1
|
|
2
|
Montemurro M, Cioffi A, Dômont J, Rutkowski P, Roth AD, von Moos R, Inauen R, Toulmonde M, Burkhard RO, Knuesli C, Bauer S, Cassier P, Schwarb H, Le Cesne A, Koeberle D, Bärtschi D, Dietrich D, Biaggi C, Prior J, Leyvraz S. Long-term outcome of dasatinib first-line treatment in gastrointestinal stromal tumor: A multicenter, 2-stage phase 2 trial (Swiss Group for Clinical Cancer Research 56/07). Cancer 2018; 124:1449-1454. [DOI: 10.1002/cncr.31234] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 12/06/2017] [Accepted: 12/15/2017] [Indexed: 11/09/2022]
Affiliation(s)
| | - Angela Cioffi
- Medical Oncology-Sarcoma; Gustave Roussy Institute; Villejuif France
| | - Julien Dômont
- Medical Oncology-Sarcoma; Gustave Roussy Institute; Villejuif France
| | - Piotr Rutkowski
- Soft Tissue/Bone Sarcoma and Melanoma; Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology; Warsaw Poland
| | - Arnaud D. Roth
- Division of Oncology; Geneva University Hospital; Geneva Switzerland
| | - Roger von Moos
- Medical Oncology and Hematology; Cantonal Hospital Graubunden; Chur Switzerland
| | - Roman Inauen
- Department of Oncology; Cantonal Hospital St. Gallen; St. Gallen Switzerland
| | | | - Roger O. Burkhard
- Oncology Center; Hirslanden Hospital and Health Care; Zurich Switzerland
| | - Claudio Knuesli
- Medical Oncology; Hospital St. Claraspital; Basel Switzerland
| | - Sebastian Bauer
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center; University of Duisburg-Essen; Essen Germany
| | | | - Heike Schwarb
- Oncology/Internal Medicine; Cantonal Hospital Baden; Baden Switzerland
| | - Axel Le Cesne
- Medical Oncology-Sarcoma; Gustave Roussy Institute; Villejuif France
| | - Dieter Koeberle
- Department of Oncology/Hematology; Cantonal Hospital St. Gallen; St. Gallen Switzerland
| | - Daniela Bärtschi
- Coordinating Center; Swiss Group for Clinical Cancer Research; Bern Switzerland
| | - Daniel Dietrich
- Coordinating Center; Swiss Group for Clinical Cancer Research; Bern Switzerland
| | - Christine Biaggi
- Coordinating Center; Swiss Group for Clinical Cancer Research; Bern Switzerland
| | - John Prior
- Nuclear Medicine and Molecular Imaging; University Hospital of Lausanne; Lausanne Switzerland
| | - Serge Leyvraz
- Medical Oncology; University Hospital of Lausanne; Lausanne Switzerland
| |
Collapse
|
3
|
Lian J, Lin D, Xie X, Xu Y, Xu L, Meng L, Zhu Y. NVP-AUY922, a novel HSP90 inhibitor, inhibits the progression of malignant pheochromocytoma in vitro and in vivo. Onco Targets Ther 2017; 10:2219-2226. [PMID: 28458565 PMCID: PMC5403128 DOI: 10.2147/ott.s130236] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose Malignant pheochromocytoma (PCC) is a rare tumor with a very poor prognosis and no effective treatments. The aim of this study was to assess the efficacy of a novel second-generation synthetic heat-shock protein 90 (HSP90) inhibitor, NVP-AUY922, to treat malignant PCC in vitro and in vivo. Materials and methods Cell Counting Kit-8 (CCK-8) and Transwell assays were used to assess the effects of NVP-AUY922 on the proliferation and migration of the PCC cell line PC12. Flow cytometry was used to determine the effects of NVP-AUY922 on apoptosis and cell-cycle progression. Activation of phosphatidylinositol-3-kinase (PI3K)/protein kinase B (PKB/AKT) and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling was measured using a Western blot analysis. In vivo, a mouse xenograft model was used to test the effects of intraperitoneal injection of NVP-AUY922 on tumor growth. Results NVP-AUY922 was found to be cytotoxic in PC12 cells at lower concentrations compared with 17-allylamino-17-demethoxygeldanamcyin (17-AAG). NVP-AUY922 inhibited the proliferation of PC12 cells in a time- and concentration-dependent manner and decreased the rate of migration of PC12 cells. Furthermore, we found that HSP90 inhibition induced cell-cycle arrest and apoptosis. In vivo, administration of NVP-AUY922 reduced PCC tumor growth without significant weight loss. Finally, we observed the modulation of MEK/ERK and PI3K/AKT signaling in response to NVP-AUY922 exposure. Conclusion NVP-AUY922 exhibits potent anti-PCC activities in vitro and in vivo and represents a promising therapeutic small molecule for treating malignant PCC.
Collapse
Affiliation(s)
| | | | - Xing Xie
- Department of Urology, Ruijin Hospital
| | - Yunze Xu
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Lieyu Xu
- Department of Urology, Ruijin Hospital
| | - Li Meng
- Department of Urology, Ruijin Hospital
| | - Yu Zhu
- Department of Urology, Ruijin Hospital
| |
Collapse
|
4
|
Mir O, Cropet C, Toulmonde M, Cesne AL, Molimard M, Bompas E, Cassier P, Ray-Coquard I, Rios M, Adenis A, Italiano A, Bouché O, Chauzit E, Duffaud F, Bertucci F, Isambert N, Gautier J, Blay JY, Pérol D. Pazopanib plus best supportive care versus best supportive care alone in advanced gastrointestinal stromal tumours resistant to imatinib and sunitinib (PAZOGIST): a randomised, multicentre, open-label phase 2 trial. Lancet Oncol 2016; 17:632-41. [PMID: 27068858 DOI: 10.1016/s1470-2045(16)00075-9] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/19/2016] [Accepted: 01/26/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND Gastrointestinal stromal tumours (GIST) are the most common mesenchymal neoplasms of the gastrointestinal tract. Imatinib followed by sunitinib and regorafenib is the standard sequence of treatment for advanced disease. Pazopanib is effective in soft tissue sarcomas but has never been assessed in advanced GIST in a randomised trial. We aimed to assess the efficacy and safety of pazopanib in patients with previously treated advanced GIST. METHODS In this randomised, open-label phase 2 study, we enrolled adults (aged ≥18 years) with advanced GIST resistant to imatinib and sunitinib from 12 comprehensive cancer centres or university hospitals in France and randomly assigned them 1:1 using an interactive web-based centralised platform to 800 mg oral pazopanib once daily in 4-week cycles plus best supportive care or best supportive care alone. Randomisation was stratified by the number of previous treatment regimens (2 vs ≥3); no-one was masked to treatment group allocation. Upon disease progression, patients in the best supportive care group were allowed to switch to pazopanib as compassionate treatment. The primary endpoint was investigator-assessed progression-free survival, analysed by intention-to-treat. All randomised participants who received at least one dose of pazopanib were included in the safety analysis. This study is registered with ClinicalTrials.gov, number NCT01323400. FINDINGS Between April 12, 2011, and Dec 9, 2013, 81 patients were enrolled and randomly assigned to pazopanib plus best supportive care (n=40) or best supportive care alone (n=41). The median follow-up was 26·4 months (IQR 22·0-37·8) in the pazopanib plus best supportive care group and 28·9 months (22·0-35·2) in the best supportive care group. 4-month investigator-assessed progression-free survival was 45·2% (95% CI 29·1-60·0) in the pazopanib plus best supportive care group versus 17·6% (7·8-30·8) in the best supportive care group (hazard ratio [HR] 0·59, 95% CI 0·37-0·96; p=0·029). Median progression-free survival was 3·4 months (95% CI 2·4-5·6) with pazopanib plus best supportive care and 2·3 months (2·1-3·3) with best supportive care alone (HR 0·59 [0·37-0·96], p=0·03). 36 (88%) of the patients originally assigned to the best supportive care group switched to pazopanib following investigator-assessed disease progression; these patients had a median progression-free survival from pazopanib initiation of 3·5 months (95% CI 2·2-5·2). 55 (72%) of the 76 pazopanib-treated patients had pazopanib-related grade 3 or worse adverse events, the most common of which was hypertension (15 [38%] in the pazopanib plus best supportive care group and 13 [36%] in the best supportive care group). 20 (26%) patients had pazopanib-related serious adverse events (14 [35%] in the pazopanib plus best supportive care group and six [17%] in the best supportive care group), including pulmonary embolism in eight (9%) patients (five [13%] in the pazopanib plus best supportive care group and three [7%] in the best supportive care group). Three pazopanib-related deaths occurred (two pulmonary embolisms [one in each group] and one hepatic cytolysis [in the best supportive care group]). Three adverse event-related but not pazopanib-related deaths occurred in the best supportive care group after switch to pazopanib; these deaths were from hyperammonaemic encephalopathy, pneumopathy, and respiratory failure. INTERPRETATION Pazopanib plus best supportive care improves progression-free survival compared with best supportive care alone in patients with advanced GIST resistant to imatinib and sunitinib, with a toxicity profile similar to that reported for other sarcomas. This trial provides reference outcome data for future studies of targeted inhibitors in the third-line setting for these patients. FUNDING GlaxoSmithKline, French National Cancer Institute, EuroSARC (FP7-278742), Centre Léon Bérard.
Collapse
Affiliation(s)
- Olivier Mir
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Claire Cropet
- Direction of Clinical Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Maud Toulmonde
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Axel Le Cesne
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Mathieu Molimard
- Pharmacology Department, Université de Bordeaux INSERM U657, Bordeaux, France
| | - Emmanuelle Bompas
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest René Gauducheau, Saint Herblain, France
| | - Philippe Cassier
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | | | - Maria Rios
- Department of Medical Oncology, Institut de Cancérologie de Lorraine-Alexis Vautrin, Nancy, France
| | - Antoine Adenis
- Department of Medical Oncology, Centre Oscar Lambret, Lille, France
| | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Olivier Bouché
- Department of Medical Oncology, Centre Hospitalier Universitaire, Reims, France
| | - Emmanuelle Chauzit
- Pharmacology Department, Université de Bordeaux INSERM U657, Bordeaux, France
| | | | - François Bertucci
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Nicolas Isambert
- Department of Medical Oncology, Centre George-François Leclerc, Dijon, France
| | - Julien Gautier
- Direction of Clinical Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France.
| | - David Pérol
- Direction of Clinical Research and Innovation, Centre Léon Bérard, Lyon, France
| | | |
Collapse
|
5
|
Islam B, Stadlbauer P, Krepl M, Koca J, Neidle S, Haider S, Sponer J. Extended molecular dynamics of a c-kit promoter quadruplex. Nucleic Acids Res 2015; 43:8673-93. [PMID: 26245347 PMCID: PMC4605300 DOI: 10.1093/nar/gkv785] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/21/2015] [Indexed: 01/29/2023] Open
Abstract
The 22-mer c-kit promoter sequence folds into a parallel-stranded quadruplex with a unique structure, which has been elucidated by crystallographic and NMR methods and shows a high degree of structural conservation. We have carried out a series of extended (up to 10 μs long, ∼50 μs in total) molecular dynamics simulations to explore conformational stability and loop dynamics of this quadruplex. Unfolding no-salt simulations are consistent with a multi-pathway model of quadruplex folding and identify the single-nucleotide propeller loops as the most fragile part of the quadruplex. Thus, formation of propeller loops represents a peculiar atomistic aspect of quadruplex folding. Unbiased simulations reveal μs-scale transitions in the loops, which emphasizes the need for extended simulations in studies of quadruplex loops. We identify ion binding in the loops which may contribute to quadruplex stability. The long lateral-propeller loop is internally very stable but extensively fluctuates as a rigid entity. It creates a size-adaptable cleft between the loop and the stem, which can facilitate ligand binding. The stability gain by forming the internal network of GA base pairs and stacks of this loop may be dictating which of the many possible quadruplex topologies is observed in the ground state by this promoter quadruplex.
Collapse
Affiliation(s)
- Barira Islam
- Central European Institute of Technology (CEITEC), Masaryk University, Campus Bohunice, Kamenice 5, 625 00 Brno, Czech Republic
| | - Petr Stadlbauer
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65 Brno, Czech Republic
| | - Miroslav Krepl
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65 Brno, Czech Republic
| | - Jaroslav Koca
- Central European Institute of Technology (CEITEC), Masaryk University, Campus Bohunice, Kamenice 5, 625 00 Brno, Czech Republic National Center for Biomolecular Research, Faculty of Science, Masaryk University, Campus Bohunice, Kamenice 5, 625 00 Brno, Czech Republic
| | - Stephen Neidle
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Shozeb Haider
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Jiri Sponer
- Central European Institute of Technology (CEITEC), Masaryk University, Campus Bohunice, Kamenice 5, 625 00 Brno, Czech Republic Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65 Brno, Czech Republic
| |
Collapse
|
6
|
Steinmann S, Gali-Muhtasib H, Huebner K, Al-Halabi R, Abou Merhi R, Aman P, Agaimy A, Haller F, Schneider-Stock R. Hsp90 inhibition by AUY922 as an effective treatment strategy against myxoid liposarcoma. Cancer Lett 2015. [PMID: 26225840 DOI: 10.1016/j.canlet.2015.07.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Liposarcoma is one of the most common soft tissue sarcomas in adults. Recognized histological subtypes include well differentiated/dedifferentiated liposarcoma (WD/DDLS), myxoid liposarcoma (MLS) and pleomorphic liposarcoma. Currently, there are no proper subtype-specific treatments due to the genetic, histological and clinical heterogeneity of the liposarcoma subentities. In the past decade, the rising understanding of the various genetic and molecular aberrations in liposarcoma led to the development of novel alternative therapeutic strategies. One such therapy is the inhibition of the heat shock protein 90 (Hsp90) which is overexpressed in liposarcomas. In this study, we dissect the functional role of a novel potent Hsp90 inhibitor NVP-AUY922 (AUY922) in different cell lines of myxoid (MLS402, MLS1765) and undifferentiated (SW872) liposarcomas. We show that compared with 17-AAG treatment, lower concentrations of AUY922 achieve markedly cytotoxic effects on tumor cell viability. Combination treatment of AUY922 (20 nM) with Doxorubicin (300 nM) yielded a further reduction in cell viability in comparison to Doxorubicin alone. In vivo, we document an inhibition of tumor growth after AUY922 treatment. Further analyses revealed that Hsp90-inhibition induces apoptotic cell death and cell cycle arrest. In addition, we report striking perturbations of subtype-specific pattern in Raf/MEK/ERK and PI3K signaling after AUY922 application. In conclusion, our results provide evidence that Hsp90-inhibition by AUY922 may be a promising alternative therapeutic strategy for myxoid liposarcoma patients.
Collapse
Affiliation(s)
- Sara Steinmann
- Experimental Tumor Pathology, University Hospital Erlangen of Friedrich-Alexander University Erlangen-Nuremberg, Universitaetsstrasse 22, 91054 Erlangen, Germany; Institute of Pathology, University Hospital Erlangen of Friedrich-Alexander University Erlangen-Nuremberg, Krankenhausstr. 8-10, 91054 Erlangen, Germany
| | | | - Kerstin Huebner
- Experimental Tumor Pathology, University Hospital Erlangen of Friedrich-Alexander University Erlangen-Nuremberg, Universitaetsstrasse 22, 91054 Erlangen, Germany; Institute of Pathology, University Hospital Erlangen of Friedrich-Alexander University Erlangen-Nuremberg, Krankenhausstr. 8-10, 91054 Erlangen, Germany
| | - Racha Al-Halabi
- Genomic and Health Laboratory, Faculty of Sciences, R. Hariri Campus, Lebanese University, Hadath, Lebanon
| | - Raghida Abou Merhi
- Genomic and Health Laboratory, Faculty of Sciences, R. Hariri Campus, Lebanese University, Hadath, Lebanon
| | - Pierre Aman
- Sahlgrenska Cancer Center, Department of Pathology, Institute of Biomedicine, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Abbas Agaimy
- Institute of Pathology, University Hospital Erlangen of Friedrich-Alexander University Erlangen-Nuremberg, Krankenhausstr. 8-10, 91054 Erlangen, Germany
| | - Florian Haller
- Institute of Pathology, University Hospital Erlangen of Friedrich-Alexander University Erlangen-Nuremberg, Krankenhausstr. 8-10, 91054 Erlangen, Germany
| | - Regine Schneider-Stock
- Experimental Tumor Pathology, University Hospital Erlangen of Friedrich-Alexander University Erlangen-Nuremberg, Universitaetsstrasse 22, 91054 Erlangen, Germany; Institute of Pathology, University Hospital Erlangen of Friedrich-Alexander University Erlangen-Nuremberg, Krankenhausstr. 8-10, 91054 Erlangen, Germany.
| |
Collapse
|
7
|
Zhu JQ, Ou WB. Therapeutic targets in gastrointestinal stromal tumors. World J Transl Med 2015; 4:25-37. [DOI: 10.5528/wjtm.v4.i1.25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 09/14/2014] [Accepted: 12/01/2014] [Indexed: 02/05/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common type of mesenchymal tumor of the gastrointestinal tract. The tumorigenesis of GISTs is driven by gain-of-function mutations in KIT or platelet-derived growth factor receptor α (PDGFRA), resulting in constitutive activation of the tyrosine kinase and its downstream signaling pathways. Oncogenic KIT or PDGFRA mutations are compelling therapeutic targets for the treatment of GISTs, and the KIT/PDGFRA inhibitor imatinib is the standard of care for patients with metastatic GISTs. However, most GIST patients develop clinical resistance to imatinib and other tyrosine kinase inhibitors. Five mechanisms of resistance have been characterized: (1) acquisition of a secondary point mutation in KIT or PDGFRA; (2) genomic amplification of KIT; (3) activation of an alternative receptor tyrosine kinase; (4) loss of KIT oncoprotein expression; and (5) wild-type GIST. Currently, sunitinib is used as a second-line treatment for patients after imatinib failure, and regorafenib has been approved for patients whose disease is progressing on both imatinib and sunitinib. Phase II/III trials are currently in progress to evaluate novel inhibitors and immunotherapies targeting KIT, its downstream effectors such as phosphatidylinositol 3-kinase, protein kinase B and mammalian target of rapamycin, heat shock protein 90, and histone deacetylase inhibitor. Other candidate targets have been identified, including ETV1, AXL, insulin-like growth factor 1 receptor, KRAS, FAS receptor, protein kinase c theta, ANO1 (DOG1), CDC37, and aurora kinase A. These candidates warrant clinical evaluation as novel therapeutic targets in GIST.
Collapse
|
8
|
Tornillo L. Gastrointestinal stromal tumor - an evolving concept. Front Med (Lausanne) 2014; 1:43. [PMID: 25593916 PMCID: PMC4291900 DOI: 10.3389/fmed.2014.00043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/17/2014] [Indexed: 12/18/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most frequent mesenchymal tumors of the gastrointestinal tract. The discovery that these tumors, formerly thought of smooth muscle origin, are indeed better characterized by specific activating mutation in genes coding for the receptor tyrosine kinases (RTKs) CKIT and PDGFRA and that these mutations are strongly predictive for the response to targeted therapy with RTK inhibitors has made GISTs the typical example of the integration of basic molecular knowledge in the daily clinical activity. The information on the mutational status of these tumors is essential to predict (and subsequently to plan) the therapy. As resistant cases are frequently wild type, other possible oncogenic events, defining other "entities," have been discovered (e.g., succinil dehydrogenase mutation/dysregulation, insuline growth factor expression, and mutations in the RAS-RAF-MAPK pathway). The classification of disease must nowadays rely on the integration of the clinico-morphological characteristics with the molecular data.
Collapse
Affiliation(s)
- Luigi Tornillo
- Institute of Pathology, University of Basel , Basel , Switzerland
| |
Collapse
|
9
|
Barik S, Bhuniya A, Banerjee S, Das A, Sarkar M, Paul T, Ghosh T, Ghosh S, Roy S, Pal S, Bose A, Baral R. Neem leaf glycoprotein is superior than cisplatin and sunitinib malate in restricting melanoma growth by normalization of tumor microenvironment. Int Immunopharmacol 2013; 17:42-9. [PMID: 23747315 DOI: 10.1016/j.intimp.2013.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 04/27/2013] [Accepted: 05/07/2013] [Indexed: 12/21/2022]
Abstract
We have observed earlier that therapeutic treatment with neem leaf glycoprotein (NLGP) inhibits murine B16-melanoma growth in vivo and improves survivability of treated mice. Anti-tumor effect of NLGP is directly associated with enhanced CD8(+) T cell activity and downregulation of suppressive cellular functions. Objective of this present study is to know the efficacy of NLGP in comparison to two popular drugs, Cisplatin and Sunitinib malate (Sutent) in relation to the modulation of tumor microenvironment (TME). Analysis of cytokine milieu within TME revealed IL-10, TGFβ, IL-6 rich type 2 characters was significantly switched to type 1 microenvironment with dominance of IFNγ and IL-2 within NLGP-TME, which was not found in other cases; however Cisplatin-TME appeared better in type 2 to type 1 conversion than Sutent-TME as evidenced by RT-PCR, ELISA and immunohistochemical analysis. NLGP-TME educated CD8(+) T cells exhibited greater cytotoxicity to B16 Melanoma cells in vitro and these cells showed comparatively higher expression of cytotoxicity related molecules, perforin and granzyme B than Cisplatin-TME and Sutent-TME educated T cells. Adoptive transfer of NLGP-TME exposed T cells, but not PBS-TME exposed cells in mice, is able to significantly inhibit the growth of melanoma in vivo. Such tumor growth inhibition was in significantly lower extent when therapeutic CD8(+) T cells were exposed to either Cisplatin-TME or Sutent-TME or control-TME. Accumulated evidences strongly suggest that non toxic NLGP normalized TME allows T cells to perform optimally than other TMEs under study to inhibit the melanoma growth.
Collapse
Affiliation(s)
- Subhasis Barik
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), 37, S. P. Mukherjee Road, Kolkata 700026, India
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Kulkarni H, Göring HHH, Curran JE, Diego V, Dyer TD, Cole S, Walder KR, Collier GR, Blangero J, Carless MA. Genetic basis for the increased expression of vacuolar H+ translocating ATPase genes upon imatinib treatment in human lymphoblastoid cells. Cancer Chemother Pharmacol 2013; 71:1095-100. [PMID: 23420437 DOI: 10.1007/s00280-013-2110-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 02/05/2013] [Indexed: 12/01/2022]
Abstract
PURPOSE The role of v-ATPases in cancer biology is being increasingly recognized. Yeast studies indicate that the tyrosine kinase inhibitor imatinib may interact with the v-ATPase genes and alter the course of cancer progression. Data from humans in this regard are lacking. METHODS We constructed 55 lymphoblastoid cell lines from pedigreed, cancer-free human subjects and treated them with IC20 concentration of imatinib mesylate. Using these cell lines, we (i) estimated the heritability and differential expression of 19 genes encoding several subunits of the v-ATPase protein in response to imatinib treatment; (ii) estimated the genetic similarity among these genes; and (iii) conducted a high-density scan to find cis-regulating genetic variation associated with differential expression of these genes. RESULTS We found that the imatinib response of the genes encoding v-ATPase subunits is significantly heritable and can be clustered to identify novel drug targets in imatinib therapy. Further, five of these genes were significantly cis-regulated and together represented nearly half-log fold change in response to imatinib (p = 0.0107) that was homogenous (p = 0.2598). CONCLUSIONS Our results proffer support to the growing view that personalized regimens using proton pump inhibitors or v-ATPase inhibitors may improve outcomes of imatinib therapy in various cancers.
Collapse
Affiliation(s)
- Hemant Kulkarni
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX 78227, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Montemurro M, Gelderblom H, Bitz U, Schütte J, Blay JY, Joensuu H, Trent J, Bauer S, Rutkowski P, Duffaud F, Pink D. Sorafenib as third- or fourth-line treatment of advanced gastrointestinal stromal tumour and pretreatment including both imatinib and sunitinib, and nilotinib: A retrospective analysis. Eur J Cancer 2012; 49:1027-31. [PMID: 23140824 DOI: 10.1016/j.ejca.2012.10.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 09/30/2012] [Accepted: 10/10/2012] [Indexed: 01/15/2023]
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKI) improve the outcome of patients with advanced gastrointestinal stromal tumour (GIST), but treatment failure is frequent, and prognosis then bleak. Smaller trials in this setting suggested activity for sorafenib, a multikinase inhibitor of receptor tyrosine kinases and RAF serine/threonine kinases. PATIENTS AND METHODS We retrospectively evaluated the efficacy of sorafenib, starting dose 400mg twice daily, in a large community-based cohort of 124 patients treated in 12 European and one United States (U.S.) cancer centre. All but one patient had a WHO performance score 0-2. All had failed both imatinib and sunitinib, 68 patients nilotinib and 26 had failed investigational therapy, too. RESULTS Twelve (10%) patients responded to sorafenib and 70 (57%) patients achieved disease stabilisation. Sorafenib was moderately tolerated, and toxicity reported in 56% of the patients. Rash, hand-foot-syndrome and diarrhea occurred frequently. Sorafenib dosage was reduced in a third of patients, but this did not have an impact on progression-free survival (PFS) (p=0.15). Median PFS was 6.4 months (95% confidence interval [CI], 4.6-8.0 months) and median overall survival (OS) 13.5 months (95% CI, 10.0-21.0 months). Patients with a good performance status and those who responded to sorafenib had a significant better PFS. CONCLUSION We conclude that sorafenib is active in GIST resistant to imatinib, sunitinib and nilotinib. These results warrant further investigation of sorafenib or similar molecules in GIST.
Collapse
Affiliation(s)
- M Montemurro
- Centre Pluridisciplinaire d' Oncologie, University Hospital Lausanne, Lausanne, Switzerland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Zaydfudim V, Okuno SH, Que FG, Nagorney DM, Donohue JH. Role of operative therapy in treatment of metastatic gastrointestinal stromal tumors. J Surg Res 2012; 177:248-54. [PMID: 22831567 DOI: 10.1016/j.jss.2012.07.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 05/24/2012] [Accepted: 07/03/2012] [Indexed: 12/16/2022]
Abstract
BACKGROUND Operative resection of metastatic gastrointestinal stromal tumors (GIST) is controversial. Current treatment strategies rely on the response to tyrosine kinase inhibitors (TKIs), with resultant individualization of operative intervention. We investigated the role of operative therapy in patients with metastatic GIST. METHODS This retrospective cohort study included all consecutive patients treated for metastatic and/or recurrent GIST from January 2002 to June 2011. The patients were stratified by the use of operative therapy and disease response to TKI therapy. Kaplan-Meier survival analyses with log-rank comparisons tested the effects of operative therapy and the response to TKIs on survival. RESULTS Of the 438 patients treated for GIST during the study period, 87 (median age 61 y, interquartile range 50-71; 55% male) had metastatic GIST (84% metastatic, 3% recurrent, and 13% metastatic and recurrent). Of these patients, 54 (62%) underwent operative exploration. Subtotal resection for palliative debulking (R2 resection) were performed in 19 patients; 32 patients underwent R0 resection. Operative intervention was associated with improved overall survival (OS) compared with systemic therapy alone (1 y OS, 98% versus 80% and 5-y OS, 65% versus 11%, respectively; P < 0.001). A TKI was used before resection in 32 patients. The disease response was partial in 13 patients, stable in 10, and progressive in 9. The 1- and 5-y OS and progression-free survival were strongly associated with the preoperative response to TKI and an R0 resection (all P ≤ 0.002). CONCLUSIONS Among patients with metastatic GIST, preoperative response to TKI therapy and margin-negative resection were strongly associated with improved progression-free and OS.
Collapse
Affiliation(s)
- Victor Zaydfudim
- Division of Gastroenterologic and General Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | | | |
Collapse
|
13
|
Abstract
Soft tissue sarcomas are an uncommon and diverse group of more than 50 mesenchymal malignancies. The pathogenesis of many of these is poorly understood, but others have begun to reveal the secrets of their underlying mechanisms. With considerable effort over recent years, soft tissue sarcomas have increasingly been classified on the basis of underlying molecular alterations. In turn, this has allowed the development and application of targeted agents in several specific, molecularly defined, sarcoma subtypes. This review will focus on the rationale for targeted therapy in sarcoma, with emphasis on the relevance of specific molecular factors and pathways in both translocation-associated sarcomas and in genetically complex tumors. In addition, we will address some of the early successes in sarcoma-targeted therapy as well as a few challenges and disappointments in this field. Finally, we will discuss several possible opportunities represented by poorly understood, but potentially promising new therapeutic targets, as well as several novel biological agents currently in preclinical and early phase I/II trials. This will provide the reader with the context for understanding the current state of this field and a sense of where it may be headed in the coming years.
Collapse
Affiliation(s)
- Elizabeth G Demicco
- Department of Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, 77030-4009, USA
| | | | | | | |
Collapse
|
14
|
Hauber AB, Gonzalez JM, Coombs J, Sirulnik A, Palacios D, Scherzer N. Patient preferences for reducing toxicities of treatments for gastrointestinal stromal tumor (GIST). Patient Prefer Adherence 2011; 5:307-14. [PMID: 21792302 PMCID: PMC3140312 DOI: 10.2147/ppa.s20445] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Indexed: 11/23/2022] Open
Abstract
PURPOSE To quantify gastrointestinal stromal tumor (GIST) patients' preferences for reducing treatment toxicities and the likely effect of toxicities on patients' stated adherence. METHODS English-speaking members of the Life Raft Group, a GIST patient advocacy and research organization, aged 18 years and older, completed a web-enabled survey including a series of treatment-choice questions, each presenting a pair of hypothetical GIST medication toxicity profiles. Each profile was defined by common or concerning toxicities verified via pretest interviews including: severity of edema, diarrhea, nausea, fatigue, rash, hand-foot syndrome, and heart failure; and risk of serious infection. Each subject answered 13 choice-format questions based on a predetermined experimental design with known statistical properties. Subjects were asked to rate the likelihood that they would miss or skip doses of medications with different toxicity profiles. Random-parameters logit was used to estimate a relative preference weight for each level of toxicity. RESULTS 173 subjects completed the survey. Over the ranges of toxicity levels included in the study, heart failure was the most important toxicity. Edema was the least important. For all toxicities, reducing severity from severe to moderate was more important to subjects than reducing severity from moderate to mild. Reducing heart failure from moderate to mild and diarrhea from severe to moderate had the largest effects on subjects' evaluation of adherence. CONCLUSIONS All toxicities included in the study are important to patients. Treating or reducing severe toxicities is much more important to patients than treating or reducing moderate toxicities. Focused reductions of certain toxicities may improve treatment adherence.
Collapse
Affiliation(s)
- A Brett Hauber
- RTI Health Solutions, Research Triangle Park, NC, USA
- Correspondence: A Brett Hauber, RTI Health Solutions, 3040 Cornwallis Drive, PO Box 12194, Research Triangle Park, NC 27709-2194, USA, Tel +1 215 345 6519, Fax +1 919 541 7222, Email
| | | | - John Coombs
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | - David Palacios
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | |
Collapse
|