1
|
Seo JM, Baek SY, Jeong WK, Song KD. Correction of stomach cancer CT attenuation values for variations due to differences in CT imaging conditions through repeated CT scans. PLoS One 2025; 20:e0321085. [PMID: 40273222 PMCID: PMC12021269 DOI: 10.1371/journal.pone.0321085] [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: 11/04/2024] [Accepted: 03/01/2025] [Indexed: 04/26/2025] Open
Abstract
PURPOSE To develop methods for correcting variations in CT attenuation values of advanced gastric cancer (AGC) due to differences in CT imaging conditions using repeated pre-treatment CT scans. METHODS A total 211 patients (146 men) with AGC who underwent pre-treatment CT twice were included in this retrospective study. The Pearson correlation between the difference in tumor attenuation values measured on both CT scans and the difference in attenuation values of other organs was analyzed. A formula to correct tumor CT attenuation values was developed using univariate linear regression analysis. RESULTS The Pearson correlation coefficient was the highest between the difference in tumor attenuation values and that of the main portal vein (MPV) attenuation values (0.86, P <.01). The formula to correct tumor attenuation values was as follows: calculated tumor attenuation value on CT scan 2 = tumor attenuation value on CT scan 1 - (-3.5 + 0.4 x (MPV attenuation value on CT scan 1 - MPV attenuation value on CT scan 2)). The mean difference between calculated and actual tumor attenuation values was 1.6 HU (SD, 8.7; range -22.5-24.72), with a Pearson correlation coefficient of 0.95 (P <.01). CONCLUSION Utilizing the attenuation value of the MPV allows for correction of variations in tumor attenuation values caused by different CT imaging conditions, enabling the prediction of reproducible tumor attenuation in patients with AGC. Future studies are needed to validate these findings and address the study's limitations, including its retrospective design and the absence of unenhanced CT data. ADVANCES IN KNOWLEDGE The attenuation value of the MPV can be used to predict reproducible tumor attenuation values in gastric cancer.
Collapse
Affiliation(s)
- Jeong Min Seo
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea
| | - Sun Young Baek
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea
- Biomedical Statistics Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Woo Kyoung Jeong
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea
| | - Kyoung Doo Song
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
2
|
Wang R, Kang W, Liu Z, Zheng Y, Sui H, Li L, Wang J, Xiang J, Peng X, Chen X, Zhu Z, Zhang J. Head-to-Head Comparison of [ 68Ga]Ga-NOTA-RM26 and [ 18F]FDG PET/CT in Patients with Gastrointestinal Stromal Tumors: A Prospective Study. J Nucl Med 2025; 66:201-206. [PMID: 39448271 DOI: 10.2967/jnumed.124.267810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common stromal tumors in the gastrointestinal tract. This study was designed to evaluate a gastrin-releasing peptide receptor antagonist PET tracer, [68Ga]Ga-NOTA-RM26, and compare it with [18F]FDG PET/CT in the assessment of patients with GISTs. Methods: With institutional review board approval and informed consent, 30 patients with suspected or proven GISTs based on abdominal CT or gastroscopy were recruited. All patients underwent [68Ga]Ga-NOTA-RM26 and [18F]FDG PET/CT scans. Pathology and other patient information were collected. Results: No radiopharmaceutical-related adverse events were observed in the patients. In total, 18 lesions in 16 patients were diagnosed as GIST, 3 patients were diagnosed with schwannoma, and 4 patients were diagnosed with leiomyoma. In 18 GISTs, the mean SUVmax of [68Ga]Ga-NOTA-RM26 PET was significantly higher than that of [18F]FDG PET (17.07 ± 19.57 vs. 2.28 ± 1.65; P < 0.01), and [68Ga]Ga-NOTA-RM26 PET/CT had a higher tumor detection rate than did [18F]FDG PET/CT (88.9% vs. 50%; P < 0.01). The uptake of [68Ga]Ga-NOTA-RM26 in GISTs was significantly higher than that in 2 other benign tumors (leiomyoma or schwannoma) (17.07 ± 19.57 vs. 4.23 ± 1.77; P = 0.014). With the SUVmax cutoff value of 6.0, the sensitivity of 68Ga-NOTA-RM26 PET/CT in diagnosing GISTs is 72% and the specificity is 85.7%. Conclusion: Compared with [18F]FDG PET/CT, [68Ga]Ga-NOTA-RM26 PET/CT is a promising and effective imaging modality for the detection of GISTs.
Collapse
Affiliation(s)
- Rongxi Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, Helios, Singapore
| | - Weiming Kang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhen Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Yumin Zheng
- Department of Nuclear Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Huimin Sui
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Linlin Li
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jiarou Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jialin Xiang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xingtong Peng
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, Helios, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Departments Chemical and Biomolecular Engineering, and Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore; and
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Proteos, Singapore
| | - Zhaohui Zhu
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China;
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore;
- Theranostics Center of Excellence, Yong Loo Lin School of Medicine, National University of Singapore, Helios, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
3
|
Tripathi M, Purwar R, Sinha R, Singh P, Pandey M. Gastrointestinal Stromal Tumor of the Rectum: Report of a Case With Long-Term Imatinib Treatment. Cureus 2024; 16:e74269. [PMID: 39717333 PMCID: PMC11663722 DOI: 10.7759/cureus.74269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
Gastrointestinal stromal tumors (GIST) are rare in the rectum. These usually present with symptoms produced by compression of pelvic organs or bleeding. Surgery is the treatment of choice, however, at times the surgery can be mutilating and organ preservation may not be possible. In such cases imatinib can be used. We report a case of rectal GIST that presented with urinary obstruction and obstipation treated with long-term imatinib with very good response and relief of symptoms, which prompted the patient to refuse surgery and resulted in organ preservation.
Collapse
Affiliation(s)
- Madhumita Tripathi
- Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| | - Roli Purwar
- Obstetrics and Gynaecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| | - Richie Sinha
- Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| | - Pooja Singh
- Oncological Research, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| | - Manoj Pandey
- Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, IND
| |
Collapse
|
4
|
Beecroft JR, Brar S, Feng X, Hamilton T, Han-Lee C, Henning JW, Josephy PD, Khalili K, Ko YJ, Lemieux C, Liu DM, MacDonald DB, Noujaim J, Pollett A, Salawu A, Saleh R, Smrke A, Warren BE, Zbuk K, Razak AA. Pan-Canadian consensus recommendations for GIST management in high- and low-throughput centres across Canada. Ther Adv Med Oncol 2024; 16:17588359241266179. [PMID: 39386314 PMCID: PMC11461906 DOI: 10.1177/17588359241266179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/18/2024] [Indexed: 10/12/2024] Open
Abstract
Gastrointestinal stromal tumours (GISTs) are mesenchymal tumours that originate from the interstitial cells of Cajal. GISTs are mainly driven by gain-of-function mutations in receptor tyrosine kinase or platelet-derived growth factor receptor alpha. Surgical resection is the only curative treatment for localized tumours and all currently approved medical GIST treatments are based on orally available tyrosine kinase inhibitors. Recent discoveries in the molecular and clinical features of GISTs have greatly impacted GIST management. Due to the provincially rather than nationally administered Canadian healthcare system, there have been inconsistencies in the treatment of GISTs across the country. Therefore, guidance on the latest knowledge, clinical management and treatment of GIST is needed to standardize the approach to GIST management nationwide. To establish pan-Canadian guidance, provide up-to-date data and harmonize the clinical practice of GIST management in high- and low-throughput centres across Canada; a panel of 20 physicians with extensive clinical experience in GIST management reviewed relevant literature. This included radiologists, pathologists, interventional radiologists, surgeons and medical oncologists across Canada. The structured literature focused on seven key domains: molecular profiling, radiological techniques/reporting, targeted localized therapy, intricacies of systemic treatments, emerging tests, multidisciplinary care and patient advocacy. This literature review, along with clinical expertise and opinion, was used to develop this concise and clinically relevant consensus paper to harmonize the knowledge and clinical practice on GIST management across Canada. The content presented here will help guide healthcare providers, especially in Canada, in terms of approaching and managing GIST.
Collapse
Affiliation(s)
- J. Robert Beecroft
- Division of Interventional Radiology, Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital, Toronto, ON, Canada
| | - Savtaj Brar
- Department of Surgery, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Xiaolan Feng
- Division of Medical Oncology, Tom Baker Cancer Center, Calgary, AB, Canada
| | - Trevor Hamilton
- Department of Surgery, BC Cancer, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Cheng Han-Lee
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, AB, Canada
| | - Jan-Willem Henning
- Department of Oncology, Tom Baker Cancer Centre, Cuming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Korosh Khalili
- Department of Medical Imaging, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Yoo-Joung Ko
- Department of Medicine, St. Michael’s Hospital, Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Christopher Lemieux
- Division of Hematology and Medical Oncology, Centre Hospitalier Universitaire de Québec, Université Laval, Quebec City, QC, Canada
| | - David M. Liu
- Department of Radiology, University of British Columbia, School of Biomedical Engineering, Vancouver, BC, Canada
- Department of Interventional Radiology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - D. Blair MacDonald
- Department of Medical Radiology, The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Jonathan Noujaim
- Division of Medical Oncology, Hôpital Maisonneuve-Rosemont, University of Montreal, Montreal, QC, Canada
| | - Aaron Pollett
- Pathology and Laboratory Medicine, Division of Diagnostic Medical Genetics, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Abdulazeez Salawu
- Division of Medical Oncology, Princess Margaret Cancer Centre, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Ramy Saleh
- Division of Medical Oncology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Alannah Smrke
- Division of Medical Oncology, BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - Blair E. Warren
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Kevin Zbuk
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| | - Albiruni Abdul Razak
- Division of Medical Oncology, Princess Margaret Cancer Centre, Mount Sinai Hospital, University of Toronto, 610 University Ave., Toronto, ON M2G 2M9, Canada
| |
Collapse
|
5
|
Bodei L, Jayaprakasam VS, Ying Wong BZ, Aparici CM. Neuroendocrine Tumors: Beta Labeled Radiopeptides. PET Clin 2024; 19:e1-e11. [PMID: 40199623 DOI: 10.1016/j.cpet.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Peptide receptor radionuclide therapy (PRRT) consists of administrating a radiolabeled octreotide derivative that targets somatostatin receptors present on the cell membrane of neuroendocrine tumor cells. Although PRRT was initially performed with 90Y-peptides, currently 177Lu-peptides represent the predominant form of treatment. PRRT results in significant tumor and symptomatic control in patients. Like with other available systemic therapies, responses are relatively short-lived. Several new peptides and strategies to improve the efficacy and tolerability of PRRT have been proposed. A critical step is individualizing treatments based on specific dosimetric estimates for the tumor and normal organs, and determining tissue radiosensitivity.
Collapse
Affiliation(s)
- Lisa Bodei
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Cornell Medical College of Cornell University, New York, NY, USA.
| | - Vetri Sudar Jayaprakasam
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | | | - Carina Mari Aparici
- Division of Nuclear Medicine, Department of Radiology, University of Stanford, Stanford, CA, USA
| |
Collapse
|
6
|
Li J, Huang S, Zhu H, Shou C, Lin T, Yin X, Zhu Q, Sun D, Li X, Shen L, Li J, Kou Y, Zhou Y, Zhang B, Qian H, Yu J, Zhou Y, Tang L, Zhang X. CT features combined with RECIST 1.1 criteria improve progression assessments of sunitinib-treated gastrointestinal stromal tumors. Eur Radiol 2024; 34:3659-3670. [PMID: 37947835 DOI: 10.1007/s00330-023-10383-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/14/2023] [Accepted: 09/07/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVES To explore the auxiliary value of combining CT features with existing response evaluation criteria in the prediction of progressive disease (PD) in gastrointestinal stromal tumors (GIST) patients treated with sunitinib. MATERIAL AND METHODS Eighty-one patients with GISTs who received sunitinib were included in this retrospective multicenter study and divided into training and external validation cohorts. Progression at six months was determined as a reference standard. The predictive performance of the RECIST 1.1 and Choi criteria was compared. CT features at baseline and the first follow-up were analyzed. Logistic regression analyses were used to determine the most significant predictors and develop modified criteria. RESULTS A total of 216 lesions showed a good response and 107 showed a poor response in 81 patients. The RECIST 1.1 criteria performed better than the Choi criteria in predicting progression (AUC, 0.75 vs. 0.69, p = 0.04). The expanded/intensified high-enhancement area, blurred tumor-tissue interface, and progressive enlarged vessels feeding or draining the mass (EVFDM) differed significantly between lesions with good and poor responses in the training cohort (p = 0.001, 0.003, and 0.000, respectively). Multivariate analysis revealed that the expanded/intensified high-enhancement area (p = 0.001), progressive EVFDM (p = 0.000), and RECIST PD (p = 0.000) were independent predictive factors. Modified RECIST (mRECIST) criteria were developed and showed significantly higher AUCs in the training and external validation cohorts than the RECIST 1.1 criteria (training: 0.81 vs. 0.73, p = 0.002; validation: 0.82 vs. 0.77, p = 0.04). CONCLUSION The mRECIST criteria, combining CT features with the RECIST 1.1 criteria, demonstrated superior performance in the prediction of early progression in GIST patients receiving sunitinib. CLINICAL RELEVANCE STATEMENT The mRECIST criteria, which combine CT features with the RECIST 1.1 criteria, may facilitate the early detection of progressive disease in GIST patients treated with sunitinib, thereby potentially guiding the timely switch to late-line medications or combination with surgical excision. KEY POINTS • The RECIST 1.1 criteria outperformed the Choi criteria in identifying progression of GISTs in patients treated with sunitinib. • GISTs displayed different morphologic features on CT depending on how they responded to sunitinib. • Combining CT morphologic features with the RECIST 1.1 criteria allowed for the prompt and accurate identification of progressing GIST lesions.
Collapse
Affiliation(s)
- Jiazheng Li
- Department of Radiology, Peking University Cancer Hospital and Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing, China
| | - Shaoqing Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hui Zhu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chunhui Shou
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianyu Lin
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaonan Yin
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Quanjian Zhu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Dongmei Sun
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoting Li
- Department of Radiology, Peking University Cancer Hospital and Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing, China
| | - Jian Li
- Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing, China.
| | - Youwei Kou
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yongjian Zhou
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Bo Zhang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China.
| | - Haoran Qian
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jiren Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Ye Zhou
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Lei Tang
- Department of Radiology, Peking University Cancer Hospital and Institute, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing, China.
| | - Xinhua Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
7
|
Guglielmo P, Alongi P, Baratto L, Abenavoli E, Buschiazzo A, Celesti G, Conte M, Filice R, Gorica J, Jonghi-Lavarini L, Lanzafame H, Laudicella R, Librando M, Linguanti F, Mattana F, Miceli A, Olivari L, Piscopo L, Romagnolo C, Santo G, Vento A, Volpe F, Evangelista L. Head-to-Head Comparison of FDG and Radiolabeled FAPI PET: A Systematic Review of the Literature. Life (Basel) 2023; 13:1821. [PMID: 37763225 PMCID: PMC10533171 DOI: 10.3390/life13091821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
FAPI-based radiopharmaceuticals are a novel class of tracers, mainly used for PET imaging, which have demonstrated several advantages over [18F]FDG, especially in the case of low-grade or well-differentiated tumors. We conducted this systematic review to evaluate all the studies where a head-to-head comparison had been performed to explore the potential utility of FAPI tracers in clinical practice. FAPI-based radiopharmaceuticals have shown promising results globally, in particular in detecting peritoneal carcinomatosis, but studies with wider populations are needed to better understand all the advantages of these new radiopharmaceuticals.
Collapse
Affiliation(s)
| | - Pierpaolo Alongi
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico, Di Cristina e Benfratelli, 90127 Palermo, Italy;
| | - Lucia Baratto
- Department of Radiology, Division of Pediatric Radiology, Lucile Packard Children’s Hospital, Stanford University, Stanford, CA 94304, USA;
| | - Elisabetta Abenavoli
- Nuclear Medicine Unit, Careggi University Hospital, Largo Brambilla 3, 50134 Florence, Italy;
| | - Ambra Buschiazzo
- Nuclear Medicine Division, Santa Croce and Carle Hospital, 12100 Cuneo, Italy;
| | - Greta Celesti
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98122 Messina, Italy; (G.C.); (M.L.)
| | - Miriam Conte
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00185 Rome, Italy; (M.C.); (J.G.)
| | - Rossella Filice
- Unit of Nuclear Medicine, Biomedical Department of Internal and Specialist Medicine, University of Palermo, 90133 Palermo, Italy; (R.F.); (R.L.)
| | - Joana Gorica
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza University of Rome, 00185 Rome, Italy; (M.C.); (J.G.)
| | - Lorenzo Jonghi-Lavarini
- Department of Nuclear Medicine, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy;
| | - Helena Lanzafame
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, 45147 Essen, Germany;
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, 45147 Essen, Germany
| | - Riccardo Laudicella
- Unit of Nuclear Medicine, Biomedical Department of Internal and Specialist Medicine, University of Palermo, 90133 Palermo, Italy; (R.F.); (R.L.)
| | - Maria Librando
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98122 Messina, Italy; (G.C.); (M.L.)
| | - Flavia Linguanti
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | - Francesco Mattana
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCSS, 20141 Milan, Italy;
| | - Alberto Miceli
- Nuclear Medicine Unit, Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy;
| | - Laura Olivari
- Nuclear Medicine Unit, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar, Italy;
| | - Leandra Piscopo
- Department of Advanced Biomedical Sciences, University Federico II, 80138 Naples, Italy; (L.P.); (F.V.)
| | - Cinzia Romagnolo
- Department of Nuclear Medicine, “Ospedali Riuniti” Hospital, 60126 Ancona, Italy;
| | - Giulia Santo
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy;
| | - Antonio Vento
- Nuclear Medicine Department, ASP 1-P.O. San Giovanni di Dio, 92100 Agrigento, Italy;
| | - Fabio Volpe
- Department of Advanced Biomedical Sciences, University Federico II, 80138 Naples, Italy; (L.P.); (F.V.)
| | - Laura Evangelista
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy;
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| |
Collapse
|
8
|
Neuroendocrine Tumor Therapy Response Assessment. PET Clin 2023; 18:267-286. [PMID: 36858748 DOI: 10.1016/j.cpet.2022.11.009] [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: 03/03/2023]
Abstract
Peptide receptor radionuclide therapy has become an integral part of management of neuroendocrine neoplasms. Gallium-68- and lutetium-177-labeled somatostatin receptor analogues have replaced yttrium-90- and 111-indium-based tracers. Several newer targeted therapies are also being used in clinical and research settings. It is imperative to accurately evaluate the response to these agents. The characteristics of NENs and the response patterns of the targeted therapies make response assessment in this group challenging. This article provides an overview of the strengths and weaknesses of the various biomarkers available for response assessment.
Collapse
|
9
|
Jaleel J, Subudhi TK, Sagar S, Yadav R, Tripathi M, Bal C. Incidentally Detected Gastrointestinal Stromal Tumor in a Patient with Carcinoma Prostate: 68Ga-Prostate-Specific Membrane Antigen Versus 18F-Fluorodeoxyglucose Positron Emission Tomography/Computed Tomography. Indian J Nucl Med 2023; 38:67-68. [PMID: 37180184 PMCID: PMC10171767 DOI: 10.4103/ijnm.ijnm_105_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/08/2022] [Indexed: 02/25/2023] Open
Abstract
Uptake of 68Ga-prostate-specific membrane antigen (PSMA) in various nonprostatic tumors is well documented in the literature. We present a case of a gastrointestinal stromal tumor, incidentally detected on 68Ga-PSMA positron emission tomography/computed tomography imaging in a patient who underwent imaging for a suspected recurrence of carcinoma prostate.
Collapse
Affiliation(s)
- Jasim Jaleel
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Tumulu Kishan Subudhi
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Sambit Sagar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Rajni Yadav
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Madhavi Tripathi
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Chandrasekhar Bal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
10
|
Wu C, Zhang X, Zeng Y, Wu R, Ding L, Xia Y, Chen Z, Zhang X, Wang X. [ 18F]FAPI-42 PET/CT versus [ 18F]FDG PET/CT for imaging of recurrent or metastatic gastrointestinal stromal tumors. Eur J Nucl Med Mol Imaging 2022; 50:194-204. [PMID: 36040490 DOI: 10.1007/s00259-022-05955-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE PET has been important for monitoring recurrence and metastasis of Gastrointestinal Stromal Tumors (GISTs) and the selection of therapeutic strategies. A significant portion of GISTs lesions show negative FDG uptake and therefore calls for more tumor-specific imaging biomarkers. This study compared the imaging performance of [18F]FAPI-42 PET/CT and [18F]FDG PET/CT in recurrent or metastatic gastrointestinal stromal tumors (R/M GISTs). METHODS This study retrospectively included 35 patients with R/M GISTs who underwent both FAPI PET/CT and FDG PET/CT. The definite diagnosis was confirmed by pathology or follow-up drug treatment effects. The differences in detection rates and tumor-to-background SUVmax ratio (SUVTBR) of different locations between dual-tracer PET/CT were compared. Factors including tumor size, degree of enhancement, type of gene mutation, and targeted treatment potentially influencing the uptake of both tracers were assessed. The excised lesions (n = 3) underwent immunohistochemical staining to verify FAP expression in the tissue. RESULTS A total of 106 lesions in 35 patients were identified, out of which 38/106 (35.8%) lesions (FAPI + /FDG -) were additionally detected by FAPI PET/CT as compared to that by FDG, including 26 liver metastases, ten peritoneal metastases, one gastrointestinal recurrence, and one bone metastasis. The positive detection rate of FAPI PET/CT for recurrent or metastatic GISTs was higher than that of FDG (80.2% vs. 53.8%, P< 0.001), especially in liver metastases (87.5% vs. 33.3%, P< 0.001). Moreover, the SUVTBR of liver metastases of GISTs in FAPI PET/CT was higher than that in FDG [2.4 (0.3 to 11.2) vs. 0.9 (0.3 to 6.5), P< 0.001]. The longest diameter of tumors in the FDG-positive group was higher than that of the FDG-negative group (P= 0.005); still, it did not differ between the FAPI-positive group and the FAPI-negative group. No difference in the degree of enhancement was observed between both tracers' positive and negative groups. Besides, the SUVTBR of FDG but not FAPI differed significantly among various gene mutations (P< 0.001) as well as the targeted therapy and no targeted therapy groups (P= 0.001). FAP was expressed in R/M GISTs, and the uptake of FAPI corresponded to the level of FAP expression. CONCLUSION In conclusion, FAPI for imaging of R/M GISTs could be superior to FDG, specifically for liver metastases. The uptake of FAPI could reflect the level of FAP expression, and it was independent of tumor size, degree of enhancement, type of gene mutation, and targeted therapy as compared to FDG.
Collapse
Affiliation(s)
- Chunhui Wu
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, 58 2nd Zhongshan Road, Guangzhou, 510080, Guangdong Province, China
| | - Xinhua Zhang
- Center of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou , Guangdong Province, China
| | - Yu Zeng
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, 58 2nd Zhongshan Road, Guangzhou, 510080, Guangdong Province, China
| | - Renbo Wu
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, 58 2nd Zhongshan Road, Guangzhou, 510080, Guangdong Province, China
| | - Li Ding
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou , Guangdong Province, China
| | - Yanzhe Xia
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou , Guangdong Province, China
| | - Zhifeng Chen
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, 58 2nd Zhongshan Road, Guangzhou, 510080, Guangdong Province, China
| | - Xiangsong Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, 58 2nd Zhongshan Road, Guangzhou, 510080, Guangdong Province, China.
| | - Xiaoyan Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Sun Yat-Sen University, 58 2nd Zhongshan Road, Guangzhou, 510080, Guangdong Province, China.
| |
Collapse
|
11
|
Weeda YA, Kalisvaart GM, van Velden FHP, Gelderblom H, van der Molen AJ, Bovee JVMG, van der Hage JA, Grootjans W, de Geus-Oei LF. Early Prediction and Monitoring of Treatment Response in Gastrointestinal Stromal Tumors by Means of Imaging: A Systematic Review. Diagnostics (Basel) 2022; 12:2722. [PMID: 36359564 PMCID: PMC9689665 DOI: 10.3390/diagnostics12112722] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 05/11/2025] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are rare mesenchymal neoplasms. Tyrosine kinase inhibitor (TKI) therapy is currently part of routine clinical practice for unresectable and metastatic disease. It is important to assess the efficacy of TKI treatment at an early stage to optimize therapy strategies and eliminate futile ineffective treatment, side effects and unnecessary costs. This systematic review provides an overview of the imaging features obtained from contrast-enhanced (CE)-CT and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) PET/CT to predict and monitor TKI treatment response in GIST patients. PubMed, Web of Science, the Cochrane Library and Embase were systematically screened. Articles were considered eligible if quantitative outcome measures (area under the curve (AUC), correlations, sensitivity, specificity, accuracy) were used to evaluate the efficacy of imaging features for predicting and monitoring treatment response to various TKI treatments. The methodological quality of all articles was assessed using the Quality Assessment of Diagnostic Accuracy Studies, v2 (QUADAS-2) tool and modified versions of the Radiomics Quality Score (RQS). A total of 90 articles were included, of which 66 articles used baseline [18F]FDG-PET and CE-CT imaging features for response prediction. Generally, the presence of heterogeneous enhancement on baseline CE-CT imaging was considered predictive for high-risk GISTs, related to underlying neovascularization and necrosis of the tumor. The remaining articles discussed therapy monitoring. Clinically established imaging features, including changes in tumor size and density, were considered unfavorable monitoring criteria, leading to under- and overestimation of response. Furthermore, changes in glucose metabolism, as reflected by [18F]FDG-PET imaging features, preceded changes in tumor size and were more strongly correlated with tumor response. Although CE-CT and [18F]FDG-PET can aid in the prediction and monitoring in GIST patients, further research on cost-effectiveness is recommended.
Collapse
Affiliation(s)
- Ylva. A. Weeda
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Gijsbert M. Kalisvaart
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Aart. J. van der Molen
- Department of Radiology, Section of Abdominal Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Judith V. M. G. Bovee
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jos A. van der Hage
- Department of Surgical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Willem Grootjans
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
- Department of Radiation Science & Technology, Technical University of Delft, 2629 JB Delft, The Netherlands
| |
Collapse
|
12
|
Soft Tissue Sarcomas: The Role of Quantitative MRI in Treatment Response Evaluation. Acad Radiol 2022; 29:1065-1084. [PMID: 34548230 DOI: 10.1016/j.acra.2021.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/29/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Although curative surgery remains the cornerstone of the therapeutic strategy in patients with soft tissue sarcomas (STS), neoadjuvant radiotherapy and chemotherapy (NART and NACT, respectively) are increasingly used to improve operability, surgical margins and patient outcome. The best imaging modality for locoregional assessment of STS is MRI but these tumors are mostly evaluated in a qualitative manner. OBJECTIVE After an overview of the current standard of care regarding treatment for patients with locally advanced STS, this review aims to summarize the principles and limitations of (i) the current methods used to evaluate response to neoadjuvant treatment in clinical practice and clinical trials in STS (RECIST 1.1 and modified Choi criteria), (ii) quantitative MRI sequences (i.e., diffusion weighted imaging and dynamic contrast enhanced MRI), and (iii) texture analyses and (delta-) radiomics.
Collapse
|
13
|
Kyriazoglou A, Jespers P, Vandecavaye V, Mir O, Kasper B, Papai Z, Blay JY, Italiano A, Zaffaroni F, Litière S, Nzokirantevye A, Schöffski P. Exploratory analysis of tumor imaging in a Phase 2 trial with cabozantinib in gastrointestinal stromal tumor: lessons learned from study EORTC STBSG 1317 'CaboGIST'. Acta Oncol 2022; 61:663-668. [PMID: 35481400 DOI: 10.1080/0284186x.2022.2068967] [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: 11/01/2022]
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are malignant mesenchymal tumors arising in the gastrointestinal tract. Their systemic treatment is based on the use of tyrosine kinase inhibitors (TKIs) with imatinib, sunitinib, and regorafenib being the preferred agents. Assessment of tumor response to TKI treatment in GISTs is traditionally done according the Response Evaluation Criteria in Solid Tumors (RECIST), while Choi criteria have also been proposed as alternative tool assessing both volumetric and density changes on computer tomography (CT) scans. EORTC STBSG 1317 'CaboGIST' was a single-arm prospective Phase 2 trial which met its primary endpoint, as 60% of patients previously treated with imatinib and sunitinib were progression-free at 12 weeks (95% CI 45-74%) based on local RECIST assessment. MATERIALS AND METHODS We report here an exploratory analysis of local versus central RECIST version 1.1 assessment and a comparison of RECIST version 1.1 versus Choi criteria. RESULTS Comparisons between local and central RECIST version 1.1 at week 12 revealed discrepancies in 17/43 evaluable cases (39.5%). When comparing Choi with local and central RECIST version 1.1, discrepancies were observed in 27/43 (62.8%) and 21/43 (48.8%) cases, respectively. A total of 68% of evaluable patients were progression-free and alive at week 12 based on local RECIST, 84% according to central RECIST analysis and 81% when applying Choi criteria. Central assessment upgraded the treatment response both with RECIST version 1.1 and Choi. CONCLUSIONS The results of this exploratory analysis support the conclusion that cabozantinib is active in patients with metastatic or recurrent GIST after treatment with imatinib and sunitinib and confirm once again the limitations of RECIST to capture response to TKI in GIST, and the importance to include density changes in the response evaluation in this setting. Clinical trial number: EORTC 1317, NCT02216578.
Collapse
Affiliation(s)
- Anastasios Kyriazoglou
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
- Second Propaedeutic Department of Medicine, Attikon University Hospital, Athens, Greece
| | - Pieter Jespers
- European Organization for Research and Treatment of Cancer, Brussels, Belgium
| | - Vincent Vandecavaye
- Department of Radiology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Olivier Mir
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Bernd Kasper
- Mannheim University Medical Center, Mannheim Cancer Center (MCC), University of Heidelberg, Mannheim, Germany
| | | | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard & Université Claude Bernard Lyon I, Lyon, France
| | | | - Facundo Zaffaroni
- European Organization for Research and Treatment of Cancer, Brussels, Belgium
| | - Saskia Litière
- European Organization for Research and Treatment of Cancer, Brussels, Belgium
| | | | - Patrick Schöffski
- Second Propaedeutic Department of Medicine, Attikon University Hospital, Athens, Greece
| |
Collapse
|
14
|
Mesenchymal tumors of the stomach: radiologic and pathologic correlation. Abdom Radiol (NY) 2022; 47:1988-2003. [PMID: 35347384 DOI: 10.1007/s00261-022-03498-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/09/2022] [Accepted: 03/14/2022] [Indexed: 11/01/2022]
Abstract
Mesenchymal tumors of the stomach are uncommon, with gastrointestinal stromal tumor (GIST) being the most common among them. Majority of the tumors may arise from cells of Cajal, smooth muscle cells, neural cells, totipotent stem cells, adipocytes or fibroblasts. Imaging plays an important role not only in staging but also in characterizing these tumors. Many of these tumors have characteristic imaging features. GISTs usually present as large cavitating and necrotic tumors with exophytic component. Presence of fat tissue within the tumor suggests a lipoma or a teratoma, early phase hyperenhancement indicates glomus tumor and hemangioma, and delayed contrast enhancement is seen in schwannoma. Their differentiation from epithelial tumors like carcinoma and neuroendocrine tumors is often possible based on the location (mesenchymal tumors are intramural), spread, morphological appearance and enhancement patterns. However, overlapping features exist between these tumors with imaging often being only suggestive. A biopsy is necessary for a definitive diagnosis in many cases.
Collapse
|
15
|
Gupta M, Choudhury PS, Jain P, Sharma M, Koyyala VPB, Goyal S, Agarwal C, Jajodia A, Pasricha S, Sharma A, Batra U. Molecular Response Assessment with Immune Adaptive PERCIST in Lung Cancer Patients Treated with Nivolumab: Is It Better Than iRECIST? World J Nucl Med 2022; 21:34-43. [PMID: 35502277 PMCID: PMC9056126 DOI: 10.1055/s-0042-1744201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Aims
We compared the immune response evaluation criteria in solid tumors (iRECIST) with immune adaptive positron emission tomography response criteria in solid tumors (imPERCIST) in lung cancer patients treated with nivolumab.
Materials and Methods
Twenty lung cancer patients underwent fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT) scan at baseline (PET-0), after four cycles (PET-1) and six to eight cycles (PET-2) of nivolumab were included. Kappa coefficient (
k
) was derived to see the level of agreement in two response criteria. Progression-free survival (PFS) curves were computed by the Kaplan–Meier method and compared with the Log Rank test. Univariate and multivariate regression for the percentage change in the sum of diameters (SoD), standard uptake value maximum (SUVmax), sum of metabolic tumor volume (SoMTV), and sum of total lesion glycolysis (SoTLG) was computed. A
p
-value less than 0.05 was considered significant.
Results
Kappa coefficient showed a substantial level of agreement (k 0.769) in two response criteria. Mean PFS in partial response, stable disease, and progressive disease (PD) patients in iRECIST and imPERCIST was 27.3, 17.7, 4.2, and 23.3, 18.8, 3.8 months, respectively. The Kaplan–Meier method with the log rank test showed a significant difference in PFS on intracomparison within both criteria; however, it was not significant on intercomparison. On univariate analysis, the percentage change in SoD, SoMTV, SoTLG was significant. However, on multivariate analysis, only percentage change in SoD was a significant predictor.
Conclusions
We concluded that imPERCIST was equally effective as currently recommended criteria iRECIST for response evaluation of nivolumab in lung cancer patients.
Collapse
Affiliation(s)
- Manoj Gupta
- Department of Nuclear Medicine, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Partha S. Choudhury
- Department of Nuclear Medicine, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Parveen Jain
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Manish Sharma
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Venkata P. B. Koyyala
- Department of Medical Oncology, Homi Bhabha Cancer Hospital and Research Centre, Visakhapatnam, Andhra Pradesh, India
| | - Sumit Goyal
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Chaturbhuj Agarwal
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Ankush Jajodia
- Department of Radiology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Sunil Pasricha
- Department of Pathology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Anurag Sharma
- Department of Research, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Ullas Batra
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| |
Collapse
|
16
|
Hui C, Sum R. Hepatic GIST metastases: an illustrative case series. BJR Case Rep 2022; 8:20210166. [PMID: 36177254 PMCID: PMC9499438 DOI: 10.1259/bjrcr.20210166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 11/05/2022] Open
Abstract
Gastrointestinal stromal tumours (GISTs) are uncommon mesenchymal tumours affecting the gastrointestinal tract. The liver is one of the most common sites for metastatic disease from GISTs and may exhibit a variety of CT and MR imaging appearances. These imaging features can vary prior to and following treatment with tyrosine kinase inhibitors. We report on the spectrum of imaging appearances of hepatic GIST metastases on multiphase contrast CT imaging and hepatocyte-specific contrast enhanced MR. To our knowledge, there are no published series specifically focusing on the appearances of liver metastases from GISTs. An awareness of the protean appearances and pitfalls on CT and MRI of hepatic GIST metastases, prior to and at different times along the treatment pathway, will assist in early diagnosis of liver metastases, accurate assessment of tumour response and detection of recurrent metastatic disease.
Collapse
Affiliation(s)
- Cathryn Hui
- Department of Diagnostic Imaging, Monash Medical Centre, Melbourne, Australia
- Monash University, Melbourne, Australia
| | - Reuben Sum
- Department of Diagnostic Imaging, Monash Medical Centre, Melbourne, Australia
| |
Collapse
|
17
|
Foo T, Goldstein D, Segelov E, Shapiro J, Pavlakis N, Desai J, Yip D, Zalcberg J, Price TJ, Nagrial A, Chantrill L, Burge M, Karapetis CS, Tebbutt N, Roy AC. The Management of Unresectable, Advanced Gastrointestinal Stromal Tumours. Target Oncol 2022; 17:95-110. [PMID: 35290591 PMCID: PMC8995292 DOI: 10.1007/s11523-022-00869-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2022] [Indexed: 12/11/2022]
Abstract
Gastrointestinal stromal tumours (GISTs) are the most common gastrointestinal tract mesenchymal tumours. Tyrosine kinase inhibitors (TKIs) have transformed the management of advanced GIST. Imatinib was the first TKI to gain approval as management for patients with advanced GIST, establishing a new standard of care. Since then, as a result of several trials including the GRID and INVICTUS studies, we now have five lines of approved targeted therapy, including imatinib, sunitinib, regorafenib, ripretinib and avapritinib for the treatment of unresectable, advanced GISTs. In this review, the Australasian Gastrointestinal Trials Group (AGITG) provide an overview of the key trials that have changed clinical practice, discuss the molecular drivers of GISTs, the importance of molecular testing and directing therapy according to molecular targets, as well as future strategies in the management of advanced GISTs.
Collapse
Affiliation(s)
- Tiffany Foo
- Flinders Centre for Innovation in Cancer/Flinders University, Bedford Park, SA, 5042, Australia
| | - David Goldstein
- Department of Medical Oncology, Prince of Wales Hospital, University of NSW, Sydney, NSW, Australia
| | - Eva Segelov
- Department of Medical Oncology, School of Clinical Sciences, Monash University and Monash Health, Melbourne, VIC, Australia
| | - Jeremy Shapiro
- Cabrini Health, Monash University, Melbourne, VIC, Australia
| | - Nick Pavlakis
- Department of Medical Oncology, Royal North Shore Hospital, University of Sydney, Sydney, NSW, Australia
| | - Jayesh Desai
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Desmond Yip
- Department of Medical Oncology, Canberra Region Cancer Centre, The Canberra Hospital, Canberra, ACT, Australia
| | - John Zalcberg
- Alfred Health, Monash University, Melbourne, VIC, Australia
| | - Timothy J Price
- The Queen Elizabeth Hospital/University of Adelaide, Adelaide, SA, Australia
| | - Adnan Nagrial
- Department of Medical Oncology, Westmead and Blacktown Hospitals, University of Sydney, Sydney, NSW, Australia
| | - Lorraine Chantrill
- Department of Medical Oncology, Wollongong Hospital, Illawarra Shoalhaven Local Health District, Illawarra, NSW, Australia
| | - Matt Burge
- Department of Cancer Care Services, Royal Brisbane Hospital, University of Queensland, Herston, QLD, Australia
| | - Christos S Karapetis
- Flinders Centre for Innovation in Cancer/Flinders University, Bedford Park, SA, 5042, Australia
| | - Niall Tebbutt
- Department of Medical Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Austin Health, Heidelberg, VIC, Australia
| | - Amitesh C Roy
- Flinders Centre for Innovation in Cancer/Flinders University, Bedford Park, SA, 5042, Australia.
| |
Collapse
|
18
|
Karthikeyan M, Kolandasamy C, Naganath Babu OL. Malignant Gastrointestinal Stromal Tumor of Rectum: A Case Report and Review of Literature. Surg J (N Y) 2022; 8:e60-e64. [PMID: 35187230 PMCID: PMC8850004 DOI: 10.1055/s-0042-1742778] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 12/22/2021] [Indexed: 11/07/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are rare tumors of the gastrointestinal tract accounting for less than 1% of all gut tumors. GISTs occurring in the rectum are extremely rare and these usually present at an advanced stage compared with other sites. We report a case of a middle-aged female who presented with features of anemia and subacute obstruction due to a large rectal tumor and underwent abdominoperineal resection. The histopathological examination confirmed the diagnosis of high-grade malignant GIST with multiple lymph nodal metastasis. She was started on adjuvant imatinib therapy and is on follow-up without any evidence of recurrence. The authors conclude that GIST must be included in the differential diagnosis of a rectal tumor. Diagnosis is established by biopsy and immunohistochemistry studies. Surgical resection with histological negative margins is the standard curative treatment. Adjuvant targeted therapy can reduce long-term recurrence in high-risk cases.
Collapse
Affiliation(s)
- Mohan Karthikeyan
- Institute of Surgical Gastroenterology, Madras Medical College and Rajiv Gandhi Government General Hospital, Chennai, Tamil Nadu, India
| | - Chinnusamy Kolandasamy
- Institute of Surgical Gastroenterology, Madras Medical College and Rajiv Gandhi Government General Hospital, Chennai, Tamil Nadu, India
| | - Obla L Naganath Babu
- Institute of Surgical Gastroenterology, Madras Medical College and Rajiv Gandhi Government General Hospital, Chennai, Tamil Nadu, India
| |
Collapse
|
19
|
Fournier L, de Geus-Oei LF, Regge D, Oprea-Lager DE, D’Anastasi M, Bidaut L, Bäuerle T, Lopci E, Cappello G, Lecouvet F, Mayerhoefer M, Kunz WG, Verhoeff JJC, Caruso D, Smits M, Hoffmann RT, Gourtsoyianni S, Beets-Tan R, Neri E, deSouza NM, Deroose CM, Caramella C. Twenty Years On: RECIST as a Biomarker of Response in Solid Tumours an EORTC Imaging Group - ESOI Joint Paper. Front Oncol 2022; 11:800547. [PMID: 35083155 PMCID: PMC8784734 DOI: 10.3389/fonc.2021.800547] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
Response evaluation criteria in solid tumours (RECIST) v1.1 are currently the reference standard for evaluating efficacy of therapies in patients with solid tumours who are included in clinical trials, and they are widely used and accepted by regulatory agencies. This expert statement discusses the principles underlying RECIST, as well as their reproducibility and limitations. While the RECIST framework may not be perfect, the scientific bases for the anticancer drugs that have been approved using a RECIST-based surrogate endpoint remain valid. Importantly, changes in measurement have to meet thresholds defined by RECIST for response classification within thus partly circumventing the problems of measurement variability. The RECIST framework also applies to clinical patients in individual settings even though the relationship between tumour size changes and outcome from cohort studies is not necessarily translatable to individual cases. As reproducibility of RECIST measurements is impacted by reader experience, choice of target lesions and detection/interpretation of new lesions, it can result in patients changing response categories when measurements are near threshold values or if new lesions are missed or incorrectly interpreted. There are several situations where RECIST will fail to evaluate treatment-induced changes correctly; knowledge and understanding of these is crucial for correct interpretation. Also, some patterns of response/progression cannot be correctly documented by RECIST, particularly in relation to organ-site (e.g. bone without associated soft-tissue lesion) and treatment type (e.g. focal therapies). These require specialist reader experience and communication with oncologists to determine the actual impact of the therapy and best evaluation strategy. In such situations, alternative imaging markers for tumour response may be used but the sources of variability of individual imaging techniques need to be known and accounted for. Communication between imaging experts and oncologists regarding the level of confidence in a biomarker is essential for the correct interpretation of a biomarker and its application to clinical decision-making. Though measurement automation is desirable and potentially reduces the variability of results, associated technical difficulties must be overcome, and human adjudications may be required.
Collapse
Affiliation(s)
- Laure Fournier
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Université de Paris, Assistance Publique–Hôpitaux de Paris (AP-HP), Hopital europeen Georges Pompidou, Department of Radiology, Paris Cardiovascular Research Center (PARCC) Unité Mixte de Recherche (UMRS) 970, Institut national de la santé et de la recherche médicale (INSERM), Paris, France
| | - Lioe-Fee de Geus-Oei
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, Netherlands
| | - Daniele Regge
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Surgical Sciences, University of Turin, Turin, Italy
- Radiology Unit, Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia-Istituto Di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), Turin, Italy
| | - Daniela-Elena Oprea-Lager
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology & Nuclear Medicine, Cancer Centre Amsterdam, Amsterdam University Medical Centers [Vrije Universiteit (VU) University], Amsterdam, Netherlands
| | - Melvin D’Anastasi
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Medical Imaging Department, Mater Dei Hospital, University of Malta, Msida, Malta
| | - Luc Bidaut
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- College of Science, University of Lincoln, Lincoln, United Kingdom
| | - Tobias Bäuerle
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Egesta Lopci
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine Unit, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) – Humanitas Research Hospital, Milan, Italy
| | - Giovanni Cappello
- Department of Surgical Sciences, University of Turin, Turin, Italy
- Radiology Unit, Candiolo Cancer Institute, Fondazione del Piemonte per l’Oncologia-Istituto Di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), Turin, Italy
| | - Frederic Lecouvet
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Marius Mayerhoefer
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Wolfgang G. Kunz
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, University Hospital, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Joost J. C. Verhoeff
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Damiano Caruso
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Marion Smits
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, Netherlands
- Brain Tumour Centre, Erasmus Medical Centre (MC) Cancer Institute, Rotterdam, Netherlands
| | - Ralf-Thorsten Hoffmann
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Institute and Policlinic for Diagnostic and Interventional Radiology, University Hospital, Carl-Gustav-Carus Technical University Dresden, Dresden, Germany
| | - Sofia Gourtsoyianni
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, School of Medicine, National and Kapodistrian University of Athens, Areteion Hospital, Athens, Greece
| | - Regina Beets-Tan
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- School For Oncology and Developmental Biology (GROW) School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Emanuele Neri
- European Society of Oncologic Imaging (ESOI), European Society of Radiology, Vienna, Austria
- Diagnostic and Interventional Radiology, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Nandita M. deSouza
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, London, United Kingdom
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Quantitative Imaging Biomarkers Alliance, Radiological Society of North America, Oak Brook, IL, United States
| | - Christophe M. Deroose
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Caroline Caramella
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Radiology Department, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph Centre International des Cancers Thoraciques, Université Paris-Saclay, Le Plessis-Robinson, France
| |
Collapse
|
20
|
Gupta M, Choudhury P, Jain P, Sharma M, Babu Koyyala V, Goyal S, Agarwal C, Jajodia A, Pasricha S, Sharma A, Batra U. Molecular response assessment with immune adaptive positron emission tomography response criteria in solid tumors in lung cancer patients treated with nivolumab: Is it better than immune response evaluation criteria in solid tumors? World J Nucl Med 2022. [DOI: 10.4103/wjnm.wjnm_58_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
21
|
Ruchalski K, Dewan R, Sai V, McIntosh LJ, Braschi-Amirfarzan M. Imaging response assessment for oncology: An algorithmic approach. Eur J Radiol Open 2022; 9:100426. [PMID: 35693043 PMCID: PMC9184854 DOI: 10.1016/j.ejro.2022.100426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 01/04/2023] Open
Abstract
Treatment response assessment by imaging plays a vital role in evaluating changes in solid tumors during oncology therapeutic clinical trials. Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 is the reference standard imaging response criteria and provides details regarding image acquisition, image interpretation and categorical response classification. While RECIST 1.1 is applied for the majority of clinical trials in solid tumors, other criteria and modifications have been introduced when RECIST 1.1 outcomes may be incomplete. Available criteria beyond RECIST 1.1 can be explored in an algorithmic fashion dependent on imaging modality, tumor type and method of treatment. Positron Emission Tomography Response Criteria in Solid Tumors (PERCIST) is available for use with PET/CT. Modifications to RECIST 1.1 can be tumor specific, including mRECIST for hepatocellular carcinoma and mesothelioma. Choi criteria for gastrointestinal stromal tumors incorporate tumor density with alterations to categorical response thresholds. Prostate Cancer Working Group 3 (PCWG3) imaging criteria combine RECIST 1.1 findings with those of bone scans. In addition, multiple response criteria have been created to address atypical imaging responses in immunotherapy.
Collapse
|
22
|
Ishida T, Takahashi T, Nishida T, Ohnishi H, Tsuboyama T, Sato S, Nakahara Y, Miyazaki Y, Takeno A, Kurokawa Y, Saito T, Yamashita K, Tanaka K, Yamamoto K, Makino T, Yamasaki M, Motoori M, Kimura Y, Nakajima K, Eguchi H, Doki Y. New response evaluation criteria using early morphological change in imatinib treatment for patients with gastrointestinal stromal tumor. Gastric Cancer 2022; 25:218-225. [PMID: 34417657 DOI: 10.1007/s10120-021-01234-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 08/11/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND The introduction of molecularly targeted drugs, including imatinib, has greatly improved the prognosis of gastrointestinal stromal tumor (GIST), and based on the different response image, the methods of response evaluation have been established for GISTs. Furthrmore, the best response evaluation using them has been reported to be associated with progression-free survival (PFS) in imatinib treatment. However, since it is more important to predict the clinical outcomes of imatinib treatment in "early treatment phase", new predicting factor in earlier stage is desired to work out the whole strategy of each patient. Early morphological change (EMC) was previously reported as a predictive marker for molecularly targeted drugs in metastatic colorectal cancer. The purpose of the present study was to verify the efficacy of EMC in predicting the outcome in patients with GIST receiving imatinib at early evaluation. METHODS We retrospectively reviewed 66 patients. EMC in computed tomography (CT) image was evaluated, and the patients were categorized into two groups: active MR (morphological response) (+) group and active MR (-) group. We investigated the association between the presence of active MR and clinical outcomes. RESULTS Forty-five patients had active MR ( +). The median progression-free survival (PFS) in patients with/without active MR was 49/23 months (P = 0.0039). CONCLUSION The evaluation criteria based on EMC could be a sensitive method to predict the clinical outcome of imatinib treatment for patients with unresectable GIST.
Collapse
Affiliation(s)
- Tomo Ishida
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2-E2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2-E2, Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Toshirou Nishida
- Department of Surgery, Japan Community Health Care Organization Osaka Hospital, 4-2-78, Fukushima, Fukushima-ku, Osaka, Japan
| | - Hiromitsu Ohnishi
- Department of Radiology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takahiro Tsuboyama
- Department of Radiology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shinsuke Sato
- Department of Gastroenterological Surgery, Shizuoka General Hospital, 4-27-1, Kita Ando Aoi-ku, Shizuoka, Japan
| | - Yujiro Nakahara
- Department of Gastroenterological Surgery, Osaka Police Hospital, 10-31Kitayama-choTennouji-ku, Osaka, Japan
| | - Yasuhiro Miyazaki
- Department of Surgery, Osaka General Medical Center, 3-1-56, Bandai-Higashi, Sumiyoshi-ku, Osaka, Japan
| | - Atsushi Takeno
- Departmrent of Surgery, Kansai Rosai Hospital, 3-1-69, Inabaso, Amagasaki, Hyogo, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2-E2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takuro Saito
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2-E2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kotaro Yamashita
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2-E2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Koji Tanaka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2-E2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kazuyoshi Yamamoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2-E2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tomoki Makino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2-E2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Makoto Yamasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2-E2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masaaki Motoori
- Department of Surgery, Osaka General Medical Center, 3-1-56, Bandai-Higashi, Sumiyoshi-ku, Osaka, Japan
| | - Yutaka Kimura
- Department of Surgery, Kinki University Faculty of Medicine, 377-2, Ohno-Higashi, Sayama, Osaka, Japan
| | - Kiyokazu Nakajima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2-E2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2-E2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2-E2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
23
|
Van den Abbeele AD, Sakellis CG, George S. PET imaging of Gastrointestinal Stromal Tumors (GIST). Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00110-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
24
|
Jin L, Ma X, Zhang N, Zhang Q, Chen X, Zhang Z, Ding G, Yu H. Targeting Oncogenic miR-181a-2-3p Inhibits Growth and Suppresses Cisplatin Resistance of Gastric Cancer. Cancer Manag Res 2021; 13:8599-8609. [PMID: 34815714 PMCID: PMC8605795 DOI: 10.2147/cmar.s332713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/17/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose This study aimed to explore the value of miR-181a-2-3p in cisplatin (DDP) treatment effectiveness prediction, and to reveal the function underlying the reversal of DDP resistance in patients with gastric cancer (GC). Methods miRNA expression dataset of three DDP-resistant GC cell lines and their DDP-sensitive parental cell lines obtained from GEO DataSets and GenBank, and functional miRNAs were annotated by bioinformatics analyses. Serum specimens and tumor samples were collected from 91 GC patients for understanding of the interrelation between chemotherapy response and miRNA expression. RT-qPCR validated these miRNAs at the transcriptional level in both gastric cancer cells and 91 gastric cancer patients. The correlation between the miRNAs expression and clinical parameters of the patients were analyzed. Receiver operating characteristics (ROC) analysis has been utilized to assess the diagnostic performance. The MTT and colony formation assays were performed to assess cell proliferation. Flow cytometry was conducted to detect cell apoptosis. DDP-resistant GC cells and their DDP-sensitive parental cells were transfected with miRNA mimic or inhibitor vector to overexpress or downregulate miRNA expression. Results miR-181a-2-3p as a unique miRNA was found in the common differentially expressed-miRNAs (DE-miRNAs) after miRNA screening and validation from three DDP-resistant and DDP-sensitive gastric cancer cell lines. Clinical data analysis displayed that miR-181a-2-3p expression was apparently increased in larger tumor size (≥5 cm), higher T stage (T4), and chemotherapy resistance. miR-181a-2-3p (AUC=0.926, SE=0.028, 95% CI: 0.872-0.980, p< 0.0001) differentiated chemosensitive GC patients from chemoresistant GC patients. miR-181a-2-3p presented a higher level in gastric cancer, and could serve as a valid biomarker to predict the overall survival of GC patients. Upregulation of miR-181a-2-3p rendered the apoptosis-inducing and anti-proliferative effects of DDP, while downregulating it decreased these effects. Conclusion miR-181a-2-3p can function as a therapeutic target and a tumor biomarker. Targeting oncogenic miR-181a-2-3p inhibits growth and suppresses cisplatin resistance of gastric cancer.
Collapse
Affiliation(s)
- Lei Jin
- Department of Vascular Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xuemei Ma
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Nan Zhang
- Department of Radiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, People's Republic of China
| | - Qian Zhang
- Clinical Epidemiology and EBM Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xueming Chen
- Department of Vascular Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Guoqian Ding
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hongzhi Yu
- Department of Vascular Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
25
|
Li Q, Che F, Wei Y, Jiang HY, Zhang Y, Song B. Role of noninvasive imaging in the evaluation of intrahepatic cholangiocarcinoma: from diagnosis and prognosis to treatment response. Expert Rev Gastroenterol Hepatol 2021; 15:1267-1279. [PMID: 34452581 DOI: 10.1080/17474124.2021.1974294] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/26/2021] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Intrahepatic cholangiocarcinoma is the second most common liver cancer. Desmoplastic stroma may be revealed as distinctive histopathologic findings favoring intrahepatic cholangiocarcinoma. Meanwhile, a range of imaging manifestations is often accompanied with rich desmoplastic stroma in intrahepatic cholangiocarcinoma, which can indicate large bile duct ICC, and a higher level of cancer-associated fibroblasts with poor prognosis and weak treatment response. AREAS COVERED We provide a comprehensive review of current state-of-the-art and recent advances in the imaging evaluation for diagnosis, staging, prognosis and treatment response of intrahepatic cholangiocarcinoma. In addition, we discuss precursor lesions, cells of origin, molecular mutation, which would cause the different histological classification. Moreover, histological classification and tumor microenvironment, which are related to the proportion of desmoplastic stroma with many imaging manifestations, would be also discussed. EXPERT OPINION The diagnosis, prognosis, treatment response of intrahepatic cholangiocarcinoma may be revealed as the presence and the proportion of desmoplastic stroma with a range of imaging manifestations. With the utility of radiomics and artificial intelligence, imaging is helpful for ICC evaluation. Multicentre, large-scale, prospective studies with external validation are in need to develop comprehensive prediction models based on clinical data, imaging findings, genetic parameters, molecular, metabolic, and immune biomarkers.
Collapse
Affiliation(s)
- Qian Li
- Department of Radiology, Sichuan University West China Hospital, Chengdu, China
| | - Feng Che
- Department of Radiology, Sichuan University West China Hospital, Chengdu, China
| | - Yi Wei
- Department of Radiology, Sichuan University West China Hospital, Chengdu, China
| | - Han-Yu Jiang
- Department of Radiology, Sichuan University West China Hospital, Chengdu, China
| | - Yun Zhang
- Department of Radiology, Sichuan University West China Hospital, Chengdu, China
| | - Bin Song
- Department of Radiology, Sichuan University West China Hospital, Chengdu, China
| |
Collapse
|
26
|
A Multicenter Randomized Controlled Prospective Study to Assess Efficacy of Laparoscopic Electrochemotherapy in the Treatment of Locally Advanced Pancreatic Cancer. J Clin Med 2021; 10:jcm10174011. [PMID: 34501459 PMCID: PMC8432461 DOI: 10.3390/jcm10174011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/03/2021] [Accepted: 09/03/2021] [Indexed: 12/21/2022] Open
Abstract
Background: Eighty percent of patients with pancreatic adenocarcinoma present a locally advanced or metastatic disease at diagnosis and are not eligible for surgery if not with palliative intent. In cases of locally advanced disease (LAPC), the combination of chemo and radiotherapy is the only therapeutic option and correlates with a median survival of 15 months (10 months without treatment), with partial remission of disease in 50% of cases. The feasibility and safety of Electrochemotherapy (ECT) have been demonstrated in the treatment of deep tumors. Aim: The aim of the study is to evaluate the efficacy of electrochemotherapy (ECT) followed by conventional systemic treatment compared to the only conventional systemic treatment in LAPC in terms of objective response and overall survival. Patients and Methods: This study is a phase IIb prospective multicenter randomized controlled trial with two arms. The study will include 90 patients: 45 in the control group and 45 in the experimental group. Patients with LAPC in the control arm will receive conventional chemotherapy (FOLFOXIRI). Patients with LAPC in the experimental arm will be subjected to Electrochemotherapy and subsequently to FOLFOXIRI. The objective response at 30, 90, and 180 days from treatment will be based on the computed tomography (CT), magnetic resonance (MR), and positron emission tomography/CT response (PET/CT). The objective long-term treatment response will be evaluated with the modified response evaluation criteria in solid tumors (m-RECIST) criteria, which will take into account the difference in vascularization, determined by the images obtained by CT and MR of the tumor treated before and after ECT. Conclusions: Not resectable liver metastasis, pancreatic tumors, and locally advanced renal carcinomas can be treated with laparoscopic electrodes. ECT could represent an effective therapeutic option for patients not eligible for surgery susceptible to be managed only with palliative therapies.
Collapse
|
27
|
Treatment response assessment following transarterial radioembolization for hepatocellular carcinoma. Abdom Radiol (NY) 2021; 46:3596-3614. [PMID: 33909092 DOI: 10.1007/s00261-021-03095-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/01/2021] [Accepted: 04/10/2021] [Indexed: 12/17/2022]
Abstract
Transarterial radioembolization with yttrium-90 microspheres is an established therapy for hepatocellular carcinoma. Post-procedural imaging is important for the assessment of both treatment response and procedural complications. A variety of challenging treatment-specific imaging phenomena complicate imaging assessment, such as changes in tumoral size, tumoral and peritumoral enhancement, and extrahepatic complications. A review of the procedural steps, emerging variations, and timelines for post-treatment tumoral and extra-tumoral imaging changes are presented, which may aid the reporting radiologist in the interpretation of post-procedural imaging. Furthermore, a description of post-procedural complications and their significance is provided.
Collapse
|
28
|
Ko CC, Yeh LR, Kuo YT, Chen JH. Imaging biomarkers for evaluating tumor response: RECIST and beyond. Biomark Res 2021; 9:52. [PMID: 34215324 PMCID: PMC8252278 DOI: 10.1186/s40364-021-00306-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
Response Evaluation Criteria in Solid Tumors (RECIST) is the gold standard for assessment of treatment response in solid tumors. Morphologic change of tumor size evaluated by RECIST is often correlated with survival length and has been considered as a surrogate endpoint of therapeutic efficacy. However, the detection of morphologic change alone may not be sufficient for assessing response to new anti-cancer medication in all solid tumors. During the past fifteen years, several molecular-targeted therapies and immunotherapies have emerged in cancer treatment which work by disrupting signaling pathways and inhibited cell growth. Tumor necrosis or lack of tumor progression is associated with a good therapeutic response even in the absence of tumor shrinkage. Therefore, the use of unmodified RECIST criteria to estimate morphological changes of tumor alone may not be sufficient to estimate tumor response for these new anti-cancer drugs. Several studies have reported the low reliability of RECIST in evaluating treatment response in different tumors such as hepatocellular carcinoma, lung cancer, prostate cancer, brain glioma, bone metastasis, and lymphoma. There is an increased need for new medical imaging biomarkers, considering the changes in tumor viability, metabolic activity, and attenuation, which are related to early tumor response. Promising imaging techniques, beyond RECIST, include dynamic contrast-enhanced computed tomography (CT) or magnetic resonance imaging (MRI), diffusion-weight imaging (DWI), magnetic resonance spectroscopy (MRS), and 18 F-fluorodeoxyglucose (FDG) positron emission tomography (PET). This review outlines the current RECIST with their limitations and the new emerging concepts of imaging biomarkers in oncology.
Collapse
Affiliation(s)
- Ching-Chung Ko
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, Taiwan.,Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Lee-Ren Yeh
- Department of Radiology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yu-Ting Kuo
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, Taiwan.,Department of Medical Imaging, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Jeon-Hor Chen
- Department of Radiology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan. .,Tu & Yuan Center for Functional Onco-Imaging, Department of Radiological Sciences, University of California, 164 Irvine Hall, Irvine, CA, 92697 - 5020, USA.
| |
Collapse
|
29
|
Dercle L, Lu L, Schwartz LH, Qian M, Tejpar S, Eggleton P, Zhao B, Piessevaux H. Radiomics Response Signature for Identification of Metastatic Colorectal Cancer Sensitive to Therapies Targeting EGFR Pathway. J Natl Cancer Inst 2021; 112:902-912. [PMID: 32016387 DOI: 10.1093/jnci/djaa017] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/05/2019] [Accepted: 01/24/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The authors sought to forecast survival and enhance treatment decisions for patients with liver metastatic colorectal cancer by using on-treatment radiomics signature to predict tumor sensitiveness to irinotecan, 5-fluorouracil, and leucovorin (FOLFIRI) alone (F) or in combination with cetuximab (FC). METHODS We retrospectively analyzed 667 metastatic colorectal cancer patients treated with F or FC. Computed tomography quality was classified as high (HQ) or standard (SD). Four datasets were created using the nomenclature (treatment) - (quality). Patients were randomly assigned (2:1) to training or validation sets: FCHQ: 78:38, FCSD: 124:62, FHQ: 78:51, FSD: 158:78. Four tumor-imaging biomarkers measured quantitative radiomics changes between standard of care computed tomography scans at baseline and 8 weeks. Using machine learning, the performance of the signature to classify tumors as treatment sensitive or treatment insensitive was trained and validated using receiver operating characteristic (ROC) curves. Hazard ratio and Cox regression models evaluated association with overall survival (OS). RESULTS The signature (area under the ROC curve [95% confidence interval (CI)]) used temporal decrease in tumor spatial heterogeneity plus boundary infiltration to successfully predict sensitivity to antiepidermal growth factor receptor therapy (FCHQ: 0.80 [95% CI = 0.69 to 0.94], FCSD: 0.72 [95% CI = 0.59 to 0.83]) but failed with chemotherapy (FHQ: 0.59 [95% CI = 0.44 to 0.72], FSD: 0.55 [95% CI = 0.43 to 0.66]). In cetuximab-containing sets, radiomics signature outperformed existing biomarkers (KRAS-mutational status, and tumor shrinkage by RECIST 1.1) for detection of treatment sensitivity and was strongly associated with OS (two-sided P < .005). CONCLUSIONS Radiomics response signature can serve as an intermediate surrogate marker of OS. The signature outperformed known biomarkers in providing an early prediction of treatment sensitivity and could be used to guide cetuximab treatment continuation decisions.
Collapse
Affiliation(s)
- Laurent Dercle
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY, USA.,Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Lin Lu
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY, USA
| | - Lawrence H Schwartz
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY, USA
| | - Min Qian
- Department of Biostatistics, Columbia University Medical Center, New York, NY, USA
| | - Sabine Tejpar
- Molecular Digestive Oncology, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| | | | - Binsheng Zhao
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY, USA
| | - Hubert Piessevaux
- Department of Hepato-Gastroenterology, Cliniques Universitaires Saint-Luc, UCLouvain Brussels, Brussels, Belgium
| |
Collapse
|
30
|
Colagrande S, Calistri L, Campani C, Dragoni G, Lorini C, Nardi C, Castellani A, Marra F. CT volume of enhancement of disease (VED) can predict the early response to treatment and overall survival in patients with advanced HCC treated with sorafenib. Eur Radiol 2021; 31:1608-1619. [PMID: 32827266 PMCID: PMC7880966 DOI: 10.1007/s00330-020-07171-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/22/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To analyse the predictive value of the volume of enhancement of disease (VED), based on the CT arterial enhancement coefficient (ΔArt%), in the evaluation of the sorafenib response in patients with advanced hepatocellular carcinoma (HCC). METHODS Patients with sorafenib-treated advanced HCC, who underwent a multiphase contrast-enhanced CT before (T0) and after 60-70 days of starting therapy (T1), were included. The same target lesions utilised for the response evaluation according to modified Response Evaluation Criteria in Solid Tumors criteria were retrospectively used for the ΔArt% calculation ([(HUarterial phase - HUunenhanced phase) / HUunenhanced phase] × 100). ΔArt% was weighted for the lesion volume to obtain the VED. We compared VEDT0 and VEDT1 values in patients with clinical benefit (CB) or progressive disease (PD). The impact of VED, ancillary imaging findings, and blood chemistries on survival probability was evaluated. RESULTS Thirty-two patients (25 men, mean age 65.8 years) analysed between 2012 and 2016 were selected. At T1, 8 patients had CB and 24 had PD. VEDT0 was > 70% in 8/8 CB patients compared with 12/24 PD patients (p = 0.011). Patients with VEDT0 > 70% showed a significantly higher median survival than those with lower VEDT0 (451.5 days vs. 209.5 days, p = 0.032). Patients with VEDT0 > 70% and alpha-fetoproteinT0 ≤ 400 ng/ml had significantly longer survival than all other three combinations. In multivariate analysis, VEDT0 > 70% emerged as the only factor independently associated with survival (p = 0.037). CONCLUSION In patients with advanced HCC treated with sorafenib, VED is a novel radiologic parameter obtained by contrast-enhanced CT, which could be helpful in selecting patients who are more likely to respond to sorafenib, and with a longer survival. KEY POINTS • To achieve the best results of treatment with sorafenib in advanced HCC, a strict selection of patients is needed. • New radiologic parameters predictive of the response to sorafenib would be essential. • Volume of enhancement of disease (VED) is a novel radiologic parameter obtained by contrast-enhanced CT, which could be helpful in selecting patients who are more likely to respond to therapy, and with a longer survival.
Collapse
Affiliation(s)
- S Colagrande
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy.
| | - L Calistri
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy
| | - C Campani
- Department of Experimental and Clinical Medicine, University of Florence, 50134, Florence, Italy
| | - G Dragoni
- Department of Experimental and Clinical Medicine, University of Florence, 50134, Florence, Italy
| | - C Lorini
- Department of Health Science, University of Florence, Viale Morgagni 48, 50134, Florence, Italy
| | - C Nardi
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy
| | - A Castellani
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134, Florence, Italy
| | - F Marra
- Department of Experimental and Clinical Medicine, University of Florence, 50134, Florence, Italy
- Research Centre Denothe, University of Florence, Florence, Italy
| |
Collapse
|
31
|
Role of Key Guidelines in an Era of Precision Oncology: A Primer for the Radiologist. AJR Am J Roentgenol 2021; 216:1112-1125. [PMID: 33502227 DOI: 10.2214/ajr.20.23025] [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: 11/18/2022]
Abstract
OBJECTIVE. The purpose of this article is to familiarize radiologists with the evidence-based imaging guidelines of major oncologic societies and organizations and to discuss approaches to effective implementation of the most recent guidelines in daily radiology practice. CONCLUSION. In an era of precision oncology, radiologists in practice and radiologists in training are key stakeholders in multidisciplinary care, and their awareness and understanding of society guidelines is critically important.
Collapse
|
32
|
Cancer Detection and Quantification of Treatment Response Using Diffusion-Weighted MRI. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00068-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
33
|
Lacroix M, Mulé S, Herin E, Pigneur F, Richard P, Zegai B, Baranes L, Djabbari M, Brunetti F, de'Angelis N, Laurent A, Tacher V, Kobeiter H, Luciani A. Virtual unenhanced imaging of the liver derived from 160-mm rapid-switching dual-energy CT (rsDECT): Comparison of the accuracy of attenuation values and solid liver lesion conspicuity with native unenhanced images. Eur J Radiol 2020; 133:109387. [PMID: 33166833 DOI: 10.1016/j.ejrad.2020.109387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/06/2020] [Accepted: 10/27/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVES To evaluate the reliability of attenuation values of the liver parenchyma and focal liver lesions on virtual unenhanced images from arterial (VUEart) and portal venous phases (VUEport) compared to native unenhanced (NU) attenuation values in patients referred for assessment of malignant liver lesions. METHODS Seventy-three patients with confirmed primary or metastatic liver tumors who underwent a multiphase contrast-enhanced rapid-switching kVp dual-energy CT (rsDECT) were included in this IRB-approved retrospective study. Both qualitative and quantitative analyses - including the lesion-to-liver contrast-to-noise ratio (LL-CNR) - were performed and compared between NU and both VUEart and VUEport images. RESULTS The mean liver attenuation values were significantly lower in VUEart images (56.7 ± 6.7 HU) than in NU images (59.6 ± 7.5 HU, p = 0.008), and were comparable between VUEart and VUEport images (57.9 ± 6 UH, p = 0.38) and between VUEport and NU images (p = 0.051). The mean liver lesions attenuation values were comparable between NU, VUEart and VUEport images (p = 0.60). Strong and significant correlations values were found both in liver lesions and tumor-free parenchyma (r = 0.82-0.91, p < 0.01). The mean LL-CNR was significantly higher in VUEart and VUEport images than in NU images (1.7 ± 1 and 1.6 ± 1.1 vs 0.9 ± 0.6; p < 0.001), but was comparable between VUEart and VUEport images (p > 0.9). Lesion conspicuity was significantly higher in VUEport images than in NU images (p < 0.001). CONCLUSION VUEport images derived from 3rd generation rsDECT could confidently replace NU images in patients undergoing assessment for malignant liver lesions. These images provide comparable attenuation values in both liver lesions and liver parenchyma while reducing the radiation dose and scanning time.
Collapse
Affiliation(s)
- Maxime Lacroix
- Service d'Imagerie Médicale, AP-HP, Hôpitaux Universitaires Henri Mondor, 94010 Créteil, France.
| | - Sébastien Mulé
- Service d'Imagerie Médicale, AP-HP, Hôpitaux Universitaires Henri Mondor, 94010 Créteil, France; Faculté de Médecine de Créteil, Université Paris Est Créteil, 94000 Créteil, France; INSERM IMRB, U 955, Equipe 18, Créteil, France
| | - Edouard Herin
- Service d'Imagerie Médicale, AP-HP, Hôpitaux Universitaires Henri Mondor, 94010 Créteil, France
| | - Frédéric Pigneur
- Service d'Imagerie Médicale, AP-HP, Hôpitaux Universitaires Henri Mondor, 94010 Créteil, France
| | | | - Benhalima Zegai
- Service d'Imagerie Médicale, AP-HP, Hôpitaux Universitaires Henri Mondor, 94010 Créteil, France
| | - Laurence Baranes
- Service d'Imagerie Médicale, AP-HP, Hôpitaux Universitaires Henri Mondor, 94010 Créteil, France
| | - Marjan Djabbari
- Service d'Imagerie Médicale, AP-HP, Hôpitaux Universitaires Henri Mondor, 94010 Créteil, France
| | - Francesco Brunetti
- Service de chirurgie digestive, AP-HP, Hôpital Henri Mondor, 94010 Créteil, France
| | - Nicola de'Angelis
- Faculté de Médecine de Créteil, Université Paris Est Créteil, 94000 Créteil, France; Service de chirurgie digestive, AP-HP, Hôpital Henri Mondor, 94010 Créteil, France
| | - Alexis Laurent
- Faculté de Médecine de Créteil, Université Paris Est Créteil, 94000 Créteil, France; Service de chirurgie digestive, AP-HP, Hôpital Henri Mondor, 94010 Créteil, France
| | - Vania Tacher
- Service d'Imagerie Médicale, AP-HP, Hôpitaux Universitaires Henri Mondor, 94010 Créteil, France
| | - Hicham Kobeiter
- Service d'Imagerie Médicale, AP-HP, Hôpitaux Universitaires Henri Mondor, 94010 Créteil, France; Faculté de Médecine de Créteil, Université Paris Est Créteil, 94000 Créteil, France
| | - Alain Luciani
- Service d'Imagerie Médicale, AP-HP, Hôpitaux Universitaires Henri Mondor, 94010 Créteil, France; Faculté de Médecine de Créteil, Université Paris Est Créteil, 94000 Créteil, France; INSERM IMRB, U 955, Equipe 18, Créteil, France
| |
Collapse
|
34
|
Bergamini C, Cavalieri S, Cascella T, Lanocita R, Alfieri S, Resteghini C, Platini F, Orlandi E, Locati LD, Marchianò A, Licitra L. Local therapies for liver metastases of rare head and neck cancers: a monoinstitutional case series. TUMORI JOURNAL 2020; 107:188-195. [PMID: 32924878 DOI: 10.1177/0300891620952844] [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] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Transarterial chemoembolization (TACE) and radiofrequency ablation (RFA) are established procedures for treating hepatocellular cancer and selected malignancies with liver metastasis. The aim of this study is to describe a monoinstitutional case series of local approaches in patients with liver metastases from rare head and neck cancers (HNCs). METHODS This is a retrospective series of adult patients with HNC treated with liver locoregional approaches (TACE or RFA) at our institution from 2007 to 2018. In case of chemoembolization, the preferred chemotherapeutic drug was doxorubicin. Response according to RECIST (Response Evaluation Criteria in Solid Tumors) was assessed with contrast-enhanced computed tomography scans. RESULTS Thirty-four patients were treated (20 men, median age 58 years) with TACE (27), transarterial embolization (2), or RFA (7). Primary tumours were salivary gland (21), thyroid (6), nasopharyngeal (5), and sinonasal cancers (2). Seventeen patients (50%) had a single metastatic liver nodule; 70% of the remaining 17 patients had at least three liver metastases. The median diameter of the metastatic liver mass undergoing treatment was 39 mm (range 11-100 mm). Median follow-up was 27.6 months. Response rate was 35% (3% complete, 32% partial response). Median progression-free survival and overall survival were 6.9 and 19.6 months, respectively. Treatment-related adverse events occurred in 59% of patients (21% grade ⩾ 3; no grade 5). DISCUSSION This retrospective case series demonstrates that locoregional radiologic approaches for rare HNCs with liver metastases are feasible. These procedures deserve further prospective studies before being considered safe and active in these malignancies where the availability of effective systemic treatments is lacking.
Collapse
Affiliation(s)
- Cristiana Bergamini
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Stefano Cavalieri
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Tommaso Cascella
- Radiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Rodolfo Lanocita
- Radiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Salvatore Alfieri
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Carlo Resteghini
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Francesca Platini
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Ester Orlandi
- Radiotherapy 1-2, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Laura Deborah Locati
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Alfonso Marchianò
- Radiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Lisa Licitra
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
35
|
Valenzuela RF, Kundra V, Madewell JE, Costelloe CM. Advanced Imaging in Musculoskeletal Oncology: Moving Away From RECIST and Embracing Advanced Bone and Soft Tissue Tumor Imaging (ABASTI) - Part I - Tumor Response Criteria and Established Functional Imaging Techniques. Semin Ultrasound CT MR 2020; 42:201-214. [PMID: 33814106 DOI: 10.1053/j.sult.2020.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
According to the Revised Response Evaluation Criteria in Solid Tumors (RECIST) 1.1, the majority of bone metastases are considered to be nonmeasurable disease. Traditional response criteria rely on physical measurements. New criteria would be valuable if they incorporated newly developed imaging features in order to provide a more comprehensive assessment of oncological status. Advanced magnetic resonance imaging (MRI) sequences such as diffusion-weighted imaging (DWI) and perfusion-weighted imaging (PWI) with dynamic contrast-enhanced (DCE) perfusion imaging are reviewed in the context of the initial and post-therapeutic assessment of musculoskeletal tumors. Particular attention is directed to the pseudoprogression phenomenon in which a successfully treated tumor enlarges from the pretherapeutic baseline, followed by regression without a change in therapy.
Collapse
Affiliation(s)
- Raul Fernando Valenzuela
- The University of Texas MD Anderson Cancer Center, Department of Musculoskeletal Imaging, Houston, Texas.
| | - Vikas Kundra
- The University of Texas MD Anderson Cancer Center, Department of Musculoskeletal Imaging, Houston, Texas
| | - John E Madewell
- The University of Texas MD Anderson Cancer Center, Department of Musculoskeletal Imaging, Houston, Texas
| | - Colleen M Costelloe
- The University of Texas MD Anderson Cancer Center, Department of Musculoskeletal Imaging, Houston, Texas
| |
Collapse
|
36
|
Ambrosini R, Balli MC, Laganà M, Bertuletti M, Bottoni L, Vaccher F, Cosentini D, Di Terlizzi M, Sigala S, Grisanti S, Tiberio GAM, Berruti A, Grazioli L. Adrenocortical Carcinoma and CT Assessment of Therapy Response: The Value of Combining Multiple Criteria. Cancers (Basel) 2020; 12:1395. [PMID: 32481732 PMCID: PMC7352253 DOI: 10.3390/cancers12061395] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 12/19/2022] Open
Abstract
We evaluated tumor response at Computed Tomography (CT) according to three radiologic criteria: RECIST 1.1, CHOI and tumor volume in 34 patients with metastatic adrenocortical carcinoma (ACC) submitted to standard chemotherapy. These three criteria agreed in defining partial response, stable or progressive disease in 24 patients (70.5%). Partial response (PR) was observed in 29.4%, 29.4% and 41.2% of patients according to RECIST 1.1, CHOI and tumor volume, respectively. It was associated with a favorable prognosis, regardless of the criterion adopted. The concordance of all the 3 criteria in defining the disease response identified 8 patients (23.5%) which displayed a very good prognosis: median progression free survival (PFS) and overall survival (OS) 14.9 and 37.7 months, respectively. Seven patients (20.6%) with PR assessed by one or two criteria, however, still had a better prognosis than non-responding patients, both in terms of PFS: median 12.3 versus 9.9 months and OS: 21 versus 12.2, respectively. In conclusions, the CT assessment of disease response of ACC patients to chemotherapy with 3 different criteria is feasible and allows the identification of a patient subset with a more favorable outcome. PR with at least one criterion can be useful to early identify patients that deserve continuing the therapy.
Collapse
Affiliation(s)
- Roberta Ambrosini
- I Radiology Unit, ASST Spedali Civili, 25123 Brescia, Italy; (M.C.B.); (M.B.); (L.B.); (F.V.); (M.D.T.); (L.G.)
| | - Maria Carolina Balli
- I Radiology Unit, ASST Spedali Civili, 25123 Brescia, Italy; (M.C.B.); (M.B.); (L.B.); (F.V.); (M.D.T.); (L.G.)
| | - Marta Laganà
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia at ASST Spedali Civili, 25123 Brescia, Italy; (M.L.); (D.C.); (S.G.); (A.B.)
| | - Martina Bertuletti
- I Radiology Unit, ASST Spedali Civili, 25123 Brescia, Italy; (M.C.B.); (M.B.); (L.B.); (F.V.); (M.D.T.); (L.G.)
| | - Luca Bottoni
- I Radiology Unit, ASST Spedali Civili, 25123 Brescia, Italy; (M.C.B.); (M.B.); (L.B.); (F.V.); (M.D.T.); (L.G.)
| | - Filippo Vaccher
- I Radiology Unit, ASST Spedali Civili, 25123 Brescia, Italy; (M.C.B.); (M.B.); (L.B.); (F.V.); (M.D.T.); (L.G.)
| | - Deborah Cosentini
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia at ASST Spedali Civili, 25123 Brescia, Italy; (M.L.); (D.C.); (S.G.); (A.B.)
| | - Marco Di Terlizzi
- I Radiology Unit, ASST Spedali Civili, 25123 Brescia, Italy; (M.C.B.); (M.B.); (L.B.); (F.V.); (M.D.T.); (L.G.)
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Salvatore Grisanti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia at ASST Spedali Civili, 25123 Brescia, Italy; (M.L.); (D.C.); (S.G.); (A.B.)
| | - Guido Alberto Massimo Tiberio
- Surgical Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili di Brescia, 25123 Brescia, Italy;
| | - Alfredo Berruti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia at ASST Spedali Civili, 25123 Brescia, Italy; (M.L.); (D.C.); (S.G.); (A.B.)
| | - Luigi Grazioli
- I Radiology Unit, ASST Spedali Civili, 25123 Brescia, Italy; (M.C.B.); (M.B.); (L.B.); (F.V.); (M.D.T.); (L.G.)
| |
Collapse
|
37
|
Patil PG, Reddy P, Rawat S, Ananthasivan R, Sinha R. Multimodality Approach in Detection and Characterization of Hepatic Metastases. JOURNAL OF GASTROINTESTINAL AND ABDOMINAL RADIOLOGY 2020. [DOI: 10.1055/s-0039-3402100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
AbstractEarly detection of liver metastases is important in patients with known primary malignancies. This plays an important role in treatment planning and impacts on further management of certain primary malignancies.Magnetic resonance imaging (MRI), computed tomography (CT), and positron emission tomography-computed tomography scans are reported to have high accuracy in the diagnosis of intrahepatic lesions. MRI in particular has the advantages of its high tissue sensitivity and its multiparametric approach.Hepatic metastatic lesions have considerable overlap in their radiological appearance, and in this article the imaging appearance of various hepatic metastasis and approach is described.
Collapse
Affiliation(s)
- Pooja G. Patil
- Department of Radiology, Manipal Hospital, Bangalore, Karnataka, India
| | - Pramesh Reddy
- Department of Radiology, Manipal Hospital, Bangalore, Karnataka, India
| | - Sudarshan Rawat
- Department of Radiology, Manipal Hospital, Bangalore, Karnataka, India
| | - Rupa Ananthasivan
- Department of Radiology, Manipal Hospital, Bangalore, Karnataka, India
| | - Rakesh Sinha
- Department of Radiology, South Warwickshire NHS Foundation Trust, Warwick, United Kingdom
| |
Collapse
|
38
|
Jin L, Ma XM, Wang TT, Yang Y, Zhang N, Zeng N, Bai ZG, Yin J, Zhang J, Ding GQ, Zhang ZT. Psoralen Suppresses Cisplatin-Mediated Resistance and Induces Apoptosis of Gastric Adenocarcinoma by Disruption of the miR196a-HOXB7-HER2 Axis. Cancer Manag Res 2020; 12:2803-2827. [PMID: 32368152 PMCID: PMC7185648 DOI: 10.2147/cmar.s248094] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 04/02/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose The present study aimed to investigate the impact of psoralen on miR-196a-5p expression and function, and to reveal the mechanism underlying miR-196a-5p-mediated inhibition and the reversal of cisplatin (DDP) resistance. Methods Serum samples were collected from 50 patients with gastric cancer (GC), and the association between miR-196a-5p expression and the response to chemotherapy was assessed. A DDP-resistant GC cell line was also established to determine the effects of miR-196a-5p and psoralen on DDP resistance. MGC803 cells were transfected with miR-196a-5p mimic and inhibitor vectors for the overexpression and downregulation of miR-196a-5p, respectively. Results Clinical data analysis showed that the lower expression levels of miR-196a-5p were significantly associated with chemoresistance in patients with GC. Upregulation of miR-196a-5p significantly enhanced the anti-proliferative effect, apoptosis and sensitivity to DDP by regulating the protein expression levels of HOXB7, HER2, Bcl-2 and G1/S-specific cyclin-D1 (CCND1). Furthermore, psoralen reversed miR-196a-5p-induced DDP resistance and reduced the expression levels of HOXB7, HER2, Bcl-2 and CCND1. Conclusion miR-196a-5p may be a novel biomarker of chemotherapeutic success in patients with GC and may also influence the sensitivity of GC cells to DDP. Moreover, psoralen can increase chemotherapeutic sensitivity by upregulating miR-196a-5p and then downregulating HOXB7-HER2 signaling axis.
Collapse
Affiliation(s)
- Lei Jin
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| | - Xue-Mei Ma
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| | - Ting-Ting Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| | - Yao Yang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| | - Nan Zhang
- National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China.,Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Na Zeng
- National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China.,Clinical Epidemiology and EBM Center, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Zhi-Gang Bai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| | - Jie Yin
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| | - Jun Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| | - Guo-Qian Ding
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| | - Zhong-Tao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.,National Clinical Research Center for Digestive Diseases, Beijing, People's Republic of China
| |
Collapse
|
39
|
Jin L, Zhang N, Zhang Q, Ding G, Yang Z, Zhang Z. Serum microRNAs as potential new biomarkers for cisplatin resistance in gastric cancer patients. PeerJ 2020; 8:e8943. [PMID: 32328349 PMCID: PMC7164432 DOI: 10.7717/peerj.8943] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 03/18/2020] [Indexed: 12/19/2022] Open
Abstract
Background microRNAs (miRNAs) have been studied for their role in the early detection of several diseases. However, there is no current information on the systematic screening of serum-derived cisplatin resistance biomarkers in gastric cancer (GC). Methods Cisplatin-resistant GC cell lines were screened for dysregulated miRNAs using small RNA sequencing (sRNA-seq) and miRNAs were functionally annotated using bioinformatics analyses. Real-time quantitative polymerase chain reaction (RT-qPCR) was used to validate the miRNA-relative transcription levels in GC cells and in 74 GC patients. We analyzed the associations between the clinical characteristics of the patients and their miRNA expression. Receiver operating characteristic (ROC) analysis was used to evaluate the diagnostic value for serum-derived cisplatin resistance. Results Seven miRNAs were identified from 35 differentially expressed miRNAs between the MGC803/DDP and MGC803 cells in a public database. We found four miRNA candidates (miR-9-3p, miR-9-5p, miR-146a-5p, and miR-433-3p) that were significantly associated with chemotherapy responses in GC cells and patients. miR-9-5p (AUC = 0.856, 95% CI [0.773–0.939], p < 0.0001) and a combined group (miR-9-5p + miR-9-3p + miR-433-3p) (AUC = 0.915, 95% CI [0.856–0.975], P < 0.0001) distinguished chemoresistant GC patients from chemosensitive GC patients. Conclusions Our study reveals the potential therapeutic use of two serum-based biomarkers, miR-9-5p and a combined group (miR-9-5p + miR-9-3p + miR-433-3p), as indicators for the successful use of cisplatin in GC patients.
Collapse
Affiliation(s)
- Lei Jin
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Nan Zhang
- National Clinical Research Center for Digestive Diseases, Beijing, China.,Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Qian Zhang
- National Clinical Research Center for Digestive Diseases, Beijing, China.,Clinical Epidemiology and EBM Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Guoqian Ding
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhenghan Yang
- National Clinical Research Center for Digestive Diseases, Beijing, China.,Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
40
|
Hahn F, Müller L, Jungmann F, Mähringer-Kunz A, Tanyildizi Y, Düber C, Galle PR, Weinmann A, Kloeckner R. Survival prediction for patients with non-resectable intrahepatic cholangiocarcinoma undergoing chemotherapy: a retrospective analysis comparing the tumor marker CA 19-9 with cross-sectional imaging. J Cancer Res Clin Oncol 2020; 146:1883-1890. [PMID: 32232655 PMCID: PMC7256028 DOI: 10.1007/s00432-020-03200-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/24/2020] [Indexed: 12/13/2022]
Abstract
Purpose Carbohydrate antigen (CA) 19-9 has been established as the main serum marker for patients with intrahepatic cholangiocarcinoma (ICC). The aim of this study was to compare the prognostic value of CA 19-9 changes versus response determined by imaging in patients with ICC undergoing chemotherapy. Methods Between 2003 and 2018, 151 patients with histopathologically confirmed ICC underwent chemotherapy at our tertiary care center for non-resectable or recurrent ICC, of whom 121 were included in this study. Serum CA 19-9 levels and imaging were retrospectively evaluated during chemotherapy. Log-rank testing and optimal stratification were used to classify patients into risk groups. Results Prior to chemotherapy, baseline serum CA 19-9 levels above the previously published cut-off of 37 U/ml were associated with poor survival (median OS 8.7 vs. 12.4 months, p = 0.003). After the beginning of chemotherapy, an increase in CA 19-9 of more than 40 U/ml resulted in impaired residual survival (median OS 5.0 vs. 12.1 months, p < 0.001). However, progressive disease at the first follow-up imaging proved the strongest predictor for poor outcome (median OS 4.6 vs. 15.5 months, p < 0.001). In contrast to prior studies, our data did not show statistically relevant differences in survival time with respect to absolute or relative decreases in serum CA 19-9 levels. Conclusion In our study, the disease control rate—that is, the absence of progressive disease—was the strongest predictor of prolonged residual OS. To this end, both CA 19-9 changes and progressive disease on initial follow-up showed remarkable discriminatory power, with the latter slightly outperforming the former. Therefore, imaging should remain the mainstay of patient evaluation during follow-up. Electronic supplementary material The online version of this article (10.1007/s00432-020-03200-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Felix Hahn
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckst. 1, 55131, Mainz, Germany
| | - Lukas Müller
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckst. 1, 55131, Mainz, Germany
| | - Florian Jungmann
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckst. 1, 55131, Mainz, Germany
| | - Aline Mähringer-Kunz
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckst. 1, 55131, Mainz, Germany
| | - Yasemin Tanyildizi
- Department of Neuroradiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Christoph Düber
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckst. 1, 55131, Mainz, Germany
| | - Peter R Galle
- Department of Internal Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Arndt Weinmann
- Department of Internal Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Clinical Registry Unit (CRU), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Roman Kloeckner
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckst. 1, 55131, Mainz, Germany.
| |
Collapse
|
41
|
Spina JC, Hume I, Pelaez A, Peralta O, Quadrelli M, Garcia Monaco R. Expected and Unexpected Imaging Findings after 90Y Transarterial Radioembolization for Liver Tumors. Radiographics 2020; 39:578-595. [PMID: 30844345 DOI: 10.1148/rg.2019180095] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Transarterial radioembolization (TARE), also called radioembolization or selective internal radiation therapy, is an interventional radiology technique used to treat primary liver tumors and liver metastases. The aim of this therapy is to deliver tumoricidal doses of radiation to liver tumors while selecting a safe radiation dose limit for nontumoral liver and lung tissue. Hence, correct treatment planning is essential to obtaining good results. However, this treatment invariably results in some degree of irradiation of normal liver parenchyma, inducing different radiologic findings that may affect follow-up image interpretation. When evaluating treatment response, the treated area size, tumor necrosis, devascularization, and changes seen at functional MRI must be taken into account. Unlike with other interventional procedures, with TARE, it can take several months for the tumor response to become evident. Ideally, responding lesions will show reduced size and decreased enhancement 3-6 months after treatment. In addition, during follow-up, there are many imaging findings related to the procedure itself (eg, peritumoral edema, inflammation, ring enhancement, hepatic fibrosis, and capsular retraction) that can make image interpretation and response evaluation difficult. Possible complications, either hepatic or extrahepatic, also can occur and include biliary injuries, hepatic abscess, radioembolization-induced liver disease, and radiation pneumonitis or dermatitis. A complete understanding of these possible posttreatment changes is essential for correct radiologic interpretations during the follow-up of patients who have undergone TARE. ©RSNA, 2019.
Collapse
Affiliation(s)
- Juan C Spina
- From the Departments of Radiology (J.C.S., A.P., O.P., M.Q., R.G.M.) and Nuclear Medicine (I.H.), Hospital Italiano, Tte Gral Juan Domingo Perón 4230, C1199ABH CABA, Buenos Aires, Argentina
| | - Isabel Hume
- From the Departments of Radiology (J.C.S., A.P., O.P., M.Q., R.G.M.) and Nuclear Medicine (I.H.), Hospital Italiano, Tte Gral Juan Domingo Perón 4230, C1199ABH CABA, Buenos Aires, Argentina
| | - Ana Pelaez
- From the Departments of Radiology (J.C.S., A.P., O.P., M.Q., R.G.M.) and Nuclear Medicine (I.H.), Hospital Italiano, Tte Gral Juan Domingo Perón 4230, C1199ABH CABA, Buenos Aires, Argentina
| | - Oscar Peralta
- From the Departments of Radiology (J.C.S., A.P., O.P., M.Q., R.G.M.) and Nuclear Medicine (I.H.), Hospital Italiano, Tte Gral Juan Domingo Perón 4230, C1199ABH CABA, Buenos Aires, Argentina
| | - Marcos Quadrelli
- From the Departments of Radiology (J.C.S., A.P., O.P., M.Q., R.G.M.) and Nuclear Medicine (I.H.), Hospital Italiano, Tte Gral Juan Domingo Perón 4230, C1199ABH CABA, Buenos Aires, Argentina
| | - Ricardo Garcia Monaco
- From the Departments of Radiology (J.C.S., A.P., O.P., M.Q., R.G.M.) and Nuclear Medicine (I.H.), Hospital Italiano, Tte Gral Juan Domingo Perón 4230, C1199ABH CABA, Buenos Aires, Argentina
| |
Collapse
|
42
|
Zhang Y, Wang J, Li H, Zheng T, Jiang H, Li M, Song B. Performance of LI-RADS version 2018 CT treatment response algorithm in tumor response evaluation and survival prediction of patients with single hepatocellular carcinoma after radiofrequency ablation. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:388. [PMID: 32355832 PMCID: PMC7186681 DOI: 10.21037/atm.2020.03.120] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 03/02/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND The Liver Imaging Reporting and Data System treatment response algorithm (LI-RADS TRA) was developed to evaluate the tumor response of patients with hepatocellular carcinoma (HCC) after locoregional treatments. This study aimed to evaluate the performance of LI-RADS computed tomography (CT) TRA version 2018 in tumor response assessment and survival prediction of patients with single HCC after radiofrequency ablation (RFA). METHODS Forty patients who underwent RFA for single HCC between 2010 and 2016 were included in this retrospective study. The overall survival (OS) data from all the patients after the first therapy was collected. Two readers independently assessed the pretreatment (within 7 d) and posttreatment (within 90 d after RFA) CT manifestations using the LI-RADS version 2018 CT TRA. Inter-reader agreement was assessed. Another radiologist re-evaluated any divergent results and came to the final conclusion. The performance of LI-RADS version 2018 CT TRA for tumor response assessment and predicting survival of patients with single HCC after RFA was evaluated. RESULTS Interobserver agreement was moderate between the 2 readers [κ=0.602, 95% confidence interval (CI): 0.390-0.814] when using LI-RADS version 2018 TRA to evaluate tumor response for patients with single HCC after RFA. Patients classified as LR-TR viable had significantly lower OS than those classified as LR-TR nonviable (P=0.005) and LR-TR equivocal (P=0.036). However, the OS between LR-TR nonviable and LR-TR equivocal did not differ significantly (P=0.901). CONCLUSIONS LI-RADS version 2018 CT TRA can be applied to predict viable or nonviable HCC after RFA. Patients with LR-TR viable had significantly lower OS than those with LR-TR nonviable and LR-TR equivocal. More research is needed to validate the performance of LI-RADS version 2018 TRA in HCC tumor response evaluation, to better grasp the use of the tie-breaking rule, and to improve the accuracy of prediction for tumor viability.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Radiology, Sichuan University West China Hospital, Chengdu 610041, China
| | - Jinju Wang
- Department of Liver Surgery & Liver Transplantation Center, West China Hospital, Chengdu 610041, China
| | - Hui Li
- Department of Liver Surgery & Liver Transplantation Center, West China Hospital, Chengdu 610041, China
| | - Tianying Zheng
- Department of Radiology, Sichuan University West China Hospital, Chengdu 610041, China
| | - Hanyu Jiang
- Department of Radiology, Sichuan University West China Hospital, Chengdu 610041, China
| | - Mou Li
- Department of Radiology, Sichuan University West China Hospital, Chengdu 610041, China
| | - Bin Song
- Department of Radiology, Sichuan University West China Hospital, Chengdu 610041, China
| |
Collapse
|
43
|
Rubin DL, Ugur Akdogan M, Altindag C, Alkim E. ePAD: An Image Annotation and Analysis Platform for Quantitative Imaging. ACTA ACUST UNITED AC 2020; 5:170-183. [PMID: 30854455 PMCID: PMC6403025 DOI: 10.18383/j.tom.2018.00055] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Medical imaging is critical for assessing the response of patients to new cancer therapies. Quantitative lesion assessment on images is time-consuming, and adopting new promising quantitative imaging biomarkers of response in clinical trials is challenging. The electronic Physician Annotation Device (ePAD) is a freely available web-based zero-footprint software application for viewing, annotation, and quantitative analysis of radiology images designed to meet the challenges of quantitative evaluation of cancer lesions. For imaging researchers, ePAD calculates a variety of quantitative imaging biomarkers that they can analyze and compare in ePAD to identify potential candidates as surrogate endpoints in clinical trials. For clinicians, ePAD provides clinical decision support tools for evaluating cancer response through reports summarizing changes in tumor burden based on different imaging biomarkers. As a workflow management and study oversight tool, ePAD lets clinical trial project administrators create worklists for users and oversee the progress of annotations created by research groups. To support interoperability of image annotations, ePAD writes all image annotations and results of quantitative imaging analyses in standardized file formats, and it supports migration of annotations from various propriety formats. ePAD also provides a plugin architecture supporting MATLAB server-side modules in addition to client-side plugins, permitting the community to extend the ePAD platform in various ways for new cancer use cases. We present an overview of ePAD as a platform for medical image annotation and quantitative analysis. We also discuss use cases and collaborations with different groups in the Quantitative Imaging Network and future directions.
Collapse
Affiliation(s)
- Daniel L Rubin
- Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA
| | - Mete Ugur Akdogan
- Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA
| | - Cavit Altindag
- Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA
| | - Emel Alkim
- Department of Biomedical Data Science, Radiology, and Medicine (Biomedical Informatics Research), Stanford University, Stanford, CA
| |
Collapse
|
44
|
Ma J, Dercle L, Lichtenstein P, Wang D, Chen A, Zhu J, Piessevaux H, Zhao J, Schwartz LH, Lu L, Zhao B. Automated Identification of Optimal Portal Venous Phase Timing with Convolutional Neural Networks. Acad Radiol 2020; 27:e10-e18. [PMID: 31151901 DOI: 10.1016/j.acra.2019.02.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/25/2019] [Accepted: 02/26/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To develop a deep learning-based algorithm to automatically identify optimal portal venous phase timing (PVP-timing) so that image analysis techniques can be accurately performed on post contrast studies. METHODS 681 CT-scans (training: 479 CT-scans; validation: 202 CT-scans) from a multicenter clinical trial in patients with liver metastases from colorectal cancer were retrospectively analyzed for algorithm development and validation. An additional external validation was performed on a cohort of 228 CT-scans from gastroenteropancreatic neuroendocrine cancer patients. Image acquisition was performed according to each centers' standard CT protocol for single portal venous phase, portal venous acquisition. The reference gold standard for the classification of PVP-timing as either optimal or nonoptimal was based on experienced radiologists' consensus opinion. The algorithm performed automated localization (on axial slices) of the portal vein and aorta upon which a novel dual input Convolutional Neural Network calculated a probability of the optimal PVP-timing. RESULTS The algorithm automatically computed a PVP-timing score in 3 seconds and reached area under the curve of 0.837 (95% CI: 0.765, 0.890) in validation set and 0.844 (95% CI: 0.786, 0.889) in external validation set. CONCLUSION A fully automated, deep-learning derived PVP-timing algorithm was developed to classify scans' contrast-enhancement timing and identify scans with optimal PVP-timing. The rapid identification of such scans will aid in the analysis of quantitative (radiomics) features used to characterize tumors and changes in enhancement with treatment in a multitude of settings including quantitative response criteria such as Choi and MASS which rely on reproducible measurement of enhancement.
Collapse
Affiliation(s)
- Jingchen Ma
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China; Department of Radiology, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032
| | - Laurent Dercle
- Department of Radiology, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032; Gustave Roussy, Université Paris-Saclay, Université Paris-Saclay, Département D'imagerie Médicale, Villejuif, France
| | - Philip Lichtenstein
- Department of Radiology, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032
| | - Deling Wang
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China
| | - Aiping Chen
- Department of Radiology, First Affiliated Hospital of NanJing Medical University, Nanjing, China
| | - Jianguo Zhu
- Department of Radiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | - Jun Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Lawrence H Schwartz
- Department of Radiology, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032
| | - Lin Lu
- Department of Radiology, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032.
| | - Binsheng Zhao
- Department of Radiology, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032
| |
Collapse
|
45
|
Dercle L, Ma J, Xie C, Chen AP, Wang D, Luk L, Revel-Mouroz P, Otal P, Peron JM, Rousseau H, Lu L, Schwartz LH, Mokrane FZ, Zhao B. Using a single abdominal computed tomography image to differentiate five contrast-enhancement phases: A machine-learning algorithm for radiomics-based precision medicine. Eur J Radiol 2020; 125:108850. [PMID: 32070870 DOI: 10.1016/j.ejrad.2020.108850] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/25/2020] [Indexed: 11/29/2022]
Abstract
PURPOSE The clinical adoption of quantitative imaging biomarkers (radiomics) has established the need for high quality contrast-enhancement in medical images. We aimed to develop a machine-learning algorithm for Quality Control of Contrast-Enhancement on CT-scan (CECT-QC). METHOD Multicenter data from four independent cohorts [A, B, C, D] of patients with measurable liver lesions were analyzed retrospectively (patients:time-points; 503:3397): [A] dynamic CTs from primary liver cancer (60:2359); [B] triphasic CTs from primary liver cancer (31:93); [C] triphasic CTs from hepatocellular carcinoma (121:363); [D] portal venous phase CTs of liver metastasis from colorectal cancer (291:582). Patients from cohort A were randomized to training-set (48:1884) and test-set (12:475). A random forest classifier was trained and tested to identify five contrast-enhancement phases. The input was the mean intensity of the abdominal aorta and the portal vein measured on a single abdominal CT scan image at a single time-point. The output to be predicted was: non-contrast [NCP], early-arterial [E-AP], optimal-arterial [O-AP], optimal-portal [O-PVP], and late-portal [L-PVP]. Clinical utility was assessed in cohorts B, C, and D. RESULTS The CECT-QC algorithm showed performances of 98 %, 90 %, and 84 % for predicting NCP, O-AP, and O-PVP, respectively. O-PVP was reached in half of patients and was associated with a peak in liver malignancy density. Contrast-enhancement quality significantly influenced radiomics features deciphering the phenotype of liver neoplasms. CONCLUSIONS A single CT-image can be used to differentiate five contrast-enhancement phases for radiomics-based precision medicine in the most common liver neoplasms occurring in patients with or without liver cirrhosis.
Collapse
Affiliation(s)
- Laurent Dercle
- Columbia University Vagellos College of Physicians and Surgeons, Department of Radiology, New York, New York City, USA; Department of Radiology New York Presbyterian Hospital, USA.
| | - Jingchen Ma
- Columbia University Vagellos College of Physicians and Surgeons, Department of Radiology, New York, New York City, USA; Department of Radiology New York Presbyterian Hospital, USA
| | - Chuanmiao Xie
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Ai-Ping Chen
- Columbia University Vagellos College of Physicians and Surgeons, Department of Radiology, New York, New York City, USA; Department of Radiology New York Presbyterian Hospital, USA
| | - Deling Wang
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Lyndon Luk
- Columbia University Vagellos College of Physicians and Surgeons, Department of Radiology, New York, New York City, USA; Department of Radiology New York Presbyterian Hospital, USA
| | - Paul Revel-Mouroz
- Radiology Department, Rangueil University Hospital, Toulouse, France
| | - Philippe Otal
- Radiology Department, Rangueil University Hospital, Toulouse, France
| | - Jean-Marie Peron
- Hepatology Department, Purpan University Hospital, Toulouse, France
| | - Hervé Rousseau
- Radiology Department, Rangueil University Hospital, Toulouse, France
| | - Lin Lu
- Columbia University Vagellos College of Physicians and Surgeons, Department of Radiology, New York, New York City, USA; Department of Radiology New York Presbyterian Hospital, USA
| | - Lawrence H Schwartz
- Columbia University Vagellos College of Physicians and Surgeons, Department of Radiology, New York, New York City, USA; Department of Radiology New York Presbyterian Hospital, USA
| | - Fatima-Zohra Mokrane
- Columbia University Vagellos College of Physicians and Surgeons, Department of Radiology, New York, New York City, USA; Department of Radiology New York Presbyterian Hospital, USA; Radiology Department, Rangueil University Hospital, Toulouse, France
| | - Binsheng Zhao
- Columbia University Vagellos College of Physicians and Surgeons, Department of Radiology, New York, New York City, USA; Department of Radiology New York Presbyterian Hospital, USA
| |
Collapse
|
46
|
Wei SC, Xu L, Li WH, Li Y, Guo SF, Sun XR, Li WW. Risk stratification in GIST: shape quantification with CT is a predictive factor. Eur Radiol 2020; 30:1856-1865. [PMID: 31900704 PMCID: PMC7062662 DOI: 10.1007/s00330-019-06561-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/19/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022]
Abstract
Background Tumor shape is strongly associated with some tumor’s genomic subtypes and patient outcomes. Our purpose is to find the relationship between risk stratification and the shape of GISTs. Methods A total of 101 patients with primary GISTs were confirmed by pathology and immunohistochemistry and underwent enhanced CT examination. All lesions’ pathologic sizes were 1 to 10 cm. Points A and B were the extremities of the longest diameter (LD) of the tumor and points C and D the extremities of the small axis, which was the longest diameter perpendicular to AB. The four angles of the quadrangle ABCD were measured and each angle named by its summit (A, B, C, D). For regular lesions, we took angles A and B as big angle (BiA) and small angle (SmA). For irregular lesions, we compared A/B ratio and D/C ratio and selected the larger ratio for analysis. The chi-square test, t test, ROC analysis, and hierarchical or binary logistic regression analysis were used to analyze the data. Results The BiA/SmA ratio was an independent predictor for risk level of GISTs (p = 0.019). With threshold of BiA at 90.5°, BiA/SmA ratio at 1.35 and LD at 6.15 cm, the sensitivities for high-risk GISTs were 82.4%, 85.3%, and 83.8%, respectively; the specificities were 87.1%, 71%, and 77.4%, respectively; and the AUCs were 0.852, 0.818, and 0.844, respectively. LD could not effectively distinguish between intermediate-risk and high-risk GISTs, but BiA could (p < 0.05). Shape and Ki-67 were independent predictors of the mitotic value (p = 0.036 and p < 0.001, respectively), and the accuracy was 87.8%. Conclusions Quantifying tumor shape has better predictive efficacy than LD in predicting the risk level and mitotic value of GISTs, especially for high-risk grading and mitotic value > 5/50HPF. Key Points • The BiA/SmA ratio was an independent predictor affecting the risk level of GISTs. LD could not effectively distinguish between intermediate-risk and high-risk GISTs, but BiA could. • Shape and Ki-67 were independent predictors of the mitotic value. • The method for quantifying the tumor shape has better predictive efficacy than LD in predicting the risk level and mitotic value of GISTs.
Collapse
Affiliation(s)
- Sheng-Cai Wei
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No 440 Jiyan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Liang Xu
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No 440 Jiyan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Wan-Hu Li
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No 440 Jiyan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Yun Li
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No 440 Jiyan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Shou-Fang Guo
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No 440 Jiyan Road, Jinan, 250117, Shandong Province, People's Republic of China
| | - Xiao-Rong Sun
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No 440 Jiyan Road, Jinan, 250117, Shandong Province, People's Republic of China.
| | - Wen-Wu Li
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No 440 Jiyan Road, Jinan, 250117, Shandong Province, People's Republic of China.
| |
Collapse
|
47
|
Ameli S, Shaghaghi M, Kamel IR, Zaheer A. Therapy Response Imaging in Hepatobiliary and Pancreatic Malignancies. MEDICAL RADIOLOGY 2020:117-137. [DOI: 10.1007/978-3-030-31171-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
48
|
PET in Gastrointestinal, Pancreatic, and Liver Cancers. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
49
|
Habbel VSA, Zeile M, Stavrou GA, Wacker F, Brüning R, Oldhafer KJ, Rodt T. Correlation between SACE (Subjective Angiographic Chemoembolization Endpoint) score and tumor response and its impact on survival after DEB-TACE in patients with hepatocellular carcinoma. Abdom Radiol (NY) 2019; 44:3463-3479. [PMID: 31332502 DOI: 10.1007/s00261-019-02128-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE To asses angiographic and computed tomographic success criteria during and after transcatheter arterial drug-eluting bead chemoembolization (DEB-TACE) in patients with hepatocellular carcinoma (HCC) and its impact on progression-free survival (PFS) and overall survival (OS). METHODS In this retrospective single-center study, 50 patients with unresectable HCC having undergone DEB-TACE from January 2010 to July 2015 were assessed. The angiographic endpoint was classified by Subjective Angiographic Chemoembolization Endpoint (SACE) scale. Relative tumor density in arterial (DArt) and portal venous phase (DPV) computed tomography post- versus pre-DEB-TACE were calculated, respectively. Tumor response according to modified Response Criteria in Solid Tumors (mRECIST) was assessed. Univariate Kaplan-Meier and Cox regression analysis were carried out. RESULTS SACE scores I, II, III, and IV were found in 1 (2%), 20 (40%), 15 (30%), and 14 (28%) patients, respectively. Median OS and PFS were 14.2 and 5.5 months, respectively. Death rates at 6 months, 1 year and 2 years were 24%, 38%, and 52%, respectively. SACE score during DEB-TACE significantly correlated with local and overall mRECIST results (local: p < 0.001, r = 0.49, overall: p = 0.042, r = 0.29) and inversely correlated with DPV (p = 0.005, r = - 0.40). In univariate analysis, progressive disease (PD) according to mRECIST and increase of DArt and DPV were associated with significantly shorter PFS. Modified RECIST independently predicted OS (hazard ratio for complete remission vs. PD = 0.15, 95% confidence interval 0.03-0.68, p = 0.014). CONCLUSIONS A direct impact of SACE on PFS or OS could not be shown. However, SACE significantly correlated with local and overall mRECIST tumor response that again significantly predicted OS. We therefore postulate an indirect impact of SACE on OS. Consequently, complete embolization should be attempted.
Collapse
|
50
|
Doi T, Kurokawa Y, Sawaki A, Komatsu Y, Ozaka M, Takahashi T, Naito Y, Ohkubo S, Nishida T. Efficacy and safety of TAS-116, an oral inhibitor of heat shock protein 90, in patients with metastatic or unresectable gastrointestinal stromal tumour refractory to imatinib, sunitinib and regorafenib: a phase II, single-arm trial. Eur J Cancer 2019; 121:29-39. [PMID: 31536852 DOI: 10.1016/j.ejca.2019.08.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/08/2019] [Accepted: 08/14/2019] [Indexed: 12/31/2022]
Abstract
AIM We evaluated the efficacy and safety of TAS-116, a novel class of an orally active selective inhibitor of heat shock protein 90, in patients with advanced gastrointestinal stromal tumour (GIST) after failure of three or more lines of standard treatment with imatinib, sunitinib and regorafenib. METHODS In this single-arm phase II study, patients received 160 mg/day oral TAS-116 for five consecutive days, followed by a 2-day rest. The primary end-point was centrally assessed progression-free survival (PFS). The secondary end-points were objective response rate, disease control rate, overall survival (OS), metabolic response rate, safety, pharmacokinetics and pharmacogenomics. RESULTS Forty-one patients were enrolled in Japan, and 40 patients underwent efficacy and safety evaluation. At the cut-off date, the median PFS was 4.4 months (95% confidence interval [CI], 2.8-6.0) and 12-week progression-free rate was 73.4% (95% CI, 58.1-88.7). Thirty-four patients (85.0%) had stable disease for ≥ 6 weeks. The median OS was 11.5 months (95% CI, 7.0-not reached). All patients experienced at least one treatment-related adverse event (AE), including diarrhoea (80.0%), decreased appetite (45.0%) and increase in blood creatinine level (42.5%). Grade ≥3 AEs and treatment-related grade ≥3 AEs occurred in 23 (57.5%) and 21 (52.5%) patients, respectively. All AEs resolved after dose modification, and no TAS-116-related AEs led to treatment discontinuation. CONCLUSION TAS-116 showed significant activity in advanced GIST refractory to standard treatment. Further development of TAS-116 is warranted. TRIAL REGISTRATION JapicCTI-163182.
Collapse
Affiliation(s)
| | | | - Akira Sawaki
- Fujita Health University Hospital, Nagoya, Japan
| | | | - Masato Ozaka
- The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | - Yoichi Naito
- National Cancer Center Hospital East, Chiba, Japan
| | | | | |
Collapse
|