 0.0% (95% CI 0.0%-67.4%), P = .73). CONCLUSION High-quality evidence is lacking, and few studies met the inclusion criteria of this systematic review. The sensitivity of PET using a SUVmax reduction threshold of 35% was suboptimal and varied widely. However, specificity was consistent across studies with a pooled value of 75.0%, suggesting early PET assessment is a better predictor of treatment resistance than of pathological response. Further research is required to define optimal PET-guided treatment decisions in OAC.
Collapse
Affiliation(s)
- Kieran G Foley
- Royal Glamorgan Hospital & Velindre Cancer Centre, Cardiff, UK
| | | | - Clare Hannon
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Kevin M Bradley
- Wales Diagnostic and Research Positron Emission Tomography Imaging Centre, Cardiff University, Cardiff, UK
| | - Elizabeth Smyth
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
36
|
Lex JR, Kurisunkal V, Kaneuchi Y, Fujiwara T, Sherriff J, Wigley C, Stevenson JD, Parry MC, Jeys LM. Pelvic Ewing sarcoma: Should all patients receive pre-operative radiotherapy, or should it be delivered selectively? Eur J Surg Oncol 2021; 47:2618-2626. [PMID: 34030919 DOI: 10.1016/j.ejso.2021.05.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Locally recurrent disease following surgical resection of Ewing sarcoma (ES) confers a poor prognosis. Limited evidence is available evaluating non-selective use of pre-operative radiotherapy (RT) for patients with pelvic ES and its effect on local control and survival. PATIENTS AND METHODS 49 consecutive patients with pelvic ES were identified retrospectively from a prospectively collated database. Patients either received non-selective pre-operative RT and surgery (n = 27), or selective post-operative RT (n = 22) (surgery alone (n = 11) or surgery and post-operative RT (n = 11)). RESULTS Patients who had non-selective pre-operative radiotherapy appeared to have a higher LRFS, 88.0% compared to 66.5% in the selective RT group (p = 0.096, Kaplan Meier; p = 0.028, Chi-squared). Administration of non-selective, pre-operative RT to all patients with pelvic ES elevates the LFRS to that of the good responder group (≥90% tumour necrosis and margins, p = 0.880). There was no difference in metastasis-free survival, 60.0% and 54.5% (p = 0.728) and overall survival (OS), 57.7% and 63.6% (p = 0.893). The majority of pre-operative RT patients had both good necrosis (≥90%) (p = 0.003) and widely excised tumours, 81.5% vs 59.1% (p = 0.080). Tumour volume ≥250 ml was associated with worse LRFS (p = 0.045) and post-operative complications (p = 0.017). There may be improved LRFS (p = 0.057) with pre-operative proton-beam RT compared to surgery and selective post-operative RT. CONCLUSION Pre-operative photon or proton-beam RT to all pelvic ES may improve LRFS compared to the selective delivery of post-operative RT. Radiotherapy delivered to all patients results in a greater percentage of highly necrotic tumours at surgical excision, enabling a greater proportion of patients with wide resection margins.
Collapse
Affiliation(s)
- Johnathan R Lex
- Oncology Department, The Royal Orthopaedic Hospital, Birmingham, UK; Division of Orthopaedic Surgery, Department of Surgery, University of Toronto, Toronto, Canada.
| | - Vineet Kurisunkal
- Oncology Department, The Royal Orthopaedic Hospital, Birmingham, UK.
| | - Yoichi Kaneuchi
- Oncology Department, The Royal Orthopaedic Hospital, Birmingham, UK.
| | - Tomohiro Fujiwara
- Oncology Department, The Royal Orthopaedic Hospital, Birmingham, UK.
| | - Jenny Sherriff
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
| | - Catrin Wigley
- Oncology Department, The Royal Orthopaedic Hospital, Birmingham, UK.
| | - Jonathan D Stevenson
- Oncology Department, The Royal Orthopaedic Hospital, Birmingham, UK; Aston University Medical School, Aston University, Birmingham, UK.
| | - Michael C Parry
- Oncology Department, The Royal Orthopaedic Hospital, Birmingham, UK; Aston University Medical School, Aston University, Birmingham, UK.
| | - Lee M Jeys
- Oncology Department, The Royal Orthopaedic Hospital, Birmingham, UK; Faculty of Health Sciences, Aston University, Birmingham, UK.
| |
Collapse
|
37
|
Duclos V, Iep A, Gomez L, Goldfarb L, Besson FL. PET Molecular Imaging: A Holistic Review of Current Practice and Emerging Perspectives for Diagnosis, Therapeutic Evaluation and Prognosis in Clinical Oncology. Int J Mol Sci 2021; 22:4159. [PMID: 33923839 PMCID: PMC8073681 DOI: 10.3390/ijms22084159] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023] Open
Abstract
PET/CT molecular imaging has been imposed in clinical oncological practice over the past 20 years, driven by its two well-grounded foundations: quantification and radiolabeled molecular probe vectorization. From basic visual interpretation to more sophisticated full kinetic modeling, PET technology provides a unique opportunity to characterize various biological processes with different levels of analysis. In clinical practice, many efforts have been made during the last two decades to standardize image analyses at the international level, but advanced metrics are still under use in practice. In parallel, the integration of PET imaging with radionuclide therapy, also known as radiolabeled theranostics, has paved the way towards highly sensitive radionuclide-based precision medicine, with major breakthroughs emerging in neuroendocrine tumors and prostate cancer. PET imaging of tumor immunity and beyond is also emerging, emphasizing the unique capabilities of PET molecular imaging to constantly adapt to emerging oncological challenges. However, these new horizons face the growing complexity of multidimensional data. In the era of precision medicine, statistical and computer sciences are currently revolutionizing image-based decision making, paving the way for more holistic cancer molecular imaging analyses at the whole-body level.
Collapse
Affiliation(s)
- Valentin Duclos
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270 Le Kremlin-Bicêtre, France; (V.D.); (A.I.); (L.G.)
| | - Alex Iep
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270 Le Kremlin-Bicêtre, France; (V.D.); (A.I.); (L.G.)
| | - Léa Gomez
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270 Le Kremlin-Bicêtre, France; (V.D.); (A.I.); (L.G.)
| | - Lucas Goldfarb
- Service Hospitalier Frédéric Joliot-CEA, 91401 Orsay, France;
| | - Florent L. Besson
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270 Le Kremlin-Bicêtre, France; (V.D.); (A.I.); (L.G.)
- Université Paris Saclay, CEA, CNRS, Inserm, BioMaps, 91401 Orsay, France
- School of Medicine, Université Paris Saclay, 94720 Le Kremlin-Bicêtre, France
| |
Collapse
|
38
|
Manning MA, Shafa S, Mehrotra AK, Grenier RE, Levy AD. Role of Multimodality Imaging in Gastroesophageal Reflux Disease and Its Complications, with Clinical and Pathologic Correlation. Radiographics 2021; 40:44-71. [PMID: 31917657 DOI: 10.1148/rg.2020190029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Gastroesophageal reflux disease (GERD) is a common condition and impairs the quality of life for millions of patients, accounts for considerable health care spending, and is a primary risk factor for esophageal adenocarcinoma. There have been substantial advances in understanding the pathogenesis of GERD and its complications and much progress in diagnosis and management of GERD; however, these have not been comprehensively discussed in the recent radiology literature. Understanding the role of imaging in GERD and its complications is important to aid in multidisciplinary treatment of GERD. GERD results from prolonged or recurrent reflux of gastric contents into the esophagus. Common symptoms include heartburn or regurgitation. Prolonged reflux of gastric contents into the esophagus can cause erosive esophagitis. Over time, the inflammatory response related to esophagitis can lead to deposition of fibrous tissue and development of strictures. Alternatively, the esophageal mucosa can undergo metaplasia (Barrett esophagus), a precursor to dysplasia (which can lead to adenocarcinoma). Conventional barium esophagography has long been considered the primary imaging modality for the esophagus, and the fluoroscopic findings for diagnosis of GERD have been well established. Multimodality imaging has a clear role in detection and assessment of the complications of GERD, specifically reflux esophagitis and Barrett esophagus; differentiation of benign and malignant strictures; and detection, staging, and posttreatment surveillance of esophageal adenocarcinoma. Given the dramatic changes in utilization of abdominal imaging during the past 2 decades, with significantly declining volume of fluoroscopic procedures and concomitant increase in CT and MRI studies, it is crucial that modern radiologists appreciate the value of barium esophagography in the workup of GERD and recognize the key imaging features of GERD and its complications at CT and MRI.
Collapse
Affiliation(s)
- Maria A Manning
- From the American Institute for Radiologic Pathology, 1100 Wayne Ave, Suite 1020, Silver Spring, MD 20910 (M.A.M.); Department of Radiology (M.A.M., A.D.L.) and Division of Gastroenterology and Hepatology (S.S.), MedStar Georgetown University Hospital, Washington, DC; the Joint Pathology Center, Silver Spring, Md (A.K.M.); and Georgetown University School of Medicine, Washington, DC (R.E.G.)
| | - Shervin Shafa
- From the American Institute for Radiologic Pathology, 1100 Wayne Ave, Suite 1020, Silver Spring, MD 20910 (M.A.M.); Department of Radiology (M.A.M., A.D.L.) and Division of Gastroenterology and Hepatology (S.S.), MedStar Georgetown University Hospital, Washington, DC; the Joint Pathology Center, Silver Spring, Md (A.K.M.); and Georgetown University School of Medicine, Washington, DC (R.E.G.)
| | - Anupamjit K Mehrotra
- From the American Institute for Radiologic Pathology, 1100 Wayne Ave, Suite 1020, Silver Spring, MD 20910 (M.A.M.); Department of Radiology (M.A.M., A.D.L.) and Division of Gastroenterology and Hepatology (S.S.), MedStar Georgetown University Hospital, Washington, DC; the Joint Pathology Center, Silver Spring, Md (A.K.M.); and Georgetown University School of Medicine, Washington, DC (R.E.G.)
| | - Rachel E Grenier
- From the American Institute for Radiologic Pathology, 1100 Wayne Ave, Suite 1020, Silver Spring, MD 20910 (M.A.M.); Department of Radiology (M.A.M., A.D.L.) and Division of Gastroenterology and Hepatology (S.S.), MedStar Georgetown University Hospital, Washington, DC; the Joint Pathology Center, Silver Spring, Md (A.K.M.); and Georgetown University School of Medicine, Washington, DC (R.E.G.)
| | - Angela D Levy
- From the American Institute for Radiologic Pathology, 1100 Wayne Ave, Suite 1020, Silver Spring, MD 20910 (M.A.M.); Department of Radiology (M.A.M., A.D.L.) and Division of Gastroenterology and Hepatology (S.S.), MedStar Georgetown University Hospital, Washington, DC; the Joint Pathology Center, Silver Spring, Md (A.K.M.); and Georgetown University School of Medicine, Washington, DC (R.E.G.)
| |
Collapse
|
39
|
Tumor Regression in Lymph Node Metastases of Esophageal Adenocarcinomas after Neoadjuvant Therapy. GASTROINTESTINAL DISORDERS 2020. [DOI: 10.3390/gidisord2040036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Tumor regression following neoadjuvant treatment can be observed in lymph node (LN) metastases similar to the primary tumor in esophageal adenocarcinomas (EAC). We evaluated the prognostic significance of tumor regression in LN metastases of locally advanced EAC of 239 patients treated with neoadjuvant radiochemotherapy (RCTX) or chemotherapy (CTX) followed by esophagectomy. We examined retrospectively the LN for histopathologic signs of regression, i.e., nodular fibrosis and acellular mucin. LN classification was performed according to two parameters: presence (−) or absence (+) of residual tumor and regression characteristics in the LN, resulting in four categories: LN−/REG−, LN−/REG+, LN+/REG+, LN+/REG−. In total, LN metastases with residual tumor were detectable in 117/239 (49%) cases. Regression in LN were observed in 85/239 cases (35.5%). The distribution of the LN/REG categories were as follows: 97 patients (40.6%) were LN−/REG−. A total of 25 patients (10.5%) were LN−/REG+. A total of 60 (25.1%) were LN+/REG+ and 57 (23.8%) LN+/REG−. The LN/Reg categorization had a significant prognostic value in univariate analysis (p < 0.001) and multivariate analysis (HR = 1.326; p = 0.002) with similar results for the subgroups of patients treated with RCTX or CTX. The prognosis of LN−/REG+ was worse than LN−/REG− but better than both LN+ categories, which was demonstrated in the Kaplan–Meier curves but did not reach statistical significance (p = 0.104 and p = 0.090, respectively). In contrast, there was no difference between LN+/REG+ and LN+/REG− (p = 0.802). In summary, regression in LN metastases of EAC can be observed in a significant number of patients after neoadjuvant therapy. Complete regression of former LN metastases in comparison to “true” negative LN seems to be of prognostic relevance but additional studies are needed to confirm this trend seen in our study.
Collapse
|
40
|
Raggi D, Bandini M, Giannatempo P, Farè E, Marandino L, Colecchia M, Calareso G, Padovano B, Serafini G, Alessi A, Necchi A. Prognostic Role of Early Interim Fluorodeoxyglucose Positron Emission Tomography in Patients With Advanced Seminoma Undergoing Standard Treatment. Clin Genitourin Cancer 2020; 19:237-245.e2. [PMID: 32980271 DOI: 10.1016/j.clgc.2020.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 01/12/2023]
Abstract
BACKGROUND Patients with advanced seminoma have an exceedingly favorable prognosis. Studies aiming to reduce the total treatment burden and side effects in patients with well-defined disease and very good prognosis are warranted. PATIENTS AND METHODS In a prospective observational study, patients with advanced stage seminoma were treated with bleomycin, etoposide, and cisplatin (BEP) or EP according to guidelines. Fluorodeoxyglucose with positron emission tomography and computed tomography (FDG-PET/CT) examinations were performed at baseline, after 2 cycles (PET/CT2) in all patients, and after chemotherapy at the physician's discretion. Disease response to treatment assessed by PET/CT was qualitatively evaluated by 2 independent nuclear medicine physicians. Contrast-enhanced CT scans were also performed according to guidelines (at baseline, after treatment, during follow-up). The study's primary endpoint was to evaluate the association between PET/CT2 findings and relapse-free survival. RESULTS From January 2009 to January 2017, a total of 75 consecutive patients were enrolled, of whom 70 were included for analysis. The clinical disease stage was IIA-B and IIC-III in 40% and 60%, respectively. By local assessment, 46 PET/CT2 scans (65.7%) were reported as negative, and 46% of these patients had stage IIC-III disease. Five-year relapse-free survival of PET/CT2-positive patients was 75% (95% confidence interval, 60-95) compared to 97.8% (95% confidence interval, 93.7-100) of PET/CT2-negative patients (P = .002). In univariate analyses, PET/CT2 was significantly associated with relapse-free survival (P = .02). CONCLUSIONS No residual FDG uptake after 2 cycles of conventional chemotherapy is prognostic in advanced seminoma, but it may be useful to optimize the standard prognostic risk groups and may be tested within larger prospective clinical trials of chemotherapy deescalation.
Collapse
Affiliation(s)
- Daniele Raggi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco Bandini
- Unit of Urology, Urological Research Institute (URI), IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Patrizia Giannatempo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Farè
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Laura Marandino
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maurizio Colecchia
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppina Calareso
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Barbara Padovano
- Department of Nuclear Medicine, PET Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Gianluca Serafini
- Department of Nuclear Medicine, PET Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandra Alessi
- Department of Nuclear Medicine, PET Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Necchi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
41
|
Fu J, Tang L, Li ZY, Li XT, Zhu HF, Sun YS, Ji JF. Diffusion kurtosis imaging in the prediction of poor responses of locally advanced gastric cancer to neoadjuvant chemotherapy. Eur J Radiol 2020; 128:108974. [PMID: 32416553 DOI: 10.1016/j.ejrad.2020.108974] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/15/2020] [Accepted: 03/19/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE To assess the efficacy of diffusion kurtosis imaging (DKI) in the prediction of the treatment response to neoadjuvant chemotherapy in patients with locally advanced gastric cancer (LAGC). METHODS A total of 31 LAGC patients were enrolled in this prospective study. All patients underwent diffusion-weighted MRI examination (with b = 01, 2001, 5001, 8002, 10004, 15004, 20006 s/mm2, the subscript denotes the number of signal averages) before and after chemotherapy. DKI and mono-exponential (b = 0, 800 s/mm2) models were built. Apparent diffusion coefficient (ADC), mean diffusivity (MD) and mean kurtosis (MK) of the LAGC tumors were measured. The absolute change values (ΔX) and percentage change values (%ΔX) of the above parameters post neoadjuvant chemotherapy (NACT) were calculated. The response was evaluated according to the pathological tumor regression grade scores (effective response group: TRG 0-2, poor response group: TRG 3). Mann-Whitney U test and receiver operating characteristic (ROC) curves were applicated for statistical analysis. RESULTS There were 17 patients in the effective response group (ERG), and 14 patients in the poor response group (PRG). The MKpre and MKpost values in PRG were significantly higher than those in ERG [(0.671 ± 0.026) and (0.641 ± 0.019) vs. (0.584 ± 0.023) and (0.519 ± 0.018), p < 0.001]. ADCpost and MDpost in PRG were significantly lower than those in ERG (p = 0.005, p =0.001). Significant differences were also observed for % ΔMK, ΔMD and ΔMK between the two groups (p < 0.05). The area under the curve (AUC) for the prediction of PRG was highest for MKpost (AUC = 0.958, cutoff value = 0.614). The MKpre and MKpost had the highest sensitivity (91.70 %) and specificity (93.80 %) in the prediction of PRG, respectively. CONCLUSION Both DKI and ADC values show potential for the prediction of the PRG in LAGC patients. The DKI parameters, especially MKpost displayed the best performance.
Collapse
Affiliation(s)
- Jia Fu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Radiology Department, Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China; Department of Radiology, Civil Aviation General Hospital, No. 1 Gaojingjia, Chaoyang Road, Chaoyang District, Beijing 100123, China.
| | - Lei Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Radiology Department, Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China.
| | - Zi-Yu Li
- Department of Gastrointestinal Cancer Center Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China.
| | - Xiao-Ting Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Radiology Department, Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China.
| | - Hai-Feng Zhu
- Department of Radiology, Civil Aviation General Hospital, No. 1 Gaojingjia, Chaoyang Road, Chaoyang District, Beijing 100123, China.
| | - Ying-Shi Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Radiology Department, Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China.
| | - Jia-Fu Ji
- Department of Gastrointestinal Cancer Center Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, No. 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China.
| |
Collapse
|
42
|
Roy AC, Shapiro J, Burge M, Karapetis CS, Pavlakis N, Segelov E, Chau I, Lordick F, Chen LT, Barbour A, Tebbutt N, Price T. Management of early-stage gastro-esophageal cancers: expert perspectives from the Australasian Gastrointestinal Trials Group (AGITG) with invited international faculty. Expert Rev Anticancer Ther 2020; 20:305-324. [PMID: 32202178 DOI: 10.1080/14737140.2020.1746185] [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: 10/24/2022]
Abstract
Introduction: A multimodal approach in operable early-stage oesophago-gastric (OG) cancer has evolved in the last decade, leading to improvement in overall outcomes.Areas covered: A review of the published literature and conference abstracts was undertaken on the topic of optimal adjunctive chemotherapy or chemoradiotherapy in early-stage OG cancers. This review article focuses on the current evidence pertaining to neoadjuvant and perioperative strategies in curable OG cancers including the evolving landscape of immunotherapy and targeted drugs in this setting.Expert commentary: Adjunctive therapies in the form of preoperative chemo-radiotherapy (CRT) or chemotherapy and perioperative chemotherapy over surgery alone improve outcomes in patients with operable OG cancer. Although there are variations in practice around the world, a multi-disciplinary approach to patient care is of paramount importance. Immunotherapy and on treatment functional imaging are two examples of emerging strategies to improve the outcome for early-stage patients. A better understanding of the molecular biology of this disease may help overcome the problem of tumor heterogeneity and enable more rationally designed and targeted therapeutic interventions in the future.
Collapse
Affiliation(s)
- Amitesh C Roy
- Flinders Centre for Innovation in Cancer, Flinders University, Bedford Park, Australia
| | | | - Matt Burge
- Department of Cancer Care Services, Royal Brisbane Hospital, University Of Queensland, Herston, Australia
| | - Christos S Karapetis
- Flinders Centre for Innovation in Cancer, Flinders University, Bedford Park, Australia
| | - Nick Pavlakis
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, Australia
| | - Eva Segelov
- Department of Medical Oncology, Monash University and Monash Health, Melbourne, Australia
| | - Ian Chau
- Department of Medical Oncology, Royal Marsden Hospital, Institute of Cancer Research, Surrey, London, UK
| | - Florian Lordick
- Leipzig University Medical Centre, University Cancer Centre Leipzig, Leipzig, Germany
| | - Li-Tong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Andrew Barbour
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Australia
| | - Niall Tebbutt
- Department of Medical Oncology, Austin Health, Heidelberg, Australia
| | - Tim Price
- Queen Elizabeth Hospital, University of Adelaide, Adelaide, Australia
| |
Collapse
|
43
|
Abstract
The use of 18F-fluorodeoxyglucose (FDG) positron emission tomography combined with computed tomography (PET/CT) is well established in the evaluation of alimentary tract malignancies. This review of the literature and demonstration of correlative images focuses on the current role of PET/CT in the diagnosis (including pathologic/clinical staging) and post-therapy follow-up of esophageal, gastric, and colorectal cancers. PET/CT provides utility in the management of esophageal cancer, including detection of distant disease prior to resection. In gastric cancer, PET/CT is useful in detecting solid organ metastases and in characterizing responders vs. non-responders after neoadjuvant chemotherapy, the latter of which have poorer overall survival. In patients with GIST tumors, PET/CT also determines response to imatinib therapy with greater expedience as compared to CECT. For colorectal cancer, PET/CT has proven helpful in detecting hepatic and other distant metastases, treatment response, and differentiating post-radiation changes from tumor recurrence. Our review also highlights several pitfalls in PET/CT interpretation of alimentary tract lesions.
Collapse
|
44
|
Kadota T, Abe S, Yoda Y, Yoshinaga S, Oda I, Kojima T, Kato K, Daiko H, Yano T. Clinical outcomes according to the modified endoscopic criteria for neoadjuvant chemotherapy in resectable esophageal squamous cell carcinoma. Dig Endosc 2020; 32:337-345. [PMID: 31295769 DOI: 10.1111/den.13483] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/03/2019] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Neoadjuvant chemotherapy (nCT) followed by surgery is one of the standard treatments for resectable esophageal squamous cell carcinoma (ESCC). According to the Response Evaluation Criteria in Solid Tumors, endoscopic evaluation of a primary lesion is not recommended during nCT because of reduced objectivity. This study aimed to develop and validate endoscopic evaluation criteria for nCT. METHODS This study retrospectively investigated patients with T2/3 ESCC who underwent nCT followed by radical esophagectomy across two institutions (test and validation sets). We retrospectively estimated the therapeutic effect by classifying patients according to degree of tumor shrinkage (evaluated with endoscopy) as follows: marked reduction (MR), half reduction (HR), insufficient reduction (IR), and progressive disease (PD). Three endoscopists evaluated patients in the test set. Another three endoscopists evaluated patients in the validation set. We analyzed recurrence-free survival (RFS) 3 years after surgery. RESULTS Of 129 patients in the test set, 44 had MR, 35 had HR, 44 had IR, and six had PD. The 3-year RFS rates were 55% (overall), 79% (MR), 54% (HR), 35% (IR), and 33% (PD). Of 91 patients in the validation set, 22 had MR, 49 had HR, 18 had IR, and two had PD. The 3-year RFS rates were 54% (overall), 77% (MR), 55% (HR), 22% (IR), and 50% (PD). CONCLUSIONS Our endoscopic criteria for nCT predicted prognosis; however, future studies are needed to further investigate our criteria before general application in the clinical setting.
Collapse
Affiliation(s)
- Tomohiro Kadota
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Chiba, Japan
| | - Seiichiro Abe
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan
| | - Yusuke Yoda
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Chiba, Japan
| | | | - Ichiro Oda
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan
| | - Takashi Kojima
- Department of Gastroenterology, Gastrointestinal Oncology Division, National Cancer Center Hospital East, Chiba, Japan
| | - Ken Kato
- Gastrointestinal Medical Oncology Divisions, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroyuki Daiko
- Department of Esophageal Surgery, National Cancer Center Hospital East, Chiba, Japan.,Esophageal Surgery Division, National Cancer Center Hospital, Tokyo, Japan
| | - Tomonori Yano
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Chiba, Japan
| |
Collapse
|
45
|
Lordick F, Herrmann K. Metabolic response assessment and PET-guided treatment of esophageal cancer. Ann Oncol 2020; 31:163-164. [DOI: 10.1016/j.annonc.2019.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022] Open
|
46
|
Barbour AP, Walpole ET, Mai GT, Barnes EH, Watson DI, Ackland SP, Martin JM, Burge M, Finch R, Karapetis CS, Shannon J, Nott LM, Varma S, Marx G, Falk GL, Gebski V, Oostendorp M, Wilson K, Thomas J, Lampe G, Zalcberg JR, Simes J, Smithers BM. Preoperative cisplatin, fluorouracil, and docetaxel with or without radiotherapy after poor early response to cisplatin and fluorouracil for resectable oesophageal adenocarcinoma (AGITG DOCTOR): results from a multicentre, randomised controlled phase II trial. Ann Oncol 2020; 31:236-245. [PMID: 31959340 DOI: 10.1016/j.annonc.2019.10.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Patients with oesophageal/gastro-oesophageal junction adenocarcinoma (EAC) not showing early metabolic response (EMR) to chemotherapy have poorer survival and histological response rates <5%. We investigated whether tailoring neoadjuvant therapy can improve outcomes in these patients. PATIENTS AND METHODS Patients with resectable EAC were enrolled and randomised into two single-arm, multicentre phase II trials. After induction cisplatin and 5-fluorouracil (CF), all were assessed by day 15 positron emission tomography (PET). Patients with an EMR [maximum standardised uptake values (SUVmax) ≥35% reduction from baseline to day 15 PET] received a second CF cycle then oesophagectomy. Non-responders were randomised 1 : 1 to two cycles of CF and docetaxel (DCF, n = 31) or DCF + 45 Gy radiotherapy (DCFRT, n = 35) then oesophagectomy. The primary end point was major histological response (<10% residual tumour) in the oesophagectomy specimen; secondary end points were overall survival (OS), progression-free survival (PFS), and locoregional recurrence (LR). RESULTS Of 124 patients recruited, major histological response was achieved in 3/45 (7%) with EMR, 6/30 (20%) DCF, and 22/35 (63%) DCFRT patients. Grade 3/4 toxicities occurred in 12/45 (27%) EMR (CF), 13/31 (42%) DCF, and 25/35 (71%) DCFRT patients. No treatment-related deaths occurred. LR by 3 years was seen in 5/45 (11%) EMR, 10/31 (32%) DCF, and 4/35 (11%) DCFRT patients. PFS [95% confidence interval (CI)] at 36 months was 47% (31% to 61%) for EMR, 29% (15% to 45%) for DCF, and 46% (29% to 61%) for DCFRT patients. OS (95% CI) at 60 months was 53% (37% to 67%) for EMR, 31% (16% to 48%) for DCF, and 46% (29% to 61%) for DCFRT patients. CONCLUSIONS EMR is associated with favourable OS, PFS, and low LR. For non-responders, the addition of docetaxel augmented histological response rates, but OS, PFS, and LR remained inferior compared with responders. DCFRT improved histological response and PFS/LR outcomes, matching the EMR group. Early PET/CT has the potential to tailor therapy for patients not showing an early response to chemotherapy. TRIAL REGISTRATION ACTRN12609000665235.
Collapse
Affiliation(s)
- A P Barbour
- Divisions of Surgery and Cancer Services, Princess Alexandra Hospital, Brisbane, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| | - E T Walpole
- Divisions of Surgery and Cancer Services, Princess Alexandra Hospital, Brisbane, Australia
| | - G T Mai
- Divisions of Surgery and Cancer Services, Princess Alexandra Hospital, Brisbane, Australia
| | - E H Barnes
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - D I Watson
- Department of Surgery, Flinders Medical Centre, Adelaide, Australia; School of Medicine, Flinders University, Adelaide, Australia
| | - S P Ackland
- School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - J M Martin
- School of Medicine and Public Health, University of Newcastle, Newcastle, Australia
| | - M Burge
- Faculty of Medicine, The University of Queensland, Brisbane, Australia; Cancer Care Services, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - R Finch
- Cancer Care Services, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - C S Karapetis
- Department of Surgery, Flinders Medical Centre, Adelaide, Australia; School of Medicine, Flinders University, Adelaide, Australia
| | - J Shannon
- Nepean Cancer Centre, Nepean Hospital, Sydney, Australia
| | - L M Nott
- Medical Oncology, Royal Hobart Hospital, Hobart, Australia
| | - S Varma
- Cancer Services, Townsville Hospital, Townsville, Australia
| | - G Marx
- Clinical Trials Unit, Sydney Adventist Hospital, Sydney, Australia
| | - G L Falk
- Clinical Trials Unit, Sydney Adventist Hospital, Sydney, Australia
| | - V Gebski
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - M Oostendorp
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - K Wilson
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - J Thomas
- Divisions of Surgery and Cancer Services, Princess Alexandra Hospital, Brisbane, Australia; Clinical Trials, Mater Research Institute, Brisbane, Australia
| | - G Lampe
- Divisions of Surgery and Cancer Services, Princess Alexandra Hospital, Brisbane, Australia
| | - J R Zalcberg
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - J Simes
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - B M Smithers
- Divisions of Surgery and Cancer Services, Princess Alexandra Hospital, Brisbane, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Australia
| |
Collapse
|
47
|
Prätherapeutische Fehlklassifikationen bei Ösophaguskarzinomen und Adenokarzinomen des ösophagogastralen Übergangs. Chirurg 2020; 91:41-50. [DOI: 10.1007/s00104-019-1011-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
48
|
Ku GY, Ilson DH. Cancer of the Esophagus. ABELOFF'S CLINICAL ONCOLOGY 2020:1174-1196.e6. [DOI: 10.1016/b978-0-323-47674-4.00071-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
49
|
PET in Gastrointestinal, Pancreatic, and Liver Cancers. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
50
|
Greally M, Ilson DH. The Multidisciplinary Management of Early Distal Esophageal and Gastroesophageal Junction Cancer. ESOPHAGEAL CANCER 2020:251-273. [DOI: 10.1007/978-3-030-29832-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|