1
|
Manlubatan SIT, Reyes JAS, Lopez MPJ, Iraola-Davoco KJ. Monophasic synovial sarcoma arising from the rectum presenting as intractable anal pain. BMJ Case Rep 2025; 18:e261817. [PMID: 39809494 DOI: 10.1136/bcr-2024-261817] [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: 01/16/2025] Open
Abstract
Synovial sarcoma is a rare malignant mesenchymal tumour typically found in the extremities, but it can also develop in the gastrointestinal tract, with the upper rectum being the most common site. We describe a case of a man in his 60s diagnosed with monophasic synovial sarcoma in the lower rectum, presenting with severe, intractable anal pain.
Collapse
|
2
|
Mousavi S, Ono Y, VanderLaan PA, Guzmán-Arocho YD. Gastrointestinal stromal tumors in fine-needle aspiration biopsies. Diagn Cytopathol 2024; 52:575-581. [PMID: 38396207 DOI: 10.1002/dc.25285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024]
Abstract
Gastrointestinal stromal tumors (GISTs), although rare, are the most common mesenchymal neoplasms of the gastrointestinal tract. Their potential for malignancy underscores the significance of identifying them through cytomorphologic findings and pertinent immunohistochemical markers. GISTs can emerge anywhere along the gastrointestinal tract with a predilection for the stomach. The clinical manifestations vary from nonspecific abdominal symptoms to incidental discovery during diagnostic interventions for unrelated signs and symptoms. Cytologically, GIST aspirates contain spindle or epithelioid cells with immunoreactivity for CD117/c-KIT, DOG-1, and CD34. Molecularly, KIT or PDGFRA mutations are prevalent, guiding targeted therapy with tyrosine kinase inhibitors. Distinct subtypes like succinate dehydrogenase-deficient GISTs pose challenges, often affecting younger individuals and displaying unique features. Histologically, GISTs are graded by mitotic rates, aiding prognostication. Distinguishing GISTs from similar entities is pivotal, necessitating attention to their immunostaining patterns for making an accurate diagnosis and molecular alterations for effectively planning treatment. Common differential diagnoses include leiomyoma, schwannoma, and solitary fibrous tumor. This article presents a classic GIST case and showcases relatively simple diagnostic clues for identifying similar lesions that may occur in diverse locations.
Collapse
Affiliation(s)
- Seyedreza Mousavi
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Yuho Ono
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Paul A VanderLaan
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Yaileen D Guzmán-Arocho
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
van der Burg SJ, Bleckman RF, van der Sluis PC, Hartgrink HH, Reyners AK, Bonenkamp JJ, van Sandick JW, Wouters MW, van Houdt WJ, Schrage YM. Improvement of perioperative outcomes of gastric gastrointestinal stromal tumour (GIST) resections and the influence of minimal invasive surgery. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108479. [PMID: 38901292 DOI: 10.1016/j.ejso.2024.108479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/28/2024] [Accepted: 06/10/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Safety of minimally invasive surgery (MIS) for gastrointestinal stromal tumours (GISTs) is still under debate since it might increase the risk of tumour rupture, especially in larger tumours. The aim of this study was to investigate trends in treatment and perioperative outcomes of patients undergoing resections of gastric GISTs over time. METHODS This was a multicentre retrospective study of consecutive patients who underwent wedge resection or partial gastrectomy for localized gastric GIST at five GIST reference centres between January 2009 and January 2022. To evaluate changes in treatment and perioperative outcomes over time, patients were divided into four equal periods. Perioperative outcomes were analysed separately and as a novel composite measure textbook outcome (TO). RESULTS In total 385 patients were included. Patient and tumour characteristics did not change over time, except for median age (62-65-68-68 years, p = 0.002). The proportion of MIS increased (4.0%-9.8%-37.4%-53.0 %, p < 0.001). Postoperative complications (Clavien Dindo ≥2; 22%-15%-11%-10 %, p = 0.146), duration of admission (6-6-5-4 days, p < 0.001) and operating time (92-94-77-73 min, p = 0.007) decreased over time while TO increased (54.0%- 52.7%-65.9%-76.0 %, p < 0.001). No change was seen in perioperative ruptures (6.0%- 3.6%-1.6%-3.0 %, p = 0.499). MIS was correlated with less CD ≥ 2 complications (p = 0.006), shorter duration of admission (p < 0.001) and more TO (p < 0.001). Similar results were observed in tumours ≤5 cm and >5 cm. CONCLUSION A larger percentage of gastric GIST were treated with MIS over time. MIS was correlated with less complications, shorter duration of admission and more TO. Tumour rupture rates remained low over time.
Collapse
Affiliation(s)
- Stijn Jc van der Burg
- The Netherlands Cancer Institute, Department of Surgical Oncology, Amsterdam, the Netherlands
| | - Roos F Bleckman
- University of Groningen, University Medical Centre Groningen, Department of Medical Oncology, Groningen, the Netherlands
| | - Pieter C van der Sluis
- Erasmus MC Cancer Institute, Department of Surgical Oncology, Rotterdam, the Netherlands
| | - Henk H Hartgrink
- Leiden University Medical Centre, Department of Surgical Oncology, Leiden, the Netherlands
| | - An Kl Reyners
- University of Groningen, University Medical Centre Groningen, Department of Medical Oncology, Groningen, the Netherlands
| | - Johannes J Bonenkamp
- Radboud University Medical Centre, Department of Surgical Oncology, Nijmegen, the Netherlands
| | - Johanna W van Sandick
- The Netherlands Cancer Institute, Department of Surgical Oncology, Amsterdam, the Netherlands
| | - Michel Wjm Wouters
- The Netherlands Cancer Institute, Department of Surgical Oncology, Amsterdam, the Netherlands
| | - Winan J van Houdt
- The Netherlands Cancer Institute, Department of Surgical Oncology, Amsterdam, the Netherlands
| | - Yvonne M Schrage
- The Netherlands Cancer Institute, Department of Surgical Oncology, Amsterdam, the Netherlands.
| |
Collapse
|
4
|
Ali RH, Alsaber AR, Mohanty AK, Alnajjar A, Mohammed EMA, Alateeqi M, Jama H, Almarzooq A, Benobaid N, Alqallaf Z, Ahmed AA, Bahzad S, Alkandari M. Molecular Profiling of KIT/PDGFRA-Mutant and Wild-Type Gastrointestinal Stromal Tumors (GISTs) with Clinicopathological Correlation: An 18-Year Experience at a Tertiary Center in Kuwait. Cancers (Basel) 2024; 16:2907. [PMID: 39199677 PMCID: PMC11352935 DOI: 10.3390/cancers16162907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
In gastrointestinal stromal tumors (GISTs), identifying prototypical mutations in the KIT/PDGFRA oncogenes, or in rare alternate genes, is essential for prognostication and predicting response to tyrosine kinase inhibitors. Conversely, wild-type GISTs (WT-GIST), which lack known mutations, have limited treatment options. Data on the mutational landscape of GISTs and their impact on disease progression are very limited in Kuwait. Using a targeted next-generation sequencing panel, we investigated the spectrum and frequency of KIT, PDGFRA, and RAS-pathway-related mutations in 95 out of 200 GISTs diagnosed at Kuwait Cancer Center from 2005 to 2023 and assessed their correlation with clinicopathological parameters. Among the 200 tumors (median age 55 years; 15-91), 54% originated in the stomach, 33% in the small bowel, 7% in the colorectum, 1.5% in the peritoneum, and 4.5% had an unknown primary site. Of the 95 molecularly profiled cases, 88% had a mutation: KIT (61%), PDGFRA (25%), NF1 (2%), and one NTRK1 rearrangement. Ten WT-GISTs were identified (stomach = 6, small bowel = 2, and colorectum = 2). WT-GISTs tended to be smaller (median 4.0 cm; 0.5-8.0) (p = 0.018), with mitosis ≤5/5 mm2, and were of lower risk (p = 0.019). KIT mutations were an adverse indicator of disease progression (p = 0.049), while wild-type status did not significantly impact progression (p = 0.934). The genetic landscape in this cohort mirrors that of global studies, but regional collaborations are needed to correlate outcomes with genetic variants.
Collapse
Affiliation(s)
- Rola H. Ali
- Department of Pathology, College of Medicine, Kuwait University, Safat 13110, Kuwait
- Histopathology Laboratory, Sabah Hospital, Sabah Medical District, Safat 13001, Kuwait
| | - Ahmad R. Alsaber
- Department of Management, College of Business and Economics, American University of Kuwait, Safat 13034, Kuwait;
| | - Asit K. Mohanty
- Department of Medical Oncology, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (A.K.M.); (A.A.)
| | - Abdulsalam Alnajjar
- Department of Medical Oncology, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (A.K.M.); (A.A.)
| | - Eiman M. A. Mohammed
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Mona Alateeqi
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Hiba Jama
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Ammar Almarzooq
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Noelle Benobaid
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Zainab Alqallaf
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Amir A. Ahmed
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Shakir Bahzad
- Molecular Genetics Laboratory, Kuwait Cancer Center, Sabah Medical District, Safat 13001, Kuwait; (E.M.A.M.); (M.A.); (H.J.); (A.A.); (N.B.); (Z.A.); (A.A.A.); (S.B.)
| | - Mohammad Alkandari
- Histopathology Laboratory, Farwaniya Hospital, Sabah Al Nasser Area 92426, Kuwait;
| |
Collapse
|
5
|
Tonni G, Palicelli A, Bassi MC, Torricelli F, Vacca I, Aguzzoli L, Mandato VD. Gastrointestinal Stromal Tumors (GISTs) Mimicking Primary Ovarian Tumors or Metastasizing to the Ovaries: A Systematic Literature Review. Cancers (Basel) 2024; 16:2305. [PMID: 39001368 PMCID: PMC11240519 DOI: 10.3390/cancers16132305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
Background: Gastrointestinal stromal tumors (GISTs) are a rare neoplasm, sometimes mimicking primary ovarian tumors (OTs) and/or metastasizing to the ovaries (M-OT). We performed a systematic literature review (SLR) of OTs and M-OTs, investigating differences in recurrence-free and overall survival. Methods: Our SLR was performed according to PRISMA guidelines, searching in Pubmed, Scopus, and Web of Science databases from inception until 21 April 2024. Results: Overall, 59 OTs (Group 1) and 21 M-OTs (Group 2) were retrieved. The absence of residual disease after surgery was achieved significantly in a higher percentage of patients with Group 1 GISTs (91.5%) compared with Group 2 GISTs (57.1%). Chemotherapy was more frequently administered to Group 2 patients (33% vs. 0%). Recurrence and deaths for disease were significantly more frequent in Group 2 than Group 1 cases (54.5% vs. 6.8%, and 37.5% vs. 9.8%, respectively). Conclusions: GISTs can rarely mimic primary ovarian cancers or even more rarely metastasize to the ovaries. Group 1 GISTs occurred in younger women, were not associated with elevated tumor markers, and had a better prognosis. In contrast, Group 2 GISTs occurred in older women, may exhibit elevated tumor markers, and presented a worse prognosis. However, no significant statistical difference for survival between the two studied groups was detected. Computed tomography scans can define the size of GISTs, which correlate to stage and prognostic risk classes. The gold standard treatment is complete surgical resection, which was achieved in almost all cases of Group 1 GISTs and in half of Group 2. Histopathology and immunohistochemistry are essential for the final diagnosis and guide chemotherapy treatment.
Collapse
Affiliation(s)
- Gabriele Tonni
- Department of Obstetrics and Neonatology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Azienda USL-IRCCS di Reggio Emilia, Via Amendola 2, 42123 Reggio Emilia, Italy
| | - Andrea Palicelli
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Maria Chiara Bassi
- Senior Librarian, Biblioteca Medica, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Federica Torricelli
- Translational Research Laboratory, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Ilaria Vacca
- Department of Obstetrics and Gynecologic Oncology, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Lorenzo Aguzzoli
- Department of Obstetrics and Gynecologic Oncology, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Vincenzo Dario Mandato
- Department of Obstetrics and Gynecologic Oncology, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| |
Collapse
|
6
|
D’Ambrosio L, Fumagalli E, De Pas TM, Nannini M, Bertuzzi A, Carpano S, Boglione A, Buonadonna A, Comandini D, Gasperoni S, Vincenzi B, Brunello A, Badalamenti G, Maccaroni E, Baldi GG, Merlini A, Mogavero A, Ligorio F, Pennacchioli E, Conforti F, Manessi G, Aliberti S, Tolomeo F, Fiore M, Sbaraglia M, Dei Tos AP, Stacchiotti S, Pantaleo MA, Gronchi A, Grignani G. Guideline-Based Follow-Up Outcomes in Patients With Gastrointestinal Stromal Tumor With Low Risk of Recurrence: A Report From the Italian Sarcoma Group. JAMA Netw Open 2023; 6:e2341522. [PMID: 37930700 PMCID: PMC10628737 DOI: 10.1001/jamanetworkopen.2023.41522] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
Importance Gastrointestinal stromal tumor (GIST) follow-up is recommended by international guidelines, but data on the role of follow-up in patients with low relapse risk are missing. For these patients, the potential benefit of anticipating recurrence detection should be weighed against psychological burden and radiologic examination loads in terms of costs and radiation exposure. Objective To evaluate the outcomes of guideline-based follow-up in low-risk GIST. Design, Setting, and Participants This multi-institutional retrospective cohort study involving Italian Sarcoma Group reference institutions evaluated patients with GIST who underwent surgery between January 2001 and June 2019. Median follow-up time was 69.2 months. Data analysis was performed from December 15, 2022, to March 20, 2023. Patients with GIST at low risk according to Armed Forces Institute of Pathology criteria were included provided adequate clinical information was available: primary site, size, mitotic index, surgical margins, and 2 or more years of follow-up. Exposures All patients underwent follow-up according to European Society for Medical Oncology (ESMO) guidelines. Main Outcomes and Measures The primary outcome was the number of tests needed to identify a relapse according to ESMO guidelines follow-up plan. Secondary outcomes included relapse rate, relapse timing, disease-free survival (DFS), overall survival (OS), GIST-specific survival (GIST-SS), postrelapse OS, secondary tumor rates, and theoretical ionizing radiation exposure. An exploratory end point, new follow-up schedule proposal for patients with low-risk GIST according to the observed results, was also assessed. Results A total of 737 patients (377 men [51.2%]; median age at diagnosis, 63 [range, 18-86] years) with low-risk GIST were included. Estimated 5-year survival rates were 95.5% for DFS, 99.8% for GIST-SS, and 96.1% for OS. Estimated 10-year survival rates were 93.4% for DFS, 98.1% for GIST-SS, and 91.0% for OS. Forty-two patients (5.7%) experienced disease relapse during follow-up (9 local, 31 distant, 2 both), of which 9 were detected after 10 or more years. This translated into approximately 1 relapse detected for every 170 computed tomography scans performed, with a median radiation exposure of 80 (IQR, 32-112) mSv per patient. Nongastric primary tumor (hazard ratio [HR], 2.09; 95% CI, 1.14-3.83; P = .02), and KIT mutation (HR, 2.77; 95% CI, 1.05-7.27; P = .04) were associated with a higher risk of relapse. Second tumors affected 187 of 737 patients (25%), of which 56 were detected during follow-up and represented the primary cause of death in these patients. Conclusions and Relevance In this cohort study on patients affected by low-risk GISTs, the risk of relapse was low despite a follow-up across 10 or more years. These data suggest the need to revise follow-up schedules to reduce the anxiety, costs, and radiation exposure of currently recommended follow-up strategy.
Collapse
Affiliation(s)
- Lorenzo D’Ambrosio
- Department of Medical Oncology, University of Turin, Turin, Italy
- San Luigi Gonzaga University Hospital, Orbassano, Italy
| | - Elena Fumagalli
- Medical Oncology Unit 2, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Tommaso Martino De Pas
- Medical Oncology Division, Cliniche Humanitas Gavazzeni, Bergamo, Italy
- Previously at Unit of Sarcomas and Thymomas, European Institute of Oncology, Milan, Italy
| | - Margherita Nannini
- Oncology Unit. Department of Medical and Surgical Sciences, University of Bologna, 40138, Bologna, Italy
| | - Alexia Bertuzzi
- Medical Oncology, Humanitas Cancer Center, Rozzano (MI), Italy
| | - Silvia Carpano
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Angela Buonadonna
- Sarcoma and gastrointestinal tumors Unit, Centro di Riferimento Oncologico, Aviano, Italy
| | - Danila Comandini
- Medical Oncology 1, Ospedale Policlinico San Martino, University of Genova, Genova, Italy
| | - Silvia Gasperoni
- Clinical Oncology Unit, Oncology Department and Robotic Surgery, AOU Careggi, Florence, Italy
| | - Bruno Vincenzi
- Medical Oncology, Università Campus Bio-Medico, Rome, Italy
| | | | - Giuseppe Badalamenti
- Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Elena Maccaroni
- Department of Oncology, Azienda Ospedaliero-Universitaria delle Marche, 60126 Ancona, Italy
| | | | - Alessandra Merlini
- Department of Medical Oncology, University of Turin, Turin, Italy
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Andrea Mogavero
- Department of Medical Oncology, University of Turin, Turin, Italy
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Francesca Ligorio
- Medical Oncology Unit 2, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | | | - Fabio Conforti
- Medical Oncology Division, Cliniche Humanitas Gavazzeni, Bergamo, Italy
- Previously at Unit of Sarcomas and Thymomas, European Institute of Oncology, Milan, Italy
| | - Giulia Manessi
- Department of Medical Oncology, University of Turin, Turin, Italy
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Sandra Aliberti
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Francesco Tolomeo
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Marco Fiore
- Sarcoma Service, Surgical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Marta Sbaraglia
- Department of Medicine, University of Padua School of Medicine, Padua, Italy
| | | | | | - Maria Abbondanza Pantaleo
- Oncology Unit. Department of Medical and Surgical Sciences, University of Bologna, 40138, Bologna, Italy
| | - Alessandro Gronchi
- Sarcoma Service, Surgical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Giovanni Grignani
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
- Medical Oncology 2, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| |
Collapse
|
7
|
Mohamed N, Khan M, Hosler G, Tumminello K. Primary vulvar extragastrointestinal stromal tumor in a 77-year-old woman. J Cutan Pathol 2023. [PMID: 37127848 DOI: 10.1111/cup.14432] [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: 10/26/2022] [Revised: 03/07/2023] [Accepted: 03/23/2023] [Indexed: 05/03/2023]
Abstract
Extragastrointestinal stromal tumors (EGISTs) carry the same morphological, immunohistochemical and molecular features as gastrointestinal stromal tumors (GISTs) and involve extragastrointestinal tract soft tissue. The majority of reported EGIST cases arise from intraabdominal, retroperitoneal, or pelvic soft tissue. A significant subset of such tumors originates from the gastrointestinal muscle layer, grows in an exophytic manner, then loses attachment to the gastrointestinal tract. Consequently, true EGISTs are exceedingly rare. Herein, we are reporting a case of a vulvar EGIST. A 77-year-old woman presented with a painless subcutaneous nodule on the right perineum. An excisional biopsy showed a fairly circumscribed bland spindle cell lesion in the dermis. The tumor cells were positive for CD117 and ANO1/DOG-1 and negative for smooth muscle myosin, smooth muscle actin, STAT6, low- and high-molecular-weight cytokeratins, SOX10, MART-1, CD10, S-100 protein, and estrogen and progesterone receptors. A diagnosis of EGIST was made and complete excision was recommended. Superficial/subcutaneous EGISTs are extremely rare, and it is important for dermatopathologists to be aware of this entity as it can be misdiagnosed as more common spindle cell neoplasms, both benign and malignant, including but not limited to smooth muscle neoplasms (leiomyoma/leiomyosarcoma), spindle cell melanoma, and sarcomatoid squamous cell carcinoma.
Collapse
Affiliation(s)
- Nada Mohamed
- Department of Pathology, Texas A&M College of Medicine-Baylor Scott & White Health, Temple, Texas, USA
| | | | - Gregory Hosler
- ProPath, Dallas, Texas, USA
- Department of Dermatology, University of Texas Southwestern, Dallas, Texas, USA
| | | |
Collapse
|
8
|
Catalano F, Cremante M, Dalmasso B, Pirrone C, Lagodin D’Amato A, Grassi M, Comandini D. Molecular Tailored Therapeutic Options for Advanced Gastrointestinal Stromal Tumors (GISTs): Current Practice and Future Perspectives. Cancers (Basel) 2023; 15:cancers15072074. [PMID: 37046734 PMCID: PMC10093725 DOI: 10.3390/cancers15072074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are one of the most common mesenchymal tumors characterized by different molecular alterations that lead to specific clinical presentations and behaviors. In the last twenty years, thanks to the discovery of these mutations, several new treatment options have emerged. This review provides an extensive overview of GISTs’ molecular pathways and their respective tailored therapeutic strategies. Furthermore, current treatment strategies under investigation and future perspectives are analyzed and discussed.
Collapse
Affiliation(s)
- Fabio Catalano
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Malvina Cremante
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Bruna Dalmasso
- Genetica dei Tumori Rari, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Chiara Pirrone
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | | | - Massimiliano Grassi
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence:
| | - Danila Comandini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
9
|
Campora M, Paudice M, Gambella A, Comandini D, Parente P, Sbaraglia M, Dei Tos AP, Grillo F, Mastracci L. Counting mitoses in gastrointestinal stromal tumours (GISTs): variable practices in the real-world setting and their clinical implications. Virchows Arch 2023; 482:589-594. [PMID: 36416965 PMCID: PMC10033612 DOI: 10.1007/s00428-022-03454-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/03/2022] [Accepted: 11/08/2022] [Indexed: 11/24/2022]
Abstract
Mitotic count (MC) is an important prognostic indicator in gastrointestinal stromal tumours (GISTs). Though MC evaluation was initially proposed in 50 HPFs, recent international guidelines recommend that MC be performed on 5 mm2 because HPFs may have different areas depending on the ocular field number (FN) of the utilized light microscope. Performing MC on different areas leads to a non-standardized evaluation and erroneous risk stratification. The aim of the study was to audit real-life MC practices with special emphasis on possible risk stratification errors. A survey was administered to Italian pathologists to evaluate the following: method used for MC (5 mm2 versus 50 HPF); FN of the light microscope; prognostic scheme for risk stratification. Based on the results of the survey, 100 GISTs (25/risk class using Miettinen prognostic scheme) were retrieved and MC performed using 5 mm2 versus the corresponding mm2 area sizes of 50 HPFs with variable FNs (18, 20, 22). The survey demonstrated that the majority of pathologists (64.5%) use 50 HPFs with various FNs leading to excessive area size. The most frequently used prognostic scheme is that by Miettinen. Using this prognostic scheme and counting mitoses in 5 mm2 versus 50 HPFs with FNs 18, 20 and 22, a change in risk class was identified ranging from 10 to 41%, depending on FN. In conclusion, this study demonstrates that MC is still frequently performed on 50 HPF, with area sizes exceeding the specified 5 mm2 by far.
Collapse
Affiliation(s)
| | - Michele Paudice
- IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | | | - Danila Comandini
- Oncology Unit, Department of Oncology and Haematology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Paola Parente
- Fondazione IRCCS Casa Sollievo della Sofferenza, Pathology Unit, San Giovanni Rotondo (FG), Italy
| | - Marta Sbaraglia
- Department of Pathology, Azienda Ospedale-Università Padova, Padua, Italy
| | - Angelo Paolo Dei Tos
- Department of Pathology, Azienda Ospedale-Università Padova, Padua, Italy
- Department of Medicine (DIMED), University of Padua School of Medicine, Padua, Italy
| | - Federica Grillo
- IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Luca Mastracci
- IRCCS Ospedale Policlinico San Martino, Largo Benzi 10, 16132, Genoa, Italy.
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy.
| |
Collapse
|
10
|
Hornick JL, Webster F, Dei Tos AP, Hemmings C, Miettinen M, Oda Y, Raut CP, Rubin BP, Von Mehren M, Wardelmann E, Fletcher CDM. Dataset for reporting of gastrointestinal stromal tumours: recommendations from the International Collaboration on Cancer Reporting (ICCR). Histopathology 2023; 82:376-384. [PMID: 36073677 DOI: 10.1111/his.14791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 09/04/2022] [Accepted: 09/05/2022] [Indexed: 01/20/2023]
Abstract
Gastrointestinal stromal tumours (GISTs) are the most common mesenchymal tumours of the gastrointestinal tract and are among the most frequent sarcomas. Accurate diagnosis, classification, and reporting are critical for prognostication and patient management, including selection of appropriate targeted therapy. Here we report on international consensus-based datasets for the pathology reporting of biopsy and resection specimens of GIST. The datasets were produced under the auspices of the International Collaboration on Cancer Reporting (ICCR), a global alliance of major international pathology and cancer organizations. An international expert panel consisting of pathologists, a surgical oncologist, and a medical oncologist produced a set of core and noncore data items for biopsy and resection specimens based on a critical review and discussion of current evidence. All professionals involved were subspecialized soft tissue tumour experts and affiliated with tertiary referral centres. Commentary was provided for each data item to explain its clinical relevance and the rationale for selection as a core or noncore element. Following international public consultation, the datasets, which include synoptic reporting guides, were finalized and ratified, and published on the ICCR website. These first international datasets for GIST are intended to promote high-quality, standardised pathology reporting. Their widespread adoption will improve consistency of reporting, facilitate multidisciplinary communication, and enhance comparability of data, all of which will ultimately help to improve the management of patients with GIST. All the ICCR datasets, including these on GIST, are freely available worldwide on the ICCR website (www.iccr-cancer.org/datasets).
Collapse
Affiliation(s)
- Jason L Hornick
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Fleur Webster
- International Collaboration on Cancer Reporting, Sydney, NSW, Australia
| | - Angelo Paolo Dei Tos
- Department of Pathology, Azienda Ospedaliera Universitaria di Padova, Padova, Italy.,Department of Medicine, University of Padua School of Medicine, Padua, Italy
| | - Chris Hemmings
- Department of Anatomic Pathology, Canterbury Health Laboratories, Christchurch, New Zealand.,Department of Pathology and Biomedical Science, Christchurch Clinical School, University of Otago School of Medicine, Christchurch, New Zealand
| | - Markku Miettinen
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health Bethesda, Bethesda, MD, USA
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Chandrajit P Raut
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA.,Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Brian P Rubin
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Margaret Von Mehren
- Department of Hematology and Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, University of Münster, Münster, Germany
| | | |
Collapse
|
11
|
Muacevic A, Adler JR, Alderazi AE, Almahari SAI, Alawadhi AM. Gastrointestinal Stromal Tumor Recurrence Presenting as a Small Bowel Obstruction: A Case Report. Cureus 2023; 15:e33806. [PMID: 36655152 PMCID: PMC9841066 DOI: 10.7759/cureus.33806] [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: 01/15/2023] [Indexed: 01/18/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the alimentary tract in adults. The most common site is the stomach, followed by the small intestine. The clinical presentation varies from an incidental finding in asymptomatic patients to a large palpable mass causing complications such as bowel obstruction or viscus perforation. The best imaging modality is a CT scan of the abdomen. Treatment is determined by the size and location of the GISTs. Surgical intervention is considered for resectable tumors, while tyrosine kinase inhibitor therapy is considered for irresectable, metastatic, or recurrent GISTs. In this case report, we present a 30-year-old female who is a known case of gastric GIST and liver metastases. She presented to the emergency department with intestinal obstruction secondary to a recurrent GIST abdominal mass and underwent emergency laparotomy for mass resection. Following surgery, the patient developed aspiration pneumonia, which was treated with proper antibiotics. She was discharged in stable clinical condition with a recommendation to start alternative tyrosine kinase therapy. GISTs are difficult to diagnose preoperatively, as most patients are asymptomatic, and they may present with complications, as in our case, a small bowel obstruction. A proper imaging modality will guide the physician toward the diagnosis, but the final diagnosis will be achieved by biopsy. The diagnosis may be challenging, as small bowel obstruction has many causes, although GISTs should be kept in mind as one of the deferential diagnoses.
Collapse
|
12
|
Yang W, Shou C, Yu J, Wang X, Zhang Q, Yu H, Lin X. Elevated preoperative controlling nutritional status (CONUT) scores as a predictor of postoperative recurrence in gastrointestinal stromal tumors. J Surg Oncol 2022; 126:1191-1198. [PMID: 35912441 DOI: 10.1002/jso.27042] [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: 03/29/2022] [Revised: 06/28/2022] [Accepted: 07/19/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND The controlling nutritional status (CONUT) score is associated with the postoperative outcomes in various types of tumors, and its prognostic role in gastrointestinal stromal tumors (GISTs) needs to be clarified. METHODS Patients with completely resected primary GISTs in the absence of imatinib adjuvant therapy were included. Recurrence-free survival (RFS) was estimated with the Kaplan-Meier method and compared using log-rank test. Prognostic factors were compared using Cox proportional hazards model. RESULTS A total of 455 patients were included. The median follow-up time was 132.0 months (range: 7.0-253.0). Recurrence/metastasis developed in 92 (20.2%) patients. Patients were assigned to three groups: 219 (48.1%) were in normal nutrition group (CONUT = 0-1), 196 (43.1%) were in mild malnutrition group (CONUT = 2-4) and 40 (8.8%) were in moderate-severe malnutrition group (CONUT ≥ 5). Nongastric primary tumor site, large tumor size, high mitotic index, tumor rupture and high CONUT score were independent prognostic factors for shorter RFS using multivariate analysis (p < 0.05). CONCLUSIONS Elevated preoperative CONUT score was a predictor of recurrence for patients with resected GIST. The clinical application of the CONUT score is simple and feasible, and might contribute to the individualized treatment of GIST patients.
Collapse
Affiliation(s)
- Weili Yang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chunhui Shou
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiren Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaodong Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qing Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hang Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xianke Lin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
13
|
Song IH, Sung YE, Kang J, Lee A, Lee SH. Molecular and immunohistochemical comparison between primary gastrointestinal mucosal melanomas and atypical gastrointestinal stromal tumors. Pathol Res Pract 2022; 240:154189. [DOI: 10.1016/j.prp.2022.154189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 11/07/2022]
|
14
|
Prognostic Nomogram for Gastrointestinal Stromal Tumors after Surgery Based on the SEER Database. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5639174. [PMID: 36420093 PMCID: PMC9678471 DOI: 10.1155/2022/5639174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/25/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022]
Abstract
We aimed to determine prognostic factors and develop an effective and practical nomogram for predicting cancer-specific survival in gastrointestinal stromal tumor (GIST) patients. Postoperative data were obtained from the SEER database (2000-2018). Patients were divided into training and validation cohorts at random (7 : 3). Prognostic factors were screened, and a prognostic nomogram was established using log-rank testing and Cox regression. We used DCA, ROC curves, C-index, and calibration curves to evaluate our model's predictive performance. The clinical value of the nomogram and the modified National Institute of Health (M-NIH) classification were compared using the NRI and IDI. The Kaplan-Meier method was applied to examine survival by risk group, and log-rank tests were applied to compare variations in survival curves. Independent prognostic risk factors associated with cancer-specific survival on multivariate Cox proportional hazards regression analysis were age, race, and tumor location, size, grade, and stage. Clinically relevant variables need to be considered in addition to statistically significant variables when developing prognostic models to aid clinical decision-making. We included two additional variables (mitotic rate and chemotherapy) when constructing the prognostic model. The C-index was 0.766 (95% confidence interval (CI): 0.737-0.794) in the training cohort and 0.795 (95% CI: 0.754-0.836) in the internal validation group suggesting robustness. The areas under the ROC curve for three-year and five-year survival were >0.700, indicating satisfactory discrimination. The calibration curves showed good agreement between the predictions of the nomogram and the actual results. The NRI (0.346 for 3-year and 0.265 for 5-year cancer-specific survival for patients with GIST (GSS) prediction; validation cohort: 0.356 for 3-year and 0.246 for 5-year GSS prediction) and IDI values (0.047 for 3-year and 0.060 for 5-year GSS prediction; validation cohort: 0.071 for 3-year and 0.084 for 5-year GSS prediction) suggested that the established nomogram performed significantly better than the M-NIH classification. The DCA indicated that the nomogram was clinically useful and had a high discriminative ability in identifying patients who were at high risk of poor outcomes. According to nomogram findings, patients were divided into three groups (high, moderate, and low risk), with significantly different prognoses in both cohorts. Our nomogram satisfactorily predicted survival in postsurgical GIST patients, which may assist clinicians to evaluate the postoperative status and guide subsequent treatments.
Collapse
|
15
|
Mathias-Machado MC, de Jesus VHF, de Carvalho Oliveira LJ, Neumann M, Peixoto RD. Current Molecular Profile of Gastrointestinal Stromal Tumors and Systemic Therapeutic Implications. Cancers (Basel) 2022; 14:5330. [PMID: 36358751 PMCID: PMC9656487 DOI: 10.3390/cancers14215330] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 07/25/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are malignant mesenchymal tumors arising from the intestinal pacemaker cells of Cajal. They compose a heterogenous group of tumors due to a variety of molecular alterations. The most common gain-of-function mutations in GISTs are either in the KIT (60-70%) or platelet-derived growth factor receptor alpha (PDGFRA) genes (10-15%), which are mutually exclusive. However, a smaller subset, lacking KIT and PDGFRA mutations, is considered wild-type GISTs and presents distinct molecular findings with the activation of different proliferative pathways, structural chromosomal and epigenetic changes, such as inactivation of the NF1 gene, mutations in the succinate dehydrogenase (SDH), BRAF, and RAS genes, and also NTRK fusions. Currently, a molecular evaluation of GISTs is imperative in many scenarios, aiding in treatment decisions from the (neo)adjuvant to the metastatic setting. Here, we review the most recent data on the molecular profile of GISTs and highlight therapeutic implications according to distinct GIST molecular subtypes.
Collapse
Affiliation(s)
| | | | | | - Marina Neumann
- Centro Paulista de Oncologia (Oncoclínicas), São Paulo 04538-132, Brazil
| | | |
Collapse
|
16
|
Abstract
Gastrointestinal stromal tumors (GISTs) are rare malignancies of the gastrointestinal tract but are the most common sarcoma. This review covers aspects of the care of patients with GIST relevant to surgeons. In particular, management of sub-2 cm GISTs, the utility of neoadjuvant and adjuvant therapy for primary GISTs, and indications for surgery in the setting of metastatic disease are discussed.
Collapse
Affiliation(s)
- Ilaria Caturegli
- Department of Surgery, Brigham and Women's Hospital, Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| | - Chandrajit P Raut
- Department of Surgery, Brigham and Women's Hospital, Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
17
|
Feng X, Li H, Fourquet J, Brahmi M, Dufresne A, Meurgey A, Ray-Coquard I, Wang Q, Bollard J, Ducimetiere F, Chibon F, Blay JY. Refining Prognosis in Localized Gastrointestinal Stromal Tumor: Clinical Significance of Phosphatase and Tensin Homolog Low Expression and Gene Loss. JCO Precis Oncol 2022; 6:e2200129. [PMID: 36001861 PMCID: PMC9489173 DOI: 10.1200/po.22.00129] [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: 11/20/2022] Open
Abstract
To investigate the use of PTEN biomarker to improve prognostic stratification in patients with localized gastrointestinal stromal tumor (GIST).
Collapse
Affiliation(s)
- Xiaolan Feng
- Department of Medicine Oncology, Tom Baker Cancer Center, Calgary, Alberta, Canada.,Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Haocheng Li
- Department of Mathematics and Statistics, University of Calgary, Calgary, Alberta, Canada
| | - Joanna Fourquet
- OncoSarc, INSERM U1037, Cancer Research Center in Toulouse, Toulouse, France
| | - Mehdi Brahmi
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Armelle Dufresne
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Alexandra Meurgey
- Department of Pathology, Institut Claudius Régaud, IUCT-Oncopole, Toulouse, France
| | | | - Qing Wang
- Department of Biopathology, Centre Léon Bérard, Lyon, France.,Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Julien Bollard
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | | | - Frederic Chibon
- OncoSarc, INSERM U1037, Cancer Research Center in Toulouse, Toulouse, France.,Department of Pathology, Institut Claudius Régaud, IUCT-Oncopole, Toulouse, France
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France.,University Claude Bernard Lyon, Lyon, France
| |
Collapse
|
18
|
Gastrointestinal Stromal Tumors Mimicking Gynecologic Disease: Clinicopathological Analysis of 20 Cases. Diagnostics (Basel) 2022; 12:diagnostics12071563. [PMID: 35885469 PMCID: PMC9319443 DOI: 10.3390/diagnostics12071563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/25/2022] [Accepted: 06/25/2022] [Indexed: 11/29/2022] Open
Abstract
Diagnosis of pelvic gastrointestinal stromal tumors (GISTs) can be challenging because of their nonspecific presentation and similarity to gynecological neoplasms. In this series, we describe the clinicopathological features of 20 GIST cases: 18 patients presented with pelvic mass and/or abdominal pain concerning gynecological disease; 2 patients presented with a posterior rectovaginal mass or an anorectal mass. Total abdominal hysterectomy and/or salpingo-oophorectomy (unilateral or bilateral) were performed in 13 cases. Gross and histological examination revealed that the ovary/ovaries were involved in three cases, the uterus in two cases, the vagina in two cases and the broad ligament in one case. Immunohistochemically, all tumors (20/20, 100%) were diffusely immunoreactive for c-KIT. The tumor cells were also diffusely positive for DOG-1 (10/10, 100%) and displayed focal to diffuse positivity for CD34 (11/12, 92%). Desmin was focally and weakly expressed in 1 of the 14 tested tumors (1/14, 7%), whereas 2 of 8 tumors (2/8, 25%) showed focal SMA positivity. At the molecular level, 7 of 8 (87.5%) GISTs with molecular analysis contained c-KIT mutations with the second and third c-KIT mutations detected in some recurrent tumors. In addition to c-KIT mutation, a pathogenic RB1 mutation was detected in two cases. We extensively discussed these cases focusing on their differential diagnosis described by the submitting pathologists during consultation. Our study emphasizes the importance of precision diagnosis of GISTs. Alertness to this entity in unusual locations, in combination with clinical history, morphological features as well as immunophenotype, is crucial in leading to a definitive classification.
Collapse
|
19
|
Intraoperative Near-Infrared Fluorescence Imaging with Indocyanine Green for Identification of Gastrointestinal Stromal Tumors (GISTs), a Feasibility Study. Cancers (Basel) 2022; 14:cancers14061572. [PMID: 35326721 PMCID: PMC8946640 DOI: 10.3390/cancers14061572] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/14/2022] [Accepted: 03/14/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Surgical resection plays a pivotal role in the treatment of GIST patients. The current study aims to explore the use of near-infrared fluorescence imaging to optimize the intraoperative tumor identification of GISTs. For this purpose, the potential and limitations of the widely used, and non-specific, tracer indocyanine green were assessed in a multicenter study including 10 patients. Our results show that GISTs typically have similar fluorescence intensity to the surrounding tissue, within several minutes after the intravenous administration of indocyanine green. These findings justify future research into specific fluorescent tracers for GISTs, and set a reference for future intraoperative imaging trials. Abstract Background: Optimal intraoperative tumor identification of gastrointestinal stromal tumors (GISTs) is important for the quality of surgical resections. This study aims to assess the potential of near-infrared fluorescence (NIRF) imaging with indocyanine green (ICG) to improve intraoperative tumor identification. Methods: Ten GIST patients, planned to undergo resection, were included. During surgery, 10 mg of ICG was intravenously administered, and NIRF imaging was performed at 5, 10, and 15 min after the injection. The tumor fluorescence intensity was visually assessed, and tumor-to-background ratios (TBRs) were calculated for exophytic lesions. Results: Eleven GIST lesions were imaged. The fluorescence intensity of the tumor was visually synchronous and similar to the background in five lesions. In one lesion, the tumor fluorescence was more intense than in the surrounding tissue. Almost no fluorescence was observed in both the tumor and healthy peritoneal tissue in two patients with GIST lesions adjacent to the liver. In three GISTs without exophytic growth, no fluorescence of the tumor was observed. The median TBRs at 5, 10, and 15 min were 1.0 (0.4–1.2), 1.0 (0.5–1.9), and 0.9 (0.7–1.2), respectively. Conclusion: GISTs typically show similar fluorescence intensity to the surrounding tissue in NIRF imaging after intraoperative ICG administration. Therefore, intraoperatively administered ICG is currently not applicable for adequate tumor identification, and further research should focus on the development of tumor-specific fluorescent tracers for GISTs.
Collapse
|
20
|
de Nonneville A, Finetti P, Picard M, Monneur A, Pantaleo MA, Astolfi A, Ostrowski J, Birnbaum D, Mamessier E, Bertucci F. CSPG4 Expression in GIST Is Associated with Better Prognosis and Strong Cytotoxic Immune Response. Cancers (Basel) 2022; 14:cancers14051306. [PMID: 35267618 PMCID: PMC8909029 DOI: 10.3390/cancers14051306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Gastrointestinal stromal tumors (GIST) are the most frequent sarcomas of the gastrointestinal tract. Identification of novel prognostic and/or therapeutic targets is a major issue to overcome tyrosine kinase inhibitors resistances. CSPG4, a cell surface proteoglycan, emerged as a potential therapeutic target for immune therapy in different cancers, including sarcomas. CSPG4 expression has never been studied in GIST. In this work we analyzed CSPG4 mRNA expression in a large series of clinical GIST samples given the scarcity of disease (n = 309 patients). We find that high CSPG4 expression is independently associated with disease-free survival, and with an immune landscape favorable to induce strong cytotoxic immune response after NK cell stimulation. Our results suggest the potential value of CSPG4-specific chimeric antigen receptor-redirected cytokine-induced killer lymphocytes treatment in GIST, notably “CSPG4-high” tumors, and calls for preclinical validation, drug testing in vivo, then in clinical trials. Abstract The treatment of gastrointestinal stromal tumors (GIST) must be improved through the development of more reliable prognostic factors and of therapies able to overcome imatinib resistance. The immune system represents an attractive tool. CSPG4, a cell surface proteoglycan, emerged as a potential therapeutic target for immune therapy in different cancers, including cell therapy based on CSPG4-specific chimeric antigen receptor (CAR)-redirected cytokine-induced killer lymphocytes (CSPG4-CAR.CIKs) in sarcomas. CSPG4 expression has never been studied in GIST. We analyzed CSPG4 mRNA expression data of 309 clinical GIST samples profiled using DNA microarrays and searched for correlations with clinicopathological and immune features. CSPG4 expression, higher in tumors than normal digestive tissues, was heterogeneous across tumors. High expression was associated with AFIP low-risk, gastric site, and localized stage, and independently with longer postoperative disease-free survival (DFS) in localized stage. The correlations between CSPG4 expression and immune signatures highlighted a higher anti-tumor immune response in “CSPG4-high” tumors, relying on both the adaptive and innate immune system, in which the boost of NK cells by CSPG4-CAR.CIKs might be instrumental, eventually combined with immune checkpoint inhibitors. In conclusion, high CSPG4 expression in GIST is associated with better DFS and offers an immune environment favorable to a vulnerability to CAR.CIKs.
Collapse
Affiliation(s)
- Alexandre de Nonneville
- Predictive Oncology Laboratory, Equipe Labellisée Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseille, France; (A.d.N.); (P.F.); (M.P.); (D.B.); (E.M.)
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, INSERM, 13009 Marseille, France;
| | - Pascal Finetti
- Predictive Oncology Laboratory, Equipe Labellisée Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseille, France; (A.d.N.); (P.F.); (M.P.); (D.B.); (E.M.)
| | - Maelle Picard
- Predictive Oncology Laboratory, Equipe Labellisée Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseille, France; (A.d.N.); (P.F.); (M.P.); (D.B.); (E.M.)
| | - Audrey Monneur
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, INSERM, 13009 Marseille, France;
| | - Maria Abbondanza Pantaleo
- Department of Specialized, Experimental and Diagnostic Medicine, Sant’Orsola-Malpighi Hospital, University of Bologna, 40138 Bologna, Italy; (M.A.P.); (A.A.)
| | - Annalisa Astolfi
- Department of Specialized, Experimental and Diagnostic Medicine, Sant’Orsola-Malpighi Hospital, University of Bologna, 40138 Bologna, Italy; (M.A.P.); (A.A.)
| | - Jerzy Ostrowski
- Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Center of Postgraduate Education, 01-813 Warsaw, Poland;
- Department of Genetics, Maria Sklodowska-Curie National Institute of Oncology, 02-781 Warsaw, Poland
| | - Daniel Birnbaum
- Predictive Oncology Laboratory, Equipe Labellisée Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseille, France; (A.d.N.); (P.F.); (M.P.); (D.B.); (E.M.)
| | - Emilie Mamessier
- Predictive Oncology Laboratory, Equipe Labellisée Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseille, France; (A.d.N.); (P.F.); (M.P.); (D.B.); (E.M.)
| | - François Bertucci
- Predictive Oncology Laboratory, Equipe Labellisée Ligue Nationale Contre Le Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, Inserm UMR1068, CNRS UMR7258, Aix-Marseille University, 13009 Marseille, France; (A.d.N.); (P.F.); (M.P.); (D.B.); (E.M.)
- Department of Medical Oncology, Institut Paoli-Calmettes, Aix-Marseille University, CNRS, INSERM, 13009 Marseille, France;
- Correspondence: ; Tel.: +33-4-91-22-35-37; Fax: +33-4-91-22-36-70
| |
Collapse
|
21
|
Cho H, Nishida T, Takahashi T, Masuzawa T, Hirota S. Impact of the KIT/PDGFRA genotype on prognosis in imatinib-naïve Japanese patients with gastrointestinal stromal tumor. Ann Gastroenterol Surg 2022; 6:241-248. [PMID: 35261949 PMCID: PMC8889858 DOI: 10.1002/ags3.12527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 11/12/2022] Open
Abstract
Background Most gastrointestinal stromal tumors (GIST) harbor a mutation in KIT or platelet-derived growth factor receptor alfa (PDGFRA). Although genotyping is a useful predictive marker of tyrosine kinase inhibitors, whether it can predict prognosis remains controversial. Methods Data on 402 patients with GIST who underwent macroscopically complete surgery and received no neoadjuvant/adjuvant therapy were selected from a prospective GIST database at the three, participating hospitals. The types and locations of KIT and PDGFRA mutations were analyzed by direct sequencing of the amplified genes. The association between the genotypic characteristics and prognosis was then examined. Results Tumor genotypes were analyzed in 398 of 402 (99%) patients, and 120 mutation patterns were identified. KIT mutations had broad malignancy potential which differed according to the type of mutation. Deletion and deletion-insertion type mutations were associated with worse RFS while duplication and substitution type mutations were associated with favorable RFS KIT deletion/deletion-insertion, including codons 557 and 558, were especially associated with worse RFS on multivariate analysis both of all the patients and those with KIT mutations. Conclusions Specific GIST genotypes were significantly associated with a risk of recurrence. Genotype analysis may be useful for predicting the prognosis and determining the indications for adjuvant imatinib in patients with GIST.
Collapse
Affiliation(s)
- Haruhiko Cho
- Department of SurgeryTokyo Metropolitan Cancer and Infectious Diseases CenterKomagome HospitalTokyoJapan
- Department of Gastrointestinal SurgeryKanagawa Cancer CenterYokohamaJapan
| | - Toshirou Nishida
- Department of SurgeryNational Cancer Center HospitalTokyoJapan
- Department of SurgeryJapan Community Health Care OrganizationOsaka HospitalOsakaJapan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuitaJapan
| | - Toru Masuzawa
- Department of SurgeryOsaka Police HospitalOsakaJapan
- Department of Gastrointestinal SurgeryKansai Rosai HospitalAmagasakiJapan
| | - Seiichi Hirota
- Department of Surgical PathologyHyogo College of MedicineNishinomiyaJapan
| |
Collapse
|
22
|
Dermawan JK, Vanderbilt CM, Chang JC, Untch BR, Singer S, Chi P, Tap WD, Antonescu CR. FGFR2::TACC2 fusion as a novel KIT-independent mechanism of targeted therapy failure in a multidrug-resistant gastrointestinal stromal tumor. Genes Chromosomes Cancer 2022; 61:412-419. [PMID: 35170141 DOI: 10.1002/gcc.23030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 11/06/2022] Open
Abstract
Genetic alterations in FGF/FGFR pathway are infrequent in gastrointestinal stromal tumors (GIST), with rare cases of quadruple wildtype GISTs harboring FGFR1 gene fusions and mutations. Additionally, FGF/FGFR overexpression was shown to promote drug resistance to kinase inhibitors in GISTs. However, FGFR gene fusions have not been directly implicated as a mechanism of drug resistance in GISTs. Herein, we report a patient presenting with a primary small bowel spindle cell GIST and concurrent peritoneal and liver metastases displaying an imatinib-sensitive KIT exon 11 in-frame deletion. After an initial 9-month benefit to imatinib, the patient experienced intraabdominal peritoneal recurrence owing to secondary KIT exon 13 missense mutation and FGFR4 amplification. Despite several additional rounds of tyrosine kinase inhibitors (TKI), the patient's disease progressed after 2 years and presented with multiple peritoneal and liver metastases, including one pericolonic mass harboring secondary KIT exon 18 missense mutation, and a concurrent transverse colonic mass with a FGFR2::TACC2 fusion and AKT2 amplification. All tumors, including primary and recurrent masses, harbored an MGA c.7272 T > G (p.Y2424*) nonsense mutation and CDKN2A/CDKN2B/MTAP deletions. The transcolonic mass showed elevated mitotic count (18/10 HPF), as well as significant decrease in CD117 and DOG1 expression, in contrast to all the other resistant nodules that displayed diffuse and strong CD117 and DOG1 immunostaining. The FGFR2::TACC2 fusion resulted from a 742 kb intrachromosomal inversion at the chr10q26.3 locus, leading to a fusion between exons 1-17 of FGFR2 and exons 7-17 TACC2, which preserves the extracellular and protein tyrosine kinase domains of FGFR2. We present the first report of a multi-drug resistant GIST patient who developed an FGFR2 gene fusion as a secondary genetic event to the selective pressure of various TKIs. This case also highlights the heterogeneous escape mechanisms to targeted therapy across various tumor nodules, spanning from both KIT-dependent and KIT-independent off-target activation pathways.
Collapse
Affiliation(s)
- Josephine K Dermawan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Chad M Vanderbilt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jason C Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Brian R Untch
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Samuel Singer
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ping Chi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - William D Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Cristina R Antonescu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
23
|
Palatresi D, Fedeli F, Danti G, Pasqualini E, Castiglione F, Messerini L, Massi D, Bettarini S, Tortoli P, Busoni S, Pradella S, Miele V. Correlation of CT radiomic features for GISTs with pathological classification and molecular subtypes: preliminary and monocentric experience. Radiol Med 2022; 127:117-128. [PMID: 35022956 DOI: 10.1007/s11547-021-01446-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/30/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE Our primary purpose was to search for computed tomography (CT) radiomic features of gastrointestinal stromal tumors (GISTs) that could potentially correlate with the risk class according to the Miettinen classification. Subsequently, assess the existence of features with possible predictive value in differentiating responder from non-responder patients to first-line therapy with Imatinib. METHODS A retrospective study design was carried out using data from June 2009 to December 2020. We analyzed all the preoperative CTs of patients undergoing surgery for GISTs. We segmented non-contrast-enhanced CT (NCECT) and contrast-enhanced venous CT (CECT) images obtained either on three different CT scans (heterogeneous cohort) or on a single CT scan (homogeneous cohort). We then divided the patients into two groups according to Miettinen classification criteria and based on the predictive value of response to first-line therapy with Imatinib. RESULTS We examined 54 patients with pathological confirmation of GISTs. For the heterogeneous cohort, we found a statistically significant relationship between 57 radiomic features for NCECT and 56 radiomic features for CECT using the Miettinen risk classification. In the homogeneous cohort, we found the same relationship between 8 features for the NCECT and 5 features for CECT, all included in the heterogeneous cohort. The various radiomic features are distributed with different values in the two risk stratification groups according to the Miettinen classification. We also found some features for groups predictive of response to first-line therapy with Imatinib. CONCLUSIONS We found radiomic features that correlate with statistical significance for both the Miettinen risk classification and the molecular subtypes of response. All features found in the homogeneous study cohort were also found in the heterogeneous cohort. CT radiomic features may be useful in assessing the risk class and prognosis of GISTs.
Collapse
Affiliation(s)
- Daniele Palatresi
- Department of Radiology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Filippo Fedeli
- Department of Radiology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Ginevra Danti
- Department of Radiology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy.
| | - Elisa Pasqualini
- Pathology Unit, Department of Health Sciences, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Francesca Castiglione
- Histopathology and Molecular Diagnostics Unit, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Luca Messerini
- Department of Experimental and Clinical Medicine, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Daniela Massi
- Pathology Unit, Department of Health Sciences, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Silvia Bettarini
- Medical Physics Department, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Paolo Tortoli
- Medical Physics Department, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Simone Busoni
- Medical Physics Department, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Silvia Pradella
- Department of Radiology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| | - Vittorio Miele
- Department of Radiology, Careggi University Hospital, Largo Brambilla 3, 50134, Florence, Italy
| |
Collapse
|
24
|
Casali PG, Blay JY, Abecassis N, Bajpai J, Bauer S, Biagini R, Bielack S, Bonvalot S, Boukovinas I, Bovee JVMG, Boye K, Brodowicz T, Buonadonna A, De Álava E, Dei Tos AP, Del Muro XG, Dufresne A, Eriksson M, Fedenko A, Ferraresi V, Ferrari A, Frezza AM, Gasperoni S, Gelderblom H, Gouin F, Grignani G, Haas R, Hassan AB, Hindi N, Hohenberger P, Joensuu H, Jones RL, Jungels C, Jutte P, Kasper B, Kawai A, Kopeckova K, Krákorová DA, Le Cesne A, Le Grange F, Legius E, Leithner A, Lopez-Pousa A, Martin-Broto J, Merimsky O, Messiou C, Miah AB, Mir O, Montemurro M, Morosi C, Palmerini E, Pantaleo MA, Piana R, Piperno-Neumann S, Reichardt P, Rutkowski P, Safwat AA, Sangalli C, Sbaraglia M, Scheipl S, Schöffski P, Sleijfer S, Strauss D, Strauss SJ, Hall KS, Trama A, Unk M, van de Sande MAJ, van der Graaf WTA, van Houdt WJ, Frebourg T, Gronchi A, Stacchiotti S. Gastrointestinal stromal tumours: ESMO-EURACAN-GENTURIS Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2022; 33:20-33. [PMID: 34560242 DOI: 10.1016/j.annonc.2021.09.005] [Citation(s) in RCA: 312] [Impact Index Per Article: 104.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 02/06/2023] Open
Affiliation(s)
- P G Casali
- Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy; Department of Oncology and Hemato-oncology University of Milan, Milan, Italy
| | - J Y Blay
- Centre Leon Berard and UCBL1, Lyon, France
| | - N Abecassis
- Instituto Portugues de Oncologia de Lisboa Francisco Gentil, EPE, Lisbon, Portugal
| | - J Bajpai
- Department of Medical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - S Bauer
- Department of Medical Oncology, Interdisciplinary Sarcoma Center, West German Cancer Center, University of Duisburg-Essen, Essen, Germany
| | - R Biagini
- Department of Oncological Orthopedics, Musculoskeletal Tissue Bank, IFO, Regina Elena National Cancer Institute, Rome, Italy
| | - S Bielack
- Klinikum Stuttgart-Olgahospital, Stuttgart, Germany
| | - S Bonvalot
- Department of Surgery, Institut Curie, Paris, France
| | | | - J V M G Bovee
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - K Boye
- Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - T Brodowicz
- Vienna General Hospital (AKH), Medizinische Universität Wien, Vienna, Austria
| | - A Buonadonna
- Centro di Riferimento Oncologico di Aviano, Aviano, Italy
| | - E De Álava
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain; Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, Seville, Spain
| | - A P Dei Tos
- Department of Pathology, Azienda Ospedale Università Padova, Padova, Italy
| | - X G Del Muro
- Integrated Unit ICO Hospitalet, HUB, Barcelona, Spain
| | - A Dufresne
- Département d'Oncologie Médicale, Centre Leon Berard, Lyon, France
| | - M Eriksson
- Skane University Hospital-Lund, Lund, Sweden
| | - A Fedenko
- P. A. Herzen Cancer Research Institute, Moscow, Russian Federation
| | - V Ferraresi
- Sarcomas and Rare Tumors Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - A Ferrari
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - A M Frezza
- Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - S Gasperoni
- Department of Oncology and Robotic Surgery, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - H Gelderblom
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, The Netherlands
| | - F Gouin
- Centre Leon-Berard Lyon, Lyon, France
| | - G Grignani
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - R Haas
- Department of Radiotherapy, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Radiotherapy, Leiden University Medical Centre, Leiden, The Netherlands
| | - A B Hassan
- Oxford University Hospitals NHS Foundation Trust and University of Oxford, Oxford, UK
| | - N Hindi
- Department of Medical Oncology, Fundación Jimenez Diaz, University Hospital, Advanced Therapies in Sarcoma Lab, Madrid, Spain
| | - P Hohenberger
- Mannheim University Medical Center, Mannheim, Germany
| | - H Joensuu
- Helsinki University Hospital (HUH) and University of Helsinki, Helsinki, Finland
| | - R L Jones
- Sarcoma Unit, Royal Marsden Hospital and Institute of Cancer Research, London, UK
| | - C Jungels
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - P Jutte
- University Medical Center Groningen, Groningen, The Netherlands
| | - B Kasper
- Mannheim University Medical Center, Mannheim, Germany
| | - A Kawai
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - K Kopeckova
- University Hospital Motol, Prague, Czech Republic
| | - D A Krákorová
- Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - A Le Cesne
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - F Le Grange
- Department of Oncology, University College London Hospitals NHS Foundation Trust (UCLH), London, UK
| | - E Legius
- Department for Human Genetics, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - A Leithner
- Department of Orthopaedics and Trauma, Medical University of Graz, Graz, Austria
| | - A Lopez-Pousa
- Medical Oncology Department, Hospital Universitario Santa Creu i Sant Pau, Barcelona, Spain
| | - J Martin-Broto
- Department of Medical Oncology, Fundación Jimenez Diaz, University Hospital, Advanced Therapies in Sarcoma Lab, Madrid, Spain
| | - O Merimsky
- Aviv Sourasky Medical Center (Ichilov), Tel Aviv, Israel
| | - C Messiou
- Department of Radiology, Royal Marsden Hospital and Institute of Cancer Research, London, UK
| | - A B Miah
- Department of Oncology, Royal Marsden Hospital and Institute of Cancer Research, London, UK
| | - O Mir
- Department of Ambulatory Cancer Care, Gustave Roussy, Villejuif, France
| | - M Montemurro
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - C Morosi
- Department of Radiology, IRCCS Foundation National Cancer Institute, Milan, Italy
| | - E Palmerini
- Department of Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - M A Pantaleo
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria, di Bologna, Bologna, Italy
| | - R Piana
- Azienda Ospedaliero, Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | | | - P Reichardt
- Helios Klinikum Berlin Buch, Berlin, Germany
| | - P Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - A A Safwat
- Aarhus University Hospital, Aarhus, Denmark
| | - C Sangalli
- Department of Radiotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - M Sbaraglia
- Department of Pathology, Azienda Ospedale Università Padova, Padova, Italy
| | - S Scheipl
- Department of Orthopaedics and Trauma, Medical University of Graz, Graz, Austria
| | - P Schöffski
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - S Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - D Strauss
- Department of Surgery, Royal Marsden Hospital, London, UK
| | - S J Strauss
- Department of Oncology, University College London Hospitals NHS Foundation Trust (UCLH), London, UK
| | - K Sundby Hall
- Department of Oncology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - A Trama
- Department of Research, Evaluative Epidemiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - M Unk
- Institute of Oncology of Ljubljana, Ljubljana, Slovenia
| | - M A J van de Sande
- Department of Orthopedic Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - W T A van der Graaf
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands; Department of Medical Oncology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - W J van Houdt
- Department of Surgical Oncology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - T Frebourg
- Department of Genetics, Normandy Center for Genomic and Personalized Medicine, Normandie University, UNIROUEN, Inserm U1245 and Rouen University Hospital, Rouen, France
| | - A Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori and University of Milan, Milan, Italy
| | - S Stacchiotti
- Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
25
|
A CT-based nomogram for predicting the malignant potential of primary gastric gastrointestinal stromal tumors preoperatively. Abdom Radiol (NY) 2021; 46:3075-3085. [PMID: 33713161 DOI: 10.1007/s00261-021-03026-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 02/19/2021] [Accepted: 02/25/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE To develop and validate a computerized tomography (CT)-based nomogram for predicting the malignant potential of primary gastric gastrointestinal stromal tumors (GISTs). METHODS The primary and validation cohorts consisted of 167 and 39 patients (single center, different time periods) with histologically confirmed primary gastric GISTs. Clinical data and preoperative CT images were reviewed. The association of CT characteristics with malignant potential was analyzed using univariate and stepwise logistic regression analyses. A nomogram based on significant CT findings was developed for predicting malignant potential. The predictive accuracy of the nomogram was determined by the concordance index (C-index) and calibration curves. External validation was performed with the validation cohort. RESULTS CT imaging features including tumor size, tumor location, tumor necrosis, growth pattern, ulceration, enlarged vessels feeding or draining the mass (EVFDM), tumor contour, mesenteric fat infiltration, and direct organ invasion showed significant differences between the low- and high-grade malignant potential groups in univariate analysis (P < 0.05). Only tumor size (> 5 cm vs ≤ 5 cm), location (cardiac/pericardial region vs other), EVFDM, and mesenteric fat infiltration (present vs absent) were significantly associated with high malignant potential in multivariate logistic regression analysis. Incorporating these four independent factors into the nomogram model achieved good C-indexes of 0.946 (95% confidence interval [CI] 0.899-0.975) and 0.952 (95% CI 0.913-0.977) in the primary and validation cohorts, respectively. The cutoff point was 0.33, with sensitivity, specificity, and diagnostic accuracy of 0.865, 0.915, and 0.780, respectively. DISCUSSION Primary gastric GISTs originating in the cardiac/pericardial region appear to be associated with higher malignant potential. The nomogram consisting of CT features, including size, location, EVFDM, and mesenteric fat infiltration, could be used to accurately predict the high malignant potential of primary gastric GISTs.
Collapse
|
26
|
Al-Maqrashi Z, Burney IA, Taqi KM, Al-Sawafi Y, Qureshi A, Lakhtakia R, Mehdi I, Al-Bahrani B, Kumar S, Al-Moundhri M. Clinicopathological Features and Outcomes of Gastrointestinal Stromal Tumours in Oman: A multi-centre study. Sultan Qaboos Univ Med J 2021; 21:e237-e243. [PMID: 34221471 PMCID: PMC8219329 DOI: 10.18295/squmj.2021.21.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/13/2020] [Accepted: 09/09/2020] [Indexed: 11/16/2022] Open
Abstract
Objectives This study aimed to report the clinicopathological features, management and long-term outcomes of patients with gastrointestinal stromal tumours (GISTs) in Oman. Methods This retrospective study was conducted on patients treated for GIST between January 2003 and December 2017 at three tertiary referral centres in Muscat, Oman. All patients with confirmed histopathological diagnoses of GIST and followed-up at the centres during this period were included. Relevant information was retrieved from hospital records until April 2019. Results A total of 44 patients were included in the study. The median age was 55.5 years and 56.8% were female. The most common primary site of disease was the stomach (63.6%) followed by the jejunum/ileum (18.2%). Two patients (4.5%) had c-Kit-negative, discovered on GIST-1-positive disease. A total of 24 patients (54.5%) presented with localised disease and eight (33.3%) were classified as being at high risk of relapse. Patients with metastatic disease received imatinib in a palliative setting, whereas those with completely resected disease in the intermediate and high-risk groups were treated with adjuvant imatinib. Of the six patients (13.6%) with progressive metastatic disease, of which four had mutations on exon 11 and one on exon 9, while one had wild-type disease. Overall, rates of progression-free survival and overall survival (OS) at 100 months were 77.4% and 80.4%, respectively. Rates of OS for patients with localised and metastatic disease were 89.9% and 80.2%, respectively. Conclusion The presenting features and outcomes of patients with GISTs in Oman were comparable to those reported in the regional and international literature.
Collapse
Affiliation(s)
| | - Ikram A Burney
- Department of Medicine, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Kadhim M Taqi
- Division of General Surgery, Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Yaqoob Al-Sawafi
- Department of General Surgery, Armed Forces Hospital, Muscat, Oman
| | - Asim Qureshi
- Department of Pathology, King's Mill Hospital, Sherwood Forest Hospitals National Health Service Foundation Trust, Mansfield, Nottinghamshire, UK.,Department of Pathology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Ritu Lakhtakia
- Department of Pathology, Mohammed bin Rashid University of Medicine & Health Sciences, Dubai, United Arab Emirates
| | - Itrat Mehdi
- National Oncology Centre, Royal Hospital, Muscat, Oman
| | | | - Shiyam Kumar
- Department of Medical Oncology, Yeovil District Hospital NHS Foundation Trust, Somerset, UK
| | - Mansour Al-Moundhri
- Department of Medicine, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
27
|
Sbaraglia M, Businello G, Bellan E, Fassan M, Dei Tos AP. Mesenchymal tumours of the gastrointestinal tract. Pathologica 2021; 113:230-251. [PMID: 34294940 PMCID: PMC8299319 DOI: 10.32074/1591-951x-309] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal tumours represent a heterogenous group of neoplasms encopassing benign, intermediate malignancy, and malignant entities. Sarcomas account for approximately 1% of human malignancies. In consideration of their rarity as well as of intrinsic complexity, diagnostic accuracy represents a major challenge. Traditionally, mesenchymal tumours are regarded as lesions the occurrence of which is mostly limited to somatic soft tissues. However, the occurrence of soft tissue tumours at visceral sites represent a well recognized event, and the GI-tract ranks among the most frequently involved visceral location. There exist entities such as gastrointestinal stromal tumours (GIST) and malignant gastointestinal neuroectodermal tumors that exhibit exquisite tropism for the GI-tract. This review will focus also on other relevant clinico-pathologic entities in which occurrence at visceral location is not at all negligible.
Collapse
Affiliation(s)
- Marta Sbaraglia
- Department of Pathology, Azienda Ospedale-Università Padova, Padua, Italy
- Department of Medicine, University of Padua School of Medicine, Padua, Italy
| | - Gianluca Businello
- Department of Medicine, University of Padua School of Medicine, Padua, Italy
| | - Elena Bellan
- Department of Medicine, University of Padua School of Medicine, Padua, Italy
| | - Matteo Fassan
- Department of Pathology, Azienda Ospedale-Università Padova, Padua, Italy
- Department of Medicine, University of Padua School of Medicine, Padua, Italy
| | - Angelo Paolo Dei Tos
- Department of Pathology, Azienda Ospedale-Università Padova, Padua, Italy
- Department of Medicine, University of Padua School of Medicine, Padua, Italy
| |
Collapse
|
28
|
Brčić I, Argyropoulos A, Liegl-Atzwanger B. Update on Molecular Genetics of Gastrointestinal Stromal Tumors. Diagnostics (Basel) 2021; 11:194. [PMID: 33525726 PMCID: PMC7912114 DOI: 10.3390/diagnostics11020194] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/21/2021] [Accepted: 01/23/2021] [Indexed: 12/14/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract. The majority are sporadic, solitary tumors that harbor mutually exclusive KIT or PDGFRA gain-of-function mutations. The type of mutation in addition to risk stratification corresponds to the biological behavior of GIST and response to treatment. Up to 85% of pediatric GISTs and 10-15% of adult GISTs are devoid of these (KIT/PDGFRA) mutations and are referred to as wild-type GISTs (wt-GIST). It has been shown that these wt-GISTs are a heterogeneous tumor group with regard to their clinical behavior and molecular profile. Recent advances in molecular pathology helped to further sub-classify the so-called "wt-GISTs". Based on their significant clinical and molecular heterogeneity, wt-GISTs are divided into a syndromic and a non-syndromic (sporadic) subgroup. Recently, the use of succinate dehydrogenase B (SDHB) by immunohistochemistry has been used to stratify GIST into an SDHB-retained and an SDHB-deficient group. In this review, we focus on GIST sub-classification based on clinicopathologic, and molecular findings and discuss the known and yet emerging prognostic and predictive genetic alterations. We also give insights into the limitations of targeted therapy and highlight the mechanisms of secondary resistance.
Collapse
Affiliation(s)
| | | | - Bernadette Liegl-Atzwanger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria; (I.B.); (A.A.)
| |
Collapse
|
29
|
Song LJ, Ge HJ, Shi XQ, Shen WW. Prognostic and predictive values of the KIT11-mutated grading system in patients with gastrointestinal stromal tumors: a retrospective study. Hum Pathol 2021; 110:31-42. [PMID: 33476644 DOI: 10.1016/j.humpath.2021.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 01/10/2023]
Abstract
The KIT11 mutation is the most frequent mutation pattern in gastrointestinal stromal tumors (GISTs). However, few studies have investigated the correlation between the KIT11-mutated grading system and imatinib mesylate (IM) sensitivity (the first choice for adjuvant treatment of GISTs). Here, we elucidated the clinical value of the KIT11-mutated grading system for prognostic prediction in patients with GISTs treated with IM. A total of 106 patients with GIST were treated with IM (8: intermediate-risk, 98: high-risk; 10: KIT9-mutated, 86: KIT11-mutated, 5: wild-type, and 5: other mutations). KIT11-mutated patients were divided into 3 grades based on the KIT11-mutated site and type. Clinical backgrounds and prognostic outcomes were retrospectively compared between the 3 groups. Of 86 KIT11-mutated patients treated with IM, 32 (37.21%) had grade 1 tumors, 37 (43.02%) had grade 2 tumors, and 17 (19.77%) had grade 3 tumors. The 5-year disease-free survival (DFS) was significantly worse in patients with grade 3 KIT11-mutated GISTs (41.96%, p = 0.001) than in those with grade 1 (93%) and grade 2 (70.64%) cases. The multivariable analysis suggested that the KIT11-mutated grading system was an independent risk factor for DFS in patients treated with IM (hazard risk, 2.512; 95% confidence interval, 1.370-4.607; p = 0.003). In conclusion, the KIT11-mutated grading system provides good prognostic stratification for DFS in patients treated with IM. Grade 1 tumors predict a favorable response to IM.
Collapse
Affiliation(s)
- Ling-Jun Song
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080 China.
| | - Hui-Juan Ge
- Department of Pathology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 PR China
| | - Xiao-Qin Shi
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080 China
| | - Wei-Wei Shen
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080 China; Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
30
|
Liang L, Li X, Li D, Liu P, Nong L, Dong Y, Liu J, Huang S, Li T. Mutational characteristics of gastrointestinal stromal tumors: A single-center analysis of 302 patients. Oncol Lett 2021; 21:174. [PMID: 33552291 PMCID: PMC7798044 DOI: 10.3892/ol.2021.12435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/20/2020] [Indexed: 11/05/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) represent a spectrum of tumors characterized by variable behaviors and activating mutations in KIT proto-oncogene, receptor tyrosine kinase (KIT) or platelet derived growth factor receptor α (PDGFRA) genes. However, whether genotype analysis should be regarded as a prognostic indicator remains unclear. In the present study, clinicopathological data and the mutation phenotypes of KIT and PDGFRA genes were assessed in a series of 302 patients with GISTs at a single center. Univariate and multivariate Cox regression analyses were performed to identify the clinicopathological and mutational factors associated with relapse-free survival (RFS) in patients who had undergone complete primary GIST resection. KIT and PDGFRA mutations were identified in 233 (77.2%) and 30 (9.9%) cases, respectively. The following clinicopathological parameters were significantly associated with a shorter RFS: Male, non-gastric tumor origin, larger tumor size (>5 cm), high mitotic activity (>5/50 high-power fields), necrosis and epithelioid morphology. Tumors at non-gastric sites, with high National Institutes of Health risk classification, high World Health Organization (WHO) grade and KIT deletion involving codons 557/558/559 exhibited a significantly higher risk of progression. In the Cox regression model, KIT deletion involving codons 557/558/559, non-gastric origin and high WHO grade were independent indicators of RFS. The adverse prognosis associated with KIT deletions involving codons 557/558/559 was also observed for gastric GISTs. Conversely, spindle morphology, KIT exon 11 substitution and PDGFRA exon 18 mutation were associated with a longer RFS and lower rate of relapse. Furthermore, the coexistence of KIT exon 11 deletion and exon 13 duplication was observed in one tumor, with adverse prognostic features. Heterogeneity affecting morphology, immunostaining and genotype was identified in 4 cases. In addition, the presence of succinate dehydrogenase-deficient GIST was found in 5 cases (3.6%). In conclusion, the tumor genotype with regard to KIT and PDGFRA mutations exhibited prognostic significance for the risk of GIST progression and may be helpful for the optimization of tailored adjuvant therapy.
Collapse
Affiliation(s)
- Li Liang
- Department of Pathology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Xin Li
- Department of Pathology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Dong Li
- Department of Pathology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Ping Liu
- Department of Pathology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Lin Nong
- Department of Pathology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Ying Dong
- Department of Pathology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Jumei Liu
- Department of Pathology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Sixia Huang
- Department of Pathology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Ting Li
- Department of Pathology, Peking University First Hospital, Beijing 100034, P.R. China
| |
Collapse
|
31
|
Palomba G, Paliogiannis P, Sini MC, Colombino M, Casula M, Manca A, Pisano M, Sotgiu G, Doneddu V, Palmieri G, Cossu A. KIT and PDGFRa mutational patterns in Sardinian patients with gastrointestinal stromal tumors. Eur J Cancer Prev 2021; 30:53-58. [PMID: 32091431 PMCID: PMC7713762 DOI: 10.1097/cej.0000000000000581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 01/20/2020] [Indexed: 12/16/2022]
Abstract
Gastrointestinal stromal tumor (GIST) is the most common mesenchymal malignancy of the gastrointestinal tract. We provide in the present article the molecular characterization of a series of primary GISTs in a cohort of Sardinian patients (Italy), with the aim to describe the patterns of KIT and PDGFRa mutations and the corresponding clinical features. Ninety-nine Sardinian patients with histologically-proven diagnosis of GIST were included in the study. Medical records and pathology reports were used to assess the demographic and clinical features of the patients and the disease at the time of the diagnosis. Formalin-fixed, paraffin-embedded tissue samples were retrieved for each case, and mutation analysis of the KIT and PDGFRa genes was performed. KIT and PDGFRa mutations were detected in 81.8% and 5% of the cases, respectively. The most common KIT mutation was W557_K558del in exon 11, while D842V in exon 18 was the most common PDGFRa genetic alteration; V561D was the only PDGFRa mutation found in exon 12. The global "wild-type" cases, with no mutations in either the KIT or PDGFRa genes, were 13 (13.1%). The mean survival of those patients was approximately 46.9 (±43.9) months. Globally, 86.9% of Sardinian patients with GIST had a KIT or PDGFRa mutation; the former were more frequent in comparison with other Italian cohorts, while PDGFRa mutations were rare. No statistical differences in survival between mutated and wild-type cases, and between KIT and PDGFRa mutated cases were detected in our study.
Collapse
Affiliation(s)
- Grazia Palomba
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR)
| | - Panagiotis Paliogiannis
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Maria C. Sini
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR)
| | - Maria Colombino
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR)
| | - Milena Casula
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR)
| | - Antonella Manca
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR)
| | - Marina Pisano
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR)
| | - Giovanni Sotgiu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Valentina Doneddu
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR)
| | - Giuseppe Palmieri
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry (ICB), National Research Council (CNR)
| | - Antonio Cossu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
32
|
Zhou G, Xiao K, Gong G, Wu J, Zhang Y, Liu X, Jiang Z, Ma C. A novel nomogram for predicting liver metastasis in patients with gastrointestinal stromal tumor: a SEER-based study. BMC Surg 2020; 20:298. [PMID: 33238982 PMCID: PMC7689971 DOI: 10.1186/s12893-020-00969-4] [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: 08/02/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Background Liver metastasis (LIM) of gastrointestinal stromal tumor (GIST) is associated with poor prognosis. The present study aimed at developing and validating nomogram to predict LIM in patients with GIST, thus helping clinical diagnosis and treatment. Methods The data of GIST patients derived from Surveillance, Epidemiology, and End Results (SEER) database from 2010 to 2016, which were then screened by univariate and multivariate logistic regression for the construction of LIM nomogram. The model discrimination of LIM nomogram was evaluated by concordance index (C-index) and calibration plots, while the predictive accuracy and clinical values were measured by decision curve analysis (DCA) and clinical impact plot. Furthermore, we validated predictive nomogram in the internal testing set. Results A total of 3797 patients were enrolled and divided randomly into training and validating groups in a 3-to-1 ratio. After logistic regression, the significant variables were sex, tumor location, tumor size, N stage and mitotic rate. The calibration curves showed the perfect agreement between nomogram predictions and actual observations, while the DCA and clinical impact plot showed the clinical utility of LIM nomogram. C-index of the nomogram was 0.812. What’s more, receiver operating characteristic curves (ROC) also showed good discrimination and calibration in the training set (AUC = 0.794, 95% CI 0.778–0.808) and the testing set (AUC = 0.775, 95% CI 0.748–0.802). Conclusion The nomogram for patients with GIST can effectively predict the individualized risk of liver metastasis and provide insightful information to clinicians to optimize therapeutic regimens.
Collapse
Affiliation(s)
- Guowei Zhou
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Keshuai Xiao
- Department of General Surgery, Xinyang Central Hospital, Xin Yang, 464000, Henan Province, China
| | - Guanwen Gong
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| | - Jiabao Wu
- Department of Pediatrics, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Ya Zhang
- Department of Gynecology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Xinxin Liu
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Zhiwei Jiang
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Chaoqun Ma
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
33
|
Lin Y, Wang M, Jia J, Wan W, Wang T, Yang W, Li C, Chen X, Cao H, Zhang P, Tao K. Development and validation of a prognostic nomogram to predict recurrence in high-risk gastrointestinal stromal tumour: A retrospective analysis of two independent cohorts. EBioMedicine 2020; 60:103016. [PMID: 32980695 PMCID: PMC7522759 DOI: 10.1016/j.ebiom.2020.103016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/05/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The risk of recurrence in localised, primary gastrointestinal stromal tumour (GIST) classified as high-risk after complete resection varies significantly. Thus, we aimed to develop a nomogram to predict the recurrence of high-risk GIST after surgery to aid patient selection. METHODS We retrospectively evaluated patients (n = 424) with high-risk GIST who underwent curative resection as the initial treatment at two high-volume medical centres, between January 2005 and September 2019. The least absolute shrinkage and selection operator (LASSO) regression model was utilised to select potentially relevant features. Multivariate Cox proportional hazards analysis was used to develop a novel nomogram. FINDINGS The nomogram comprised age, fibrinogen levels, prognostic nutritional index (PNI), platelet-lymphocyte ratio (PLR), mitotic counts and tumour size, which provided favourable calibration and discrimination in the training dataset with an AUC of 0•749 and a C-index of 0•742 (95%CI:0•689-0•804). Further, it showed acceptable discrimination in the validation cohort, with an AUC of 0•778 and C-index of 0•735 (95%CI:0•634-0•846). The time-dependant receiver operating characteristic (ROC) curves performed well throughout the observation period. Additionally, the nomogram could classify high-risk GISTs into 'very high-risk' and 'general high-risk' groups with a hazard ratio (HR) of 5•190 (95%CI: 3•202-8•414) and 5•438 (95%CI: 2•236-13•229) for the training and validation datasets, respectively. INTERPRETATION The nomogram independently predicted post-operative recurrence-free survival (RFS) in high-risk GIST and showed favourable discrimination and calibration values. It may be a useful clinical tool for identifying 'very high-risk' GIST, by allowing treatment strategy optimisation in these patients. FUNDING National Natural Science Foundation of China (No. 81702386 and 81874184).
Collapse
Affiliation(s)
- Yao Lin
- Department of Gastrointestinal Surgery, Union Hospital Tongji Medical College Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Ming Wang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Jia
- Department of Gastrointestinal Surgery, Union Hospital Tongji Medical College Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Wenze Wan
- Department of Gastrointestinal Surgery, Union Hospital Tongji Medical College Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Tao Wang
- Department of Gastrointestinal Surgery, Union Hospital Tongji Medical College Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Wenchang Yang
- Department of Gastrointestinal Surgery, Union Hospital Tongji Medical College Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Chengguo Li
- Department of Gastrointestinal Surgery, Union Hospital Tongji Medical College Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Xin Chen
- Department of Gastrointestinal Surgery, Union Hospital Tongji Medical College Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Hui Cao
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital Tongji Medical College Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China.
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital Tongji Medical College Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China.
| |
Collapse
|
34
|
Zhang X, Ning L, Hu Y, Zhao S, Li Z, Li L, Dai Y, Jiang L, Wang A, Chu X, Li Y, Yang D, Lu C, Yao L, Cui G, Lin H, Chen G, Cui Q, Guo H, Zhang H, Lun Z, Xia L, Su Y, Han G, Hui X, Wei Z, Sun Z, Shen S, Zhou Y. Prognostic Factors for Primary Localized Gastrointestinal Stromal Tumors After Radical Resection: Shandong Gastrointestinal Surgery Study Group, Study 1201. Ann Surg Oncol 2020; 27:2812-2821. [PMID: 32040699 DOI: 10.1245/s10434-020-08244-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Most previous risk-prediction models for gastrointestinal stromal tumors (GISTs) were based on Western populations. In the current study, we collected data from 23 hospitals in Shandong Province, China, and used the data to examine prognostic factors in Chinese patients and establish a new recurrence-free survival (RFS) prediction model. METHODS Records were analyzed for 5285 GIST patients. Independent prognostic factors were identified using Cox models. Receiver operating characteristic curve analysis was used to compare a novel RFS prediction model with current risk-prediction models. RESULTS Overall, 4216 patients met the inclusion criteria and 3363 completed follow-up. One-, 3-, and 5-year RFS was 94.6% (95% confidence interval [CI] 93.8-95.4), 85.9% (95% CI 84.7-87.1), and 78.8% (95% CI 77.0-80.6), respectively. Sex, tumor location, size, mitotic count, and rupture were independent prognostic factors. A new prognostic index (PI) was developed: PI = 0.000 (if female) + 0.270 (if male) + 0.000 (if gastric GIST) + 0.350 (if non-gastric GIST) + 0.000 (if no tumor rupture) + 1.259 (if tumor rupture) + 0.000 (tumor mitotic count < 6 per 50 high-power fields [HPFs]) + 1.442 (tumor mitotic count between 6 and 10 per 50 HPFs) + 2.026 (tumor mitotic count > 10 per 50 HPFs) + 0.096 × tumor size (cm). Model-predicted 1-, 3-, and 5-year RFS was S(12, X) = 0.9926exp(PI), S(36, X) = 0.9739exp(PI) and S(60, X) = 0.9471exp(PI), respectively. CONCLUSIONS Sex, tumor location, size, mitotic count, and rupture were independently prognostic for GIST recurrence. Our RFS prediction model is effective for Chinese GIST patients.
Collapse
Affiliation(s)
- Xiaoqian Zhang
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing, China
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Liang Ning
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yulong Hu
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shanfeng Zhao
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zequn Li
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yong Dai
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Lixin Jiang
- Department of Gastrointestinal Thyroid Surgery, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Ailiang Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Xianqun Chu
- Department of Gastroenterology Surgery, Jining No.1 People's Hospital, Jining, Shandong, China
| | - Yuming Li
- Department of Gastrointestinal Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Daogui Yang
- Department of Gastrointestinal Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Chunlei Lu
- Department of Laparoscopic Surgery Center, Linyi People's Hospital, Linyi, Shandong, China
| | - Linguo Yao
- Department of Gastrointestinal Surgery, Shengli Oilfield Central Hospital, Yantai, Shandong, China
| | - Gang Cui
- Department of General Surgery, Taian City Central Hospital, Taian, Shandong, China
| | - Huizhong Lin
- Department of Gastric Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Gang Chen
- Department of Gastrointestinal Surgery, Tengzhou Central People's Hospital, Tengzhou, Shandong, China
| | - Qing Cui
- Department of General Surgery, Zibo Central Hospital, Zibo, Shandong, China
| | - Hongliang Guo
- The Fourth Department of General Surgery, Shandong Cancer Hospital, Jinan, Shandong, China
| | - Huanhu Zhang
- Department of General Surgery, Weihai Municipal Hospital, Weihai, Shandong, China
| | - Zengjun Lun
- Department of General Surgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China
| | - Lijian Xia
- Department of Gastrointestinal Surgery, Shandong Province Qianfoshan Hospital, Jinan, Shandong, China
| | - Yingfeng Su
- Department of General Surgery, Dezhou People's Hospital, Dezhou, Shandong, China
| | - Guoxin Han
- Department of General Surgery, Affiliated Hospital of Taishan Medical University, Taian, Shandong, China
| | - Xizeng Hui
- Department of Surgery, People's Hospital of Rizhao, Rizhao, Shandong, China
| | - Zhixin Wei
- Department of General Surgery, Heze Municipal Hospital, Heze, Shandong, China
| | - Zuocheng Sun
- Department of Surgery, Weifang People's Hospital, Weifang, Shandong, China
| | - Shuai Shen
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yanbing Zhou
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
35
|
Zhang H, Liu Q. Prognostic Indicators for Gastrointestinal Stromal Tumors: A Review. Transl Oncol 2020; 13:100812. [PMID: 32619820 PMCID: PMC7327422 DOI: 10.1016/j.tranon.2020.100812] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 02/08/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are potentially malignancies that can occur anywhere in the digestive tract. Tyrosine kinase inhibitors (TKIs) such as imatinib have proven effective since the discovery of KIT and PDGFRA. The current version of NCNN, ESMO and EURACAN guidelines recognized that the three main prognostic factors are the mitotic rate, tumor size and tumor site. In addition, tumor rupture is also recognized as an independent risk factor. However, recent evidence shows that various types of gene mutations are associated with prognosis, and influencing factors such as gastrointestinal bleeding and high Ki67 index have been associated with poor prognosis. It shows that the current risk classification is still insufficient and controversial. With the emergence of more and more lack mutation in KIT/PDGFRA GISTs (KIT/PDGFRA wild-type GISTs) or drug resistance genes, primary and secondary drug resistance problems are caused, which makes the treatment of late or metastatic GIST face challenges. Therefore, this article will review the clinicopathological characteristics of GIST, the special molecular subtypes and other factors that may affect prognosis. We will also explore reliable prognostic markers for better postoperative management and improve the prognosis of patients with GIST.
Collapse
Affiliation(s)
- Haixin Zhang
- Department of Trauma center, The First Hospital of China Medical University, Shenyang, China
| | - Qi Liu
- Department of Trauma center, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
36
|
Jiaxin L, Peiyun Z, Zheng T, Wei Y, Shanshan S, Lei R, Zhengwen X, Yong F, Xiaodong G, Anwei X, Kuntang S, Yingyong H. Comparison of prognostic prediction models for rectal gastrointestinal stromal tumor. Aging (Albany NY) 2020; 12:11416-11430. [PMID: 32561689 PMCID: PMC7343501 DOI: 10.18632/aging.103204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/17/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Rectal gastrointestinal stromal tumors (RGISTs) are biologically characterized tumors that are relatively rare. Thus, few studies have reported a specific prognostic system for this subset of tumors but integrated it into parallel systems, such as small intestine. Our aim is to develop a new predictive staging system nomogram (named FD-ZS system) for RGISTs. RESULTS Tumor size and mitotic rate were independent risk factors for tumor recurrence in RGISTs according to univariate and multivariate survival analyses. A prognostic predictive nomogram was developed, and a cut-off value of 65 points was calculated by X-tile to discriminate risk based on tumor size and mitotic rate. The C-indices for the FD-ZS, FD-Hou, NIH, and WHO systems were 0.706, 0.693, 0.687, and 0.680, respectively. CONCLUSION In the present study, a concise two-tier grading system (FD-ZS) for prognostic prediction of RGISTs that is simpler to several reported systems was developed, and a cut-off value was established to help RGIST patients determine whether to undergo adjuvant imatinib treatment. METHODS A nomogram was employed, and its predictive accuracy and discriminative ability were determined by concordance index (C-index) and calibration curve analyses. The nomogram was then compared with three stratification systems used for GISTs (FD-Hou, NIH, and WHO).
Collapse
Affiliation(s)
- Liu Jiaxin
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Chest Hospital, Shanghai Jiao Tong University, Department of Pathology, Shanghai, China
| | - Zhou Peiyun
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Tang Zheng
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Yuan Wei
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shen Shanshan
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ren Lei
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xing Zhengwen
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fang Yong
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gao Xiaodong
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xue Anwei
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shen Kuntang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hou Yingyong
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Cuccaro F, Burgio Lo Monaco MG, Rashid I, Bisceglia L, Caputo E, Melcarne A, Palma F, Tanzarella M, Cozzi E, Coviello V. Population-based incidence of gastrointestinal stromal tumors in Puglia. TUMORI JOURNAL 2020; 107:39-45. [PMID: 32539634 DOI: 10.1177/0300891620931944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION This study presents the incidence of gastrointestinal stromal tumors (GISTs) in an Italian region of over 4 million inhabitants monitored for 10 years and is the largest incidence study of this type of cancer conducted so far in Italy. METHODS In order to ensure the registration of all GISTs, including those with nonmalignant behavior, a cancer list was integrated with the cases found through an ad hoc data mining process that covered all the pathologic reports of Puglia. Case distributions by sex, age groups, site, and prognostic groups according to Miettinem and Lasota classification and crude and age-standardized incidence rates were produced. RESULTS In the 10-year period 2006 to 2015, 708 cases of GIST were recorded in Puglia. The average crude incidence rate was 1.7 per 100,000 person-years and the age-standardized incidence rate, using 2013 European standard population, was 1.8 per 100,000 person-years (95% confidence interval [CI], 1.6-1.9). Incidence was higher in men than in women: crude incidence rate was 2.0 per 100,000 person-years and age-standardized incidence rate 2.2 per 100,000 person-years (95% CI, 2.0-2.4) in men and 1.5 per 100,000 person-years and 1.4 per 100,000 person-years (95% CI, 1.2-1.6) in women. DISCUSSION Our incidence rates are comparable with those of other international studies and they are located in the medium to high end of the range. The comparisons are affected by a different capacity of the cancer registries to intercept and record GISTs with nonmalignant behavior. Distribution of cases for sex, age groups, sites, and prognostic risk groups are consistent with previous results.
Collapse
Affiliation(s)
- Francesco Cuccaro
- Cancer Registry of the Local Health Unit of Barletta-Andria-Trani, section of the Puglia Cancer Registry, Barletta, Italy
| | | | - Ivan Rashid
- Coordination Center of the Puglia Cancer Registry, Regional Strategic Agency for Health and Social of Puglia, Bari, Italy
| | - Lucia Bisceglia
- Coordination Center of the Puglia Cancer Registry, Regional Strategic Agency for Health and Social of Puglia, Bari, Italy
| | - Enrico Caputo
- Cancer Registry of the Local Health Unit of Bari, section of the Puglia Cancer Registry, Bari, Italy
| | - Anna Melcarne
- Cancer Registry of the Local Health Unit of Lecce, section of the Puglia Cancer Registry, Lecce, Italy
| | - Fernando Palma
- Cancer Registry of the Local Health Unit of Foggia, section of the Puglia Cancer Registry, Foggia, Italy
| | - Margherita Tanzarella
- Cancer Registry of the Local Health Unit of Taranto, section of the Puglia Cancer Registry, Taranto, Italy
| | - Emma Cozzi
- Cancer Registry of the Local Health Unit of Brindisi, section of the Puglia Cancer Registry, Brindisi, Italy
| | - Vincenzo Coviello
- Cancer Registry of the Local Health Unit of Barletta-Andria-Trani, section of the Puglia Cancer Registry, Barletta, Italy
| |
Collapse
|
38
|
Rekhi B, Kattoor J, Jennifer A, Govindarajan N, Uppin S, Jambhekar NA, Jojo A, Chatterjee U, Banerjee D, Singh P. Grossing and reporting of a soft tissue tumor specimen in surgical pathology: Rationale, current evidence, and recommendations. Indian J Cancer 2020; 58:17-27. [PMID: 33402595 DOI: 10.4103/ijc.ijc_738_20] [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: 11/04/2022]
Abstract
Soft tissue tumors, including sarcomas are complex and diagnostically challenging tumors. This is as a result of their heterogeneity and overlapping clinicopathological, immunohistochemical and also molecular features, the latter to some extent. More than 80 types of sarcoma have been described. Current management, which is best offered at centers with active multidisciplinary care, is based on balancing oncologic and functional outcomes in such cases. This has transcended into the types of specimens received for grossing these rather uncommon tumors. Over the years, diagnostic specimens have reduced in their sizes from, open biopsies to core needle biopsies. These specimens need to be adequately and judiciously triaged for ancillary techniques, such as molecular testing. Conservative surgeries have led to resected specimens for marginal assessment. Lately, post neoadjuvant (chemotherapy or radiation therapy)-treated resection specimens of soft tissue sarcomas are being submitted for surgical pathology reporting. This article focuses on the grossing of soft tissue tumors, including sarcomas, in terms of types of specimens, grossing techniques including rationale, tissue triage, reporting, and recommendations from the surgical pathologists actively engaged in reporting musculoskeletal tumors, based on current evidence.
Collapse
Affiliation(s)
- Bharat Rekhi
- Department of Surgical Pathology, Bone and Soft Tissues, Disease Management Group, Tata Memorial Hospital, Homi Bhabha National Institute University, Mumbai, Maharashtra, India
| | - Jayasree Kattoor
- Department of Pathology, Regional Cancer Centre, Thiruvananthapuram, Kerala, India
| | - Anne Jennifer
- Department of Pathology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Nandini Govindarajan
- Department of Pathology, Dr Kamkashi Memorial Hospitals, Chennai, Tamil Nadu, India
| | - Shantveer Uppin
- Department of Pathology, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Nirmala A Jambhekar
- Formerly, Department of Surgical Pathology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Annie Jojo
- Department of Pathology, Institute of Post-Graduate Medical Education and Research and Seth Sukhlal Karnani Memorial Hospital, Kolkata, West Bengal, India
| | - Uttara Chatterjee
- Department of Pathology, Amrita Institute of Medical Sciences, Kochi, Kerala, India
| | - Devmalya Banerjee
- Department of Pathology, Narayana Super Speciality Hospital, Howrah, Kolkata, West Bengal, India
| | - Pradyumn Singh
- Department of Pathology, Ram Manohar Lohia Hospital, Lucknow, Uttar Pradesh, India
| |
Collapse
|
39
|
Optimal thresholds of risk parameters for gastrointestinal stromal tumors. Eur J Surg Oncol 2019; 46:180-188. [PMID: 31431322 DOI: 10.1016/j.ejso.2019.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 08/10/2019] [Accepted: 08/12/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GIST) are the most frequent mesenchymal neoplasms of the gastrointestinal tract with highly variable potential for relapse. Tumor size and mitotic index (MI) are major risk factors that predict the outcome of GIST patients. Recent risk stratification schemes include some or all of the empirical size thresholds of 2 cm, 5 cm, and 10 cm and MI thresholds of 5 per 50 high-power fields (hpf) and 10 per 50 hpf. However, data that verify these numbers are sparse. METHODS By exhaustive regression tree analysis, maximally selected rank statistics and survival difference analysis with bootstrap sampling on a naive GIST population of 161 patients with a mean follow-up of 44 months, current stratification schemes using tumor size and MI were analyzed herein. RESULTS /Conclusions: Thresholds that optimally stratify the risk of recurrence are observed at tumor sizes of 4-5 cm and 10-11 cm and at mitotic indices of about 5 per 50 hpf and 10 per 50 hpf, respectively. While these data validate the canonical thresholds for size and MI used in risk stratification of GIST, transition regions as well as differences in the implementation of these thresholds between the different classification schemes proposed in the recent years should be considered when classifying GIST.
Collapse
|
40
|
Mazzocca A, Napolitano A, Silletta M, Spalato Ceruso M, Santini D, Tonini G, Vincenzi B. New frontiers in the medical management of gastrointestinal stromal tumours. Ther Adv Med Oncol 2019; 11:1758835919841946. [PMID: 31205499 PMCID: PMC6535752 DOI: 10.1177/1758835919841946] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 03/13/2019] [Indexed: 12/11/2022] Open
Abstract
The tyrosine kinase inhibitor (TKI) imatinib has radically changed the natural history of KIT-driven gastrointestinal stromal tumours (GISTs). Approved second-line and third-line medical therapies are represented by the TKIs sunitinib and regorafenib, respectively. While imatinib remains the cardinal drug for patients with GISTs, novel therapies are being developed and clinically tested to overcome the mechanisms of resistance after treatments with the approved TKI, or to treat subsets of GISTs driven by rarer molecular events. Here, we review the therapy of GISTs, with a particular focus on the newest drugs in advanced phases of clinical testing that might soon change the current therapeutic algorithm.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bruno Vincenzi
- Medical Oncology, Università Campus Bio-Medico, Via Alvaro del Portillo 200, Rome, Italy
| |
Collapse
|
41
|
Yang J, Gu Y, Huang X, Xu J, Zhang Y, Yang X, Tian H, Zhan W. Prognostic impact of preoperative neutrophil-lymphocyte ratio for surgically resected gastrointestinal stromal tumors. Medicine (Baltimore) 2019; 98:e15319. [PMID: 31008985 PMCID: PMC6494246 DOI: 10.1097/md.0000000000015319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 03/05/2019] [Accepted: 03/23/2019] [Indexed: 12/18/2022] Open
Abstract
Neutrophil-lymphocyte ratio (NLR) was shown to be prognostic value in various malignancies. There are limited data about predictive or prognostic role of NLR during gastrointestinal stromal tumors (GISTs) patients. This study evaluated the prognostic significance of preoperative NLR in patients with GIST.We retrospectively enrolled 72 primary GIST patients who received initial curative surgical resection with or without adjuvant imatinib therapy. The preoperative NLR in the peripheral blood was calculated. Univariate and multivariate Cox proportional hazard regression models were used to identify potential predictors of tumor outcomes.The NLR cut-off value of 4.18 was selected. Multivariate analysis revealed that high NLR was associated with a unfavorable prognosis of GISTs (P < .05). Tumor size, tumor location, and age were significantly correlated with the NLR (P < .05).High NLR was an unfavorable prognostic factor of overall survival in GISTs and may be a useful preoperative biomarker of the prognosis of GISTs.
Collapse
|
42
|
Chen T, Liu S, Li Y, Feng X, Xiong W, Zhao X, Yang Y, Zhang C, Hu Y, Chen H, Lin T, Zhao M, Liu H, Yu J, Xu Y, Zhang Y, Li G. Developed and validated a prognostic nomogram for recurrence-free survival after complete surgical resection of local primary gastrointestinal stromal tumors based on deep learning. EBioMedicine 2019; 39:272-279. [PMID: 30587460 PMCID: PMC6355433 DOI: 10.1016/j.ebiom.2018.12.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/08/2018] [Accepted: 12/14/2018] [Indexed: 12/09/2022] Open
Abstract
This study aimed to develop and validate a prognostic nomogram for recurrence-free survival (RFS) after surgery in the absence of adjuvant therapy to guide the selection for adjuvant imatinib therapy based on Residual Neural Network (ResNet). The ResNet model was developed based on contrast-enhanced computed tomography (CE-CT) in a training cohort consisted of 80 patients pathologically diagnosed gastrointestinal sromal tumors (GISTs) and validated in internal and external validation cohort respectively. Independent clinicopathologic factors were integrated with the ResNet model to construct the individualized nomogram. The performance of the nomogram was evaluated in regard to discrimination, calibration, and clinical usefulness. The ResNet model was significantly associated with RFS. Integrable predictors in the individualized ResNet nomogram included the tumor site, size, and mitotic count. Compared with modified NIH, AFIP, and clinicopathologic nomogram, both ResNet nomogram and ResNet model showed a better discrimination capability with AUCs of 0·947(95%CI, 0·910-0·984) for 3-year-RFS, 0·918(0·852-0·984) for 5-year-RFS, and AUCs of 0·912 (0·851-0·973) for 3-year-RFS, 0·887(0·816-0·960) for 5-year-RFS, respectively. Calibration curve shows the good calibration of the nomogram in terms of the agreement between the estimated and the observed 3- and 5- year outcomes. Decision curve analysis showed that the ResNet nomogram had a higher overall net benefit. In conclusion, we presented a deep learning-based prognostic nomogram to predict RFS after resection of localized primary GISTs with excellent performance and could be a potential tool to select patients for adjuvant imatinib therapy.
Collapse
Affiliation(s)
- Tao Chen
- Department of General Surgery, Nanfang Hospital, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Southern Medical University, Guangdong Province, Guangzhou 510515, China.
| | - Shangqing Liu
- School of Biomedical Engineering, Southern Medical University, Guangdong Province, Guangzhou, 510515, China
| | - Yong Li
- Department of General Surgery, Guangdong Academy of Medical Science, Guangdong General Hospital, Guangdong Province, Guangzhou 510080, China
| | - Xingyu Feng
- Department of General Surgery, Guangdong Academy of Medical Science, Guangdong General Hospital, Guangdong Province, Guangzhou 510080, China
| | - Wei Xiong
- Medical Image Center, Nanfang Hospital, Guangdong Province, Southern Medical University, Guangzhou 510515, China
| | - Xixi Zhao
- Medical Image Center, Nanfang Hospital, Guangdong Province, Southern Medical University, Guangzhou 510515, China
| | - Yali Yang
- Medical Image Center, Nanfang Hospital, Guangdong Province, Southern Medical University, Guangzhou 510515, China
| | - Cangui Zhang
- Department of General Surgery, Nanfang Hospital, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Southern Medical University, Guangdong Province, Guangzhou 510515, China
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Southern Medical University, Guangdong Province, Guangzhou 510515, China
| | - Hao Chen
- Department of General Surgery, Nanfang Hospital, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Southern Medical University, Guangdong Province, Guangzhou 510515, China
| | - Tian Lin
- Department of General Surgery, Nanfang Hospital, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Southern Medical University, Guangdong Province, Guangzhou 510515, China
| | - Mingli Zhao
- Department of General Surgery, Nanfang Hospital, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Southern Medical University, Guangdong Province, Guangzhou 510515, China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Southern Medical University, Guangdong Province, Guangzhou 510515, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Southern Medical University, Guangdong Province, Guangzhou 510515, China
| | - Yikai Xu
- Medical Image Center, Nanfang Hospital, Guangdong Province, Southern Medical University, Guangzhou 510515, China.
| | - Yu Zhang
- School of Biomedical Engineering, Southern Medical University, Guangdong Province, Guangzhou, 510515, China.
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Southern Medical University, Guangdong Province, Guangzhou 510515, China.
| |
Collapse
|
43
|
Pellat A, Hautefeuille V, Coriat R. Mise au point sur les tumeurs stromales gastro-intestinales (GIST). ONCOLOGIE 2018. [DOI: 10.3166/onco-2018-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
44
|
Zhu K, Li K, Yuan DW, Xu G, Kang L, Dang CX, Zhang Y. Clinicopathological and Prognostic Significance of Expression of B-Cell-Specific Moloney Murine Leukemia Virus Insertion Site 1 (BMI-1) Gene and Protein in Gastrointestinal Stromal Tumors. Med Sci Monit 2018; 24:6414-6421. [PMID: 30209248 PMCID: PMC6149236 DOI: 10.12659/msm.909443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 05/08/2018] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Gastrointestinal stromal tumor (GIST) is an uncommon visceral sarcoma that arises predominantly in the gastrointestinal tract. Since GISTs are encountered infrequently and inflexible to traditional therapy, the aim of the present study was to explore the correlation of B-cell-specific Moloney murine leukemia virus insertion site 1 (BMI-1) mRNA and BMI-1 protein levels with the clinicopathological characteristics and prognosis significance of GISTs. MATERIAL AND METHODS GIST tissues and normal tissues were collected from 156 patients who had undergone surgical treatment. RT-qPCR and immunohistochemistry were used to measure the BMI-1 mRNA and protein levels in GIST tissues and normal tissues. Univariate survival analysis was used for determination of the factors that affect prognosis of GIST patients. Cox proportional hazards model was plotted to determine the independent risk factors for prognosis of GIST patients. RESULTS The BMI-1 mRNA and protein levels in GIST tissues were higher than those in normal tissues. BMI-1 mRNA and positive protein levels were correlated with the National Institutes of Health (NIH) risk grade, tumor diameter and infiltration, and metastasis. There was a short survival period for the patients with a positive protein level and a high mRNA level of BMI-1. The site of primary tumor, tumor diameter, NIH risk grade, infiltration, and metastasis, as well as BMI-1 mRNA and protein levels were independent risk factors for prognosis of GIST patients. CONCLUSIONS Taken together, these findings suggest there might be a relationship between BMI-1 mRNA and protein levels, and clinicopathological characteristics, including NIH risk grade, tumor size as well as infiltration and metastasis, of GIST patients. In addition, BMI-1 mRNA and protein levels were identified as independent risk factors for prognosis of GIST patients.
Collapse
Affiliation(s)
- Kun Zhu
- Department of Oncology Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, P.R. China
| | - Kang Li
- Department of Oncology Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, P.R. China
| | - Da-Wei Yuan
- Department of Oncology Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, P.R. China
| | - Gang Xu
- Department of Oncology Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, P.R. China
| | - Li Kang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, P.R. China
| | - Cheng-Xue Dang
- Department of Oncology Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, P.R. China
| | - Yong Zhang
- Department of Oncology Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, P.R. China
| |
Collapse
|
45
|
Szucs Z, Jones RL. Perspectives on the evolving state of the art management of gastrointestinal stromal tumours. Transl Gastroenterol Hepatol 2018; 3:21. [PMID: 29780899 DOI: 10.21037/tgh.2018.04.02] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 03/30/2018] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal stromal tumours (GISTs) represent a very exciting tumour entity for the medical oncologist. There has been extensive clinical and preclinical research dissecting the natural behaviour, molecular landscape and therapeutic responsiveness of this rare mesenchymal tumour. Various molecular subtypes of GIST have a differing prognostic and predictive relevance in the state of the art management of the disease. Emerging mature clinical trial data gathered over the last one and half decade provided substantial molecular profiling information in understanding the success and eventual failure of treatment. In our review of the most relevant literature we aim to guide the clinician in tailoring neoadjuvant, adjuvant and palliative treatment of GIST alongside the different, now well established molecular subgroups of GISTs.
Collapse
Affiliation(s)
- Zoltan Szucs
- Consultant Medical Oncologist, Ipswich Hospital NHS Trust, UK
| | - Robin L Jones
- Consultant Medical Oncologist, Head of Sarcoma Unit, The Royal Marsden Hospital NHS Foundation Trust/Institute of Cancer Research, Fulham Road, London, SW3 6JJ, UK
| |
Collapse
|
46
|
Nishida T, Cho H, Hirota S, Masuzawa T, Chiguchi G, Tsujinaka T. Clinicopathological Features and Prognosis of Primary GISTs with Tumor Rupture in the Real World. Ann Surg Oncol 2018; 25:1961-1969. [PMID: 29752602 PMCID: PMC5976711 DOI: 10.1245/s10434-018-6505-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Indexed: 01/21/2023]
Abstract
Background Patients with ruptured gastrointestinal stromal tumor (GIST) are recommended for imatinib adjuvant therapy; however, their clinicopathological features and prognosis in the era of imatinib are unknown. Patients and Methods The study cohort included 665 patients with histologically proven primary GISTs who underwent R0 or R1 surgery between 2003 and 2007; the validation cohort included 182 patients between 2000 and 2014. The definitions of tumor rupture in the study included perforation at tumor site, tumor fracture, piecemeal resection including open biopsy, and macroscopic injuries to the pseudocapsule. Results Tumor rupture occurred in 21 (3.2%) of 665 and 5 (2.9%) of 182 patients in the study and validation cohort, respectively. Ruptured GISTs were more symptomatic, were larger in size, and had higher mitotic count than nonruptured GISTs but were not associated with tumor location or laparoscopic surgery. GISTs with intraoperative rupture had clinicopathological features and prognostic outcomes similar to those with preoperative rupture. Recurrence rates were higher and median recurrence-free survival (RFS) and overall survival (OS) were shorter with ruptured than nonruptured GIST. Tumor rupture was one of the independent prognostic factors for RFS, but not OS, according to multivariate analysis. Conclusions Ruptured GISTs were symptomatic larger tumors with high mitotic activity, frequent relapse, and shorter RFS. Tumor rupture was an independent prognostic factor for RFS, but not for OS, in the era of imatinib. Electronic supplementary material The online version of this article (10.1245/s10434-018-6505-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Toshirou Nishida
- Department of Surgery, National Cancer Center Hospital, Tokyo, Japan. .,Department of Surgery, Osaka Police Hospital, Osaka, Japan. .,Department of Surgery, Kansai Rosai Hospital, Amagasaki, Japan.
| | - Haruhiko Cho
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan.,Department of Surgery, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Seiichi Hirota
- Department of Surgical Pathology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Toru Masuzawa
- Department of Surgery, Osaka Police Hospital, Osaka, Japan.,Department of Surgery, Kansai Rosai Hospital, Amagasaki, Japan
| | - Gaku Chiguchi
- Department of Gastroenterology, Yokohama Rosai Hospital, Yokohama, Japan
| | - Toshimasa Tsujinaka
- Department of Gastrointestinal Surgery, Kaizuka City Hospital, Kaizuka, Japan
| | | |
Collapse
|
47
|
Khoo CY, Chai X, Quek R, Teo MCC, Goh BKP. Systematic review of current prognostication systems for primary gastrointestinal stromal tumors. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2018; 44:388-394. [PMID: 29422251 DOI: 10.1016/j.ejso.2017.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 12/11/2017] [Accepted: 12/20/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND The advent of tyrosine kinase inhibitors as adjuvant therapy has revolutionized the management of GIST and emphasized the need for accurate prognostication systems. Numerous prognostication systems have been proposed for GIST but at present it remains unknown which system is superior. The present systematic review aims to summarize current prognostication systems for primary treatment-naive GIST. METHODS A literature review of the Pubmed and Embase databases was performed to identify all published articles in English, from the 1st January 2002 to 28th Feb 2017, reporting on clinical prognostication systems of GIST. RESULTS Twenty-three articles on GIST prognostication systems were included. These systems were classified as categorical systems, which stratify patients into risk groups, or continuous systems, which provide an individualized form of risk assessment. There were 16 categorical systems in total. There were 4 modifications of the National Institute of Health (NIH) system, 2 modifications of Armed Forces Institute of Pathology (AFIP) criteria and 3 modifications of Joensuu (modified NIH) criteria. Of the 7 continuous systems, there were 3 prognostic nomograms, 3 mathematical models and 1 prognostic heat/contour maps. Tumor size, location and mitotic count remain the main variables used in these systems. CONCLUSION Numerous prognostication systems have been proposed for the risk stratification of GISTs. The most widely used systems today are the NIH, Joensuu modified NIH, AFIP and the Memorial Sloan Kettering Cancer Center nomogram. More validation and comparison studies are required to determine the optimal prognostication system for GIST.
Collapse
Affiliation(s)
- Chun Yuet Khoo
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xun Chai
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Richard Quek
- Division of Medical Oncology, National Cancer Center, Singapore
| | - Melissa C C Teo
- Division of Surgical Oncology, National Cancer Center, Singapore; Duke-NUS Medical School, Singapore
| | - Brian K P Goh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore; Duke-NUS Medical School, Singapore.
| |
Collapse
|
48
|
Nishida T. Asian consensus guidelines for gastrointestinal stromal tumor: what is the same and what is different from global guidelines. Transl Gastroenterol Hepatol 2018; 3:11. [PMID: 29552662 DOI: 10.21037/tgh.2018.01.07] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 01/12/2018] [Indexed: 12/12/2022] Open
Abstract
There are some disparities between the clinical practice and profiles of cancer in Asia and those in Europe & North America. In Asia, surgical oncologists still have a major role in the multidisciplinary therapy of gastrointestinal stromal tumor (GIST), whereas medical oncologists hold this status in the West. Although the incidence of clinical GIST is considered similar between the two areas, small gastric GISTs are more frequently treated by surgery in East Asia compared with Europe & North America. The diagnosis and treatment of small submucosal tumors (SMTs), including GIST, is important in Asian clinical practice guidelines for GIST. Most items of Asian and Western GIST guidelines are very similar. There are slight differences between the two guidelines in the degree of recommendation, which may come from disparities of clinical practice and available medicines. Importantly, most clinical evidence in the GIST guidelines has been established by clinical trials conducted in Western countries, and the number of clinical trials is still limited in Asia, suggesting that Asian GIST patients may have limited access to investigational drugs after standard therapy. Finally, both Asian and Western GIST guidelines are well-harmonized in some parts, and their contents may reflect the medical circumstances of each region.
Collapse
Affiliation(s)
- Toshirou Nishida
- Department of Surgery, National Cancer Center Hospital, Chuoku, Tokyo 104-0045, Japan
| |
Collapse
|
49
|
Rossi S, Sbaraglia M, Dell'Orto MC, Gasparotto D, Cacciatore M, Boscato E, Carraro V, Toffolatti L, Gallina G, Niero M, Pilozzi E, Mandolesi A, Sessa F, Sonzogni A, Mancini C, Mazzoleni G, Romeo S, Maestro R, Dei Tos AP. Concomitant KIT/BRAF and PDGFRA/BRAF mutations are rare events in gastrointestinal stromal tumors. Oncotarget 2017; 7:30109-18. [PMID: 27097112 PMCID: PMC5058667 DOI: 10.18632/oncotarget.8768] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 04/04/2016] [Indexed: 01/13/2023] Open
Abstract
AIM The BRAF mutation is a rare pathogenetic alternative to KIT/PDGFRA mutation in GIST and causes Imatinib resistance. A recent description of KIT and BRAF mutations co-occurring in an untreated GIST has challenged the concept of their being mutually exclusive and may account for ab initio resistance to Imatinib, even in the presence of Imatinib-sensitive KIT mutations. BRAF sequencing is generally limited to KIT/PDGFRA wild-type cases. Hence, the frequency of concomitant mutations may be underestimated. METHODS We screened for KIT (exon 9, 11, 13, 17), PDGFRA (exon 12,14, 18) and BRAF (exon 15) mutations a series of 407 GIST. Additionally, we evaluated the BRAF V600E mutation-specific antibody, VE1, as a surrogate for V600E mutation, on a series of 313 GIST (24 on whole sections, 288 cases on tissue array), including 6 cases molecularly ascertained to carry the BRAF V600E mutation. RESULTS No concomitant KIT/BRAF or PDGFRA/BRAF mutations were detected. BRAF mutation was detected only in one case, wild-type for KIT/PDGFRA. All the 6 BRAF-mutant cases stained positive with the VE1 antibody. A weak VE1 expression was observed in 14/287 (4.9%) BRAF wild-type cases, as observed also in 2/6 BRAF-mutant cases. Overall in our series, sensitivity and specificity of the VE1 antobody were 100% and 95.1%, respectively. CONCLUSION The concomitance of BRAF mutation with either KIT or PDGFRA mutation is rare in GIST. In these tumors, moderate/strong VE1 immunoreactivity is a valuable surrogate for molecular analysis. Instead, genotyping is warranted in the presence of weak VE1 staining.
Collapse
Affiliation(s)
- Sabrina Rossi
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | - Marta Sbaraglia
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | - Marta Campo Dell'Orto
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | | | - Matilde Cacciatore
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | - Elena Boscato
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | - Valentina Carraro
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | - Luisa Toffolatti
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | - Giovanna Gallina
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | - Monia Niero
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, University of Rome "La Sapienza", Rome, Italy
| | - Alessandra Mandolesi
- Department of Pathology, University of Marche, Ancona School of Medicine, Ancona, Italy
| | - Fausto Sessa
- Department of Pathology, Macchi Fondation, Varese, Italy
| | | | - Cristina Mancini
- Department of Pathology, Azienda Ospedaliera-Universitaria, Parma, Italy
| | | | - Salvatore Romeo
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| | | | - Angelo P Dei Tos
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Treviso, Italy
| |
Collapse
|
50
|
Koseła-Paterczyk H, Rutkowski P. Dermatofibrosarcoma protuberans and gastrointestinal stromal tumor as models for targeted therapy in soft tissue sarcomas. Expert Rev Anticancer Ther 2017; 17:1107-1116. [DOI: 10.1080/14737140.2017.1390431] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Hanna Koseła-Paterczyk
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute Oncology Center, Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute Oncology Center, Warsaw, Poland
| |
Collapse
|