1
|
Boichuk S, Dunaev P, Galembikova A, Valeeva E. Fibroblast Growth Factor 2 (FGF2) Activates Vascular Endothelial Growth Factor (VEGF) Signaling in Gastrointestinal Stromal Tumors (GIST): An Autocrine Mechanism Contributing to Imatinib Mesylate (IM) Resistance. Cancers (Basel) 2024; 16:3103. [PMID: 39272961 PMCID: PMC11394061 DOI: 10.3390/cancers16173103] [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: 07/29/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
We showed previously that the autocrine activation of the FGFR-mediated pathway in GIST lacking secondary KIT mutations was a result of the inhibition of KIT signaling. We show here that the FGF2/FGFR pathway regulates VEGF-A/VEGFR signaling in IM-resistant GIST cells. Indeed, recombinant FGF2 increased the production of VEGF-A by IM-naive and resistant GIST cells. VEGF-A production was also increased in KIT-inhibited GIST, whereas the neutralization of FGF2 by anti-FGF2 mAb attenuated VEGFR signaling. Of note, BGJ 398, pan FGFR inhibitor, effectively and time-dependently inhibited VEGFR signaling in IM-resistant GIST T-1R cells, thereby revealing the regulatory role of the FGFR pathway in VEGFR signaling for this particular GIST cell line. This also resulted in significant synergy between BGJ 398 and VEGFR inhibitors (i.e., sunitinib and regorafenib) by enhancing their pro-apoptotic and anti-proliferative activities. The high potency of the combined use of VEGFR and FGFR inhibitors in IM-resistant GISTs was revealed by the impressive synergy scores observed for regorafenib or sunitinib and BGJ 398. Moreover, FGFR1/2 and VEGFR1/2 were co-localized in IM-resistant GIST T-1R cells, and the direct interaction between the aforementioned RTKs was confirmed by co-immunoprecipitation. In contrast, IM-resistant GIST 430 cells expressed lower basal levels of FGF2 and VEGF-A. Despite the increased expression VEGFR1 and FGFR1/2 in GIST 430 cells, these RTKs were not co-localized and co-immunoprecipitated. Moreover, no synergy between FGFR and VEGFR inhibitors was observed for the IM-resistant GIST 430 cell line. Collectively, the dual targeting of FGFR and VEGFR pathways in IM-resistant GISTs is not limited to the synergistic anti-angiogenic treatment effects. The dual inhibition of FGFR and VEGFR pathways in IM-resistant GISTs potentiates the proapoptotic and anti-proliferative activities of the corresponding RTKi. Mechanistically, the FGF2-induced activation of the FGFR pathway turns on VEGFR signaling via the overproduction of VEGF-A, induces the interaction between FGFR1/2 and VEGFR1, and thereby renders cancer cells highly sensitive to the dual inhibition of the aforementioned RTKs. Thus, our data uncovers the novel mechanism of the cross-talk between the aforementioned RTKs in IM-resistant GISTs lacking secondary KIT mutations and suggests that the dual blockade of FGFR and VEGFR signaling might be an effective treatment strategy for patients with GIST-acquired IM resistance via KIT-independent mechanisms.
Collapse
Affiliation(s)
- Sergei Boichuk
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia
- Department of Radiotherapy and Radiology, Faculty of Surgery, Russian Medical Academy of Continuous Professional Education, Moscow 125993, Russia
- "Biomarker" Research Laboratory, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Pavel Dunaev
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia
| | - Aigul Galembikova
- Department of Pathology, Kazan State Medical University, Kazan 420012, Russia
| | - Elena Valeeva
- Central Research Laboratory, Kazan State Medical University, Kazan 420012, Russia
| |
Collapse
|
2
|
Thirasastr P, Sutton TL, Joseph CP, Lin H, Amini B, Mayo SC, Araujo D, Benjamin RS, Conley AP, Livingston JA, Ludwig J, Patel S, Ratan R, Ravi V, Zarzour MA, Nassif Haddad EF, Nakazawa MS, Zhou X, Heinrich MC, Somaiah N. Outcomes of Late-Line Systemic Treatment in GIST: Does Sequence Matter? Cancers (Basel) 2024; 16:904. [PMID: 38473266 PMCID: PMC10931337 DOI: 10.3390/cancers16050904] [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: 12/30/2023] [Revised: 02/17/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Ripretinib and avapritinib have demonstrated activity in the late-line treatment of gastrointestinal stomal tumors (GISTs). We investigated whether patients previously treated with ripretinib benefit from avapritinib, and vice versa. Patients diagnosed with metastatic/unresectable GIST and treated with both drugs at two institutions in 2000-2021 were included. Patients were grouped by drug sequence: ripretinib-avapritinib (RA) or avapritinib-ripretinib (AR). Radiographic response was evaluated using RECIST 1.1. Kaplan-Meier and log-rank tests were used to compare time-to-progression (TTP) and overall survival (OS). Thirty-four patients (17 per group) were identified, with a median age of 48 years. The most common primary site was the small bowel (17/34, 50%), followed by the stomach (10/34, 29.4%). Baseline characteristics and tumor mutations were not significantly different between groups. Response rates (RRs) for ripretinib were 18% for RA and 12% for AR; RRs for avapritinib were 12% for AR and 18% for RA. Median TTPs for ripretinib were 3.65 months (95%CI 2-5.95) for RA and 4.73 months (1.87-15.84) for AR. Median TTPs for avapritinib were 5.39 months (2.86-18.99) for AR and 4.11 months (1.91-11.4) for RA. Median OS rates following RA or AR initiation were 29.63 (95%CI 13.8-50.53) and 33.7 (20.03-50.57) months, respectively. Both ripretinib and avapritinib were efficacious in the late-line treatment of GIST, with no evidence that efficacy depended on sequencing.
Collapse
Affiliation(s)
- Prapassorn Thirasastr
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - Thomas L. Sutton
- Division of Surgical Oncology, OHSU Knight Cancer Institute, Oregon Health & Science University School of Medicine, Portland, OR 97239, USA; (T.L.S.)
| | - Cissimol P. Joseph
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - Heather Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Behrang Amini
- Department of Musculoskeletal Imaging, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Skye C. Mayo
- Division of Surgical Oncology, OHSU Knight Cancer Institute, Oregon Health & Science University School of Medicine, Portland, OR 97239, USA; (T.L.S.)
| | - Dejka Araujo
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - Robert S. Benjamin
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - Anthony P. Conley
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - John A. Livingston
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - Joseph Ludwig
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - Shreyaskumar Patel
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - Ravin Ratan
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - Vinod Ravi
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - Maria A. Zarzour
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - Elise F. Nassif Haddad
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - Michael S. Nakazawa
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - Xiao Zhou
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| | - Michael C. Heinrich
- Cell and Developmental Biology, OHSU Knight Cancer Institute, Oregon Health & Science University School of Medicine, Portland, OR 97239, USA;
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.T.); (C.P.J.); (D.A.); (R.S.B.); (A.P.C.); (J.A.L.); (J.L.); (S.P.); (R.R.); (V.R.); (M.A.Z.); (E.F.N.H.); (M.S.N.); (X.Z.)
| |
Collapse
|
3
|
Nilojan JS, Gobishangar S, Sureska GM, Sarma SIT. Successful excision of a giant stomach gastrointestinal stromal tumour-A case report. Int J Surg Case Rep 2024; 115:109301. [PMID: 38277983 PMCID: PMC10839640 DOI: 10.1016/j.ijscr.2024.109301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 01/28/2024] Open
Abstract
INTRODUCTION Gastrointestinal stromal tumours are rare, but most common mesenchymal tumours originate from the gastrointestinal tract. Though surgery is the primary treatment, advanced tumours require targeted therapy in combination with surgery. CASE PRESENTATION A 62-year-old lady who presented with abdominal distention and pain was found to have a large abdominal mass. A contrast-enhanced computed tomography revealed a large abdominal mass extending from the epigastrium to the pelvis, with a solitary omental deposit. Despite receiving Imatinib for six months, the disease progressed, and she underwent open En block tumour excision with distal gastrectomy and distal pancreatectomy with Roux-en-Y gastrojejunostomy and part of omental resection after multi-disciplinary team discussions. Histological examination confirmed a spindle-type gastrointestinal stromal tumour, which arrived from the stomach's submucosa; immunohistochemistry showed strong cytoplasmic and membranous positivity for CD117. CLINICAL DISCUSSION While rare (0.1-3 % of GI malignancies), GISTs are most common in the stomach (56 %) and small bowel (32 %). Even large tumours can present with vague symptoms without obstructive features. Advanced tumours can be treated with targeted tumour therapy like Imatinib in combination with surgery. Surgical resection, usually laparoscopic, is the gold standard, but open surgery may be needed for large laparoscopically unresectable tumours. CONCLUSION Though large tumours may present with vague symptoms without obstructive features, they tend to be more aggressive and can progress despite imatinib therapy. While laparoscopic surgery is the gold standard, open surgery is preferable for large, laparoscopically unresectable tumours.
Collapse
|
4
|
Golčić M, Jones RL, Huang P, Napolitano A. Evaluation of Systemic Treatment Options for Gastrointestinal Stromal Tumours. Cancers (Basel) 2023; 15:4081. [PMID: 37627109 PMCID: PMC10452236 DOI: 10.3390/cancers15164081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Gastrointestinal stromal tumours (GIST) are the most common mesenchymal tumours of the gastrointestinal tract. Surgical treatment is recommended for the majority of localised GIST, while systemic treatment is the cornerstone of management for metastatic or unresectable disease. While a three-year regimen of imatinib is the standard of care in the adjuvant setting, there is no precise recommendation for the duration of neoadjuvant treatment, where imatinib is usually given between 4 and 12 months. Continuous treatment with imatinib at a dose of 400 mg once per day is recommended for most patients with unresectable or metastatic GIST in the first line. An exception is represented by patients with tumours harbouring the imatinib-insensitive PDGFRA D842V mutation who would be better treated with avapritinib. Targeted therapies are also recommended in the presence of NTRK rearrangements and BRAF mutations, although limited data are available. While an increase in the dose of imatinib to 800 mg is an option for the second line, sunitinib is usually considered the standard of care. Similar outcomes were reported for ripretinib in patients with tumours harbouring KIT exon 11 mutation, with significantly fewer side effects. Regorafenib and ripretinib are the standards of care in the third and fourth lines, respectively. The recent development of various systemic treatment options allows for a more personalised approach based on the molecular profile of the GIST, patient characteristics, and the profile of medications' adverse events. A multidisciplinary approach is paramount since combining systemic treatment with locoregional treatment options and supportive care is vital for long-term survival.
Collapse
Affiliation(s)
- Marin Golčić
- Department of Radiotherapy and Oncology, Clinical Hospital Center Rijeka, Krešimirova 42, 51000 Rijeka, Croatia
| | - Robin L. Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, Fulham Road, London SW3 6JJ, UK
| | - Paul Huang
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM2 5NG, UK;
| | - Andrea Napolitano
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, Fulham Road, London SW3 6JJ, UK
| |
Collapse
|
5
|
Mechahougui H, Michael M, Friedlaender A. Precision Oncology in Gastrointestinal Stromal Tumors. Curr Oncol 2023; 30:4648-4662. [PMID: 37232809 DOI: 10.3390/curroncol30050351] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
GIST (gastrointestinal stromal tumors) represent 20% of sarcomatous tumors and 1-2% of primary gastrointestinal cancers. They have an excellent prognosis when localized and resectable, though their prognosis is poor in the metastatic setting, with limited options after the second line until recently. Four lines are now standard in KIT-mutated GIST and one in PDGFRA-mutated GIST. An exponential growth of new treatments is expected in this era of molecular diagnostic techniques and systematic sequencing. Currently, the main challenge remains the emergence of resistance linked to secondary mutations caused by selective pressure induced by TKIs. Repeating biopsies to tailor treatments might be a step in the right direction, and liquid biopsies at progression may offer a non-invasive alternative. New molecules with wider KIT inhibition are under investigation and could change the catalog and the sequence of existing treatments. Combination therapies may also be an approach to overcome current resistance mechanisms. Here, we review the current epidemiology and biology of GIST and discuss future management options, with an emphasis on genome-oriented therapies.
Collapse
Affiliation(s)
- Hiba Mechahougui
- Oncology Department, Geneva University Hospital, 1205 Geneva, Switzerland
| | | | - Alex Friedlaender
- Oncology Department, Geneva University Hospital, 1205 Geneva, Switzerland
- Clinique Générale Beaulieu, 1206 Geneva, Switzerland
| |
Collapse
|
6
|
Huang S, Liu X, Guo X, Wu H, Lu H, Pan Z, Cai S, Wu X, Zhang X. Sunitinib versus imatinib dose escalation after failure of imatinib standard dose in patients with advanced Gastrointestinal stromal tumors - a real-world multi-center study. Transl Oncol 2023; 30:101641. [PMID: 36791510 PMCID: PMC9950928 DOI: 10.1016/j.tranon.2023.101641] [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: 01/10/2023] [Revised: 01/28/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Whether to escalate imatinib dosage or directly switch to sunitinib in gastrointestinal stromal tumors (GISTs) failing on standard dose 400 mg/d of imatinib is still controversial. METHODS We evaluated progression-free survival (PFS), overall survival (OS), and time to sunitinib failure (TTSF) of patients selecting imatinib dose escalation or directly switching to sunitinib after the failure of imatinib 400 mg/d therapy from 3 tertery referring centers between January 2008 to December 2016. RESULTS A total of 240 patients receiving sunitinib (37.5 mg continuous daily dose or 50 mg 4 weeks on with 2 weeks off) for at least 8 weeks were examined. After failure on imatinib 400 mg/d, 100 (49.3%) patients had dose escalation to 600 mg or 800 mg per day (IM group, imatinib group), and 103 (50.7%) directly switched to sunitinib (SU group, sunitinib group). The PFS in the SU and IM groups was 12 months and 5.0 months (P < 0.001), respectively. TTSF or OS in both groups was not statistically significantly different. CONCLUSIONS After the progression of imatinib standard-dose treatment in recurrent/metastatic GISTs, the PFS of patients directly switching to sunitinib was significantly longer compared with the PFS of patients with imatinib dose escalation. However, when the patients continued with sunitinib therapy after the failure of IM dose escalation, TTSF and OS in the IM group were similar to those in the SU group. Further exploration of the characteristics of the population benefiting from imatinib dose escalation are warranted.
Collapse
Affiliation(s)
- Shaoqing Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xing Liu
- Department of Gastric Surgery, Affiliated Union Hospital, Fujian Medical University, Fuzhou China
| | - Xiaodan Guo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hui Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huishan Lu
- Department of Colorectal Surgery, Affiliated Union Hospital, Fujian Medical University, Fuzhou China
| | - Zhizhong Pan
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shirong Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaojun Wu
- Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Xinhua Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
7
|
Vincenzi B, Napolitano A, Fiocco M, Mir O, Rutkowski P, Blay JY, Reichardt P, Joensuu H, Fumagalli E, Gennatas S, Hindi N, Nannini M, Spalato Ceruso M, Italiano A, Grignani G, Brunello A, Gasperoni S, De Pas T, Badalamenti G, Pantaleo MA, van Houdt WJ, IJzerman NS, Steeghs N, Gelderblom H, Desar IM, Falkenhorst J, Silletta M, Sbaraglia M, Tonini G, Martin-Broto J, Hohenberger P, Le Cesne A, Jones RL, Dei Tos AP, Gronchi A, Bauer S, Casali PG. Adjuvant Imatinib in Patients with GIST Harboring Exon 9 KIT Mutations: Results from a Multi-institutional European Retrospective Study. Clin Cancer Res 2022; 28:1672-1679. [PMID: 34615721 PMCID: PMC9365355 DOI: 10.1158/1078-0432.ccr-21-1665] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/14/2021] [Accepted: 09/30/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE The effect of high-dose imatinib (800 mg/day) on survival in the adjuvant treatment of patients with resected KIT exon 9-mutated gastrointestinal stromal tumors (GIST) is not established. Here, the association of dose and other clinicopathologic variables with survival was evaluated in a large multi-institutional European cohort. EXPERIMENTAL DESIGN Data from 185 patients were retrospectively collected in 23 European GIST reference centers. Propensity score matching (PSM) and inverse-probability of treatment weighting (IPTW) were used to account for confounders. Univariate and multivariate unweighted and weighted Cox proportional hazard regression models were estimated for relapse-free survival (RFS), modified-RFS (mRFS) and imatinib failure-free survival (IFFS). Univariate Cox models were estimated for overall survival. RESULTS Of the 185 patients, 131 (70.8%) received a starting dose of 400 mg/d and the remaining 54 (29.2%) a dose of 800 mg/d. Baseline characteristics were partially unbalanced, suggesting a potential selection bias. PSM and IPTW analyses showed no advantage of imatinib 800 mg/d. In the weighted multivariate Cox models, high-dose imatinib was not associated with the survival outcomes [RFS: hazard ratio (HR), 1.24; 95% confidence interval (CI), 0.79-1.94; mRFS: HR, 1.69; 95% CI, 0.92-3.10; IFFS: HR, 1.35; 95% CI, 0.79-2.28]. The variables consistently associated with worse survival outcomes were high mitotic index and nongastric tumor location. CONCLUSIONS In this retrospective series of patients with KIT exon 9-mutated GIST treated with adjuvant imatinib, a daily dose of 800 mg versus 400 mg did not show better results in terms of survival outcomes. Prospective evaluation of the more appropriate adjuvant treatment in this setting is warranted.
Collapse
Affiliation(s)
- Bruno Vincenzi
- Medical Oncology, Università Campus Bio-Medico, Rome, Italy.,Corresponding Author: Bruno Vincenzi, Medical Oncology, Università Campus Bio-Medico, Via Álvaro del Portillo 200, Rome 00128, Italy. Phone: 3906-22541-1227; E-mail:
| | - Andrea Napolitano
- Medical Oncology, Università Campus Bio-Medico, Rome, Italy.,Sarcoma Unit, Royal Marsden Hospital NHS Trust, London, United Kingdom
| | - Marta Fiocco
- Biomedical Statistics and Data Science, Mathematical Institute Leiden University, Leiden, the Netherlands
| | - Olivier Mir
- Sarcoma Group, Gustave Roussy, Villejuif, France
| | - Piotr Rutkowski
- Department of Bone/Soft Tissue Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | | | - Heikki Joensuu
- Oncology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Elena Fumagalli
- Medical Oncology Unit 2, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Spyridon Gennatas
- Sarcoma Unit, Royal Marsden Hospital NHS Trust, London, United Kingdom
| | - Nadia Hindi
- Biomedicine Institute of Seville/Virgen del Rocío University Hospital, Sevilla, Spain
| | | | | | - Antoine Italiano
- Sarcoma Unit, Institut Bergonié, Bordeaux, France.,Medical Science Faculty, University of Bordeaux, Bordeaux, France
| | - Giovanni Grignani
- Medical Oncology, Candiolo Cancer Institute-FPO-IRCCS, Candiolo, Italy
| | - Antonella Brunello
- Division of Medical Oncology, Istituto Oncologico Veneto- IRCCS, Padova, Italy
| | - Silvia Gasperoni
- Translational Oncology Unit, University Hospital Careggi, Firenze, Italy
| | - Tommaso De Pas
- Medical Oncology for Melanoma & Sarcoma, IEO - European Institute of Oncology IRCCS, Milan, Italy
| | | | | | - Winan J. van Houdt
- Surgical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nikki S. IJzerman
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.,Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Neeltje Steeghs
- Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hans Gelderblom
- Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ingrid M.E. Desar
- Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | | | | | - Marta Sbaraglia
- Pathological Anatomy, Azienda Ospedaliera di Padova, Padua, Italy
| | | | - Javier Martin-Broto
- Biomedicine Institute of Seville/Virgen del Rocío University Hospital, Sevilla, Spain
| | - Peter Hohenberger
- Division of Surgical Oncology and Thoracic Surgery, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | | | - Robin L. Jones
- Sarcoma Unit, Royal Marsden Hospital NHS Trust, London, United Kingdom.,Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | | | - Alessandro Gronchi
- Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | | | - Paolo G. Casali
- Medical Oncology Unit 2, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy.,Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
8
|
Napolitano A, Thway K, Smith MJ, Huang PH, Jones RL. KIT Exon 9-Mutated Gastrointestinal Stromal Tumours: Biology and Treatment. Chemotherapy 2022; 67:81-90. [PMID: 34983047 DOI: 10.1159/000521751] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/27/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND The majority of gastroinstestinal stromal tumours (GISTs) harbour oncogenic mutations in the gene encoding for the tyrosine kinase KIT. The most common mutations are found in exon 11, followed by mutations in exon 9. The latter mutations are associated more frequently with GISTs in extra-gastric locations and with a more aggressive clinical behaviour. SUMMARY Here, we review the unique and often poorly recognised molecular, biological and clinical characteristics that differentiate KIT exon 9-mutant GISTs from other GIST subtypes. In particular, KIT exon 9 mutations are associated to KIT mutants with retained sensitivity to stimulation by stem cell factor and localisation to the cell membrane. Moreover, KIT exon 9-mutant GISTs display significant activation of KIT-independent oncogenic pathways. These characteristics may explain the limited activity of the tyrosine kinase inhibitor imatinib in the adjuvant setting in KIT exon 9-mutant GISTs, as well as their lower sensitivity to standard dose imatinib in the advanced setting. In contranst, the multi-tyrosine kinase inhibitor sunitinib displays better activity in KIT-exon 9 mutant GISTs compared to others. Key Messages. KIT exon 9-mutant GISTs represent a subtype of GIST disctinct from others GISTs, including the more common KIT exon 11-mutant GISTs. A better understanding of the molecular biology and clinical behaviour of KIT exon 9-mutant GISTs may help identify more improved treatment options.
Collapse
Affiliation(s)
- Andrea Napolitano
- Sarcoma Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Khin Thway
- Sarcoma Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
- The Institute of Cancer Research, London, United Kingdom
| | - Myles J Smith
- Sarcoma Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
- The Institute of Cancer Research, London, United Kingdom
| | - Paul H Huang
- The Institute of Cancer Research, London, United Kingdom
| | - Robin L Jones
- Sarcoma Unit, Royal Marsden NHS Foundation Trust, London, United Kingdom
- The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
9
|
Starting Imatinib at 400 mg Daily in Patients with Gastrointestinal Stromal Tumors Harboring KIT Exon 9 Mutations: A Retrospective, Multicenter Study. Target Oncol 2021; 16:485-492. [PMID: 34089444 DOI: 10.1007/s11523-021-00820-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Retrospective analyses suggest that patients with advanced KIT exon 9-mutated gastrointestinal stromal tumors (GISTs) receiving imatinib 800 mg (rather than 400 mg) daily have better outcomes. In the adjuvant setting, the question of the optimal dose of imatinib remains unsettled. OBJECTIVE We aimed to retrospectively assess the activity of imatinib 400 mg in both the adjuvant and the advanced settings. PATIENTS AND METHODS We performed a multicenter study of patients with KIT exon 9-mutated GIST starting imatinib at 400 mg daily. We examined the relapse-free survival (RFS) among high-risk patients either receiving or not receiving adjuvant imatinib. In patients with advanced disease, progression-free survival (PFS, progression under imatinib 400 mg), time to imatinib failure (TIF, progression under imatinib 400, then 800 mg upon first progression), and overall survival (OS) were analyzed. RESULTS In the post-operative setting (n = 37), 20 patients received adjuvant imatinib. Median RFS in high-risk patients receiving adjuvant imatinib (n = 14) was not reached (95% CI 17.5-46.6) versus 13.6 months (95% CI 4.7-13.6) for those who did not (p = 0.37), after a median follow-up of 58 months. RFS at 36 months was 63% (30.3-96.6) versus 40% (95% CI 0-82.9), p = 0.2. In advanced disease (n = 28), median PFS, TIF and OS were 12.7 months (95% CI 6.1-18.2), 21.0 months (95% CI 17.4-28.1) and 47.0 months (95% CI 33.5-69.2), respectively. CONCLUSIONS Despite the limitations of a retrospective analysis and the small number of patients, the benefit of adjuvant imatinib 400 mg daily in high-risk patients appeared relevant. Patients with advanced disease receiving imatinib 400 mg with subsequent dose escalation had a TIF similar to that observed with an initial dose of 800 mg. Intra-patient dose escalation in this setting might be an option.
Collapse
|
10
|
Bertucci A, Guiramand J, Mescam L, Monneur A, Bisbal M, Chow-Chine L, Sannini A, Perrot D, De Luca V, Mokart D, Bertucci F. Successful Imatinib Treatment of an Abdominal Compartment Syndrome due to Huge Gastrointestinal Stromal Tumour. Case Rep Oncol 2019; 12:644-649. [PMID: 31572153 PMCID: PMC6751450 DOI: 10.1159/000502338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 07/23/2019] [Indexed: 11/30/2022] Open
Abstract
Gastrointestinal stromal tumours (GISTs) are the most common digestive mesenchymal tumours, whose prognosis has been revolutionised by targeted therapies such as oral imatinib. Abdomen compartment syndrome (ACS) is associated with mortality superior to 50% in adults. ACS has never been reported to date in patients with GIST. Specific anticancer treatment in critically ill patients in intensive care unit (ICU) remains a matter of debate given the high mortality rate. Here, we report the case of a 58-year-old woman with ACS related to a 40-cm huge GIST and multi-organ failure requiring mechanical ventilation, vasopressive support and haemodialysis. She was treated in emergency with imatinib via the naso-gastric tube (day 1), then at day 3 by decompressive laparotomy and “open abdomen” without any tumour removal. Imaging after 11 days imatinib showed objective tumour response. Because of improvement of multi-organ dysfunctions, the laparotomy was closed at day 14, and the resuscitation procedures were progressively stopped. After discharge from hospital, she survived nearly two years. This is the first case of successful treatment of cancer-associated ACS by targeted therapy and decompressive laparotomy. Imatinib in critically ill patients with GIST may be successful even in presence of multi-organ failure.
Collapse
Affiliation(s)
- Alexandre Bertucci
- Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille, INSERM UMR1068, CNRS UMR725, Institut Paoli-Calmettes, Marseille, France
| | - Jérôme Guiramand
- Department of Surgery, Institut Paoli-Calmettes, Marseille, France
| | - Lena Mescam
- Department of Pathology, Institut Paoli-Calmettes, Marseille, France
| | - Audrey Monneur
- Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille, INSERM UMR1068, CNRS UMR725, Institut Paoli-Calmettes, Marseille, France
| | - Magali Bisbal
- Unit of Intensive Care, Institut Paoli-Calmettes, Marseille, France
| | | | - Antoine Sannini
- Unit of Intensive Care, Institut Paoli-Calmettes, Marseille, France
| | - Delphine Perrot
- Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille, INSERM UMR1068, CNRS UMR725, Institut Paoli-Calmettes, Marseille, France
| | - Valéria De Luca
- Department of Radiology, Institut Paoli-Calmettes, Marseille, France
| | - Djamel Mokart
- Unit of Intensive Care, Institut Paoli-Calmettes, Marseille, France
| | - François Bertucci
- Department of Medical Oncology, Centre de Recherche en Cancérologie de Marseille, INSERM UMR1068, CNRS UMR725, Institut Paoli-Calmettes, Marseille, France
| |
Collapse
|
11
|
Parab TM, DeRogatis MJ, Boaz AM, Grasso SA, Issack PS, Duarte DA, Urayeneza O, Vahdat S, Qiao JH, Hinika GS. Gastrointestinal stromal tumors: a comprehensive review. J Gastrointest Oncol 2018; 10:144-154. [PMID: 30788170 DOI: 10.21037/jgo.2018.08.20] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are rare neoplasms of the gastrointestinal tract associated with high rates of malignant transformation. Most GISTs present asymptomatically. They are best identified by computed tomography (CT) scan and most stain positive for CD117 (C-Kit), CD34, and/or DOG-1. There have been many risk stratification classifications systems which are calculated based on tumor size, mitotic rate, location, and perforation. The approaches to treating GISTs are to resect primary low-risk tumors, resect high-risk primary or metastatic tumors with imatinib 400 mg daily for 12 months, or if the tumor is unresectable, neoadjuvant imatinib 400 mg daily followed by surgical resection is recommended. Sunitinib is required for KIT exon 9, 13, and 14 mutations, while ponatinib is used for exon 17 mutations and regorafenib for highly refractory tumors. High-risk tumors should be monitored for recurrence with serial abdominal CT scans. Radiofrequency ablation has shown to be effective when surgery is not suitable. Newer therapies of ipilimumab, nivolumab, and endoscopic ultrasound alcohol ablation have shown promising results. This report addresses the epidemiology, clinical presentation, diagnostic imaging, histologic diagnosis, classification and risk stratification, staging and grading, surgical treatment, adjuvant treatment, and metastasis of GISTs.
Collapse
Affiliation(s)
- Trisha M Parab
- Department of Surgery, Dignity Health California Hospital Medical Center, Los Angeles, CA, USA
| | - Michael J DeRogatis
- Department of Surgery, Dignity Health California Hospital Medical Center, Los Angeles, CA, USA
| | - Alexander M Boaz
- Department of Surgery, Dignity Health California Hospital Medical Center, Los Angeles, CA, USA
| | - Salvatore A Grasso
- Department of Surgery, Dignity Health California Hospital Medical Center, Los Angeles, CA, USA
| | - Paul S Issack
- Department of Orthopaedic Surgery, New York-Presbyterian Hospital, New York, NY, USA
| | - David A Duarte
- Department of Surgery, Dignity Health California Hospital Medical Center, Los Angeles, CA, USA
| | - Olivier Urayeneza
- Department of Surgery, Dignity Health California Hospital Medical Center, Los Angeles, CA, USA
| | - Saloomeh Vahdat
- Department of Pathology, Dignity Health California Hospital Medical Center, Los Angeles, CA, USA
| | - Jian-Hua Qiao
- Department of Pathology, Dignity Health California Hospital Medical Center, Los Angeles, CA, USA
| | - Gudata S Hinika
- Department of Surgery, Dignity Health California Hospital Medical Center, Los Angeles, CA, USA
| |
Collapse
|
12
|
Antiviral Screening of Multiple Compounds against Ebola Virus. Viruses 2016; 8:v8110277. [PMID: 27801778 PMCID: PMC5127007 DOI: 10.3390/v8110277] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/19/2016] [Accepted: 10/19/2016] [Indexed: 01/04/2023] Open
Abstract
In light of the recent outbreak of Ebola virus (EBOV) disease in West Africa, there have been renewed efforts to search for effective antiviral countermeasures. A range of compounds currently available with broad antimicrobial activity have been tested for activity against EBOV. Using live EBOV, eighteen candidate compounds were screened for antiviral activity in vitro. The compounds were selected on a rational basis because their mechanisms of action suggested that they had the potential to disrupt EBOV entry, replication or exit from cells or because they had displayed some antiviral activity against EBOV in previous tests. Nine compounds caused no reduction in viral replication despite cells remaining healthy, so they were excluded from further analysis (zidovudine; didanosine; stavudine; abacavir sulphate; entecavir; JB1a; Aimspro; celgosivir; and castanospermine). A second screen of the remaining compounds and the feasibility of appropriateness for in vivo testing removed six further compounds (ouabain; omeprazole; esomeprazole; Gleevec; D-LANA-14; and Tasigna). The three most promising compounds (17-DMAG; BGB324; and NCK-8) were further screened for in vivo activity in the guinea pig model of EBOV disease. Two of the compounds, BGB324 and NCK-8, showed some effect against lethal infection in vivo at the concentrations tested, which warrants further investigation. Further, these data add to the body of knowledge on the antiviral activities of multiple compounds against EBOV and indicate that the scientific community should invest more effort into the development of novel and specific antiviral compounds to treat Ebola virus disease.
Collapse
|
13
|
Molecular subtypes of gastrointestinal stromal tumor requiring specific treatments. Curr Opin Oncol 2016; 28:331-7. [DOI: 10.1097/cco.0000000000000303] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
14
|
Yuan S. Possible FDA-approved drugs to treat Ebola virus infection. Infect Dis Poverty 2015; 4:23. [PMID: 25984303 PMCID: PMC4432825 DOI: 10.1186/s40249-015-0055-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 04/13/2015] [Indexed: 01/24/2023] Open
Abstract
There is currently no effective treatment for the Ebola virus (EBOV) thus far. Most drugs and vaccines developed to date have not yet been approved for human trials. Two FDA-approved c-AbI1 tyrosine kinase inhibitors Gleevec and Tasigna block the release of viral particles; however, their clinical dosages are much lower than the dosages required for effective EBOV suppression. An α-1,2-glucosidase inhibitor Miglustat has been shown to inhibit EBOV particle assembly and secretion. Additionally, the estrogen receptor modulators Clomiphene and Toremifene prevent membrane fusion of EBOV and 50-90% of treated mice survived after Clomiphene/Toremifene treatments. However, the uptake efficiency of Clomiphene by oral administration is very low. Thus, I propose a hypothetical treatment protocol to treat Ebola virus infection with a cumulative use of both Miglustat and Toremifene to inhibit the virus effectively and synergistically. EBOV infection induces massive apoptosis of peripheral lymphocytes. Also, cytolysis of endothelial cells triggers disseminated intravascular coagulation (DIC) and subsequent multiple organ failures. Therefore, blood transfusions and active treatments with FDA-approved drugs to treat DIC are also recommended.
Collapse
Affiliation(s)
- Shu Yuan
- College of Resources Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| |
Collapse
|
15
|
Association of ABCG2 polymorphism with clinical efficacy of imatinib in patients with gastrointestinal stromal tumor. Cancer Chemother Pharmacol 2014; 75:173-82. [DOI: 10.1007/s00280-014-2630-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 11/14/2014] [Indexed: 12/29/2022]
|
16
|
Radons J. The role of inflammation in sarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 816:259-313. [PMID: 24818727 DOI: 10.1007/978-3-0348-0837-8_11] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sarcomas encompass a heterogenous group of tumors with diverse pathologically and clinically overlapping features. It is a rarely curable disease, and their management requires a multidisciplinary team approach. Chronic inflammation has emerged as one of the hallmarks of tumors including sarcomas. Classical inflammation-associated sarcomas comprise the inflammatory malignant fibrous histiocytoma and Kaposi sarcoma. The identification of specific chromosomal translocations and important intracellular signaling pathways such as Ras/Raf/MAPK, insulin-like growth factor, PI3K/AKT/mTOR, sonic hedgehog and Notch together with the increasing knowledge of angiogenesis has led to development of targeted therapies that aim to interrupt these pathways. Innovative agents like oncolytic viruses opened the way to design new therapeutic options with encouraging findings. Preclinical evidence also highlights the therapeutic potential of anti-inflammatory nutraceuticals as they can inhibit multiple pathways while being less toxic. This chapter gives an overview of actual therapeutic standards, newest evidence-based studies and exciting options for targeted therapies in sarcomas.
Collapse
Affiliation(s)
- Jürgen Radons
- Department of Radiotherapy and Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Straße 22, 81675, Munich, Germany,
| |
Collapse
|
17
|
Dual targeting of insulin and venus kinase Receptors of Schistosoma mansoni for novel anti-schistosome therapy. PLoS Negl Trop Dis 2013; 7:e2226. [PMID: 23696913 PMCID: PMC3656120 DOI: 10.1371/journal.pntd.0002226] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 04/09/2013] [Indexed: 11/21/2022] Open
Abstract
Background Chemotherapy of schistosomiasis relies on a single drug, Praziquantel (PZQ) and mass-use of this compound has led to emergence of resistant strains of Schistosoma mansoni, therefore pointing out the necessity to find alternative drugs. Through their essential functions in development and metabolism, receptor tyrosine kinases (RTK) could represent valuable drug targets for novel anti-schistosome chemotherapies. Taking advantage of the similarity between the catalytic domains of S. mansoni insulin receptors (SmIR1 and SmIR2) and Venus Kinase Receptors (SmVKR1 and SmVKR2), we studied the possibility to fight schistosomes by targeting simultaneously the four receptors with a single drug. Methodology/Principal Findings Several commercial RTK inhibitors were tested for their potential to inhibit the kinase activities of SmIR1, SmIR2, SmVKR1 and SmVKR2 intracellular domains (ICD) expressed in Xenopus oocytes. We measured the inhibitory effect of chemicals on meiosis resumption induced by the active ICD of the schistosome kinases in oocytes. The IR inhibitor, tyrphostin AG1024, was the most potent inhibitory compound towards SmIR and SmVKR kinases. In vitro studies then allowed us to show that AG1024 affected the viability of both schistosomula and adult worms of S. mansoni. At micromolar doses, AG1024 induced apoptosis and caused schistosomula death in a dose-dependent manner. In adult worms, AG1024 provoked alterations of reproductive organs, as observed by confocal laser scanner microscopy. With 5 µM AG1024, parasites were no more feeding and laying eggs, and they died within 48 h with 10 µM. Conclusion/Significance IRs and VKRs are essential in S. mansoni for key biological processes including glucose uptake, metabolism and reproduction. Our results demonstrate that inhibiting the kinase potential and function of these receptors by a single chemical compound AG1024 at low concentrations, leads to death of schistosomula and adult worms. Thus, AG1024 represents a valuable hit compound for further design of anti-kinase drugs applicable to anti-schistosome chemotherapy. Schistosomiasis is a chronic, debilitating disease that affects over 200 million people in the world. The pathology of schistosomiasis is caused mainly by host immune responses to parasite eggs and due to the formation of granulomas in liver and other tissues. There is no vaccine for schistosomiasis and treatment relies essentially on a single drug, Praziquantel. However, reduced susceptibility of schistosome isolates to Praziquantel has been reported, raising serious concerns about the need to develop new drugs against schistosomes. Receptor tyrosine kinases (RTKs) control many cellular and developmental processes and they are important targets in cancer therapy. In this paper, we have investigated the possibility to fight schistosomes by targeting with a single drug, insulin receptors (IRs) involved in parasite growth and metabolism and Venus Kinase Receptors (VKRs) which are unusual IR-like RTKs expressed in the parasite reproductive organs of Schistosoma mansoni. Diverse RTK inhibitors have been tested on kinase activities of these RTKs. The well-known IR inhibitor, tyrphostin AG1024, was demonstrated to be a potent inhibitor of both S. mansoni VKRs and IRs, able to induce in vitro death of larvae and adult worms at micromolar doses. AG1024 could represent a good hit compound for the development of novel drugs against schistosomes.
Collapse
|
18
|
Kong SH, Yang HK. Surgical treatment of gastric gastrointestinal stromal tumor. J Gastric Cancer 2013; 13:3-18. [PMID: 23610714 PMCID: PMC3627804 DOI: 10.5230/jgc.2013.13.1.3] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 03/09/2013] [Accepted: 03/10/2013] [Indexed: 12/13/2022] Open
Abstract
Gastrointestinal stromal tumor is the most common mesenchymal tumor in the gastrointestinal tract and is most frequently developed in the stomach in the form of submucosal tumor. The incidence of gastric gastrointestinal stromal tumor is estimated to be as high as 25% of the population when all small and asymptomatic tumors are included. Because gastric gastrointestinal stromal tumor is not completely distinguished from other submucosal tumors, a surgical excisional biopsy is recommended for tumors >2 cm. The surgical principles of gastrointestinal stromal tumor are composed of an R0 resection with a normal mucosa margin, no systemic lymph node dissection, and avoidance of perforation, which results in peritoneal seeding even in cases with otherwise low risk profiles. Laparoscopic surgery has been indicated for gastrointestinal stromal tumors <5 cm, and the indication for laparoscopic surgery is expanded to larger tumors if the above mentioned surgical principles can be maintained. A simple exogastric resection and various transgastric resection techniques are used for gastrointestinal stromal tumors in favorable locations (the fundus, body, greater curvature side). For a lesion at the gastroesophageal junction in the posterior wall of the stomach, enucleation techniques have been tried preserve the organ's function. Those methods have a theoretical risk of seeding a ruptured tumor, but this risk has not been evaluated by well-designed clinical trials. While some clinical trials are still on-going, neoadjuvant imatinib is suggested when marginally unresectable or multiorgan resection is anticipated to reduce the extent of surgery and the chance of incomplete resection, rupture or bleeding.
Collapse
Affiliation(s)
- Seong-Ho Kong
- Department of Surgery, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | | |
Collapse
|
19
|
Salinomycin as a drug for targeting human cancer stem cells. J Biomed Biotechnol 2012; 2012:950658. [PMID: 23251084 PMCID: PMC3516046 DOI: 10.1155/2012/950658] [Citation(s) in RCA: 250] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 10/04/2012] [Indexed: 12/17/2022] Open
Abstract
Cancer stem cells (CSCs) represent a subpopulation of tumor cells that possess self-renewal and tumor initiation capacity and the ability to give rise to the heterogenous lineages of malignant cells that comprise a tumor. CSCs possess multiple intrinsic mechanisms of resistance to chemotherapeutic drugs, novel tumor-targeted drugs, and radiation therapy, allowing them to survive standard cancer therapies and to initiate tumor recurrence and metastasis. Various molecular complexes and pathways that confer resistance and survival of CSCs, including expression of ATP-binding cassette (ABC) drug transporters, activation of the Wnt/β-catenin, Hedgehog, Notch and PI3K/Akt/mTOR signaling pathways, and acquisition of epithelial-mesenchymal transition (EMT), have been identified recently. Salinomycin, a polyether ionophore antibiotic isolated from Streptomyces albus, has been shown to kill CSCs in different types of human cancers, most likely by interfering with ABC drug transporters, the Wnt/β-catenin signaling pathway, and other CSC pathways. Promising results from preclinical trials in human xenograft mice and a few clinical pilote studies reveal that salinomycin is able to effectively eliminate CSCs and to induce partial clinical regression of heavily pretreated and therapy-resistant cancers. The ability of salinomycin to kill both CSCs and therapy-resistant cancer cells may define the compound as a novel and an effective anticancer drug.
Collapse
|
20
|
Soares PB, Jeremias TS, Alvarez-Silva M, Licinio MA, Santos-Silva MC, Vituri CL. In vitro inhibitory effects of imatinib mesylate on stromal cells and hematopoietic progenitors from bone marrow. Braz J Med Biol Res 2012; 46:39-51. [PMID: 23011404 PMCID: PMC3854343 DOI: 10.1590/s0100-879x2012007500157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2011] [Accepted: 09/11/2012] [Indexed: 11/22/2022] Open
Abstract
Imatinib mesylate (IM) is used to treat chronic myeloid leukemia (CML) because it selectively inhibits tyrosine kinase, which is a hallmark of CML oncogenesis. Recent studies have shown that IM inhibits the growth of several non-malignant hematopoietic and fibroblast cells from bone marrow (BM). The aim of the present study was to evaluate the effects of IM on stromal and hematopoietic progenitor cells, specifically in the colony-forming units of granulocyte/macrophage (CFU-GM), using BM cultures from 108 1.5- to 2-month-old healthy Swiss mice. The results showed that low concentrations of IM (1.25 µM) reduced the growth of CFU-GM in clonogenic assays. In culture assays with stromal cells, fibroblast proliferation and α-SMA expression by immunocytochemistry analysis were also reduced in a concentration-dependent manner, with a survival rate of approximately 50% with a dose of 2.5 µM. Cell viability and morphology were analyzed using MTT and staining with acrydine orange/ethidium bromide. Most cells were found to be viable after treatment with 5 µM IM, although there was gradual growth inhibition of fibroblastic cells while the number of round cells (macrophage-like cells) increased. At higher concentrations (15 µM), the majority of cells were apoptotic and cell growth ceased completely. Oil red staining revealed the presence of adipocytes only in untreated cells (control). Cell cycle analysis of stromal cells by flow cytometry showed a blockade at the G0/G1 phases in groups treated with 5-15 µM. These results suggest that IM differentially inhibits the survival of different types of BM cells since toxic effects were achieved.
Collapse
Affiliation(s)
- P B Soares
- Laboratório de Análises Clínicas, Hospital Universitário de Santa Maria, Universidade Federal de Santa Maria, Santa Maria, RS, Brasil.
| | | | | | | | | | | |
Collapse
|
21
|
Malignant Transformation of Mastocytoma Developed on Skin Mastocytosis Into Cutaneous Mast Cell Sarcoma. Am J Surg Pathol 2012; 36:779-82. [DOI: 10.1097/pas.0b013e31824c0d92] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
Sereg M, Buzogány I, Gonda G, Sápi Z, Csöregh E, Jakab Z, Rácz K, Tóth M. Gastrointestinal stromal tumor presenting as a hormonally inactive adrenal mass. Endocrine 2011; 39:1-5. [PMID: 21061093 DOI: 10.1007/s12020-010-9406-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 09/07/2010] [Indexed: 02/07/2023]
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract. Rarely, tumors with the same histological and immunohistochemical features arise in organs having no connection to the tubular gastrointestinal tract designated as extra-gastrointestinal stromal tumors (EGISTs). In this article, we report the first case of an EGIST presenting as a hormonally inactive adrenal mass. A 68-year-old woman with a 3-month history of right abdominal pain was clinically diagnosed as having a hormonally inactive right adrenal tumor sizing 15 cm in diameter. This mass and the tightly fixed right adrenal gland were resected en bloc.Histologically, the tumor was composed primarily of monomorphic spindle cells. Mitotic figure was 2–3 per 50 high power fields. Immunohistochemical analysis revealed strong positivity for CD117 (c-KIT) and smooth muscle actin (a-SMA), but negativity for beta-catenin, CD34, pan-keratin,S-100, desmin, and H-caldesmon. Genetic analysis showed no mutations in KIT gene exons 9, 11, 13, and 17,and in exon 18 of the platelet-derived growth factor-2 receptor gene (PDGFR). The patient proved to be tumor-free at the 18-month follow-up. This case under study demonstrates that EGIST should be included in the differential diagnosis of hormonally inactive adrenal tumors.CD117 (c-KIT) immunohistochemistry should be applied in the pathological workup of soft tissue adrenal tumors.This case is an additional example suggesting that the prognosis of even a very large EGIST is not definitely grave.
Collapse
Affiliation(s)
- Márta Sereg
- 2nd Department of Medicine, Semmelweis University, Szentkirályi u. 46, Budapest, Hungary
| | | | | | | | | | | | | | | |
Collapse
|