1
|
Xin X, Tang Y, Lu M, Huang J, Shang J, Yang L, Dai L, Yin J, Li J, Leng Q, Tang H, Zhong X. Prognostic value of diffusion-weighted imaging to cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy for patients with gastric cancer and peritoneal metastases. BMC Cancer 2025; 25:616. [PMID: 40188022 PMCID: PMC11972487 DOI: 10.1186/s12885-025-14008-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND To investigate the prognostic value of the apparent diffusion coefficient (ADC) calculated from diffusion-weighted imaging (DWI) to cytoreductive surgery (CRS), with or without hyperthermic intraperitoneal chemotherapy (HIPEC), for gastric cancer (GC) patients with peritoneal metastasis (PM). METHODS Between May 2016 and December 2020, 95 newly diagnosed GC patients with PM who underwent CRS combined with HIPEC (CRS + HIPEC group, n = 61) and CRS alone (CRS group, n = 34) were retrospectively included. All patients underwent abdominal 3.0 T MRI scan, including DWI, and the mean ADC (ADCmean), minimum ADC (ADCmin), and maximum ADC (ADCmax) values of the whole-volume tumor were measured. The prognostic value of the ADC parameters and clinical and histopathological characteristics were investigated by univariate and multivariate Cox analyses. RESULTS The median overall survival (OS) periods of the CRS + HIPEC and CRS groups were 18 and 9 months, respectively ([hazard ratio (HR) = 0.44 [95% CI: 0.27-0.71], P<0.001). The ADCmean and ADCmin values were positively correlated with OS in all patients (Spearman's rho [R] = 0.361 and 0.470), as well as in the CRS + HIPEC (R = 0.369 and 0.417) and CRS (R = 0.192 and 0.409) groups. The multivariate Cox analysis demonstrated that the ADCmean ≤ 1.39 × 10- 3 mm2/s and ADCmin ≤ 0.77 × 10- 3 mm2/s were significantly associated with a negative prognosis in the total population (HR = 1.68 [95% CI: 1.02-2.75] and 2.48 [95% CI: 1.51-4.08], P all < 0.05) and the CRS + HIPEC group (HR = 2.22 [95% CI: 1.19-4.14] and 2.37 [95% CI: 1.26-4.37], P all < 0.05), along with pathologic T and N stages. Only the ADCmin ≤ 0.77 × 10- 3 mm2/s was identified as an independent prognostic factor in the CRS group (HR = 3.49 [95% CI: 1.19-10.20], P = 0.022). CONCLUSIONS The minimum ADC was identified as a strong independent prognostic factor for GC patients with PM who underwent CRS, with or without HIPEC.
Collapse
Affiliation(s)
- Xin Xin
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yongfang Tang
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Man Lu
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jie Huang
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jian Shang
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lidan Yang
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lihuan Dai
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jinxue Yin
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiansheng Li
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qibin Leng
- Department of Oncology Institute, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Hongsheng Tang
- Department of Abdominal Surgery, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Xi Zhong
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Li X, Yuan F, Ni L, Li X. Meta-Analysis of MRI in Predicting Early Response to Radiotherapy and Chemotherapy in Esophageal Cancer. Acad Radiol 2025; 32:798-812. [PMID: 39266443 DOI: 10.1016/j.acra.2024.08.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/20/2024] [Accepted: 08/26/2024] [Indexed: 09/14/2024]
Abstract
RATIONALE AND OBJECTIVES At present, the application of magnetic resonance imaging (MRI) in the prediction of response to neoadjuvant therapy and concurrent chemoradiotherapy for the treatment of esophageal cancer still needs to be further explored, and its early differential value remains controversial, thus we carried out this systematic review with a meta-analysis. In the application, different MRI sequences and corresponding parameters are used for the differential diagnosis of the response to neoadjuvant therapy and concurrent chemoradiotherapy. METHODS All relevant studies evaluated the efficacy and response to MRI in neoadjuvant therapy or concurrent chemoradiotherapy for esophageal cancer on Pubmed, Embase, Cohrane Library, and Web of Science databases published before October 10, 2023 (inclusive) were systematically searched. A revised tool was used to assess the quality of diagnostic accuracy studies (QUADAS-2) to assess the risk of bias in the included original studies. A subgroup analysis of MRI sequences diffusion weighted imaging (DWI), dynamic contrast enhanced (DCE) and their corresponding different parameters, as well as the acquisition timepoints (before and after treatment) for different parameters, was performed during the meta-analysis. The bivariate mixed-effects model was used for meta-analysis. RESULTS 21 studies were finally included, involving 1128 patients with esophageal cancer. The sensitivity, specificity, and area under receiver operating characteristic curve (ROC curve) of DWI sequence for identifying response to concurrent chemoradiotherapy were 0.82 (95% CI: 0.74-0.87), 0.81 (95% CI: 0.72-0.87) and 0.88 (95% CI: 0.56-0.98), respectively. The sensitivity, specificity, and area under ROC curve of DCE sequence for identifying response to concurrent chemoradiotherapy were 0.78 (95% CI: 0.70-0.84), 0.65 (95% CI: 0.59-0.70) and 0.73 (95% CI: 0.50-0.88), respectively. In patients with esophageal cancer, the sensitivity, specificity, and area under the ROC curve of DWI sequences for identifying response to neoadjuvant therapy were 0.80 (95% CI: 0.69 - 0.88), 0.81 (95% CI: 0.69 - 0.89), and 0.88 (95% CI: 0.34 - 0.99), respectively; the sensitivity, specificity, and area under the ROC curve of DCE sequences for identifying response to neoadjuvant therapy were 0.84 (95% CI: 0.76 - 0.90), 0.61 (95% CI: 0.53 - 0.68), and 0.70 (95% CI: 0.27 - 0.94), respectively. CONCLUSIONS Based on the available evidence, MRI had a very good value in the early identification of response to neoadjuvant therapy and concurrent chemoradiotherapy for esophageal cancer, especially DWI. Apparent diffusion coefficient (ADC) value changes before and after treatment could be used as predictors of pathological response. Also, ADC value changes before and after treatment could be used as a tool to guide clinical decision-making.
Collapse
Affiliation(s)
- Xinyu Li
- lmaging Center, The First Affiliated Hospital College of Clinical Medicine of Henan University of Science and Technology, Henan Luoyang 471000, China (X.L., F.Y., L.N., X.L.).
| | - Fang Yuan
- lmaging Center, The First Affiliated Hospital College of Clinical Medicine of Henan University of Science and Technology, Henan Luoyang 471000, China (X.L., F.Y., L.N., X.L.)
| | - Li Ni
- lmaging Center, The First Affiliated Hospital College of Clinical Medicine of Henan University of Science and Technology, Henan Luoyang 471000, China (X.L., F.Y., L.N., X.L.)
| | - Xiaopan Li
- lmaging Center, The First Affiliated Hospital College of Clinical Medicine of Henan University of Science and Technology, Henan Luoyang 471000, China (X.L., F.Y., L.N., X.L.)
| |
Collapse
|
3
|
Li J, Xu S, Wang Y, Ma F, Chen X, Qu J. Spectral CT vs. diffusion-weighted imaging for the quantitative prediction of pathologic response to neoadjuvant chemotherapy in locally advanced gastric cancer. Eur Radiol 2024; 34:6193-6204. [PMID: 38345605 DOI: 10.1007/s00330-024-10642-6] [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: 12/13/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 08/31/2024]
Abstract
OBJECTIVES To compare the performance of spectral CT and diffusion-weighted imaging (DWI) for predicting pathologic response after neoadjuvant chemotherapy (NAC) in locally advanced gastric cancer (LAGC). MATERIALS AND METHODS This was a retrospective analysis drawn from a prospective dataset. Sixty-five patients who underwent baseline concurrent triple-phase enhanced spectral CT and DWI-MRI and standard NAC plus radical gastrectomy were enrolled, and those with poor images were excluded. The tumor regression grade (TRG) was the reference standard, and patients were classified as responders (TRG 0 + 1) or non-responders (TRG 2 + 3). Quantitative iodine concentration (IC), normalized IC (nIC), and apparent diffusion coefficient (ADC) were measured by placing a freehand region of interest manually on the maximal two-dimensional plane. Their differences between responders and non-responders were compared. The performances of significant parameters were evaluated by the receiver operating characteristic analysis. The correlations between parameters and TRG status were explored through Spearman correlation coefficient test. Kaplan-Meier survival analysis was adopted to analyze their relationship with patient survival. RESULTS nICDP and ADC were associated with the TRG and yielded comparable performances for predicting TRG categories, with area under the curve (AUC) of 0.674 and 0.673, respectively. Their combination achieved a significantly increased AUC of 0.770 (p ; 0.05) and was associated with patient disease-free survival, with hazard ratio of 2.508 (1.043-6.029). CONCLUSION Spectral CT and DWI were equally useful imaging techniques for predicting pathologic response to NAC in LAGC. The combination of nICDP and ADC gained significant incremental benefits and was related to patient disease-free survival. CLINICAL RELEVANCE STATEMENT Spectral CT and DWI-based quantitative measurements are effective markers for predicting the pathologic regression outcomes of locally advanced gastric cancer patients after neoadjuvant chemotherapy. KEY POINTS • The pathologic tumor regression grade, the standard criteria for treatment response after neoadjuvant chemotherapy in gastric cancer patients, is difficult to predict early. • The quantitative parameters of normalized iodine concentration at delay phase and apparent diffusion coefficients were correlated with pathologic response; their combination demonstrated incremental benefits and was associated with patient disease-free survival. • Spectral CT and DWI are equally useful imaging modalities for predicting tumor regression grade after neoadjuvant chemotherapy in patients with locally advanced gastric cancer.
Collapse
Affiliation(s)
- Jing Li
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China.
| | - Shuning Xu
- Department of Gastrointestinal Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Yi Wang
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Fei Ma
- Department of Gastrointestinal Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Xuejun Chen
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Jinrong Qu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China.
| |
Collapse
|
4
|
Song T, Lu S, Qu J, Zhang H, Wang Z, Jia Z, Li H, Zhao Y, Qin J, Feng W, Wang S, Yan X. Intravoxel incoherent motion diffusion-weighted imaging in evaluating preoperative staging of esophageal squamous cell carcinoma : Evaluation of preoperative stage of primary tumour and prediction of lymph node metastases from esophageal cancer using IVIM: a prospective study. Cancer Imaging 2024; 24:116. [PMID: 39210470 PMCID: PMC11363402 DOI: 10.1186/s40644-024-00765-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The aim of this research is to prospectively investigate the diagnostic performance of intravoxel incoherent motion (IVIM) using the integrated slice-specific dynamic shimming (iShim) technique in staging primary esophageal squamous cell carcinoma (ESCC) and predicting presence of lymph node metastases from ESCC. METHODS Sixty-three patients with ESCC were prospectively enrolled from April 2016 to April 2019. MR and IVIM using iShim technique (b = 0, 25, 50, 75, 100, 200, 400, 600, 800 s/mm2) were performed on 3.0T MRI system before operation. Primary tumour apparent diffusion coefficient (ADC) and IVIM parameters, including true diffusion coefficient (D), pseudodiffusion coefficient (D*), pseudodiffusion fraction (f) were measured by two independent radiologists. The differences in D, D*, f and ADC values of different T and N stages were assessed. Intraclass correlation coefficients (ICCs) were calculated to evaluate the interobserver agreement between two readers. The diagnostic performances of D, D*, f and ADC values in primary tumour staging and prediction of lymph node metastasis of ESCC were determined using receiver operating characteristic (ROC) curve analysis. RESULTS The inter-observer consensus was excellent for IVIM parameters and ADC (D: ICC = 0.922; D*: ICC = 0.892; f: ICC = 0.948; ADC: ICC = 0.958). The ADC, D, D* and f values of group T1 + T2 were significantly higher than those of group T3 + T4a [ADC: (2.55 ± 0.43) ×10- 3 mm2/s vs. (2.27 ± 0.40) ×10- 3 mm2/s, t = 2.670, P = 0.010; D: (1.82 ± 0.39) ×10- 3 mm2/s vs. (1.53 ± 0.33) ×10- 3 mm2/s, t = 3.189, P = 0.002; D*: 46.45 (30.30,55.53) ×10- 3 mm2/s vs. 32.30 (18.60,40.95) ×10- 3 mm2/s, z=-2.408, P = 0.016; f: 0.45 ± 0.12 vs. 0.37 ± 0.12, t = 2.538, P = 0.014]. The ADC, D and f values of the lymph nodes-positive (N+) group were significantly lower than those of lymph nodes-negative (N0) group [ADC: (2.10 ± 0.33) ×10- 3 mm2/s vs. (2.55 ± 0.40) ×10- 3 mm2/s, t=-4.564, P < 0.001; D: (1.44 ± 0.30) ×10- 3 mm2/s vs. (1.78 ± 0.37) ×10- 3 mm2/s, t=-3.726, P < 0.001; f: 0.32 ± 0.10 vs. 0.45 ± 0.11, t=-4.524, P < 0.001]. The combination of D, D* and f yielded the highest area under the curve (AUC) (0.814) in distinguishing group T1 + T2 from group T3 + T4a. D combined with f provided the highest diagnostic performance (AUC = 0.849) in identifying group N + and group N0 of ESCC. CONCLUSIONS IVIM may be used as an effective functional imaging technique to evaluate preoperative stage of primary tumour and predict presence of lymph node metastases from ESCC.
Collapse
Affiliation(s)
- Tao Song
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Shuang Lu
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Jinrong Qu
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
- Henan Province, 127 Dongming road, Jinshui District, Zhengzhou city, 450008, China.
| | - Hongkai Zhang
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Zhaoqi Wang
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Zhengyan Jia
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Hailiang Li
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Yan Zhao
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Jianjun Qin
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Wen Feng
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Shaoyu Wang
- MR Scientific Marketing, Siemens Healthineers, XI'an, 710065, China
| | - Xu Yan
- MR Scientific Marketing, Siemens Healthineers, Shanghai, 201318, China
| |
Collapse
|
5
|
Fiorino C, Palumbo D, Mori M, Palazzo G, Pellegrini AE, Albarello L, Belardo A, Canevari C, Cossu A, Damascelli A, Elmore U, Mazza E, Pavarini M, Passoni P, Puccetti F, Slim N, Steidler S, Del Vecchio A, Di Muzio NG, Chiti A, Rosati R, De Cobelli F. Early regression index (ERI) on MR images as response predictor in esophageal cancer treated with neoadjuvant chemo-radiotherapy: Interim analysis of the prospective ESCAPE trial. Radiother Oncol 2024; 194:110160. [PMID: 38369025 DOI: 10.1016/j.radonc.2024.110160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 02/20/2024]
Abstract
PURPOSE The early regression index (ERI) predicts treatment response in rectal cancer patients. Aim of current study was to prospectively assess tumor response to neoadjuvant chemo-radiotherapy (nCRT) of locally advanced esophageal cancer using ERI, based on MRI. MATERIAL AND METHODS From January 2020 to May 2023, 30 patients with esophageal cancer were enrolled in a prospective study (ESCAPE). PET-MRI was performed: i) before nCRT (tpre); ii) at mid-radiotherapy, tmid; iii) after nCRT, 2-6 weeks before surgery (tpost); nCRT delivered 41.4 Gy/23fr with concurrent carboplatin and paclitaxel. For patients that skipped surgery, complete clinical response (cCR) was assessed if patients showed no local relapse after 18 months; patients with pathological complete response (pCR) or with cCR were considered as complete responders (pCR + cCR). GTV volumes were delineated by two observers (Vpre, Vmid, Vpost) on T2w MRI: ERI and other volume regression parameters at tmid and tpost were tested as predictors of pCR + cCR. RESULTS Complete data of 25 patients were available at the time of the analysis: 3/25 with complete response at imaging refused surgery and 2/3 were cCR; in total, 10/25 patients showed pCR + cCR (pCR = 8/22). Both ERImid and ERIpost classified pCR + cCR patients, with ERImid showing better performance (AUC:0.78, p = 0.014): A two-variable logistic model combining ERImid and Vpre improved performances (AUC:0.93, p < 0.0001). Inter-observer variability in contouring GTV did not affect the results. CONCLUSIONS Despite the limited numbers, interim analysis of ESCAPE study suggests ERI as a potential predictor of complete response after nCRT for esophageal cancer. Further validation on larger populations is warranted.
Collapse
Affiliation(s)
- C Fiorino
- Medical Physics, IRCCS San Raffaele Hospital, Milano, Italy.
| | - D Palumbo
- Radiology, IRCCS San Raffaele Hospital, Milano, Italy
| | - M Mori
- Medical Physics, IRCCS San Raffaele Hospital, Milano, Italy
| | - G Palazzo
- Medical Physics, IRCCS San Raffaele Hospital, Milano, Italy
| | | | - L Albarello
- Pathology, IRCCS San Raffaele Hospital, Milano, Italy
| | - A Belardo
- Medical Physics, IRCCS San Raffaele Hospital, Milano, Italy
| | - C Canevari
- Nuclear Medicine, IRCCS San Raffaele Hospital, Milano, Italy
| | - A Cossu
- Gastric Surgery, IRCCS San Raffaele Hospital, Milano, Italy
| | - A Damascelli
- Radiology, IRCCS San Raffaele Hospital, Milano, Italy
| | - U Elmore
- Gastric Surgery, IRCCS San Raffaele Hospital, Milano, Italy
| | - E Mazza
- Oncology, IRCCS San Raffaele Hospital, Milano, Italy
| | - M Pavarini
- Medical Physics, IRCCS San Raffaele Hospital, Milano, Italy
| | - P Passoni
- Radiotherapy, IRCCS San Raffaele Hospital, Milano, Italy
| | - F Puccetti
- Gastric Surgery, IRCCS San Raffaele Hospital, Milano, Italy
| | - N Slim
- Radiotherapy, IRCCS San Raffaele Hospital, Milano, Italy
| | - S Steidler
- Radiology, IRCCS San Raffaele Hospital, Milano, Italy
| | - A Del Vecchio
- Medical Physics, IRCCS San Raffaele Hospital, Milano, Italy
| | - N G Di Muzio
- Radiotherapy, IRCCS San Raffaele Hospital, Milano, Italy; Vita-Salute University, Milano, Italy
| | - A Chiti
- Nuclear Medicine, IRCCS San Raffaele Hospital, Milano, Italy; Vita-Salute University, Milano, Italy
| | - R Rosati
- Gastric Surgery, IRCCS San Raffaele Hospital, Milano, Italy; Vita-Salute University, Milano, Italy
| | - F De Cobelli
- Radiology, IRCCS San Raffaele Hospital, Milano, Italy; Vita-Salute University, Milano, Italy
| |
Collapse
|
6
|
Wei YY, Cai JY, Wang LL, Yang J, Li YL, Li XT, Zhang XT, Shi YJ, Tang L. Dynamic change in the peritoneal cancer index based on CT after chemotherapy in the overall survival prediction of gastric cancer patients with peritoneal metastasis. J Cancer Res Clin Oncol 2024; 150:222. [PMID: 38687350 PMCID: PMC11061045 DOI: 10.1007/s00432-024-05707-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 03/15/2024] [Indexed: 05/02/2024]
Abstract
PURPOSE The purpose of this research was to investigate the efficacy of the CT-based peritoneal cancer index (PCI) to predict the overall survival of patients with peritoneal metastasis in gastric cancer (GCPM) after two cycles of chemotherapy. METHODS This retrospective study registered 112 individuals with peritoneal metastasis in gastric cancer in our hospital. Abdominal and pelvic enhanced CT before and after chemotherapy was independently analyzed by two radiologists. The PCI of peritoneal metastasis in gastric cancer was evaluated according to the Sugarbaker classification, considering the size and distribution of the lesions using CT. Then we evaluated the prognostic performance of PCI based on CT, clinical characteristics, and imaging findings for survival analysis using multivariate Cox proportional hazard regression. RESULTS The PCI change ratio based on CT after treatment (ΔPCI), therapy lines, and change in grade of ascites were independent factors that were associated with overall survival (OS). The area under the curve (AUC) value of ΔPCI for predicting OS with 0.773 was higher than that of RECIST 1.1 with 0.661 (P < 0.05). Patients with ΔPCI less than -15% had significantly longer OS. CONCLUSION CT analysis after chemotherapy could predict OS in patients with GCPM. The CT-PCI change ratio could contribute to the determination of an appropriate strategy for gastric cancer patients with peritoneal metastasis.
Collapse
Affiliation(s)
- Yi-Yuan Wei
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital and Institute, No. 52 Fucheng Road, Hai Dian District, Beijing, 100142, China
| | - Jie-Yuan Cai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, No. 52 Fucheng Road, Hai Dian District, Beijing, 100142, China
| | - Lin-Lin Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital and Institute, No. 52 Fucheng Road, Hai Dian District, Beijing, 100142, China
| | - Jie Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital and Institute, No. 52 Fucheng Road, Hai Dian District, Beijing, 100142, China
| | - Yan-Ling Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital and Institute, No. 52 Fucheng Road, Hai Dian District, Beijing, 100142, China
| | - Xiao-Ting Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital and Institute, No. 52 Fucheng Road, Hai Dian District, Beijing, 100142, China
| | - Xiao-Tian Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, No. 52 Fucheng Road, Hai Dian District, Beijing, 100142, China.
| | - Yan-Jie Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital and Institute, No. 52 Fucheng Road, Hai Dian District, Beijing, 100142, China
| | - Lei Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital and Institute, No. 52 Fucheng Road, Hai Dian District, Beijing, 100142, China
| |
Collapse
|
7
|
Deng J, Zhang W, Xu M, Liu X, Ren T, Li S, Sun Q, Xue C, Zhou J. Value of spectral CT parameters in predicting the efficacy of neoadjuvant chemotherapy for gastric cancer. Clin Radiol 2024; 79:51-59. [PMID: 37914603 DOI: 10.1016/j.crad.2023.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/26/2023] [Accepted: 08/30/2023] [Indexed: 11/03/2023]
Abstract
AIM To investigate the value of pre-chemotherapy spectral computed tomography (CT) parameters in predicting neoadjuvant chemotherapy (NAC) response in gastric cancer (GC). MATERIALS AND METHODS Sixty patients with GC who received NAC and underwent spectral CT examination before chemotherapy were enrolled retrospectively and divided into a responsive group and a non-responsive group according to the postoperative pathological tumour regression grade. Clinical characteristics were collected. The iodine concentration (IC), water concentration (WC), and effective atomic number (Eff-Z) of the portal venous phases were measured before chemotherapy, and IC was normalised to that of the aorta to provide the normalised IC (NIC). An independent samples t-test, Mann-Whitney U-test, or chi-square test was used to analyse the differences between the two groups, and the receiver operating curve (ROC) was used to evaluate the predictive performance of different variables. RESULTS The neutrophil-to-lymphocyte ratio (NLR) was lower in the responsive group than in the non-responsive group (p<0.05). IC, NIC, and Eff-Z values were significantly higher in the responsive group than in the non-responsive group (p<0.01). The areas under the ROC curves for the NLR, IC, NIC, and Eff-Z were 0.694, 0.688, 0.799, and 0.690, respectively. The combination of NIC, Eff-Z, and NLR values showed good diagnostic performance in predicting response to NAC in GC, with an area under the ROC curve of 0.857, 76.92% sensitivity, 80% accuracy, and 85.71% specificity. CONCLUSION Spectral CT parameters may serve as non-invasive tools for predicting the response to NAC in patients with GC.
Collapse
Affiliation(s)
- J Deng
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - W Zhang
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - M Xu
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - X Liu
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - T Ren
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - S Li
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - Q Sun
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - C Xue
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China
| | - J Zhou
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China; Second Clinical School, Lanzhou University, Lanzhou, 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, 730030, China.
| |
Collapse
|
8
|
Deng J, Zhang W, Xu M, Zhou J. Imaging advances in efficacy assessment of gastric cancer neoadjuvant chemotherapy. Abdom Radiol (NY) 2023; 48:3661-3676. [PMID: 37787962 DOI: 10.1007/s00261-023-04046-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/31/2023] [Accepted: 09/03/2023] [Indexed: 10/04/2023]
Abstract
Effective neoadjuvant chemotherapy (NAC) can improve the survival of patients with locally progressive gastric cancer, but chemotherapeutics do not always exhibit good efficacy in all patients. Therefore, accurate preoperative evaluation of the effect of neoadjuvant therapy and the appropriate selection of surgery time to minimize toxicity and complications while prolonging patient survival are key issues that need to be addressed. This paper reviews the role of three imaging methods, morphological, functional, radiomics, and artificial intelligence (AI)-based imaging, in evaluating NAC pathological reactions for gastric cancer. In addition, the advantages and disadvantages of each method and the future application prospects are discussed.
Collapse
Affiliation(s)
- Juan Deng
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China
- Second Clinical School, Lanzhou University, Lanzhou, 730030, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China
- Gansu International Scientifific and Technological Cooperation Base of Medical Imaging Artifificial Intelligence, Lanzhou, 730030, China
| | - Wenjuan Zhang
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China
- Second Clinical School, Lanzhou University, Lanzhou, 730030, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China
- Gansu International Scientifific and Technological Cooperation Base of Medical Imaging Artifificial Intelligence, Lanzhou, 730030, China
| | - Min Xu
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China
- Second Clinical School, Lanzhou University, Lanzhou, 730030, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China
- Gansu International Scientifific and Technological Cooperation Base of Medical Imaging Artifificial Intelligence, Lanzhou, 730030, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, China.
- Second Clinical School, Lanzhou University, Lanzhou, 730030, China.
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, 730030, China.
- Gansu International Scientifific and Technological Cooperation Base of Medical Imaging Artifificial Intelligence, Lanzhou, 730030, China.
| |
Collapse
|
9
|
Shi J, Li J, Li Z, Li Y, Xu L, Zhang Y. Prediction of pathological response grading for esophageal squamous carcinoma after neoadjuvant chemoradiotherapy based on MRI imaging using PDX. Front Oncol 2023; 13:1160815. [PMID: 37377911 PMCID: PMC10292012 DOI: 10.3389/fonc.2023.1160815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Introduction To confirm the efficacy of magnetic resonance-diffusion weighted imaging (MR-DWI) in esophageal squamous cell carcinoma (ESCC) early pathological response prediction and assessment to neoadjuvant chemoradiotherapy (nCRT) using patient-derived xenografts (PDXs). Methods PDX-bearing mice were randomly divided into two groups: the experimental group receiving cisplatin combined with radiotherapy, whereas the control group receiving normal saline. MRI scans were performed in treatment groups in the before, middle, and end of treatment. The correlations between tumor volumes, ADC values and tumor pathological response at different time nodes were explored. Then, expression of proliferation marker and apoptotic marker were detected using immunohistochemistry, and apoptosis rate was detected by TUNEL assay to further verify the results observed in the PDX models. Results The ADC values of the experimental group were significantly higher than the control group in the both middle and end stage of treatment (all P< 0.001), however, significant difference was only observed in tumor volume at the end stage of treatment (P< 0.001). Furthermore, the △ADCmid-pre in our study may able to identify tumors with or without pCR to nCRT at an early stage, due to these changes were prior to the changes of tumor volume after treatment. Finally, TUNEL results also showed that the apoptosis rate of the experiment groups increased the most in the middle stage of treatment, especially the groups with pCR, but the highest apoptosis rate occurred in the end of the treatment. Further, the two PDX models with pCR exhibited the highest levels of apoptotic marker (Bax), and lowest levels of proliferation marker (PCNA and Ki-67) in the both middle and end stage of the treatment. Conclusions ADC values could be used to determine the tumor's response to nCRT, especially in the middle stages of treatment and before the tumor tissue morphology changes, and further, the ADC values were consistent with the potential biomarkers reflecting histopathological changes. Therefore, we suggest that radiation oncologists could refer to the ADC values in the middle stages of treatment when predicting the tumor histopathological response to n CRT in patients with ESCC.
Collapse
Affiliation(s)
- Jingzhen Shi
- Department of Oncology, The Second Hospital of Tianjin Medical University, Tianjin, China
- School of Medicine, Shandong University, Jinan, China
| | - Jianbin Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhenxiang Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yankang Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Liang Xu
- Department of Medical Imaging, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yingjie Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
10
|
John NO, Irodi A, Thomas HMT, Abraham V, Sasidharan BK, John S, Pavamani SP. Utility of Mid-treatment DWI in Selecting Pathological Responders to Neoadjuvant Chemoradiotherapy in Locally Advanced Esophageal Cancer. J Gastrointest Cancer 2023; 54:447-455. [PMID: 35347663 DOI: 10.1007/s12029-022-00818-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE Pathological complete response correlates with better clinical outcomes in locally advanced esophageal cancer (LA-EC). However, there is lack of prognostic markers to identify patients in the current setting of neoadjuvant chemoradiotherapy (NACRT) followed by surgery. This study evaluates the utility of mid-treatment diffusion-weighted imaging (DWI) in identifying pathological responders of NACRT. METHODS Twenty-four patients with LA-EC on NACRT were prospectively recruited and underwent three MRI (baseline, mid-treatment, end-of-RT) scans. DWI-derived apparent diffusion coefficient (ADC) mean and minimum were used as a surrogate to evaluate the treatment response, and its correlation to pathological response was assessed. RESULTS Mid-treatment ADC mean was significantly higher among patients with pathological response compared to non-responders (p = 0.011). ADC difference (ΔADC) between baseline and mid-treatment correlated with tumor response (p = 0.007). ADC at other time points did not correlate to pathological response. CONCLUSION In this study, mid-treatment ADC values show potential to be a surrogate for tumor response in NACRT. However, larger trials are required to establish DW-MRI as a definite biomarker for tumor response.
Collapse
Affiliation(s)
- Neenu Oliver John
- Department of Radiation Oncology, Ida B. Scudder Cancer Centre, Christian Medical College, Tamil Nadu, Vellore, 632004, India
| | - Aparna Irodi
- Division of Clinical Radiology, Department of Radiodiagnosis, Christian Medical College, Tamil Nadu, Vellore, 632004, India
| | - Hannah Mary T Thomas
- Department of Radiation Oncology, Ida B. Scudder Cancer Centre, Christian Medical College, Tamil Nadu, Vellore, 632004, India
| | - Vijay Abraham
- Department of Surgery, Christian Medical College, Tamil Nadu, Vellore, 632004, India
- Department of Upper GI Surgery, The Queen Elizabeth Hospital, Woodville South, Adelaide, 5011, Australia
| | - Balu Krishna Sasidharan
- Department of Radiation Oncology, Ida B. Scudder Cancer Centre, Christian Medical College, Tamil Nadu, Vellore, 632004, India
| | - Subhashini John
- Department of Radiation Oncology, Ida B. Scudder Cancer Centre, Christian Medical College, Tamil Nadu, Vellore, 632004, India
| | - Simon P Pavamani
- Department of Radiation Oncology, Ida B. Scudder Cancer Centre, Christian Medical College, Tamil Nadu, Vellore, 632004, India.
| |
Collapse
|
11
|
Dynamic contrast-enhanced and diffusion-weighted MR imaging in early prediction of pathologic response to neoadjuvant chemotherapy in locally advanced gastric cancer. ABDOMINAL RADIOLOGY (NEW YORK) 2022; 47:3394-3405. [PMID: 35916943 DOI: 10.1007/s00261-022-03623-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 01/18/2023]
Abstract
PURPOSE To investigate the efficacy of diffusion-weighted imaging (DWI) and dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) for the early prediction of the pathologic response to neoadjuvant chemotherapy (NAC) in patients with locally advanced gastric cancer (LAGC). METHODS Fifty patients with LAGC who were treated with NAC followed by radical gastrectomy were enrolled. Uncontrasted and DCE-MRI were performed within 1 week before NAC. According to tumor regression grading (TRG), patients were labeled as responders (TRG = 0 + 1) and non-responders (TRG = 2 + 3). Apparent diffusion coefficients (ADC) and DCE-MRI kinetics (Ktrans, Ve, and Kep) were compared between the two groups. Logistic regression analysis was performed to screen independent factors to predict the NAC efficacy. The relationship between MRI parameters and TRG was studied by Spearman's correlation analysis. Receiver-operating characteristic curve analyses were applied to evaluate the efficacy. RESULTS ADC, Ktrans, and Kep values were higher in responders than in non-responders (p < 0.05) and correlated with TRG (p < 0.05). The ADC and Kep values were independent markers for predicting TRG. The area under the curve, sensitivities, specificities of ADC, Ktrans, Kep, and ADC + Kep were 0.813, 0.699, 0.709, 0.886;73.64%, 65.54%, 63.21%, 70.37%; 86.47%, 54.97%, 79.47%, 95.65%; respectively. ADC + Kep demonstrated a higher efficacy than Ktrans and Kep (p = 0.012, 0.011), but without improvement compared with ADC (p > 0.05). CONCLUSION Both DWI and DCE-MRI can effectively predict the pathologic response to NAC in LAGC. A combination of ADC and Kep increased the efficacy, and ADC is the most valuable imaging parameter.
Collapse
|
12
|
Withey SJ, Goh V, Foley KG. State-of-the-art imaging in oesophago-gastric cancer. Br J Radiol 2022; 95:20220410. [PMID: 35671095 PMCID: PMC10996959 DOI: 10.1259/bjr.20220410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 11/05/2022] Open
Abstract
Radiological investigations are essential in the management of oesophageal and gastro-oesophageal junction cancers. The current multimodal combination of CT, 18F-fluorodeoxyglucose positron emission tomography combined with CT (PET/CT) and endoscopic ultrasound (EUS) has limitations, which hinders the prognostic and predictive information that can be used to guide optimum treatment decisions. Therefore, the development of improved imaging techniques is vital to improve patient management. This review describes the current evidence for state-of-the-art imaging techniques in oesophago-gastric cancer including high resolution MRI, diffusion-weighted MRI, dynamic contrast-enhanced MRI, whole-body MRI, perfusion CT, novel PET tracers, and integrated PET/MRI. These novel imaging techniques may help clinicians improve the diagnosis, staging, treatment planning, and response assessment of oesophago-gastric cancer.
Collapse
Affiliation(s)
- Samuel J Withey
- Department of Radiology, The Royal Marsden NHS Foundation
Trust, London,
UK
| | - Vicky Goh
- Cancer Imaging, School of Biomedical Engineering & Imaging
Sciences, King’s College London,
London, UK
- Department of Radiology, Guy’s and St Thomas’ NHS
Foundation Trust, London,
UK
| | - Kieran G Foley
- Division of Cancer & Genetics, School of Medicine, Cardiff
University, Wales,
UK
- Department of Radiology, Velindre Cancer Centre,
Cardiff, UK
| |
Collapse
|
13
|
Enhanced CT-based radiomics predicts pathological complete response after neoadjuvant chemotherapy for advanced adenocarcinoma of the esophagogastric junction: a two-center study. Insights Imaging 2022; 13:134. [PMID: 35976518 PMCID: PMC9385906 DOI: 10.1186/s13244-022-01273-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/20/2022] [Indexed: 01/19/2023] Open
Abstract
Purpose This study aimed to develop and validate CT-based models to predict pathological complete response (pCR) after neoadjuvant chemotherapy (NAC) for advanced adenocarcinoma of the esophagogastric junction (AEG). Methods Pre-NAC clinical and imaging data of AEG patients who underwent surgical resection after preoperative-NAC at two centers were retrospectively collected from November 2014 to September 2020. The dataset included training (n = 60) and external validation groups (n = 32). Three models, including CT-based radiomics, clinical and radiomics–clinical combined models, were established to differentiate pCR (tumor regression grade (TRG) = grade 0) and nonpCR (TRG = grade 1–3) patients. For the radiomics model, tumor-region-based radiomics features in the arterial and venous phases were extracted and selected. The naïve Bayes classifier was used to establish arterial- and venous-phase radiomics models. The selected candidate clinical factors were used to establish a clinical model, which was further incorporated into the radiomics–clinical combined model. ROC analysis, calibration and decision curves were used to assess the model performance. Results For the radiomics model, the AUC values obtained using the venous data were higher than those obtained using the arterial data (training: 0.751 vs. 0.736; validation: 0.768 vs. 0.750). Borrmann typing, tumor thickness and degree of differentiation were utilized to establish the clinical model (AUC-training: 0.753; AUC-validation: 0.848). The combination of arterial- and venous-phase radiomics and clinical factors further improved the discriminatory performance of the model (AUC-training: 0.838; AUC-validation: 0.902). The decision curve reflects the higher net benefit of the combined model. Conclusion The combination of CT imaging and clinical factors pre-NAC for advanced AEG could help stratify potential responsiveness to NAC. Supplementary Information The online version contains supplementary material available at 10.1186/s13244-022-01273-w.
Collapse
|
14
|
Tham E, Sestito M, Markovich B, Garland-Kledzik M. Current and future imaging modalities in gastric cancer. J Surg Oncol 2022; 125:1123-1134. [PMID: 35481912 DOI: 10.1002/jso.26875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 12/24/2022]
Abstract
Gastric adenocarcinoma treatment can include endoscopic mucosal resection, surgery, chemotherapy, radiation, and palliative measures depending on staging. Both invasive and noninvasive staging techniques have been used to dictate the best treatment pathway. Here, we review the current imaging modalities used in gastric cancer as well as novel techniques to accurately stage and screen these patients.
Collapse
Affiliation(s)
- Elwin Tham
- Department of Surgical Oncology, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Michael Sestito
- Department of Surgical Oncology, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Brian Markovich
- Department of Diagnostic Radiology, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Mary Garland-Kledzik
- Department of Surgical Oncology, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| |
Collapse
|
15
|
An D, Cao Q, Su N, Li W, Li Z, Liu Y, Zhang Y, Li B. Response Prediction to Concurrent Chemoradiotherapy in Esophageal Squamous Cell Carcinoma Using Delta-Radiomics Based on Sequential Whole-Tumor ADC Map. Front Oncol 2022; 12:787489. [PMID: 35392222 PMCID: PMC8982070 DOI: 10.3389/fonc.2022.787489] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/21/2022] [Indexed: 12/24/2022] Open
Abstract
Purpose The purpose of this study was to investigate the association between the radiomics features (RFs) extracted from a whole-tumor ADC map during the early treatment course and response to concurrent chemoradiotherapy (cCRT) in patients with esophageal squamous cell carcinoma (ESCC). Methods Patients with ESCC who received concurrent chemoradiotherapy were enrolled in two hospitals. Whole-tumor ADC values and RFs were extracted from sequential ADC maps before treatment, after the 5th radiation, and after the 10th radiation, and the changes of ADC values and RFs were calculated as the relative difference between different time points. RFs were selected and further imported to a support vector machine classifier for building a radiomics signature. Radiomics signatures were obtained from both RFs extracted from pretreatment images and three sets of delta-RFs. Prediction models for different responders based on clinical characteristics and radiomics signatures were built up with logistic regression. Results Patients (n=76) from hospital 1 were randomly assigned to training (n=53) and internal testing set (n=23) in a ratio of 7 to 3. In addition, to further test the performance of the model, data from another institute (n=17) were assigned to the external testing set. Neither ADC values nor delta-ADC values were correlated with treatment response in the three sets. It showed a predictive effect to treatment response that the AUC values of the radiomics signature built from delta-RFs over the first 2 weeks were 0.824, 0.744, and 0.742 in the training, the internal testing, and the external testing set, respectively. Compared with the evaluated response, the performance of response prediction in the internal testing set was acceptable (p = 0.048). Conclusions The ADC map-based delta-RFs during the early course of treatment were effective to predict the response to cCRT in patients with ESCC.
Collapse
Affiliation(s)
- Dianzheng An
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong University, Jinan, China
| | - Qiang Cao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Na Su
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Wanhu Li
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhe Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yanxiao Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yuxing Zhang
- Department of Imaging, Anyang Tumor Hospital, The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang, China
| | - Baosheng Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
16
|
Saiz Martínez R, Dromain C, Vietti Violi N. Imaging of Gastric Carcinomatosis. J Clin Med 2021; 10:5294. [PMID: 34830575 PMCID: PMC8624519 DOI: 10.3390/jcm10225294] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/06/2021] [Accepted: 11/11/2021] [Indexed: 01/17/2023] Open
Abstract
Diagnosing the absence or presence of peritoneal carcinomatosis in patients with gastric cancer, including its extent and distribution, is an essential step in patients' therapeutic management. Such diagnosis still remains a radiological challenge. In this article, we review the strengths and weaknesses of the different imaging techniques for the diagnosis of peritoneal carcinomatosis of gastric origin as well as the techniques' imaging features. We also discuss the assessment of response to treatment and present recommendations for the follow-up of patients with complete surgical resection according to the presence of risk factors of recurrence, as well as discussing future directions for imaging improvement.
Collapse
Affiliation(s)
| | - Clarisse Dromain
- Department of Radiology, Lausanne University Hospital and University of Lausanne, 1015 Lausanne, Switzerland;
| | - Naik Vietti Violi
- Department of Radiology, Lausanne University Hospital and University of Lausanne, 1015 Lausanne, Switzerland;
| |
Collapse
|
17
|
An updated review of the TNM classification system for cancer of the oesophagus and its complications. RADIOLOGIA 2021; 63:445-455. [PMID: 34625200 DOI: 10.1016/j.rxeng.2020.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/07/2020] [Indexed: 11/21/2022]
Abstract
Cancer of the esophagus is an aggressive cancer with high mortality. Because of the esophagus's lack of serosa and its peculiar lymphatic drainage, esophageal cancer is diagnosed in advanced stages. The eighth edition of the TNM (2017) aims to standardize care for esophageal cancer throughout the world; it includes not only patients treated with esophagectomy alone, but also those receiving neoadjuvant chemotherapy and/or radiotherapy. One new development in the eighth edition is that it establishes separate classifications for different time periods, with pathologic stage groups for prior to treatment (cTNM), after esophagectomy (pTNM), and after neoadjuvant therapy (ypTNM). The combined use of endoscopic ultrasound, CT, PET-CT, and MRI provides the greatest accuracy in determining the clinical stage, and these techniques are essential for planning treatment and for evaluating the response to neoadjuvant treatment. Esophagectomy continues to be the main treatment; it is also the elective gastrointestinal surgery that has the highest mortality, and it carries the risk of multiple complications, including anastomotic leaks, pulmonary complications, technical complications, and functional complications.
Collapse
|
18
|
Wang Z, Xiong B, Kang N, Pan X, Wang C, Su L, Xing Z, Hong J. The Value of MR-DWI and T1 Mapping in Indicating Radiation-Induced Soft Tissue Injury. Front Oncol 2021; 11:651637. [PMID: 34123802 PMCID: PMC8190401 DOI: 10.3389/fonc.2021.651637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/29/2021] [Indexed: 12/23/2022] Open
Abstract
Objective To explore the value of MR-DWI and T1 mapping in predicting radiation-induced soft tissue fibrosis and its correlation with radiation inflammation. Methods ① a total of 30 C57BL/6 mice were randomly divided into a control group (Nor group), irradiation group (IR group) and irradiation plus glycyrrhetinic acid group (GA group). The IR group and GA group were treated with 6MV X-rays to irradiate the right hind limbs of mice for 30 Gy in a single shot. MRI examinations were performed before and on the 7th day after irradiation to measure the apparent diffusion coefficient (ADC) value and the longitudinal relaxation time (T1) value of the hind limb muscles of the mice. On the 90th day after irradiation, the hind limb contracture was measured, and the right hind limb muscle was taken for HE staining, masson staining, immunohistochemical staining and Western blot analysis to detect the expression of a-SMA and Fibronectin. ② The other 30 mice were grouped randomly as above. On the 7th day after irradiation, the right hind limbs of the mice were examined by MRI to measure the ADC value and T1 value of the thigh muscles, and then the right hind thigh muscles were immediately sacrificed to detect IL-1β, IL-6, TNF-a and TGF-β1 expression with ELISA. Results On the 7th day after irradiation, the ADC values of right hind thigh muscles of mice in Nor group, IR group and GA group were (1.35 ± 0.11)*10-3mm2/s, (1.48 ± 0.07) *10-3mm2/s and (1.36 ± 0.13)*10-3mm2/s, respectively, by which the differences between the IR group and Nor group (P=0.008) and that between IR group and GA group (P=0.013) were statistically significant; T1 values were (1369.7 ± 62.7)ms, (1483.7 ± 127.7)ms and (1304.1 ± 82.3)ms, respectively, with which the differences in the T1 value between the IR group and Nor group (P=0.012) and between IR group and GA group (P<0.001) were also statistically significant. On the 90th day after irradiation, the contracture lengths of the right hind limbs of the three groups of mice were (0.00 ± 0.07)cm, (2.08 ± 0.32)cm, and (1.49 ± 0.70) cm, respectively. There were statistically significant differences in the IR group compared with the Nor group (P<0.001) and the GA group (P=0.030). The ADC value (r=0.379, P=0.039) and T1 value (r=0.377, P=0.040) of the mice's hindlimbs on Day 7 after irradiation were correlated with the degree of contracture on Day 90 after irradiation; the ADC value (r=0.496, P=0.036) and T1 value (r=0.52, P=0.027) were positively correlated with the Masson staining results and with the expression of α-SMA and Fibronectin. While the ADC value was positively correlated with IL-6 (r=0.553, P=0.002), there was no obvious correlation with IL-1β, TNF-a and TGF-β1; the T1 value was positively correlated with IL-1β (r=0.419, P=0.021), IL-6 (r=0.535, P=0.002) and TNF-a (r=0.540, P=0.002) but not significantly related to TGF-β1 (r=0.155, P=0.413). Conclusion The MR-DWI and T1 mapping values on the 7th day after irradiation can reflect the early condition of tissue inflammation after the soft tissue is irradiated, and the values have a certain correlation with the degree of radiofibrosis of the soft tissue in the later period and may be used as an index to predict radiofibrosis.
Collapse
Affiliation(s)
- Zeng Wang
- Central Laboratory, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.,Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Bowen Xiong
- National Health Commission Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China.,Department of Radiation Oncology, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
| | - Nannan Kang
- Department of Radiology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Xiaoxian Pan
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Caihong Wang
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Li Su
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.,Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Department of Radiotherapy, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhen Xing
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.,Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jinsheng Hong
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.,Department of Radiotherapy, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
19
|
Abstract
Esophageal cancer is the sixth most common cause of cancer related mortality worldwide. Advances in treatment have translated into steadily improving survival rates. Accurate preoperative staging of esophageal cancer is imperative in order to provide an accurate prognosis and direct patients to the most appropriate treatment. Current preoperative staging relies on imaging, most commonly endoscopic ultrasound (EUS), computed tomography (CT) and positron emission tomography (PET). A combination of these modalities should be used in preoperative staging, as each has advantages over another. Magnetic resonance imaging (MRI) has always shown promise in its ability to accurately stage esophageal cancer, though it has not been consistently adopted as a common tool for this purpose. Recent research has demonstrated that MRI can become an integral part of esophageal cancer clinical staging. Advances in MR technology that utilize radial sampling allow for shorter, free breathing techniques without degradation of image quality, resulting in improved capability for T and N staging of esophageal cancer. MRI enhanced with superparamagnetic iron oxide (SPIO) and ultrasmall SPIO (USPIO) nanoparticles has been shown to be useful for the detection of metastatic disease in lymph nodes. This article will review the current evidence in the role that imaging plays in staging esophageal cancer.
Collapse
Affiliation(s)
- Eric J Schmidlin
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ritu R Gill
- Department of Radiology, Beth Israel Deaconness Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
20
|
Ao S, Wang Y, Song Q, Ye Y, Lyu G. Current status and future perspectives on neoadjuvant therapy in gastric cancer. Chin J Cancer Res 2021; 33:181-192. [PMID: 34158738 PMCID: PMC8181872 DOI: 10.21147/j.issn.1000-9604.2021.02.06] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer, with high morbidity and mortality rates, is one of the most heterogeneous tumors. Radical gastrectomy and postoperative chemotherapy are the standard treatments. However, the safety and efficacy of neoadjuvant therapy (NAT) need to be confirmed by many trials before implementation, creating a bottleneck in development. Although clinical benefits of NAT have been observed, a series of problems remain to be solved. Before therapy, more contributing factors should be offered for choice in the intended population and ideal regimens. Enhanced computed tomography (CT) scanning is usually applied to evaluate effectiveness according to Response Evaluation Criteria in Solid Tumors (RECIST), yet CT scanning results sometimes differ from pathological responses. After NAT, the appropriate time for surgery is still empirically defined. Our review aims to discuss the abovementioned issues regarding NAT for GC, including indications, selection of regimens, lesion assessment and NAT-surgery interval time.
Collapse
Affiliation(s)
- Sheng Ao
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, China
- Department of Gastrointestinal Surgery, Peking University People’s Hospital, Beijing 100044, China
| | - Yuchen Wang
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, China
| | - Qingzhi Song
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People’s Hospital, Beijing 100044, China
| | - Guoqing Lyu
- Department of Gastrointestinal Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, China
| |
Collapse
|
21
|
Foley KG, Pearson B, Riddell Z, Taylor SA. Opportunities in cancer imaging: a review of oesophageal, gastric and colorectal malignancies. Clin Radiol 2021; 76:748-762. [PMID: 33579518 DOI: 10.1016/j.crad.2021.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/13/2021] [Indexed: 02/07/2023]
Abstract
The incidence of gastrointestinal (GI) malignancy is increasing worldwide. In particular, there is a concerning rise in incidence of GI cancer in younger adults. Direct endoscopic visualisation of luminal tumour sites requires invasive procedures, which are associated with certain risks, but remain necessary because of limitations in current imaging techniques and the continuing need to obtain tissue for diagnosis and genetic analysis; however, management of GI cancer is increasingly reliant on non-invasive, radiological imaging to diagnose, stage, and treat these malignancies. Oesophageal, gastric, and colorectal malignancies require specialist investigation and treatment due to the complex nature of the anatomy, biology, and subsequent treatment strategies. As cancer imaging techniques develop, many opportunities to improve tumour detection, diagnostic accuracy and treatment monitoring present themselves. This review article aims to report current imaging practice, advances in various radiological modalities in relation to GI luminal tumour sites and describes opportunities for GI radiologists to improve patient outcomes.
Collapse
Affiliation(s)
- K G Foley
- Department of Clinical Radiology, Royal Glamorgan Hospital, Llantrisant, UK.
| | - B Pearson
- National Imaging Academy Wales (NIAW), Pencoed, UK
| | - Z Riddell
- National Imaging Academy Wales (NIAW), Pencoed, UK
| | - S A Taylor
- Centre for Medical Imaging, UCL, London, UK
| |
Collapse
|
22
|
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
|
23
|
Zhu Y, Zhou Y, Zhang W, Xue L, Li Y, Jiang J, Zhong Y, Wang S, Jiang L. Value of quantitative dynamic contrast-enhanced and diffusion-weighted magnetic resonance imaging in predicting extramural venous invasion in locally advanced gastric cancer and prognostic significance. Quant Imaging Med Surg 2021; 11:328-340. [PMID: 33392032 DOI: 10.21037/qims-20-246] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Extramural venous invasion (EMVI) has been found to be related to poor prognosis in gastric cancer. Preoperative diagnosis of EMVI is challenging, as it can only be detected by surgical pathology. The present study aimed to investigate the value of quantitative dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and diffusion-weighted imaging (DWI) in predicting EMVI preoperatively, and to determine the relationship between prediction results and prognosis in patients with locally advanced gastric cancer (LAGC). Methods Between January, 2015, and June, 2017, 79 LAGC patients underwent MRI preoperatively were enrolled in this study. Pathological EMVI (pEMVI) was used as the gold standard for diagnosis. The differences in quantitative DCE-MRI and DWI parameters between groups with different pEMVI status were analyzed. Multivariate logistic regression was used to build the combined prediction model for pEMVI with statistically significant quantitative parameters. The performance of the model for predicting pEMVI was evaluated using receiver operating characteristic (ROC) analysis. Patients were grouped based on MRI-predicted EMVI (mrEMVI). Kaplan-Meier analysis was used to investigate the relationship between mrEMVI and 2-year recurrence-free survival (RFS). Results Of the 79 LAGC patients who underwent MRI, 29 were pEMVI positive and 50 were pEMVI negative. Among the patients' clinical and pathological characteristics, only postoperative staging showed a significant difference between the 2 groups (P=0.015). The pEMVI-positive group had higher volume transfer constant (Ktrans) and rate constant (kep), and lower apparent diffusion coefficient (ADC) values than the negative group (0.189 vs. 0.082 min-1, 0.687 vs. 0.475 min-1, and 1.230×10-3 vs. 1.463×10-3 mm2/s, respectively; P<0.05). Quantitative parameters, Ktrans and kep, and ADC values, were independently associated with pEMVI which odds ratio values were 3.66, 2.65, and 0.30 (P<0.05), respectively, using multivariate logistic regression. ROC analysis showed that the area under the curve, sensitivity, specificity, positive predictive value, and negative predictive value in predicting pEMVI using combined Ktrans, kep, and ADC values were 0.879, 72.4%, 96%, 91.3%, and 85.7%, respectively. A total of 23 cases were considered to be mrEMVI positive, and 56 cases were considered to be mrEMVI negative, according to the predictive results. The median RFS of the mrEMVI-positive group was significant lower than the negative group (21.7 vs. 31.2 months), and the 2-year RFS rate in the mrEMVI-positive group was significantly lower than that of the negative group (43.6% vs. 72.5%, P=0.010). Conclusions The quantitative DCE-MRI parameters, Ktrans and kep, and DWI parameter, ADC, are independent predictors of pEMVI in LAGC; mrEMVI was confirmed to be a poor prognostic predictor for RFS.
Collapse
Affiliation(s)
- Yongjian Zhu
- Department of Imaging Diagnosis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yutao Zhou
- Department of Imaging Diagnosis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Li
- Department of Imaging Diagnosis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Jiang
- Department of Imaging Diagnosis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxin Zhong
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sicong Wang
- GE Healthcare, Life Sciences, Beijing, China
| | - Liming Jiang
- Department of Imaging Diagnosis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
24
|
López Sala P, Alberdi Aldasoro N, Fuertes Fernández I, Sáenz Bañuelos J. An updated review of the TNM classification system for cancer of the esophagus and its complications. RADIOLOGIA 2020. [PMID: 33268136 DOI: 10.1016/j.rx.2020.09.003] [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: 11/25/2022]
Abstract
Cancer of the esophagus is an aggressive cancer with high mortality. Because of the esophagus's lack of serosa and its peculiar lymphatic drainage, esophageal cancer is diagnosed in advanced stages. The eighth edition of the TNM (2017) aims to standardize care for esophageal cancer throughout the world; it includes not only patients treated with esophagectomy alone, but also those receiving neoadjuvant chemotherapy and/or radiotherapy. One new development in the eighth edition is that it establishes separate classifications for different time periods, with pathologic stage groups for prior to treatment (cTNM), after esophagectomy (pTNM), and after neoadjuvant therapy (ypTNM). The combined use of endoscopic ultrasound, CT, PET-CT, and MRI provides the greatest accuracy in determining the clinical stage, and these techniques are essential for planning treatment and for evaluating the response to neoadjuvant treatment. Esophagectomy continues to be the main treatment; it is also the elective gastrointestinal surgery that has the highest mortality, and it carries the risk of multiple complications, including anastomotic leaks, pulmonary complications, technical complications, and functional complications.
Collapse
Affiliation(s)
- P López Sala
- Residente del servicio de Radiodiagnóstico, Complejo Hospitalario de Navarra, Pamplona, España.
| | - N Alberdi Aldasoro
- Residente del servicio de Radiodiagnóstico, Complejo Hospitalario de Navarra, Pamplona, España
| | - I Fuertes Fernández
- FEA del servicio de Radiodiagnóstico, Complejo Hospitalario de Navarra, Pamplona, España
| | - J Sáenz Bañuelos
- FEA del servicio de Radiodiagnóstico, Complejo Hospitalario de Navarra, Pamplona, España
| |
Collapse
|
25
|
Onal Y, Samanci C. The Role of Diffusion-weighted Imaging in Patients with Gastric Wall Thickening. Curr Med Imaging 2020; 15:965-971. [PMID: 32013813 DOI: 10.2174/1573405614666181115120109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 10/18/2018] [Accepted: 10/26/2018] [Indexed: 01/20/2023]
Abstract
BACKGROUND Gastric cancer is the second leading cause of cancer death worldwide. AIMS In the benign and malign gastric pathologies, we measured the Apparent Diffusion Coefficient (ADC) value from the thickened section of the stomach wall. We assessed the diagnostic value of ADC and we wanted to see whether this value could be used to diagnose gastric pathologies. STUDY DESIGN This study has a prospective study design. METHODS A total of 90 patients, 27 with malign gastric pathologies 63 with benign gastric pathologies with Gastric Wall (GW) thickening in multidector CT, were evaluated by T2 weighted axial MR imaging and Diffusion-Weighted Imaging (DWI). Measurements were made both from the thickened wall and from the normal GW. Also, a new method called GW/spine ADC ratio was performed in image analysis. The value found after ADC measurement from the GW was proportioned to the spinal cord ADC value in the same section. RESULTS The ADC values measured from the pathological wall in patients with gastric malignancy (1.115 ± 0.156 x10-3 mm2/s) were significantly lower than the healthy wall measurements (1.621 ± 0.292 × 10-3 mm2/s) and benign gastric diseases (1.790± 0.359 x10-3 mm2/s). GW/spine ADC ratio was also lower in gastric malignancy group. CONCLUSION ADC measurement in DWI can be used to distinguish between benign and malign gastric pathologies.
Collapse
Affiliation(s)
- Yilmaz Onal
- Department of Radiology, Sultan Abdulhamid Han Training and Research Hospital, Haydarpasa, Istanbul, Turkey
| | - Cesur Samanci
- Department of Radiology, Sultan Abdulhamid Han Training and Research Hospital, Haydarpasa, Istanbul, Turkey
| |
Collapse
|
26
|
Song T, Yao Q, Qu J, Zhang H, Zhao Y, Qin J, Feng W, Zhang S, Han X, Wang S, Yan X, Li H. The value of intravoxel incoherent motion diffusion-weighted imaging in predicting the pathologic response to neoadjuvant chemotherapy in locally advanced esophageal squamous cell carcinoma. Eur Radiol 2020; 31:1391-1400. [PMID: 32901300 DOI: 10.1007/s00330-020-07248-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/05/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To explore the value of intravoxel incoherent motion diffusion-weighted imaging (IVIM-DWI) for the prediction of pathologic response to neoadjuvant chemotherapy (NAC) in locally advanced esophageal squamous cell carcinoma (ESCC). MATERIAL AND METHODS Forty patients with locally advanced ESCC who were treated with NAC followed by radical resection were prospectively enrolled from September 2015 to May 2018. MRI and IVIM were performed within 1 week before and 2-3 weeks after NAC, prior to surgery. Parameters including apparent diffusion coefficient (ADC), true diffusion coefficient (D), pseudodiffusion coefficient (D*), and pseudodiffusion fraction (f) before and after NAC were measured. Pathologic response was evaluated according to the AJCC tumor regression grade (TRG) system. The changes in IVIM values before and after therapy in different TRG groups were assessed. Receiver operating characteristic (ROC) curves analysis was used to determine the best cutoff value for predicting the pathologic response to NAC. RESULTS Twenty-two patients were identified as TRG 2 (responders), and eighteen as TRG 3 (non-responders) in pathologic evaluation. The ADC, D, and f values increased significantly after NAC. The post-NAC D and ΔD values of responders were significantly higher than those of non-responders. The area under the curve (AUC) was 0.722 for post-NAC D and 0.859 for ΔD in predicting pathologic response. The cutoff values of post-NAC D and ΔD were 1.685 × 10-3 mm2/s and 0.350 × 10-3 mm2/s, respectively. CONCLUSION IVIM-DWI may be used as an effective functional imaging technique to predict pathologic response to NAC in locally advanced ESCC. KEY POINTS • The optimal cutoff values of post-NAC D and ΔD for predicting pathologic response to NAC in locally advanced ESCC were 1.685 × 10-3 mm2/s and 0.350 × 10-3 mm2/s, respectively. • Pathologic response to NAC in locally advanced ESCC was favorable in patients with post-NAC D and ΔD values that were higher than the optimal cutoff values. • IVIM-DWI can potentially be used to preoperatively predict pathologic response to NAC in esophageal carcinoma. Accurate quantification of the D value derived from IVIM-DWI may eventually translate into an effective and non-invasive marker to predict therapeutic efficacy.
Collapse
Affiliation(s)
- Tao Song
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| | - Qi Yao
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| | - Jinrong Qu
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China.
| | - Hongkai Zhang
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| | - Yan Zhao
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| | - Jianjun Qin
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Wen Feng
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Shouning Zhang
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| | - Xianhua Han
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| | - Shaoyu Wang
- MR Scientific Marketing, Siemens Healthineers, XI'an, 710065, China
| | - Xu Yan
- MR Scientific Marketing, Siemens Healthineers, Shanghai, 201318, China
| | - Hailiang Li
- Department of Radiology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming road, Jinshui District, Zhengzhou city, Henan Province, China
| |
Collapse
|
27
|
Elsherif SB, Andreou S, Virarkar M, Soule E, Gopireddy DR, Bhosale PR, Lall C. Role of precision imaging in esophageal cancer. J Thorac Dis 2020; 12:5159-5176. [PMID: 33145093 PMCID: PMC7578477 DOI: 10.21037/jtd.2019.08.15] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Esophageal cancer is a major cause of morbidity and mortality worldwide. Recent advancements in the management of esophageal cancer have allowed for earlier detection, improved ability to monitor progression, and superior treatment options. These innovations allow treatment teams to formulate more customized management plans and have led to an increase in patient survival rates. For example, in order for the most effective management plan to be constructed, accurate staging must be performed to determine tumor resectability. This article reviews the multimodality imaging approach involved in making a diagnosis, staging, evaluating treatment response and detecting recurrence in esophageal cancer.
Collapse
Affiliation(s)
- Sherif B Elsherif
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL, USA.,Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sonia Andreou
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Mayur Virarkar
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Erik Soule
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | | | - Priya R Bhosale
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chandana Lall
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL, USA
| |
Collapse
|
28
|
Locoregional Residual Esophageal Cancer after Neo-adjuvant Chemoradiotherapy and Surgery Regarding Anatomic Site and Radiation Target Fields: A Histopathologic Evaluation Study. Ann Surg 2020; 275:e759-e765. [PMID: 32740246 DOI: 10.1097/sla.0000000000004242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Neoadjuvant chemoradiotherapy followed by surgery establishes a considerable pathologic complete response (pCR) in EC. The aim was to determine site of residual tumor and its prognostic impact. SUMMARY BACKGROUND DATA High rates of residual tumor in the adventitial region even inside the radiation fields will influence current decision-making. METHODS We evaluated resection specimens with marked target fields from 151 consecutive EC patients treated with carboplatin/paclitaxel and 41.4Gy between 2009 and 2018. RESULTS In radically resected (R0) specimens 19.8% (27/136) had a pCR (ypT0N0) and 14% nearly no response (tumor regression grade: tumor regression grade 4-5). Residual tumor commonly extended in or restricted to the adventitia (43.1%; 47/109), whereas 7.3% was in the mucosa (ypT1a), 16.5% in the submucosa (ypT1b) and 6.4% only in lymph nodes (ypT0N+). Macroscopic residues in R0-specimens of partial responders (tumor regression grade 2-3: N = 90) were found in- and outside the gross tumor volume (GTV) in 33.3% and 8.9%, and only microscopic in- and outside the clinical target volume in 58.9% and 1.1%, respectively. Residual nodal disease was observed proximally and distally to the clinical target volume in 2 and 5 patients, respectively. Disease Free Survival decreased significantly if macroscopic tumor was outside the GTV and in ypT2-4aN+. CONCLUSIONS After neoadjuvant chemoradiotherapy, pCR and ypT1aN0 were seen in a limited number of R0 resected specimens (19.8% and 7.3%, respectively), whereas 6.4% had only nodal disease (yT0N+). Disease Free Survival decreased significantly if macroscopic residue was outside the GTV and in responders with only nodal disease. Therefore, we should be cautious in applying wait and see strategies.
Collapse
|
29
|
De Cobelli F, Palumbo D, Albarello L, Rosati R, Giganti F. Esophagus and Stomach: Is There a Role for MR Imaging? Magn Reson Imaging Clin N Am 2020; 28:1-15. [PMID: 31753229 DOI: 10.1016/j.mric.2019.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
MR imaging has been increasingly used in the diagnostic work-up of benign and malignant conditions of the gastroesophageal tract. The use of an adequate MR imaging protocol is crucial, although standardization of imaging studies is still far from being implemented. Research on MR imaging-based biomarkers show promising results in assessing tumor aggressiveness and prognosis, and in the evaluation of response to treatment, both in esophageal and gastric cancers.
Collapse
Affiliation(s)
- Francesco De Cobelli
- Department of Radiology, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| | - Diego Palumbo
- Department of Radiology, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Luca Albarello
- Vita-Salute San Raffaele University, Milan, Italy; Department of Pathology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Riccardo Rosati
- Vita-Salute San Raffaele University, Milan, Italy; Department of Gastrointestinal Surgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Giganti
- Department of Radiology, University College London Hospital NHS Foundation Trust, London, UK; Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, 3(rd) Floor, Charles Bell House, 43-45 Foley Street, London W1W 7TS, UK
| |
Collapse
|
30
|
Maffazzioli L, Zilio MB, Klamt AL, Duarte JA, Mazzini GS, Campos VJ, Chedid MF, Gurski RR. ADC as a predictor of pathologic response to neoadjuvant therapy in esophageal cancer: a systematic review and meta-analysis. Eur Radiol 2020; 30:3934-3942. [PMID: 32157409 DOI: 10.1007/s00330-020-06723-x] [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: 11/20/2019] [Revised: 01/31/2020] [Accepted: 02/05/2020] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Diffusion-weighted magnetic resonance imaging (DWI) is part of clinical practice. The aim of this study was to evaluate the role of apparent diffusion coefficient (ADC) as a predictor of pathologic response to neoadjuvant therapy (nCRT) in patients with esophageal cancer (EC). METHODS The MEDLINE, Embase, and Google Scholar databases were systematically searched for studies using ADC to evaluate response to neoadjuvant therapy in patients with EC. Methodological quality of the studies was evaluated with the QUADAS tool. Data from eligible studies were extracted and evaluated by two independent reviewers. Meta-analyses were performed comparing mean ADC values between responders and non-responders to nCRT in three different scenarios: baseline (BL) absolute values; percent change between intermediate (IM) values and BL; and percent change between final follow-up (FU) value and baseline BL. RESULTS Seven studies (n = 158 patients) were included. Responders exhibited a statistically significant percent increase in ADC during nCRT (mean difference [MD] 21.06%, 95%CI = 13.04-29.09; I2 = 49%; p = 0.12). A similar increase was identified in the complete pathologic response (pCR) versus non-complete pathologic response (npCR) subgroup (MD = 25.68%, 95%CI = 18.87-32.48; I2 = 0%; p = 0.60). At the end of treatment, responders also exhibited a statistically significant percent increase in ADC (MD = 22.49%, 95%CI = 9.94-35.05; I2 = 0%; p = 0.46). BL ADC was not associated with any definition of pathologic response (MD = 0.11%, 95%CI = - 0.21-0.42; I2 = 85%; p < 0.01). CONCLUSION These results suggest that ADC can be used as a predictor of pathologic response, with a statistically significant association between percent ADC increase during and after treatment and pCR. ADC may serve as a tool to help in guiding clinical decisions. KEY POINTS • DWI is routinely included in MRI oncological protocols. • ADC can be used as a predictor of pathologic response, with a statistically significant association between percent ADC increase during and after treatment and pCR.
Collapse
Affiliation(s)
- Leticia Maffazzioli
- Division of Radiology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos 2350, 2nd Floor, Porto Alegre, 90035-903, Brazil.
- Post-Graduation Program in Medicine: Surgical Sciences, Medical School of UFRGS, Porto Alegre, Brazil.
| | - Mariana B Zilio
- Post-Graduation Program in Medicine: Surgical Sciences, Medical School of UFRGS, Porto Alegre, Brazil
- Division of Gastrointestinal Surgery, Hospital de Clínicas de Porto Alegre (HCPA), Medical School of UFRGS, Porto Alegre, Brazil
| | - Alexandre L Klamt
- Post-Graduation Program in Medicine: Surgical Sciences, Medical School of UFRGS, Porto Alegre, Brazil
- Division of Gastroenterology, Hospital de Clínicas de Porto Alegre (HCPA), Medical School of UFRGS, Porto Alegre, Brazil
| | - Juliana A Duarte
- Division of Radiology, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos 2350, 2nd Floor, Porto Alegre, 90035-903, Brazil
| | - Guilherme S Mazzini
- Division of Gastrointestinal Surgery, Hospital de Clínicas de Porto Alegre (HCPA), Medical School of UFRGS, Porto Alegre, Brazil
| | - Vinicius J Campos
- Post-Graduation Program in Medicine: Surgical Sciences, Medical School of UFRGS, Porto Alegre, Brazil
| | - Marcio F Chedid
- Post-Graduation Program in Medicine: Surgical Sciences, Medical School of UFRGS, Porto Alegre, Brazil
- Division of Gastrointestinal Surgery, Hospital de Clínicas de Porto Alegre (HCPA), Medical School of UFRGS, Porto Alegre, Brazil
| | - Richard R Gurski
- Post-Graduation Program in Medicine: Surgical Sciences, Medical School of UFRGS, Porto Alegre, Brazil
- Division of Gastrointestinal Surgery, Hospital de Clínicas de Porto Alegre (HCPA), Medical School of UFRGS, Porto Alegre, Brazil
| |
Collapse
|
31
|
Clinical response assessment on DW-MRI compared with FDG-PET/CT after neoadjuvant chemoradiotherapy in patients with oesophageal cancer. Eur J Nucl Med Mol Imaging 2020; 48:176-185. [PMID: 32572560 DOI: 10.1007/s00259-020-04917-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/07/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE In about 30% of patients treated with neoadjuvant chemoradiotherapy (nCRT) followed by surgical resection for locally advanced oesophageal cancer no vital tumour is found in the resection specimen. Accurate clinical response assessment is critical if deferral from surgery is considered in complete responders. Our study aimed to compare the performance of MRI and of FDG-PET/CT for the detection of residual disease after nCRT. METHODS Patients with oesophageal cancer eligible for nCRT and oesophagectomy were prospectively included. All patients underwent FDG-PET/CT and MRI before and between 6 and 8 weeks after nCRT. Two radiologists scored the MRI scans, and two nuclear medicine physicians scored the FDG-PET/CT scans using a 5-point score for residual disease. Histopathology after oesophagectomy represented the reference standard. Sensitivity, specificity, and area under the receiver operating characteristic curve (AUC) were calculated for detection of residual tumour (ypT+), residual nodal disease (ypN+), and any residual disease (ypT+Nx/ypT0N+). RESULTS Seven out of 33 (21%) patients had a pathological complete response. The AUCs for individual readers to detect ypT+ were 0.71/0.70 on diffusion-weighted (DW)-MRI and 0.54/0.57 on FDG-PET/CT, and to detect ypN+ were 0.89/0.81 on DW-MRI and 0.75/0.71 on FDG-PET/CT. The AUCs/sensitivities/specificities for the individual readers to detect any residual disease were 0.74/92%/57% and 0.70/96%/43% on MRI; these were 0.49/69%/29% and 0.60/69%/43% on FDG-PET/CT, respectively. CONCLUSION MRI reached higher diagnostic accuracies than FDG-PET/CT for the detection of residual tumour in oesophageal cancer patients at 6 to 8 weeks after nCRT.
Collapse
|
32
|
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
|
33
|
Abstract
Gastric cancer is the fifth most common malignancies and the third leading cause of cancer-related death worldwide, with more than 40% of new cases occurring in China. With the advancement of treatment methods, the application of adjuvant therapy and targeted drugs, the prognosis of patients with gastric cancer has been significantly improved. In recent years, more and more studies have reported that magnetic resonance imaging (MRI) showed great value in the clinical application among patients with gastric cancer, including preoperative staging, treatment response evaluation, predicting prognosis and histopathological features, treatment guidance, and molecular imaging. The remarkable research progress of MRI in gastric cancer will provide new evaluation and treatment approaches for clinical diagnosis and treatment. This article aims to review the current status of the application and research progress of MRI in patients with gastric cancer.
Collapse
Affiliation(s)
- Yingjing Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jianchun Yu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
34
|
Borggreve AS, Heethuis SE, Boekhoff MR, Goense L, van Rossum PSN, Brosens LAA, van Lier ALHMW, van Hillegersberg R, Lagendijk JJW, Mook S, Ruurda JP, Meijer GJ. Optimal timing for prediction of pathologic complete response to neoadjuvant chemoradiotherapy with diffusion-weighted MRI in patients with esophageal cancer. Eur Radiol 2020; 30:1896-1907. [PMID: 31822974 PMCID: PMC7062655 DOI: 10.1007/s00330-019-06513-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/18/2019] [Accepted: 10/14/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE This study was conducted in order to determine the optimal timing of diffusion-weighted magnetic resonance imaging (DW-MRI) for prediction of pathologic complete response (pCR) to neoadjuvant chemoradiotherapy (nCRT) for esophageal cancer. METHODS Patients with esophageal adenocarcinoma or squamous cell carcinoma who planned to undergo nCRT followed by surgery were enrolled in this prospective study. Patients underwent six DW-MRI scans: one baseline scan before the start of nCRT and weekly scans during 5 weeks of nCRT. Relative changes in mean apparent diffusion coefficient (ADC) values between the baseline scans and the scans during nCRT (ΔADC(%)) were compared between pathologic complete responders (pCR) and non-pCR (tumor regression grades 2-5). The discriminative ability of ΔADC(%) was determined based on the c-statistic. RESULTS A total of 24 patients with 142 DW-MRI scans were included. pCR was observed in seven patients (29%). ΔADC(%) from baseline to week 2 was significantly higher in patients with pCR versus non-pCR (median [IQR], 36% [30%, 41%] for pCR versus 16% [14%, 29%] for non-pCR, p = 0.004). The ΔADC(%) of the second week in combination with histology resulted in the highest c-statistic for the prediction of pCR versus non-pCR (0.87). The c-statistic of this model increased to 0.97 after additional exclusion of patients with a small tumor volume (< 7 mL, n = 3) and tumor histology of the resection specimen other than adenocarcinoma or squamous cell carcinoma (n = 1). CONCLUSION The relative change in tumor ADC (ΔADC(%)) during the first 2 weeks of nCRT is the most predictive for pathologic complete response to nCRT in esophageal cancer patients. KEY POINTS • DW-MRI during the second week of neoadjuvant chemoradiotherapy is most predictive for pathologic complete response in esophageal cancer. • A model including ΔADCweek 2was able to discriminate between pathologic complete responders and non-pathologic complete responders in 87%. • Improvements in future MRI studies for esophageal cancer may be obtained by incorporating motion management techniques.
Collapse
Affiliation(s)
- Alicia S Borggreve
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands.
- Department of Surgery, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands.
| | - Sophie E Heethuis
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Mick R Boekhoff
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Lucas Goense
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
- Department of Surgery, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Peter S N van Rossum
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Lodewijk A A Brosens
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Astrid L H M W van Lier
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Richard van Hillegersberg
- Department of Surgery, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Jan J W Lagendijk
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Stella Mook
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Jelle P Ruurda
- Department of Surgery, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Gert J Meijer
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands.
| |
Collapse
|
35
|
Borggreve AS, Goense L, van Rossum PSN, Heethuis SE, van Hillegersberg R, Lagendijk JJW, Lam MGEH, van Lier ALHMW, Mook S, Ruurda JP, van Vulpen M, Voncken FEM, Aleman BMP, Bartels-Rutten A, Ma J, Fang P, Musall BC, Lin SH, Meijer GJ. Preoperative Prediction of Pathologic Response to Neoadjuvant Chemoradiotherapy in Patients With Esophageal Cancer Using 18F-FDG PET/CT and DW-MRI: A Prospective Multicenter Study. Int J Radiat Oncol Biol Phys 2020; 106:998-1009. [PMID: 31987972 DOI: 10.1016/j.ijrobp.2019.12.038] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 11/06/2019] [Accepted: 12/26/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE Accurate preoperative prediction of pathologic response to neoadjuvant chemoradiotherapy (nCRT) in patients with esophageal cancer could enable omission of esophagectomy in patients with a pathologic complete response (pCR). This study aimed to evaluate the individual and combined value of 18F-fluorodeoxyglucose positron emission tomography with integrated computed tomography (18F-FDG PET/CT) and diffusion-weighted magnetic resonance imaging (DW-MRI) during and after nCRT to predict pathologic response in patients with esophageal cancer. METHODS AND MATERIALS In this multicenter prospective study, patients scheduled to receive nCRT followed by esophagectomy for esophageal cancer underwent 18F-FDG PET/CT and DW-MRI scanning before the start of nCRT, during nCRT, and before esophagectomy. Response to nCRT was based on histopathologic evaluation of the resection specimen. Relative changes in 18F-FDG PET/CT and DW-MRI parameters were compared between patients with pCR and non-pCR groups. Multivariable ridge regression analyses with bootstrapped c-indices were performed to evaluate the individual and combined value of 18F-FDG PET/CT and DW-MRI. RESULTS pCR was found in 26.1% of 69 patients. Relative changes in 18F-FDG PET/CT parameters after nCRT (Δ standardized uptake value [SUV]mean,postP = .016, and Δ total lesion glycolysis postP = .024), as well as changes in DW-MRI parameters during nCRT (Δ apparent diffusion coefficient [ADC]duringP = .008) were significantly different between pCR and non-pCR. A c-statistic of 0.84 was obtained for a model with ΔADCduring, ΔSUVmean,post, and histology in classifying patients as pCR (versus 0.82 for ΔADCduring and 0.79 for ΔSUVmean,post alone). CONCLUSIONS Changes on 18F-FDG PET/CT after nCRT and early changes on DW-MRI during nCRT can help identify pCR to nCRT in esophageal cancer. Moreover, 18F-FDG PET/CT and DW-MRI might be of complementary value in the assessment of pCR.
Collapse
Affiliation(s)
- Alicia S Borggreve
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands; Department of Surgery, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Lucas Goense
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands; Department of Surgery, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Peter S N van Rossum
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Sophie E Heethuis
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | | | - Jan J W Lagendijk
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Marnix G E H Lam
- Department of Nuclear Medicine, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Astrid L H M W van Lier
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Stella Mook
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Jelle P Ruurda
- Department of Surgery, University Medical Center Utrecht, Utrecht University, the Netherlands
| | | | - Francine E M Voncken
- Department of Radiation Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Berthe M P Aleman
- Department of Radiation Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Annemarieke Bartels-Rutten
- Department of Radiology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Jingfei Ma
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Penny Fang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Benjamin C Musall
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gert J Meijer
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, the Netherlands.
| |
Collapse
|
36
|
Leandri C, Soyer P, Oudjit A, Guillaumot MA, Chaussade S, Dohan A, Barret M. Contribution of magnetic resonance imaging to the management of esophageal diseases: A systematic review. Eur J Radiol 2019; 120:108684. [PMID: 31563109 DOI: 10.1016/j.ejrad.2019.108684] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/11/2019] [Accepted: 09/18/2019] [Indexed: 01/15/2023]
Abstract
PURPOSE Currently available imaging modalities used to investigate the esophagus are irradiating or limited to the analysis of the esophageal lumen. Magnetic resonance imaging (MRI) is a non-invasive and non-radiating imaging technique that provides high degrees of soft tissue contrast. Newly developed fast MRI sequences allow for both morphological and functional assessment of the esophageal body and esophagogastric junction. The purpose of this systematic review was to identify the contribution of MRI to the diagnosis and management of esophageal diseases, such as gastroesophageal reflux, esophageal motility disorders, esophageal neoplasms, and portal hypertension. METHODS We performed a systematic search of the Medline (via Ovid), EMBASE (via Ovid), PubMed and Cochrane Library databases from inception to December 2018 inclusively, using the MESH major terms "magnetic resonance imaging" AND "esophagus". RESULTS The initial search retrieved 310 references, of which 56 were found to be relevant for the study. References were analysed and classified in different subheadings: MRI protocols for the esophagus, gastroesophageal reflux disease, achalasia and other esophageal motility disorders, esophageal cancer, portal hypertension and other esophageal conditions. CONCLUSION MR Esophagography might become a non-invasive, non-irradiating technique of choice following diagnostic esophagogastroduodenoscopy for the assessment of esophageal diseases.
Collapse
Affiliation(s)
- Chloé Leandri
- Gastroenterology Department, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, France; Paris Descartes University, Paris, France.
| | - Philippe Soyer
- Paris Descartes University, Paris, France; Department of Radiology, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, France.
| | - Ammar Oudjit
- Department of Radiology, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, France.
| | - Marie-Anne Guillaumot
- Gastroenterology Department, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, France.
| | - Stanislas Chaussade
- Gastroenterology Department, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, France; Paris Descartes University, Paris, France.
| | - Anthony Dohan
- Paris Descartes University, Paris, France; Department of Radiology, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, France.
| | - Maximilien Barret
- Gastroenterology Department, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, France; Paris Descartes University, Paris, France.
| |
Collapse
|
37
|
Vollenbrock SE, Voncken FEM, Bartels LW, Beets-Tan RGH, Bartels-Rutten A. Diffusion-weighted MRI with ADC mapping for response prediction and assessment of oesophageal cancer: A systematic review. Radiother Oncol 2019; 142:17-26. [PMID: 31431376 DOI: 10.1016/j.radonc.2019.07.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE The aim was to perform a systematic review on the value of diffusion-weighted MRI (DW-MRI) with apparent diffusion coefficient (ADC) mapping in the prediction and assessment of response to chemo- and/or radiotherapy in oesophageal cancer. MATERIALS AND METHODS A systematic search was performed on Pubmed, Embase, Medline and Cochrane databases. Studies that evaluated the ADC for response evaluation before, during or after chemo- and/or radiotherapy were included. The Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) was used to assess the quality of the included studies. RESULTS Fourteen studies, comprising 516 patients, in which the response to treatment in oesophageal cancer was evaluated on ADC maps were included. Acquisition parameter settings for DW-MRI and ROI placement varied substantially. The reference standard was RECIST or endoscopic assessment in eight non-surgery studies and histopathology after surgery in six studies. A high pre-treatment ADC significantly correlated with good response in three out of 12 studies; conversely, one study reported a significantly higher pre-treatment ADC in poor responders. In five out of eight studies good responders showed a significantly larger relative increase in ADC two weeks after the onset of treatment (range 23-59%) than poor responders (range 1.5-17%). After chemo- and/or radiotherapy ADC results varied considerably, amongst others due to large variation in the interval between completion of therapy and DW-MRI. CONCLUSION DW-MRI for response evaluation to chemo- and/or radiotherapy in oesophageal cancer shows variable methods and results. A large relative ADC increase after two weeks of treatment seems most predictive for good response.
Collapse
Affiliation(s)
- Sophie E Vollenbrock
- Department of Radiology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.
| | - Francine E M Voncken
- Department of Radiation Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Lambertus W Bartels
- Image Sciences Institute, Imaging Division, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Regina G H Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Annemarieke Bartels-Rutten
- Department of Radiology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| |
Collapse
|
38
|
Hayano K, Ohira G, Hirata A, Aoyagi T, Imanishi S, Tochigi T, Hanaoka T, Shuto K, Matsubara H. Imaging biomarkers for the treatment of esophageal cancer. World J Gastroenterol 2019; 25:3021-3029. [PMID: 31293338 PMCID: PMC6603816 DOI: 10.3748/wjg.v25.i24.3021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/07/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023] Open
Abstract
Esophageal cancer is known as one of the malignant cancers with poor prognosis. To improve the outcome, combined multimodality treatment is attempted. On the other hand, advances in genomics and other "omic" technologies are paving way to the patient-oriented treatment called "personalized" or "precision" medicine. Recent advancements of imaging techniques such as functional imaging make it possible to use imaging features as biomarker for diagnosis, treatment response, and prognosis in cancer treatment. In this review, we will discuss how we can use imaging derived tumor features as biomarker for the treatment of esophageal cancer.
Collapse
Affiliation(s)
- Koichi Hayano
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan
| | - Gaku Ohira
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan
| | - Atsushi Hirata
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan
| | - Tomoyoshi Aoyagi
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan
| | - Shunsuke Imanishi
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan
| | - Toru Tochigi
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan
| | - Toshiharu Hanaoka
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan
| | - Kiyohiko Shuto
- Department of Surgery, Teikyo University Medical Center, Chiba 299-0111, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan
| |
Collapse
|
39
|
Xie T, Ye Z, Pang P, Shao G. Quantitative Multiparametric MRI May Augment the Response to Radiotherapy in Mid-Treatment Assessment of Patients with Esophageal Carcinoma. Oncol Res Treat 2019; 42:326-333. [PMID: 31064001 DOI: 10.1159/000499322] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 02/28/2019] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The purpose of this study was to assess the mid-treatment response to radiotherapy (RT) using dynamic contrast-enhanced MRI (DCE-MRI) and diffusion-weighted imaging (DWI) in patients with esophageal cancer (EC). METHODS 42 patients with squamous EC were prepared for DCE-MRI and DWI scans both before treatment (NRT) and after the fifth radiotherapy (5th RT). The patients were classified into two groups (complete response [CR] and partial response [PR]) according to tumor regression after treatment. The quantitative parameters of DCE-MRI (Ktrans, Kep, Ve, and ADC) were measured. A receiver operating characteristic curve (ROC) was used to detect the efficiency of the above parameters. RESULTS After 1 month of RT, 29 patients were classified as CR and 11 patients were classified as PR. In the NRT group, the p values of Ktrans, Kep, Ve, and ADC were 0.004, 0.078, 0.0008, and <0.0001, respectively. After the 5th RT, the p values of the above parameters were <0.001, 0.005, 0.108, and 0.365, respectively. In the NRT group, the areas under the ROC curves of Ktrans, Ve, and ADC were 0.790, 0.617, and 0.737; the sensitivity values were 89.3, 92.5, and 90.0%; the specificity values were 69.4, 27.5, and 50.0%. In the 5th RT group, the areas under the ROC curves of Ktrans and Kep were 0.816 and 0.804; the sensitivity values were 71.2 and 95.0%; the specificity values were 81.6 and 50.0%. CONCLUSION DCE-MRI combined with DWI is effective in the early prediction of radiotherapeutic response of EC after the 5th RT other than after the traditional final treatment.
Collapse
Affiliation(s)
- Tieming Xie
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhimin Ye
- Department of Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| | - Peipei Pang
- Life Sciences, GE Healthcare, Hangzhou, China
| | - Guoliang Shao
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, China,
| |
Collapse
|
40
|
Cheng B, Yu J. Predictive value of diffusion-weighted MR imaging in early response to chemoradiotherapy of esophageal cancer: a meta-analysis. Dis Esophagus 2019; 32:5054272. [PMID: 30010733 DOI: 10.1093/dote/doy065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The results of diffusion-weighted MR imaging (DW-MRI) in predicting early response to chemoradiotherapy in patients with esophageal cancer varied in different studies. We performed this meta-analysis to evaluate the predictive values of DW-MRI and compare the diagnostic efficacy of different apparent diffusion coefficients (ADCs). A comprehensive literature search was performed to identify relevant articles published before November 2017. The quality of study was assessed using Quality Assessment of Diagnostic Accuracy Studies-2. The pooled sensitivity, specificity, diagnostic odds ratio (DOR), and area under receiver operating characteristic curve of ADC values were calculated to determine the diagnostic performance. Seven studies with a total of 236 patients were included. The pooled sensitivity, specificity, DOR, and area under curve were 93% (95% CI 77%-98%), 85% (95% CI 72%-93%), 78 (95% CI 15-401), and 0.91 (95% CI 0.89-0.94), respectively, for the ▵ADC; and 75% (95% CI 62%-84%), 90% (95% CI 67%-97%), 26 (95% CI 6-110), and 0.85 (95% CI 0.82-0.88), respectively, for the post-ADC. For pre-ADC, meta-analysis was not performed because of conflicting results. In conclusions, our results demonstrate that DW-MRI has good performance for evaluating the response to chemoradiation therapy in patients with esophageal cancer. ▵ADC and post-ADC are promising reliable and valuable predictors.
Collapse
Affiliation(s)
- B Cheng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong University, Jinan, China
| | - J Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong University, Jinan, China
| |
Collapse
|
41
|
Borggreve AS, Goense L, Brenkman HJF, Mook S, Meijer GJ, Wessels FJ, Verheij M, Jansen EPM, van Hillegersberg R, van Rossum PSN, Ruurda JP. Imaging strategies in the management of gastric cancer: current role and future potential of MRI. Br J Radiol 2019; 92:20181044. [PMID: 30789792 DOI: 10.1259/bjr.20181044] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Accurate preoperative staging of gastric cancer and the assessment of tumor response to neoadjuvant treatment is of importance for treatment and prognosis. Current imaging techniques, mainly endoscopic ultrasonography (EUS), computed tomography (CT) and 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET), have their limitations. Historically, the role of magnetic resonance imaging (MRI) in gastric cancer has been limited, but with the continuous technical improvements, MRI has become a more potent imaging technique for gastrointestinal malignancies. The accuracy of MRI for T- and N-staging of gastric cancer is similar to EUS and CT, making MRI a suitable alternative to other imaging strategies. There is limited evidence on the performance of MRI for M-staging of gastric cancer specifically, but MRI is widely used for diagnosing liver metastases and shows potential for diagnosing peritoneal seeding. Recent pilot studies showed that treatment response assessment as well as detection of lymph node metastases and systemic disease might benefit from functional MRI (e.g. diffusion weighted imaging and dynamic contrast enhancement). Regarding treatment guidance, additional value of MRI might be expected from its role in better defining clinical target volumes and setup verification with MR-guided radiation treatment.
Collapse
Affiliation(s)
- Alicia S Borggreve
- 1 Department of Surgery, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands.,2 Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Lucas Goense
- 1 Department of Surgery, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands.,2 Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Hylke J F Brenkman
- 1 Department of Surgery, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Stella Mook
- 2 Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Gert J Meijer
- 2 Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Frank J Wessels
- 3 Department of Radiology, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Marcel Verheij
- 4 Department of Radiation Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek (NKI-AVL) , Amsterdam , Netherlands
| | - Edwin P M Jansen
- 4 Department of Radiation Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek (NKI-AVL) , Amsterdam , Netherlands
| | - Richard van Hillegersberg
- 1 Department of Surgery, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Peter S N van Rossum
- 2 Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| | - Jelle P Ruurda
- 1 Department of Surgery, University Medical Center Utrecht, Utrecht University , Utrecht , Netherlands
| |
Collapse
|
42
|
Early response evaluation of neoadjuvant therapy with PET/MRI to predict resectability in patients with adenocarcinoma of the esophagogastric junction. Abdom Radiol (NY) 2019; 44:836-844. [PMID: 30467723 DOI: 10.1007/s00261-018-1841-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
STUDY DESIGN AND PURPOSE Positron emission tomography (PET)/magnetic resonance imaging (MRI) is a new modality that has showed promising results for various clinical indications. Currently, evaluation of neoadjuvant therapy (NT) among patients with adenocarcinoma of the esophagogastric junction has primarily been reserved for PET/computed tomography. Our aim was to evaluate if early response evaluation by PET/MRI is a feasible method to predict resectability. METHODS AND MATERIALS Patients with untreated adenocarcinoma of the esophagogastric junction (Siewert types I/II) and fit for NT with no contraindications for PET/MRI were considered eligible. A baseline scan was performed prior to NT induction and an evaluation scan 3 weeks later. For histopathological response evaluation, the Mandard tumor regression grade score was applied. Response on PET/MRI was evaluated with Response Evaluation Criteria in Solid Tumors (RECIST 1.1), and change in ADC and SUVmax values. RESULTS Twenty-eight patients were enrolled, and 22 completed both scans and proceeded to final analyses. Seventeen patients were found resectable versus five who were found unresectable. PET/MRI response evaluation had a sensitivity 94%, specificity 80%, and AUC = 0.95 when predicting resectability in patients with adenocarcinoma of the esophagogastric junction. No association with histopathological response (tumor regression grade) was found nor was RECIST correlated with resectability. CONCLUSION Response evaluation using PET/MRI was a feasible method to predict resectability in patients with adenocarcinoma of the esophagogastric junction in this pilot study. However, larger studies are warranted to justify the use of the modality for this indication.
Collapse
|
43
|
Vollenbrock SE, Voncken FEM, van Dieren JM, Lambregts DMJ, Maas M, Meijer GJ, Goense L, Mook S, Hartemink KJ, Snaebjornsson P, Ter Beek LC, Verheij M, Aleman BMP, Beets-Tan RGH, Bartels-Rutten A. Diagnostic performance of MRI for assessment of response to neoadjuvant chemoradiotherapy in oesophageal cancer. Br J Surg 2019; 106:596-605. [PMID: 30802305 PMCID: PMC6594024 DOI: 10.1002/bjs.11094] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/12/2018] [Accepted: 11/26/2018] [Indexed: 01/03/2023]
Abstract
Background Patients with a pathological complete response (pCR) after neoadjuvant chemoradiotherapy (nCRT) for oesophageal cancer may benefit from non‐surgical management. The aim of this study was to determine the diagnostic performance of visual response assessment of the primary tumour after nCRT on T2‐weighted (T2W) and diffusion‐weighted (DW) MRI. Methods Patients with locally advanced oesophageal cancer who underwent T2W‐ and DW‐MRI (1·5 T) before and after nCRT in two hospitals, between July 2013 and September 2017, were included in this prospective study. Three radiologists evaluated T2W images retrospectively using a five‐point score for the assessment of residual tumour in a blinded manner and immediately rescored after adding DW‐MRI. Histopathology of the resection specimen was used as the reference standard; ypT0 represented a pCR. Sensitivity, specificity, area under the receiver operating characteristic (ROC) curve (AUC) and interobserver agreement were calculated. Results Twelve of 51 patients (24 per cent) had a pCR. The sensitivity and specificity of T2W‐MRI for detection of residual tumour ranged from 90 to 100 and 8 to 25 per cent respectively. Respective values for T2W + DW‐MRI were 90–97 and 42–50 per cent. AUCs for the three readers were 0·65, 0·66 and 0·68 on T2W‐MRI, and 0·71, 0·70 and 0·70 on T2W + DW‐MRI (P = 0·441, P = 0·611 and P = 0·828 for readers 1, 2 and 3 respectively). The κ value for interobserver agreement improved from 0·24–0·55 on T2W‐MRI to 0·55–0·71 with DW‐MRI. Conclusion Preoperative assessment of residual tumour on MRI after nCRT for oesophageal cancer is feasible with high sensitivity, reflecting a low chance of missing residual tumour. However, the specificity was low; this results in overstaging of complete responders as having residual tumour and, consequently, overtreatment.
Collapse
Affiliation(s)
- S E Vollenbrock
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - F E M Voncken
- Department of Radiation Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - J M van Dieren
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - D M J Lambregts
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - M Maas
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - G J Meijer
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - L Goense
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - S Mook
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - K J Hartemink
- Department of Surgery, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - P Snaebjornsson
- Department of Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - L C Ter Beek
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - M Verheij
- Department of Radiation Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - B M P Aleman
- Department of Radiation Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - R G H Beets-Tan
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - A Bartels-Rutten
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| |
Collapse
|
44
|
Zhang YH, Fischer MA, Lehmann H, Johnsson Å, Rouvelas I, Herlin G, Lundell L, Brismar TB. Computed tomography volumetry of esophageal cancer - the role of semiautomatic assessment. BMC Med Imaging 2019; 19:17. [PMID: 30767773 PMCID: PMC6377716 DOI: 10.1186/s12880-019-0317-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/28/2019] [Indexed: 01/16/2023] Open
Abstract
Background The clinical and research value of Computed Tomography (CT) volumetry of esophageal cancer tumor size remains controversial. Development in CT technique and image analysis has made CT volumetry less cumbersome and it has gained renewed attention. The aim of this study was to assess esophageal tumor volume by semi-automatic measurements as compared to manual. Methods A total of 23 esophageal cancer patients (median age 65, range 51–71), undergoing CT in the portal-venous phase for tumor staging, were retrospectively included between 2007 and 2012. One radiology resident and one consultant radiologist measured the tumor volume by semiautomatic segmentation and manual segmentation. Reproducibility of the respective measurements was assessed by intraclass correlation coefficients (ICC) and by average deviation from mean. Results Mean tumor volume was 46 ml (range 5-137 ml) using manual segmentation and 42 ml (range 3-111 ml) using semiautomatic segmentation. Semiautomatic measurement provided better inter-observer agreement than traditional manual segmentation. The ICC was significantly higher for semiautomatic segmentation in comparison to manual segmentation (0.86, 0.56, p < 0.01). The average absolute percentage difference from mean was reduced from 24 to 14% (p < 0.001) when using semiautomatic segmentation. Conclusions Semiautomatic analysis outperforms manual analysis for assessment of esophageal tumor volume, improving reproducibility.
Collapse
Affiliation(s)
- Yi-Hua Zhang
- Department of Diagnostic Radiology and Karolinska Institutet, Karolinska University Hospital, CLINTEC, Stockholm, Sweden. .,Division of Medical Imaging and Technology, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Huddinge, 141 86, Stockholm, Sweden.
| | - Michael A Fischer
- Department of Diagnostic Radiology and Karolinska Institutet, Karolinska University Hospital, CLINTEC, Stockholm, Sweden
| | - Henrik Lehmann
- Department of Diagnostic Radiology and Karolinska Institutet, Karolinska University Hospital, CLINTEC, Stockholm, Sweden
| | - Åse Johnsson
- Department of Radiology, Institute of Clinical Sciences, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ioannis Rouvelas
- Department of Surgery, Centre for Digestive Diseases and Karolinska Institutet, Karolinska University Hospital, CLINTEC, Stockholm, Sweden
| | - Gunnar Herlin
- Department of Diagnostic Radiology and Karolinska Institutet, Karolinska University Hospital, CLINTEC, Stockholm, Sweden
| | - Lars Lundell
- Department of Surgery, Centre for Digestive Diseases and Karolinska Institutet, Karolinska University Hospital, CLINTEC, Stockholm, Sweden
| | - Torkel B Brismar
- Department of Diagnostic Radiology and Karolinska Institutet, Karolinska University Hospital, CLINTEC, Stockholm, Sweden
| |
Collapse
|
45
|
Zhu Y, Fu L, Jing W, Guo D, Chen Y, Kong L, Yu J. The value of magnetic resonance imaging in esophageal carcinoma: Tool or toy? Asia Pac J Clin Oncol 2019; 15:101-107. [PMID: 30609237 DOI: 10.1111/ajco.13112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/20/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Ying Zhu
- Weifang Medical University Weifang Shandong Province China
- Department of Radiation OncologyShandong Cancer Hospital Affiliated to Shandong University Jinan Shandong Province China
| | - Lei Fu
- Department of Radiation OncologyShandong Cancer Hospital Affiliated to Shandong University Jinan Shandong Province China
| | - Wang Jing
- Department of Radiation OncologyShandong Cancer Hospital Affiliated to Shandong University Jinan Shandong Province China
| | - Dong Guo
- Weifang Medical University Weifang Shandong Province China
| | - Yan Chen
- People's Hospital of Juan Cheng County Shandong Province China
| | - Li Kong
- Department of Radiation OncologyShandong Cancer Hospital Affiliated to Shandong University Jinan Shandong Province China
- Shandong Academy of Medical Sciences Jinan Shandong Province China
| | - Jinming Yu
- Department of Radiation OncologyShandong Cancer Hospital Affiliated to Shandong University Jinan Shandong Province China
- Shandong Academy of Medical Sciences Jinan Shandong Province China
| |
Collapse
|
46
|
Novel imaging techniques in staging oesophageal cancer. Best Pract Res Clin Gastroenterol 2018; 36-37:17-25. [PMID: 30551852 DOI: 10.1016/j.bpg.2018.11.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 11/19/2018] [Indexed: 01/31/2023]
Abstract
The survival of oesophageal cancer is poor as most patients present with advanced disease. Radiological staging of oesophageal cancer is complex but is fundamental to clinical management. Accurate staging investigations are vitally important to guide treatment decisions and optimise patient outcomes. A combination of baseline computed tomography (CT), endoscopic ultrasound (EUS) and positron emission tomography (PET) are currently used for initial treatment decisions. The potential value of these imaging modalities to re-stage disease, monitor response and alter treatment is currently being investigated. This review presents an essential update on the accuracy of oesophageal cancer staging investigations, their use in re-staging after neo-adjuvant therapy and introduces evolving imaging techniques, including novel biomarkers that have clinical potential in oesophageal cancer.
Collapse
|
47
|
Borggreve AS, Mook S, Verheij M, Mul VEM, Bergman JJ, Bartels-Rutten A, Ter Beek LC, Beets-Tan RGH, Bennink RJ, van Berge Henegouwen MI, Brosens LAA, Defize IL, van Dieren JM, Dijkstra H, van Hillegersberg R, Hulshof MC, van Laarhoven HWM, Lam MGEH, van Lier ALHMW, Muijs CT, Nagengast WB, Nederveen AJ, Noordzij W, Plukker JTM, van Rossum PSN, Ruurda JP, van Sandick JW, Weusten BLAM, Voncken FEM, Yakar D, Meijer GJ. Preoperative image-guided identification of response to neoadjuvant chemoradiotherapy in esophageal cancer (PRIDE): a multicenter observational study. BMC Cancer 2018; 18:1006. [PMID: 30342494 PMCID: PMC6195948 DOI: 10.1186/s12885-018-4892-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/03/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Nearly one third of patients undergoing neoadjuvant chemoradiotherapy (nCRT) for locally advanced esophageal cancer have a pathologic complete response (pCR) of the primary tumor upon histopathological evaluation of the resection specimen. The primary aim of this study is to develop a model that predicts the probability of pCR to nCRT in esophageal cancer, based on diffusion-weighted magnetic resonance imaging (DW-MRI), dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and 18F-fluorodeoxyglucose positron emission tomography with computed tomography (18F-FDG PET-CT). Accurate response prediction could lead to a patient-tailored approach with omission of surgery in the future in case of predicted pCR or additional neoadjuvant treatment in case of non-pCR. METHODS The PRIDE study is a prospective, single arm, observational multicenter study designed to develop a multimodal prediction model for histopathological response to nCRT for esophageal cancer. A total of 200 patients with locally advanced esophageal cancer - of which at least 130 patients with adenocarcinoma and at least 61 patients with squamous cell carcinoma - scheduled to receive nCRT followed by esophagectomy will be included. The primary modalities to be incorporated in the prediction model are quantitative parameters derived from MRI and 18F-FDG PET-CT scans, which will be acquired at fixed intervals before, during and after nCRT. Secondary modalities include blood samples for analysis of the presence of circulating tumor DNA (ctDNA) at 3 time-points (before, during and after nCRT), and an endoscopy with (random) bite-on-bite biopsies of the primary tumor site and other suspected lesions in the esophagus as well as an endoscopic ultrasonography (EUS) with fine needle aspiration of suspected lymph nodes after finishing nCRT. The main study endpoint is the performance of the model for pCR prediction. Secondary endpoints include progression-free and overall survival. DISCUSSION If the multimodal PRIDE concept provides high predictive performance for pCR, the results of this study will play an important role in accurate identification of esophageal cancer patients with a pCR to nCRT. These patients might benefit from a patient-tailored approach with omission of surgery in the future. Vice versa, patients with non-pCR might benefit from additional neoadjuvant treatment, or ineffective therapy could be stopped. TRIAL REGISTRATION The article reports on a health care intervention on human participants and was prospectively registered on March 22, 2018 under ClinicalTrials.gov Identifier: NCT03474341 .
Collapse
Affiliation(s)
- A S Borggreve
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands. .,Department of Surgical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| | - S Mook
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - M Verheij
- Department of Radiation Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - V E M Mul
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GW, Groningen, The Netherlands
| | - J J Bergman
- Department of Gastroenterology, Amsterdam University Medical Centers, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - A Bartels-Rutten
- Department of Radiology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - L C Ter Beek
- Department of Radiology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - R G H Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - R J Bennink
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - M I van Berge Henegouwen
- Department of Surgical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - L A A Brosens
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - I L Defize
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.,Department of Surgical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - J M van Dieren
- Department of Gastroenterology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - H Dijkstra
- Department of Radiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GW, Groningen, The Netherlands
| | - R van Hillegersberg
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - M C Hulshof
- Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - H W M van Laarhoven
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - M G E H Lam
- Department of Nuclear Medicine, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - A L H M W van Lier
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - C T Muijs
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GW, Groningen, The Netherlands
| | - W B Nagengast
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GW, Groningen, The Netherlands
| | - A J Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - W Noordzij
- Department of Nuclear Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GW, Groningen, The Netherlands
| | - J T M Plukker
- Department of Surgical Oncology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GW, Groningen, The Netherlands
| | - P S N van Rossum
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - J P Ruurda
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - J W van Sandick
- Department of Surgical Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - B L A M Weusten
- Department of Gastroenterology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - F E M Voncken
- Department of Radiation Oncology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - D Yakar
- Department of Radiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GW, Groningen, The Netherlands
| | - G J Meijer
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | | |
Collapse
|
48
|
Giganti F, Tang L, Baba H. Gastric cancer and imaging biomarkers: Part 1 - a critical review of DW-MRI and CE-MDCT findings. Eur Radiol 2018; 29:1743-1753. [PMID: 30280246 PMCID: PMC6420485 DOI: 10.1007/s00330-018-5732-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 08/13/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022]
Abstract
Abstract The current standard of care for gastric cancer imaging includes heterogeneity in image acquisition techniques and qualitative image interpretation. In addition to qualitative assessment, several imaging techniques, including diffusion-weighted magnetic resonance imaging (DW-MRI), contrast-enhanced multidetector computed tomography (CE-MDCT), dynamic-contrast enhanced MRI and 18F-fluorodeoxyglucose positron emission tomography, can allow quantitative analysis. However, so far there is no consensus regarding the application of functional imaging in the management of gastric cancer. The aim of this article is to specifically review two promising biomarkers for gastric cancer with reasonable spatial resolution: the apparent diffusion coefficient (ADC) from DW-MRI and textural features from CE-MDCT. We searched MEDLINE/ PubMed for manuscripts published from inception to 6 February 2018. Initially, we searched for (gastric cancer OR gastric tumour) AND diffusion weighted magnetic resonance imaging. Then, we searched for (gastric cancer OR gastric tumour) AND texture analysis AND computed tomography. We collated the results from the studies related to this query. There is evidence that: (1) the ADC is a promising biomarker for the evaluation of the aggressiveness (T and N stage), treatment response and prognosis of gastric cancer; (2) textural features are related to the degree of differentiation, Lauren classification, treatment response and prognosis of gastric cancer. We conclude that these imaging biomarkers hold promise as effective additional tools in the diagnostic pathway of gastric cancer and may facilitate the multidisciplinary work between the radiologist and clinician, and across different institutions, to provide a greater biological understanding of gastric cancer. Key Points • Quantitative imaging is the extraction of quantifiable features from medical images for the assessment of normal or pathological conditions and represents a promising area for gastric cancer. • Quantitative analysis from CE-MDCT and DW-MRI allows the extrapolation of multiple imaging biomarkers. • ADC from DW-MRI and CE- MDCT-based texture features are non-invasive, quantitative imaging biomarkers that hold promise in the evaluation of the aggressiveness, treatment response and prognosis of gastric cancer.
Collapse
Affiliation(s)
- Francesco Giganti
- Department of Radiology, University College London Hospital NHS Foundation Trust, London, UK. .,Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, 3rd Floor, Charles Bell House, 43-45 Foley St, London, W1W 7TS, UK.
| | - Lei Tang
- Department of Radiology, Peking University Cancer Hospital, Beijing, China
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
49
|
Li F, Wang H, Hou J, Tang J, Lu Q, Wang L, Yu X. Utility of intravoxel incoherent motion diffusion-weighted imaging in predicting early response to concurrent chemoradiotherapy in oesophageal squamous cell carcinoma. Clin Radiol 2018; 73:756.e17-756.e26. [DOI: 10.1016/j.crad.2018.03.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 03/28/2018] [Indexed: 02/07/2023]
|
50
|
Borggreve AS, Kingma BF, Domrachev SA, Koshkin MA, Ruurda JP, Hillegersberg R, Takeda FR, Goense L. Surgical treatment of esophageal cancer in the era of multimodality management. Ann N Y Acad Sci 2018; 1434:192-209. [DOI: 10.1111/nyas.13677] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/05/2018] [Accepted: 02/23/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Alicia S. Borggreve
- Department of Surgery, University Medical Center UtrechtUtrecht University Utrecht the Netherlands
- Moscow Clinical Scientific Center Moscow Russia
| | - B. Feike Kingma
- Department of Surgery, University Medical Center UtrechtUtrecht University Utrecht the Netherlands
| | | | | | - Jelle P. Ruurda
- Department of Surgery, University Medical Center UtrechtUtrecht University Utrecht the Netherlands
| | - Richard Hillegersberg
- Department of Surgery, University Medical Center UtrechtUtrecht University Utrecht the Netherlands
| | - Flavio R. Takeda
- Sao Paulo Institute of CancerUniversity of Sao Paulo School of Medicine Sao Paulo Brazil
| | - Lucas Goense
- Department of Surgery, University Medical Center UtrechtUtrecht University Utrecht the Netherlands
| |
Collapse
|