1
|
Stecko H, Iyer S, Tsilimigras D, Pawlik TM. Demographic Trends, Co-Alterations, and Imatinib Resistance across Genomic Variants in Gastrointestinal Stromal Tumors: An AACR Project GENIE Analysis. Oncology 2024:1-14. [PMID: 39307135 DOI: 10.1159/000541454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasm of the gastrointestinal tract, the treatment of which represents a significant breakthrough in targeted cancer therapy. Given its overall rare nature, genomic differences and clinical implications between demographic groups have not been previously investigated. METHODS Anonymized demographic, clinical, and genomic data from 1,559 GIST patients in the American Association for Cancer Research Project GENIE database were analyzed using cBioPortal and custom Python scripts. Data on patient demographics, genomic alterations, and co-occurrence genetic alerations were collected and classified according to clinical implications using the OncoKB database. χ2 tests for differences in genomic alterations were used across various demographic factors and mutual exclusivity analysis was employed to identify co-mutation patterns. RESULTS Male patients demonstrated higher incidence of PDGFRA mutation (14.56% vs. 8.05%; p < 0.001), while female patients had higher likelihood of NF1 mutations (7.46% vs. 3.23%; p = 0.001). Asian patients had higher alteration rates at KIT (85.59%; p = 0.002). Co-occurrence analysis revealed KIT alterations frequently co-occurred with CDKN2A (q < 0.001), MTAP (q = 0.045), and PTEN (q = 0.056), while there was mutual exclusivity with PDGFRA (q < 0.001), NF1 (q < 0.001), and BRAF (q = 0.015). CDKN2A alterations co-occurred with MTAP (q < 0.001) and PIK3CA (q = 0.015), while being mutually exclusive with TP53 (q = 0.002) and NF1 (q = 0.007). Trends were similar among patients who had received no prior medical treatment. Imatinib-resistant mutations were more common among male patients (25.6% vs. 18.9%; p = 0.0056) and individuals under 55 (27.3% vs. 20.9%; p = 0.0228). Among patients with imatinib-resistant mutations, 77.78% had sunitinib resistance, while 70.25% maintained sensitivity to ripretinib. CONCLUSION Sex and race/ethnic differences in genomic alterations, as well as co-mutations, were prevalent among patients with GIST. Variations in mutational profiles highlight the importance of distinct genetic drivers that may be targeted to treat different patient populations.
Collapse
Affiliation(s)
- Hunter Stecko
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Sidharth Iyer
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, USA
- The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Diamantis Tsilimigras
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, Ohio, USA
| |
Collapse
|
2
|
Leong WL. From RECIST to PERCIST: navigating the landscape of tumor response assessment. Eur Radiol 2024; 34:3656-3658. [PMID: 37955672 DOI: 10.1007/s00330-023-10435-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 11/14/2023]
Affiliation(s)
- Wai Ling Leong
- Department of Biomedical Imaging, Faculty of Medicine, University Malaya Research Imaging Centre, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
3
|
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
|
4
|
Wu J, Wang C, Cui X, Liu L, Wang L, Wang J, Xue X, Dang T. MicroRNA-128 acts as a suppressor in the progression of gastrointestinal stromal tumor by targeting B-lymphoma Mo-MLV insertion region 1. Clin Transl Oncol 2024; 26:363-374. [PMID: 38103120 DOI: 10.1007/s12094-023-03354-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/11/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION The critical role of microRNA-128 (miR-128) in gastrointestinal-related diseases has been documented. In the current study, we tried to clarify the specific role miR-128 in gastrointestinal stromal tumor (GIST) and the underlying mechanism. METHODS Differentially expressed genes in GIST were identified following bioinformatics analysis. Then, expression patterns of miR-128 and B-lymphoma Mo-MLV insertion region 1 (BMI-1) in clinical tissue samples and cell lines were characterized, followed by validation of their correlation. GIST-T1 cells were selected and transfected with different mimic, inhibitor, or siRNA plasmids, after which the biological functions were assayed. RESULTS We identified low miR-128 and high BMI-1 expression in GIST tissues of 78 patients and 4 GIST cell lines. Ectopic expression of miR-128 or silencing of BMI-1 suppressed the malignant potentials of GIST-T1 cells. As a target of miR-128, BMI-1 re-expression could partly counteract the suppressive effect of miR-128 on the malignancy of GIST-T1 cells. CONCLUSION Our study provided evidence that miR-128-mediated silencing of BMI-1 could prevent malignant progression of GIST, highlighting a promising anti-tumor target for combating GIST.
Collapse
Affiliation(s)
- Jinbao Wu
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, No. 30, Hudemulin Street, Qingshan District, Baotou, 014030, Inner Mongolia Autonomous Region, People's Republic of China
| | - Changjuan Wang
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, No. 30, Hudemulin Street, Qingshan District, Baotou, 014030, Inner Mongolia Autonomous Region, People's Republic of China
| | - Xia Cui
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, No. 30, Hudemulin Street, Qingshan District, Baotou, 014030, Inner Mongolia Autonomous Region, People's Republic of China
| | - Lin Liu
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, No. 30, Hudemulin Street, Qingshan District, Baotou, 014030, Inner Mongolia Autonomous Region, People's Republic of China
| | - Lu Wang
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, No. 30, Hudemulin Street, Qingshan District, Baotou, 014030, Inner Mongolia Autonomous Region, People's Republic of China
| | - Jing Wang
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, No. 30, Hudemulin Street, Qingshan District, Baotou, 014030, Inner Mongolia Autonomous Region, People's Republic of China
| | - Xiaohui Xue
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, No. 30, Hudemulin Street, Qingshan District, Baotou, 014030, Inner Mongolia Autonomous Region, People's Republic of China
| | - Tong Dang
- Inner Mongolia Institute of Digestive Diseases, The Second Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, No. 30, Hudemulin Street, Qingshan District, Baotou, 014030, Inner Mongolia Autonomous Region, People's Republic of China.
| |
Collapse
|
5
|
Liu J, Li J, Zhu Y, Jing R, Ding S, Zhang J, Zhao L, Chen Y, Shen J. Advances in Drug Therapy for Gastrointestinal Stromal Tumour. Curr Med Chem 2024; 31:3057-3073. [PMID: 37151058 DOI: 10.2174/0929867330666230505163151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/05/2023] [Accepted: 03/03/2023] [Indexed: 05/09/2023]
Abstract
INTRODUCTION Gastrointestinal stromal tumour (GIST) is a common gastrointestinal sarcoma located in the stromal cells of the digestive tract, and molecular studies have revealed the pathogenesis of mutations in KIT and PDGFRA genes. Since imatinib opened the era of targeted therapy for GIST, tyrosine kinase inhibitors (TKIs) that can treat GIST have been developed successively. However, the lack of new drugs with satisfactory therapeutic standards has made addressing resistance a significant challenge for TKIs in the face of the resistance to first-line and second-line drugs. Therefore, we need to find as many drugs and new treatments that block mutated genes as possible. METHODS We conducted a comprehensive collection of literature using databases, integrated and analysed the selected literature based on keywords and the comprehensive nature of the articles, and finally wrote articles based on the content of the studies. RESULTS In this article, we first briefly explained the relationship between GIST and KIT/ PDGFRα and then introduced the related drug treatment. The research progress of TKIs was analyzed according to the resistance of the drugs. CONCLUSION This article describes the research progress of some TKIs and briefly introduces the currently approved TKIs and some drugs under investigation that may have better therapeutic effects, hoping to provide clues to the research of new drugs.
Collapse
Affiliation(s)
- Ju Liu
- College of Pharmacy, Liaoning University, Shenyang, Liaoning 110036, P.R. China
- API Engineering Technology Research Center of Liaoning Province, Shenyang, Liaoning 110036, P.R. China
- Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, Shenyang, Liaoning 110036, P.R. China
| | - Jiawei Li
- College of Pharmacy, Liaoning University, Shenyang, Liaoning 110036, P.R. China
| | - Yan Zhu
- College of Pharmacy, Liaoning University, Shenyang, Liaoning 110036, P.R. China
| | - Rui Jing
- College of Pharmacy, Liaoning University, Shenyang, Liaoning 110036, P.R. China
| | - Shi Ding
- College of Pharmacy, Liaoning University, Shenyang, Liaoning 110036, P.R. China
- API Engineering Technology Research Center of Liaoning Province, Shenyang, Liaoning 110036, P.R. China
- Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, Shenyang, Liaoning 110036, P. R. China
| | - Jifang Zhang
- College of Pharmacy, Liaoning University, Shenyang, Liaoning 110036, P.R. China
| | - Leyan Zhao
- College of Pharmacy, Liaoning University, Shenyang, Liaoning 110036, P.R. China
| | - Ye Chen
- College of Pharmacy, Liaoning University, Shenyang, Liaoning 110036, P.R. China
- API Engineering Technology Research Center of Liaoning Province, Shenyang, Liaoning 110036, P.R. China
- Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, Shenyang, Liaoning 110036, P.R. China
| | - Jiwei Shen
- College of Pharmacy, Liaoning University, Shenyang, Liaoning 110036, P.R. China
- API Engineering Technology Research Center of Liaoning Province, Shenyang, Liaoning 110036, P.R. China
- Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, Shenyang, Liaoning 110036, P.R. China
| |
Collapse
|
6
|
Campanella NC, Gomes INF, Alves ALV, Leal LF, Evangelista AF, Rosa MN, Melendez ME, Silva VAO, Dias RLK, Abrahão-Machado LF, Santana I, Martinho O, Guimarães DP, Faça VM, Reis RM. Biological and therapeutic implications of RKIP in Gastrointestinal Stromal Tumor (GIST): an integrated transcriptomic and proteomic analysis. Cancer Cell Int 2023; 23:256. [PMID: 37907993 PMCID: PMC10619323 DOI: 10.1186/s12935-023-03102-6] [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: 12/22/2022] [Accepted: 10/16/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GIST) represent a significant clinical challenge due to their metastatic potential and limited treatment options. Raf kinase inhibitor protein (RKIP), a suppressor of the MAPK signaling pathway, is downregulated in various cancers and acts as a metastasis suppressor. Our previous studies demonstrated low RKIP expression in GIST and its association with poor outcomes. This study aimed to expand on the previous findings and investigate the biological and therapeutic implications of RKIP loss on GIST. METHODS To validate the RKIP prognostic significance, its expression was evaluated by immunohistochemistry in 142 bona fide GIST cases. The functional role of RKIP was evaluated in vitro, using the GIST-T1 cell line, which was knocked out for RKIP. The biological and therapeutic implications of RKIP were evaluated by invasion, migration, apoptosis, and 2D / 3D viability assays. Additionally, the transcriptome and proteome of RKIP knockout cells were determined by NanoString and mass spectrometry, respectively. RESULTS Immunohistochemical analysis revealed the absence of RKIP in 25.3% of GIST cases, correlating with a tendency toward poor prognosis. Functional assays demonstrated that RKIP knockout increased GIST cells' invasion and migration potential by nearly 60%. Moreover, we found that RKIP knockout cells exhibited reduced responsiveness to Imatinib treatment and higher cellular viability in 2D and 3D in vitro models, as assessed by apoptosis-related protein expression. Through comprehensive genetic and proteomic profiling of RKIP knockout cells, we identified several putative RKIP-regulated proteins in GIST, such as COL3A1. CONCLUSIONS Using a multidimensional integrative analysis, we identified, for the first time in GIST, molecules and pathways modulated by RKIP that may potentially drive metastasis and, consequently, poor prognosis in this disease.
Collapse
Affiliation(s)
- Nathália Cristina Campanella
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
| | - Izabela Natalia Faria Gomes
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
| | - Ana Laura Vieira Alves
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
| | - Leticia Ferro Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
- School of Health Sciences Dr. Paulo Prata (FACISB), Barretos, 14785-002, Brazil
| | - Adriane Feijó Evangelista
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
| | - Marcela Nunes Rosa
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
| | - Matias Eliseo Melendez
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
- Molecular Carcinogenesis Program, National Cancer Institute, Rio de Janeiro, 20231-050, Brazil
| | - Viviane Aline Oliveira Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
- Department of Pathology, School of Medicine, Federal University of Bahia, Salvador, 40110-909, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, 40296-710, Brazil
| | - Richard Lucas Konichi Dias
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
- School of Health Sciences Dr. Paulo Prata (FACISB), Barretos, 14785-002, Brazil
| | | | - Iara Santana
- Department of Pathology, Barretos Cancer Hospital, Barretos, 14784-400, Brazil
| | - Olga Martinho
- ICVS/3B's - PT Government Associate Laboratory, Braga, 4806-909, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal
| | - Denise Peixoto Guimarães
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil
- Department of Endoscopy, Barretos Cancer Hospital, Barretos, 14784-400, Brazil
| | - Vitor Marcel Faça
- Department of Biochemistry and Immunology, Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Ribeirão Preto, 14049-900, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, CEP 14784 400, Barretos, S. Paulo, 14784-400, Brazil.
- ICVS/3B's - PT Government Associate Laboratory, Braga, 4806-909, Portugal.
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, 4710-057, Portugal.
| |
Collapse
|
7
|
Nukala SB, Jousma J, Yan G, Han Z, Kwon Y, Cho Y, Liu C, Gagnon K, Pinho S, Rehman J, Shao NY, Ong SB, Lee WH, Ong SG. Modulation of lncRNA links endothelial glycocalyx to vascular dysfunction of tyrosine kinase inhibitor. Cardiovasc Res 2023; 119:1997-2013. [PMID: 37267414 PMCID: PMC10439712 DOI: 10.1093/cvr/cvad087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 03/28/2023] [Accepted: 04/19/2023] [Indexed: 06/04/2023] Open
Abstract
AIMS Novel cancer therapies leading to increased survivorship of cancer patients have been negated by a concomitant rise in cancer therapies-related cardiovascular toxicities. Sunitinib, a first line multi-receptor tyrosine kinase inhibitor, has been reported to cause vascular dysfunction although the initiating mechanisms contributing to this side effect remain unknown. Long non-coding RNAs (lncRNAs) are emerging regulators of biological processes in endothelial cells (ECs); however, their roles in cancer therapies-related vascular toxicities remain underexplored. METHODS AND RESULTS We performed lncRNA expression profiling to identify potential lncRNAs that are dysregulated in human-induced pluripotent stem cell-derived ECs (iPSC-ECs) treated with sunitinib. We show that the lncRNA hyaluronan synthase 2 antisense 1 (HAS2-AS1) is significantly diminished in sunitinib-treated iPSC-ECs. Sunitinib was found to down-regulate HAS2-AS1 by an epigenetic mechanism involving hypermethylation. Depletion of HAS2-AS1 recapitulated sunitinib-induced detrimental effects on iPSC-ECs, whereas CRISPR-mediated activation of HAS2-AS1 reversed sunitinib-induced dysfunction. We confirmed that HAS2-AS1 stabilizes the expression of its sense gene HAS2 via an RNA/mRNA heteroduplex formation. Knockdown of HAS2-AS1 led to reduced synthesis of hyaluronic acid (HA) and up-regulation of ADAMTS5, an enzyme involved in extracellular matrix degradation, resulting in disruption of the endothelial glycocalyx which is critical for ECs. In vivo, sunitinib-treated mice showed reduced coronary flow reserve, accompanied by a reduction in Has2os and degradation of the endothelial glycocalyx. Finally, we identified that treatment with high molecular-weight HA can prevent the deleterious effects of sunitinib both in vitro and in vivo by preserving the endothelial glycocalyx. CONCLUSIONS Our findings highlight the importance of lncRNA-mediated regulation of the endothelial glycocalyx as an important determinant of sunitinib-induced vascular toxicity and reveal potential novel therapeutic avenues to attenuate sunitinib-induced vascular dysfunction.
Collapse
Affiliation(s)
- Sarath Babu Nukala
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, Chicago, IL 60607, USA
| | - Jordan Jousma
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, Chicago, IL 60607, USA
| | - Gege Yan
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, Chicago, IL 60607, USA
| | - Zhenbo Han
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, Chicago, IL 60607, USA
| | - Youjeong Kwon
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, Chicago, IL 60607, USA
| | - Yoonje Cho
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, Chicago, IL 60607, USA
| | - Chuyu Liu
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR 999078, China
| | - Keith Gagnon
- Division of Biochemistry and Molecular Biology, School of Medicine, Southern Illinois University, 1245 Lincoln Drive Carbondale, IL 62901-4413, USA
- Department of Chemistry and Biochemistry, Southern Illinois University, 1245 Lincoln Drive, Carbondale IL 62901, USA
| | - Sandra Pinho
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, Chicago, IL 60607, USA
| | - Jalees Rehman
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, Chicago, IL 60607, USA
- Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, 840 S Wood Street, Chicago, IL 60612, USA
| | - Ning-Yi Shao
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR 999078, China
| | - Sang-Bing Ong
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong (CUHK), 9/F, Lui Che Woo Clinical Sciences Building, Prince of Wales Hospital, Shatin, N.T., Hong Kong, China
- Centre for Cardiovascular Genomics and Medicine (CCGM), Lui Che Woo Institute of Innovative Medicine, 10/F, Lui Che Woo Clinical Sciences Building, Prince of Wales Hospital, Shatin, N.T., Hong Kong, China
- Hong Kong Hub of Paediatric Excellence (HK HOPE), Hong Kong Children's Hospital (HKCH), 8/F, Tower A,1 Shing Cheong Road, Kowloon Bay, Hong Kong, China
- Kunming Institute of Zoology - The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Won Hee Lee
- Department of Basic Medical Sciences, University of Arizona College of Medicine, 425 North 5th Street, Phoenix, AZ 85004, USA
| | - Sang-Ging Ong
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, 909 S Wolcott Ave, Chicago, IL 60607, USA
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong (CUHK), 9/F, Lui Che Woo Clinical Sciences Building, Prince of Wales Hospital, Shatin, N.T., Hong Kong, China
| |
Collapse
|
8
|
Tsuruta K, Takedatsu H, Yoshioka S, Yoshikai M, Tomiyasu K, Morita M, Kuwaki K, Mitsuyama K, Kawaguchi T. Symptoms Contributing to the Diagnosis of Small Bowel Tumors. Digestion 2023; 104:430-437. [PMID: 37437555 DOI: 10.1159/000531215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/22/2023] [Indexed: 07/14/2023]
Abstract
INTRODUCTION Small bowel tumors (SBTs) are difficult to diagnose because of limited opportunities and technical difficulties in evaluating the small bowel. Asymptomatic conditions or nonspecific symptoms make SBT diagnosis more challenging. In Asia, SBTs are reported to be more frequently malignant lymphoma (ML), adenocarcinoma, and gastrointestinal stromal tumor (GIST). In this study, we examined 66 patients diagnosed with SBTs and determined their clinical characteristics. METHODS This retrospective study was conducted from January 2013 to July 2020 at Kurume University Hospital. The modalities used to detect SBTs were computed tomography (CT), positron emission tomography, magnetic resonance imaging, and ultrasonography. Endoscopy was also performed in some cases to confirm SBT diagnosis. The study included 66 patients. The medical data collected included presenting symptoms, tumor location, underlying condition, diagnostic modalities, pathologic diagnosis, and treatment. RESULTS ML and adenocarcinoma were the most common tumors (22.7%), followed by GIST (21.2%) and metastatic SBT (18.2%). Symptoms that led to SBT detection were abdominal pain (44.5%), asymptomatic conditions (28.8%), hematochezia (12.1%), and anemia (10.6%). CT was the most used modality to detect SBTs. Nineteen patients were asymptomatic, and SBTs were incidentally detected in them. GISTs and benign tumors were more often asymptomatic than other malignant tumors. CONCLUSION Abdominal pain was the main symptom for SBTs in particular adenocarcinoma, ML, and metastatic SBT. In addition, GIST, which was highly prevalent in Asia, had fewer symptoms. An understanding of these characteristics may be helpful in the clinical practice of SBTs.
Collapse
Affiliation(s)
- Kozo Tsuruta
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Fukuoka, Japan,
| | - Hidetoshi Takedatsu
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Shinichiro Yoshioka
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | | | | | - Masaru Morita
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Kotaro Kuwaki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Keiichi Mitsuyama
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Fukuoka, Japan
| |
Collapse
|
9
|
Masucci MT, Motti ML, Minopoli M, Di Carluccio G, Carriero MV. Emerging Targeted Therapeutic Strategies to Overcome Imatinib Resistance of Gastrointestinal Stromal Tumors. Int J Mol Sci 2023; 24:6026. [PMID: 37046997 PMCID: PMC10094678 DOI: 10.3390/ijms24076026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 04/14/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common malignant mesenchymal neoplasms of the gastrointestinal tract. The gold standard for the diagnosis of GISTs is morphologic analysis with an immunohistochemical evaluation plus genomic profiling to assess the mutational status of lesions. The majority of GISTs are driven by gain-of-function mutations in the proto-oncogene c-KIT encoding the tyrosine kinase receptor (TKR) known as KIT and in the platelet-derived growth factor-alpha receptor (PDGFRA) genes. Approved therapeutics are orally available as tyrosine kinase inhibitors (TKIs) targeting KIT and/or PDGFRA oncogenic activation. Among these, imatinib has changed the management of patients with unresectable or metastatic GISTs, improving their survival time and delaying disease progression. Nevertheless, the majority of patients with GISTs experience disease progression after 2-3 years of imatinib therapy due to the development of secondary KIT mutations. Today, based on the identification of new driving oncogenic mutations, targeted therapy and precision medicine are regarded as the new frontiers for GISTs. This article reviews the most important mutations in GISTs and highlights their importance in the current understanding and treatment options of GISTs, with an emphasis on the most recent clinical trials.
Collapse
Affiliation(s)
- Maria Teresa Masucci
- Preclinical Models of Tumor Progression Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy
| | - Maria Letizia Motti
- Preclinical Models of Tumor Progression Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy
- Department of Movement Sciences and Wellbeing, University “Parthenope”, 80133 Naples, Italy
| | - Michele Minopoli
- Preclinical Models of Tumor Progression Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy
| | - Gioconda Di Carluccio
- Preclinical Models of Tumor Progression Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy
| | - Maria Vincenza Carriero
- Preclinical Models of Tumor Progression Unit, Istituto Nazionale Tumori IRCCS ‘Fondazione G. Pascale’, 80131 Naples, Italy
| |
Collapse
|
10
|
Safaroghli-Azar A, Emadi F, Lenjisa J, Mekonnen L, Wang S. Kinase inhibitors: Opportunities for small molecule anticancer immunotherapies. Drug Discov Today 2023; 28:103525. [PMID: 36907320 DOI: 10.1016/j.drudis.2023.103525] [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: 08/05/2022] [Revised: 01/02/2023] [Accepted: 02/07/2023] [Indexed: 03/12/2023]
Abstract
As the fifth pillar of cancer treatment, immunotherapy has dramatically changed the paradigm of therapeutic strategies by focusing on the host's immune system. In the long road of immunotherapy development, the identification of immune-modulatory effects for kinase inhibitors opened a new chapter in this therapeutic approach. These small molecule inhibitors not only directly eradicate tumors by targeting essential proteins of cell survival and proliferation but can also drive immune responses against malignant cells. This review summarizes the current standings and challenges of kinase inhibitors in immunotherapy, either as a single agent or in a combined modality.
Collapse
Affiliation(s)
- Ava Safaroghli-Azar
- Drug Discovery and Development, University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Fatemeh Emadi
- Drug Discovery and Development, University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Jimma Lenjisa
- Drug Discovery and Development, University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Laychiluh Mekonnen
- Drug Discovery and Development, University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Shudong Wang
- Drug Discovery and Development, University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia.
| |
Collapse
|
11
|
ADRB2 Regulates the Proliferation and Metastasis of Gastrointestinal Stromal Tumor Cells by Enhancing the ETV1-c-KIT Signaling. JOURNAL OF ONCOLOGY 2023; 2023:6413796. [PMID: 36778918 PMCID: PMC9918370 DOI: 10.1155/2023/6413796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/20/2022] [Accepted: 11/24/2022] [Indexed: 02/05/2023]
Abstract
Background Gastrointestinal stromal tumor (GIST) originates from a pacemaker cell, the Cajal cell. However, little is known about the cancer neuroscience in GIST. In this study, we aimed to elucidate the clinical and biological roles of adrenoceptor beta 2 (ADRB2) in GIST. Methods Immunohistochemistry was used to evaluate the expression of ADRB2 in GIST tissues. The biological effects of ADRB2 on GIST cell proliferation, migration, invasion, and apoptosis were explored using Cell Counting Kit -8, plate colony formation assay, transwell invasion assay, and flow cytometry. We also explored the growth and metastasis of xenograft tumors in nude mice. Western blotting was used to quantify protein expression and phosphorylation. Results ADRB2 is generally highly expressed in GIST. High ADRB2 expression was significantly associated with risk level, tumor size, mitotic count, and metastasis. Overexpression of ADRB2 promoted GIST cell proliferation, migration, invasion, and apoptosis, while silencing ADRB2 expression showed the opposite effects. Furthermore, we found that silencing endogenous ADRB2 inhibited GIST progression and metastasis in nude mice. ADRB2-induced ETV1 upregulation enhanced the activation of c-KIT. Conclusion ADRB2 plays an important role in the proliferation and metastasis of GIST and is expected to be a potential target for the treatment of GIST.
Collapse
|
12
|
Basu D, Pal R, Sarkar M, Barma S, Halder S, Roy H, Nandi S, Samadder A. To Investigate Growth Factor Receptor Targets and Generate Cancer Targeting Inhibitors. Curr Top Med Chem 2023; 23:2877-2972. [PMID: 38164722 DOI: 10.2174/0115680266261150231110053650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/20/2023] [Accepted: 10/02/2023] [Indexed: 01/03/2024]
Abstract
Receptor tyrosine kinase (RTK) regulates multiple pathways, including Mitogenactivated protein kinases (MAPKs), PI3/AKT, JAK/STAT pathway, etc. which has a significant role in the progression and metastasis of tumor. As RTK activation regulates numerous essential bodily processes, including cell proliferation and division, RTK dysregulation has been identified in many types of cancers. Targeting RTK is a significant challenge in cancer due to the abnormal upregulation and downregulation of RTK receptors subfamily EGFR, FGFR, PDGFR, VEGFR, and HGFR in the progression of cancer, which is governed by multiple RTK receptor signalling pathways and impacts treatment response and disease progression. In this review, an extensive focus has been carried out on the normal and abnormal signalling pathways of EGFR, FGFR, PDGFR, VEGFR, and HGFR and their association with cancer initiation and progression. These are explored as potential therapeutic cancer targets and therefore, the inhibitors were evaluated alone and merged with additional therapies in clinical trials aimed at combating global cancer.
Collapse
Affiliation(s)
- Debroop Basu
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Riya Pal
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, IndiaIndia
| | - Maitrayee Sarkar
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Soubhik Barma
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Sumit Halder
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Harekrishna Roy
- Nirmala College of Pharmacy, Vijayawada, Guntur, Andhra Pradesh, India
| | - Sisir Nandi
- Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur, 244713, India
| | - Asmita Samadder
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
- Cytogenetics and Molecular Biology Lab., Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| |
Collapse
|
13
|
Gastrointestinal stromal tumor (GIST) presenting as a multilocular cystic intra-abdominal mass in a dog. BMC Vet Res 2022; 18:415. [DOI: 10.1186/s12917-022-03504-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/03/2022] [Indexed: 11/28/2022] Open
Abstract
Abstract
Background
Gastrointestinal stromal tumor (GIST) is a malignant mesenchymal neoplasm described in humans, dogs, and cats. A hallmark of diagnosis for GISTs is positive immunohistochemical labelling with c-Kit (CD117). The differentiation of GIST from other mesenchymal neoplasms of the gastrointestinal tract is pivotal to allow for initiation of appropriate treatment. In humans, cystic GIST has been described, though this has not been reported in dogs. In humans, the cystic form of GIST has been associated with a favorable prognosis. In the present paper, we report a case of multilocular cystic GIST in a dog, which has not previously been described in this species.
Case presentation
A ten-year-old, male-entire Maltese terrier mix breed dog presented with a large cystic mural mass of the duoedenum and orad jejunum. Histopathology and positive immunohistochemical staining with CD117 confirmed a diagnosis of GIST. No evidence of metastasis was detected on routine staging with abdominal sonography and thoracic radiography at the time of diagnosis. Surgical resection was performed and toceranib therapy was initiated post-operatively. Metastasis was documented 251 days after surgery on computed tomography. Due to clinical deterioration, the patient was humanely euthanised 370 days after surgical excision.
Conclusions
There are few differential diagnoses for large multilocular cystic intra-abdominal masses in dogs. This case presents a previously undescribed presentation of gastrointestinal stromal tumor in the dog as a predominantly multilocular cystic mass. It remains unclear if the cystic form of GIST may represent a favorable prognosis in dogs.
Collapse
|
14
|
Paul D, Kales SC, Cornwell JA, Afifi MM, Rai G, Zakharov A, Simeonov A, Cappell SD. Revealing β-TrCP activity dynamics in live cells with a genetically encoded biosensor. Nat Commun 2022; 13:6364. [PMID: 36289220 PMCID: PMC9606124 DOI: 10.1038/s41467-022-33762-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/30/2022] [Indexed: 12/25/2022] Open
Abstract
The F-box protein beta-transducin repeat containing protein (β-TrCP) acts as a substrate adapter for the SCF E3 ubiquitin ligase complex, plays a crucial role in cell physiology, and is often deregulated in many types of cancers. Here, we develop a fluorescent biosensor to quantitatively measure β-TrCP activity in live, single cells in real-time. We find β-TrCP remains constitutively active throughout the cell cycle and functions to maintain discreet steady-state levels of its substrates. We find no correlation between expression levels of β-TrCP and β-TrCP activity, indicating post-transcriptional regulation. A high throughput screen of small-molecules using our reporter identifies receptor-tyrosine kinase signaling as a key axis for regulating β-TrCP activity by inhibiting binding between β-TrCP and the core SCF complex. Our study introduces a method to monitor β-TrCP activity in live cells and identifies a key signaling network that regulates β-TrCP activity throughout the cell cycle.
Collapse
Affiliation(s)
- Debasish Paul
- grid.48336.3a0000 0004 1936 8075Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892 USA
| | - Stephen C. Kales
- grid.94365.3d0000 0001 2297 5165National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850 USA
| | - James A. Cornwell
- grid.48336.3a0000 0004 1936 8075Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892 USA
| | - Marwa M. Afifi
- grid.48336.3a0000 0004 1936 8075Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892 USA
| | - Ganesha Rai
- grid.94365.3d0000 0001 2297 5165National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850 USA
| | - Alexey Zakharov
- grid.94365.3d0000 0001 2297 5165National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850 USA
| | - Anton Simeonov
- grid.94365.3d0000 0001 2297 5165National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850 USA
| | - Steven D. Cappell
- grid.48336.3a0000 0004 1936 8075Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892 USA
| |
Collapse
|
15
|
Antonarelli G, Corti C, Zucali PA, Perrino M, Manglaviti S, Lo Russo G, Varano GM, Salvini P, Curigliano G, Catania C, Conforti F, De Pas T. Continuous sunitinib schedule in advanced platinum refractory thymic epithelial neoplasms: A retrospective analysis from the ThYmic MalignanciEs (TYME) Italian collaborative group. Eur J Cancer 2022; 174:31-36. [PMID: 35970033 DOI: 10.1016/j.ejca.2022.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Thymic epithelial tumors (TETs) are rare diseases, with diverse clinical behaviour and prognosis. Intermittent dosing sunitinib represents the gold-standard systemic treatment following platinum-based chemotherapy. To ensure more homogeneous drug exposure, continuous daily dosing (CDD) sunitinib is utilised in other malignancies; however, no data exist in patients with TETs. METHODS We retrospectively examined data from patients with platinum-resistant TETs receiving CDD sunitinib 37.5 mg between 1 May 2017 and 31 May 2022 within the Italian collaborative group for ThYmic MalignanciEs. Primary end-points were median progression-free survival, overall response rate (ORR), median duration of response and major treatment-related adverse events. RESULTS A total of 20 consecutive patients (12 thymic carcinoma [TC], 6 B3, and 2 B2 thymoma) were evaluated. Among the 19 patients evaluable for response, ORR was 31.6% (95% CI, 12.5%-56.5%). Among patients with TC, one complete response, four partial responses, and four stable diseases were observed (ORR 41%).The overall median progression-free survival was 7.3 months (95% CI, 4.5-10.3): 7.3 months (95% CI, 4.4-NA) within patients with thymoma and 6.8 months (95% CI, 2.8-10.3) in patients with TC; median duration of response was 10.3 months (95% CI, 2.8-NA). CDD was associated with a manageable toxicity profile. Six patients (30%) experienced >G2 toxicity, nine required dose reduction and three discontinued treatment due to adverse events. CONCLUSIONS CDD sunitinib showed a relevant antitumor activity and confirmed a good toxicity profile. Similar effectiveness and a better toxicity profile as compared with intermittent dosing historical data suggest that this schedule should be considered.
Collapse
Affiliation(s)
- Gabriele Antonarelli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Chiara Corti
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Paolo Andrea Zucali
- Department of Oncology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| | - Matteo Perrino
- Department of Oncology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| | - Sara Manglaviti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppe Lo Russo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Gianluca Maria Varano
- Division of Interventional Radiology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Piermario Salvini
- Division of Medical Oncology, Humanitas Gavazzeni, Via Mauro Gavazzeni, 21, 24125 Bergamo, BG, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - Chiara Catania
- Division of Thoracic Medical Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Fabio Conforti
- Division of Medical Oncology, Humanitas Gavazzeni, Via Mauro Gavazzeni, 21, 24125 Bergamo, BG, Italy; Division of Medical Oncology for Melanoma, Sarcoma & Rare Tumours, European Institute of Oncology, IRCCS, Milan, Italy
| | - Tommaso De Pas
- Division of Medical Oncology, Humanitas Gavazzeni, Via Mauro Gavazzeni, 21, 24125 Bergamo, BG, Italy; Division of Medical Oncology for Melanoma, Sarcoma & Rare Tumours, European Institute of Oncology, IRCCS, Milan, Italy.
| |
Collapse
|
16
|
Wu CE, Chen CP, Huang WK, Pan YR, Aptullahoglu E, Yeh CN, Lunec J. p53 as a biomarker and potential target in gastrointestinal stromal tumors. Front Oncol 2022; 12:872202. [PMID: 35965531 PMCID: PMC9372431 DOI: 10.3389/fonc.2022.872202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 07/06/2022] [Indexed: 12/07/2022] Open
Abstract
KIT and PDGFRA play a major role in the oncogenic process in gastrointestinal stroma tumors (GIST) and small molecules have been employed with great success to target the KIT and PDGFRA pathways in this cancer. However, approximately 10% of patients with GIST are resistant to current targeted drug therapy. There is a need to explore other potential targets. Although p53 alterations frequently occur in most cancers, studies regarding p53 in GIST have been limited. The CDKN2A/MDM2/p53 axis regulates cell cycle progression and DNA damage responses, which in turn control tumor growth. This axis is the major event required for transformation from low- to high-risk GIST. Generally, p53 mutation is infrequent in GIST, but p53 overexpression has been reported to be associated with high-risk GIST and unfavorable prognosis, implying that p53 should play a critical role in GIST. Also, Wee1 regulates the cell cycle and the antitumor activity of Wee1 inhibition was reported to be p53 mutant dependent. In addition, Wee1 was reported to have potential activity in GIST through the regulation of KIT protein and this mechanism may be dependent on p53 status. In this article, we review previous reports regarding the role of p53 in GIST and propose targeting the p53 pathway as a novel additional treatment strategy for GIST.
Collapse
Affiliation(s)
- Chiao-En Wu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chiao-Ping Chen
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Wen-Kuan Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yi-Ru Pan
- Department of General Surgery and Liver Research Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Erhan Aptullahoglu
- Department of Molecular Biology and Genetics, Bilecik Seyh Edebali University, Bilecik, Turkey
| | - Chun-Nan Yeh
- Department of General Surgery and Liver Research Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
- *Correspondence: Chun-Nan Yeh, ; John Lunec,
| | - John Lunec
- Newcastle University Cancer Center, Bioscience Institute, Medical Faculty, Newcastle University, Newcastle upon Tyne, United Kingdom
- *Correspondence: Chun-Nan Yeh, ; John Lunec,
| |
Collapse
|
17
|
Ionescu C, Oprea B, Ciobanu G, Georgescu M, Bică R, Mateescu GO, Huseynova F, Barragan-Montero V. The Angiogenic Balance and Its Implications in Cancer and Cardiovascular Diseases: An Overview. Medicina (B Aires) 2022; 58:medicina58070903. [PMID: 35888622 PMCID: PMC9316440 DOI: 10.3390/medicina58070903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is the process of developing new blood vessels from pre-existing ones. This review summarizes the main features of physiological and pathological angiogenesis and those of angiogenesis activation and inhibition. In healthy adults, angiogenesis is absent apart from its involvement in female reproductive functions and tissue regeneration. Angiogenesis is a complex process regulated by the action of specific activators and inhibitors. In certain diseases, modulating the angiogenic balance can be a therapeutic route, either by inhibiting angiogenesis (for example in the case of tumor angiogenesis), or by trying to activate the process of new blood vessels formation, which is the goal in case of cardiac or peripheral ischemia.
Collapse
Affiliation(s)
- Cătălina Ionescu
- Department of Chemistry, Faculty of Sciences, University of Craiova, 107i Calea București, 200144 Craiova, Romania;
- Correspondence: (C.I.); (B.O.)
| | - Bogdan Oprea
- Histology Department, University of Medicine and Pharmacy, 2-4 Petru Rares, 200349 Craiova, Romania;
- Correspondence: (C.I.); (B.O.)
| | - Georgeta Ciobanu
- Department of Chemistry, Faculty of Sciences, University of Craiova, 107i Calea București, 200144 Craiova, Romania;
| | - Milena Georgescu
- Clinic for Plastic Surgery and Burns, County Emergency Hospital Craiova, 200642 Craiova, Romania;
| | - Ramona Bică
- General Hospital—“Victor Babes”, 281 Mihai Bravu St., Sector III, 030303 Bucharest, Romania;
| | - Garofiţa-Olivia Mateescu
- Histology Department, University of Medicine and Pharmacy, 2-4 Petru Rares, 200349 Craiova, Romania;
| | - Fidan Huseynova
- LBN, University of Montpellier, 34193 Montpellier, France; (F.H.); (V.B.-M.)
- Institute of Molecular Biology and Biotechnologies, Azerbaïjan National Academy of Sciences (ANAS), AZ1073 Baku, Azerbaijan
- Department of Histology, Cytology and Embryology, Azerbaijan Medical University, AZ1078 Baku, Azerbaijan
| | | |
Collapse
|
18
|
Arzoun H, Srinivasan M, Adam M, Thomas SS, Kuta A, Sandoval S. Evaluation of and Current Trends in the Management of Gastrointestinal Stromal Tumors: A Systematic Review. Cureus 2022; 14:e26848. [PMID: 35847170 PMCID: PMC9281617 DOI: 10.7759/cureus.26848] [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] [Accepted: 07/14/2022] [Indexed: 11/05/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are soft-tissue sarcomas that can occur anywhere in the digestive tract, with the stomach and small intestine being the most common locations. Because no imaging modalities diagnose GIST unequivocally, histological and immunohistochemical confirmation is usually required. Most GISTs are discovered by chance; hence, determining this condition's actual frequency can be challenging. Since diagnosing the tumor could be difficult, including GIST in the differential diagnosis is crucial. The objective of this review is to explore the multiple treatment options for this tumor and provide clinicians with more information on the evolving treatment modalities, which in the future could be a possible solution to cure GIST ultimately. After exploring several studies, the authors conclude that early detection is critical since the treatment depends on the tumor size, mitotic rate, and location. Medical management using targeted therapy approved by the United States Food and Drug Administration (FDA) include tyrosine kinase inhibitors such as imatinib, sunitinib, and regorafenib. Surgical resection of the tumor is also done in cases with localized tumors. Standard chemotherapy and radiotherapy are not commonly used to treat GIST patients. However, radiotherapy may be used as a palliative therapy to ease pain (such as bone pain) or control bleeding. Additional research is needed to establish potential therapeutic targets that will result in higher and longer-term response rates.
Collapse
Affiliation(s)
- Hadia Arzoun
- Internal Medicine, St. Bernards Medical Center, Jonesboro, USA
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Mirra Srinivasan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Mona Adam
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Siji S Thomas
- Internal Medicine, St. Bernards Medical Center, Jonesboro, USA
| | - Amber Kuta
- Internal Medicine, St. Bernards Medical Center, Jonesboro, USA
| | | |
Collapse
|
19
|
Liu J, Gao J, Wang A, Jiang Z, Qi S, Qi Z, Liu F, Yu K, Cao J, Chen C, Hu C, Wu H, Wang L, Wang W, Liu Q, Liu J. Nintedanib overcomes drug resistance from upregulation of FGFR signaling and imatinib-induced KIT mutations in gastrointestinal stromal tumors. Mol Oncol 2022; 16:1761-1774. [PMID: 35194937 PMCID: PMC9019892 DOI: 10.1002/1878-0261.13199] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/01/2021] [Accepted: 02/21/2022] [Indexed: 11/25/2022] Open
Abstract
Drug resistance remains a major challenge in the clinical treatment of gastrointestinal stromal tumours (GISTs). While acquired on‐target mutations of mast/stem cell growth factor receptor (KIT) kinase is the major resistance mechanism, activation of alternative signalling pathways may also play a role. Although several second‐ and third‐generation KIT kinase inhibitors have been developed that could overcome some of the KIT mutations conferring resistance, the low clinical responses and narrow safety window have limited their broad application. The present study revealed that nintedanib not only overcame resistance induced by a panel of KIT primary and secondary mutations, but also overcame ERK‐reactivation‐mediated resistance caused by the upregulation of fibroblast growth factor (FGF) activity. In preclinical models of GISTs, nintedanib significantly inhibited the proliferation of imatinib‐resistant cells, including GIST‐5R, GIST‐T1/T670I and GIST patient‐derived primary cells. In addition, it also exhibited dose‐dependent inhibition of ERK phosphorylation upon FGF ligand stimulation. In vivo antitumour activity was also observed in several xenograft GIST models. Considering the well‐documented safety and pharmacokinetic profiles of nintedanib, this finding provides evidence for the repurposing of nintedanib as a new therapy for the treatment of GIST patients with de novo or acquired resistance to imatinib.
Collapse
Affiliation(s)
- Juan Liu
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230036, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Jingjing Gao
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230036, P. R. China
| | - Aoli Wang
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Zongru Jiang
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Shuang Qi
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Ziping Qi
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Feiyang Liu
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Kailin Yu
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Jiangyan Cao
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230036, P. R. China
| | - Cheng Chen
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Chen Hu
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Hong Wu
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Li Wang
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Wenchao Wang
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| | - Qingsong Liu
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui, 230036, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,Precision Medicine Research Laboratory of Anhui Province, Hefei, Anhui, 230088, P. R. China
| | - Jing Liu
- Anhui Province Key Laboratory of Medical Physics and Technology; CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China
| |
Collapse
|
20
|
Jafari H, Atlasi Z, Mahdavinia GR, Hadifar S, Sabzi M. Magnetic κ-carrageenan/chitosan/montmorillonite nanocomposite hydrogels with controlled sunitinib release. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112042. [PMID: 33947542 DOI: 10.1016/j.msec.2021.112042] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 03/01/2021] [Accepted: 03/06/2021] [Indexed: 01/19/2023]
Abstract
This work aimed to design montmorillonite-incorporated pH-responsive and magnetic κ-carrageenan/chitosan hydrogels via a completely green route for controlled release of sunitinib anticancer drug. This was accomplished by ionic cross-linking of two biopolymers, κ-carrageenan and chitosan, in the presence of magnetic montmorillonite (mMMt) nanoplatelets. Interestingly, it was observed that the amount of mMMt affected not only the microstructure of hydrogels, but also the drug loading efficiency of nanocomposite hydrogels was noticeably increased by introducing mMMt (from 69 to 96%). The in vitro sunitinib release experiments showed that a low content of loaded sunitinib was released from all hydrogels in the buffered solution with pH 7.4. In contrast, a relatively sustained release with a high content of drug release was observed in the acidic solution of pH 5.5. During 48 h, the hydrogels nanocomposite containing a high content of mMMt showed cumulative release of 64.0 and 8.6% at pH 5.5 and 7.4, respectively. During two days, while the cumulative release of sunitinib was obtained 84.3% for the magnetic-free hydrogel, the magnetic ones showed 74.4 and 64% with the low and high contents of magnetic MMt, respectively. The developed κ-carrageenan/chitosan hydrogels with a high capacity of drug loading and subsequent pH-sensitive drug release can be considered in prolonged cancer therapy with reduced side effects.
Collapse
Affiliation(s)
- Hessam Jafari
- Polymer Research Laboratory, Department of Chemistry, Faculty of Science, University of Maragheh, 55181-83111 Maragheh, Iran
| | - Ziba Atlasi
- Polymer Research Laboratory, Department of Chemistry, Faculty of Science, University of Maragheh, 55181-83111 Maragheh, Iran
| | - Gholam Reza Mahdavinia
- Polymer Research Laboratory, Department of Chemistry, Faculty of Science, University of Maragheh, 55181-83111 Maragheh, Iran.
| | - Somayeh Hadifar
- Polymer Research Laboratory, Department of Chemistry, Faculty of Science, University of Maragheh, 55181-83111 Maragheh, Iran
| | - Mohammad Sabzi
- Department of Chemical Engineering, Faculty of Engineering, University of Maragheh, 55181-83111 Maragheh, Iran
| |
Collapse
|
21
|
Mapanao AK, Che PP, Sarogni P, Sminia P, Giovannetti E, Voliani V. Tumor grafted - chick chorioallantoic membrane as an alternative model for biological cancer research and conventional/nanomaterial-based theranostics evaluation. Expert Opin Drug Metab Toxicol 2021; 17:947-968. [PMID: 33565346 DOI: 10.1080/17425255.2021.1879047] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Introduction: Advancements in cancer management and treatment are associated with strong preclinical research data, in which reliable cancer models are demanded. Indeed, inconsistent preclinical findings and stringent regulations following the 3Rs principle of reduction, refinement, and replacement of conventional animal models currently pose challenges in the development and translation of efficient technologies. The chick embryo chorioallantoic membrane (CAM) is a system for the evaluation of treatment effects on the vasculature, therefore suitable for studies on angiogenesis. Apart from vascular effects, the model is now increasingly employed as a preclinical cancer model following tumor-grafting procedures.Areas covered: The broad application of CAM tumor model is highlighted along with the methods for analyzing the neoplasm and vascular system. The presented and cited investigations focus on cancer biology and treatment, encompassing both conventional and emerging nanomaterial-based modalities.Expert opinion: The CAM tumor model finds increased significance given the influences of angiogenesis and the tumor microenvironment in cancer behavior, then providing a qualified miniature system for oncological research. Ultimately, the establishment and increased employment of such a model may resolve some of the limitations present in the standard preclinical tumor models, thereby redefining the preclinical research workflow.
Collapse
Affiliation(s)
- Ana Katrina Mapanao
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy.,NEST-Scuola Normale Superiore, Pisa, Italy
| | - Pei Pei Che
- Department of Radiation Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center, Amsterdam, The Netherlands.,Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, The Netherlands
| | - Patrizia Sarogni
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy
| | - Peter Sminia
- Department of Radiation Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, The Netherlands.,Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per La Scienza, Pisa, Italy
| | - Valerio Voliani
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy
| |
Collapse
|
22
|
Farshidpour M, Ahmed M, Junna S, Merchant JL. Myeloid-derived suppressor cells in gastrointestinal cancers: A systemic review. World J Gastrointest Oncol 2021; 13:1-11. [PMID: 33510845 PMCID: PMC7805271 DOI: 10.4251/wjgo.v13.i1.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/01/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal (GI) cancers are one of the most common malignancies worldwide, with high rates of morbidity and mortality. Myeloid-derived suppressor cells (MDSCs) are major components of the tumor microenvironment (TME). MDSCs facilitate the transformation of premalignant cells and play roles in tumor growth and metastasis. Moreover, in patients with GI malignancies, MDSCs can lead to the suppression of T cells and natural killer cells. Accordingly, a better understanding of the role and mechanism of action of MDSCs in the TME will aid in the development of novel immune-targeted therapies.
Collapse
Affiliation(s)
- Maham Farshidpour
- Inpatient Medicine, Banner University of Medical Center, Tucson, AZ 85724, United States
| | - Monjur Ahmed
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Shilpa Junna
- Division of Gastroenterology and Hepatology, Banner University of Medical Center, Tucson, AZ 85724, United States
| | - Juanita L Merchant
- Division of Gastroenterology and Hepatology, Banner University of Medical Center, Tucson, AZ 85724, United States
| |
Collapse
|
23
|
Jaffar NFN, Muhammad Sakri MS, Jaafar H, Wan Abdul Rahman WF, Tengku Din TADAA. Evaluation of NMU-Induced Breast Cancer Treated with Sirolimus and Sunitinib on Breast Cancer Growth. Asian Pac J Cancer Prev 2020; 21:2919-2925. [PMID: 33112549 PMCID: PMC7798166 DOI: 10.31557/apjcp.2020.21.10.2919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Indexed: 12/24/2022] Open
Abstract
Objective: To analyze the effect of sirolimus and sunitinib in blocking the tumor growth and to evaluate the expressions of estrogen receptor (ER), progesterone receptor (PgR), and human epidermal growth factor receptor-2 (HER2/neu) after treated with sirolimus and sunitinib. Methods: Thirty-two female Sprague Dawley rats at age 21-days old were administered intraperitoneally with N-Methyl-N-Nitroso Urea (NMU), dosed at 70mg/kg body weight. The rats were divided into 4 groups; Group 1 (Control, n=8), Group 2 (Sirolimus, n=8), Group 3 (Sunitinib, n=8) and Group 4 (Sirolimus+Sunitinib, n=8), being treated twice when the tumor reached the size of 14.5±0.5 mm and subsequently sacrificed after 5 days. The protein expressions of ER, PgR and HER2/neu of the tumor tissues were evaluated by using immunohistochemistry analysis. Results: Treatment with sirolimus alone lowered expressions of ER and PgR of breast cancer and reduced tumor size. There was no significant difference of ER and PgR expressions between control and sunitinib treated tumor. Sunitinib treated tumors reduce in diameter after the first treatment, however the diameter increases after the second treatment. Histologically, sunitinib treated tumor did not show any aggressive invasive carcinoma of no special type (NST) histological subtypes. In addition, all NMU-induced tumors are HER2/neu-negative scoring. Conclusion: Sirolimus is neither synergistic nor additive with sunitinib for breast cancer treatment.
Collapse
Affiliation(s)
| | | | - Hasnan Jaafar
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | | |
Collapse
|
24
|
Wang S, Zhou D, Xu Z, Song J, Qian X, Lv X, Luan J. Anti-tumor Drug Targets Analysis: Current Insight and Future Prospect. Curr Drug Targets 2020; 20:1180-1202. [PMID: 30947670 DOI: 10.2174/1389450120666190402145325] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/21/2019] [Accepted: 03/22/2019] [Indexed: 12/13/2022]
Abstract
The incidence and mortality of malignant tumors are on the rise, which has become the second leading cause of death in the world. At present, anti-tumor drugs are one of the most common methods for treating cancer. In recent years, with the in-depth study of tumor biology and related disciplines, it has been gradually discovered that the essence of cell carcinogenesis is the infinite proliferation of cells caused by the disorder of cell signal transduction pathways, followed by a major shift in the concept of anti-tumor drugs research and development. The focus of research and development is shifting from traditional cytotoxic drugs to a new generation of anti-tumor drugs targeted at abnormal signaling system targets in tumor cells. In this review, we summarize the targets of anti-tumor drugs and analyse the molecular mechanisms of their effects, which lay a foundation for subsequent treatment, research and development.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Dexi Zhou
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Zhenyu Xu
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Jing Song
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Xueyi Qian
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Xiongwen Lv
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Institute for Liver Disease of Anhui Medical University, Hefei, Anhui Province, China
| | - Jiajie Luan
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| |
Collapse
|
25
|
Peng F, Liu Y. Gastrointestinal Stromal Tumors of the Small Intestine: Progress in Diagnosis and Treatment Research. Cancer Manag Res 2020; 12:3877-3889. [PMID: 32547224 PMCID: PMC7261658 DOI: 10.2147/cmar.s238227] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years, the diagnosis and treatment of gastrointestinal stromal tumors (GISTs) of the small intestine have been a hot topic due to their rarity and non-specific clinical manifestations. With the development of gene and imaging technology, surgery, and molecular targeted drugs, the diagnosis and treatment of GISTs have achieved great success. For a long time, radical resection was prioritized to treat GISTs of the small intestine. At present, preoperative tumor staging is a novel treatment for unresectable malignant tumors. In addition, karyokinesis exponent is the sole independent predictor of progression-free survival of GISTs. The DNA, miRNA, and protein of exosomes have also been found to be biomarkers with prognostic implications. The research on the treatment of GISTs has become a focus in the era of precision medicine, ushering in the use of standardized, normalized, and individualized treatment.
Collapse
Affiliation(s)
- Fangxing Peng
- Gastrointestinal Surgery, No. 2 Affiliated Hospital of North Sichuan Medical College, Mianyang, Sichuan Province 621000, People's Republic of China.,Gastrointestinal Surgery, Sichuan Mianyang 404 Hospital, Mianyang, Sichuan Province 621000, People's Republic of China
| | - Yao Liu
- Gastrointestinal Surgery, No. 2 Affiliated Hospital of North Sichuan Medical College, Mianyang, Sichuan Province 621000, People's Republic of China.,Gastrointestinal Surgery, Sichuan Mianyang 404 Hospital, Mianyang, Sichuan Province 621000, People's Republic of China
| |
Collapse
|
26
|
Jeong P, Moon Y, Lee JH, Lee SD, Park J, Lee J, Kim J, Lee HJ, Kim NY, Choi J, Heo JD, Shin JE, Park HW, Kim YG, Han SY, Kim YC. Discovery of orally active indirubin-3'-oxime derivatives as potent type 1 FLT3 inhibitors for acute myeloid leukemia. Eur J Med Chem 2020; 195:112205. [PMID: 32272419 DOI: 10.1016/j.ejmech.2020.112205] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 03/03/2020] [Accepted: 03/03/2020] [Indexed: 01/13/2023]
Abstract
FMS-like receptor tyrosine kinase-3 (FLT3) is expressed on acute leukemia cells and is implicated in the survival, proliferation and differentiation of hematopoietic cells in most acute myeloid leukemia (AML) patients. Despite recent achievements in the development of FLT3-targeted small-molecule drugs, there are still unmet medical needs related to kinase selectivity and the progression of some mutant forms of FLT3. Herein, we describe the discovery of novel orally available type 1 FLT3 inhibitors from structure-activity relationship (SAR) studies for the optimization of indirubin derivatives with biological and pharmacokinetic profiles as potential therapeutic agents for AML. The SAR exploration provided important structural insights into the key substituents for potent inhibitory activities of FLT3 and in MV4-11 cells. The profile of the most optimized inhibitor (36) showed IC50 values of 0.87 and 0.32 nM against FLT3 and FLT3/D835Y, respectively, along with potent inhibition against MV4-11 and FLT3/D835Y expressed MOLM14 cells with a GI50 value of 1.0 and 1.87 nM, respectively. With the high oral bioavailability of 42.6%, compound 36 displayed significant in vivo antitumor activity by oral administration of 20 mg/kg once daily dosing schedule for 21 days in a mouse xenograft model. The molecular docking study of 36 in the homology model of the DFG-in conformation of FLT3 resulted in a reasonable binding mode in type 1 kinases similar to the reported type 1 FLT3 inhibitors Crenolanib and Gilteritinib.
Collapse
Affiliation(s)
- Pyeonghwa Jeong
- Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Yeongyu Moon
- Bioenvironmental Science & Toxicology Division, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, Gyeongsangnam-do, 52834, Republic of Korea
| | - Je-Heon Lee
- School of Life Sciences and Center for AI-applied High Efficiency Drug Discovery, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - So-Deok Lee
- School of Life Sciences and Center for AI-applied High Efficiency Drug Discovery, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jiyeon Park
- School of Life Sciences and Center for AI-applied High Efficiency Drug Discovery, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jungeun Lee
- School of Life Sciences and Center for AI-applied High Efficiency Drug Discovery, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jiheon Kim
- School of Life Sciences and Center for AI-applied High Efficiency Drug Discovery, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Hyo Jeong Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do, 52828, Republic of Korea
| | - Na Yoon Kim
- College of Pharmacy, Dankook University, Cheonan, 330-714, Republic of Korea
| | - Jungil Choi
- Bioenvironmental Science & Toxicology Division, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, Gyeongsangnam-do, 52834, Republic of Korea
| | - Jeong Doo Heo
- Bioenvironmental Science & Toxicology Division, Gyeongnam Branch Institute, Korea Institute of Toxicology, Jinju, Gyeongsangnam-do, 52834, Republic of Korea
| | - Ji Eun Shin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yoon-Gyoon Kim
- College of Pharmacy, Dankook University, Cheonan, 330-714, Republic of Korea
| | - Sun-Young Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do, 52828, Republic of Korea.
| | - Yong-Chul Kim
- Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea; School of Life Sciences and Center for AI-applied High Efficiency Drug Discovery, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea.
| |
Collapse
|
27
|
Bergqvist C, Servy A, Valeyrie-Allanore L, Ferkal S, Combemale P, Wolkenstein P. Neurofibromatosis 1 French national guidelines based on an extensive literature review since 1966. Orphanet J Rare Dis 2020; 15:37. [PMID: 32014052 PMCID: PMC6998847 DOI: 10.1186/s13023-020-1310-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/17/2020] [Indexed: 12/13/2022] Open
Abstract
Neurofibromatosis type 1 is a relatively common genetic disease, with a prevalence ranging between 1/3000 and 1/6000 people worldwide. The disease affects multiple systems with cutaneous, neurologic, and orthopedic as major manifestations which lead to significant morbidity or mortality. Indeed, NF1 patients are at an increased risk of malignancy and have a life expectancy about 10-15 years shorter than the general population. The mainstay of management of NF1 is a patient-centered longitudinal care with age-specific monitoring of clinical manifestations, aiming at the early recognition and symptomatic treatment of complications as they occur. Protocole national de diagnostic et de soins (PNDS) are mandatory French clinical practice guidelines for rare diseases required by the French national plan for rare diseases. Their purpose is to provide health care professionals with guidance regarding the optimal diagnostic and therapeutic management of patients affected with a rare disease; and thus, harmonizing their management nationwide. PNDS are usually developed through a critical literature review and a multidisciplinary expert consensus. The purpose of this article is to present the French guidelines on NF1, making them even more available to the international medical community. We further dwelled on the emerging new evidence that might have therapeutic potential or a strong impact on NF1 management in the coming feature. Given the complexity of the disease, the management of children and adults with NF1 entails the full complement healthcare providers and communication among the various specialties.
Collapse
Affiliation(s)
- Christina Bergqvist
- Faculty of medicine, Université Paris-Est Creteil (UPEC), F-94010 Créteil Cedex, France
- Assistance Publique-Hôpital Paris (AP-HP), Hôpital Henri-Mondor, Service de Dermatologie, F-94010 Créteil, France
| | - Amandine Servy
- Assistance Publique-Hôpital Paris (AP-HP), Hôpital Henri-Mondor, Service de Dermatologie, F-94010 Créteil, France
| | - Laurence Valeyrie-Allanore
- INSERM, Centre d’Investigation Clinique 006, Referral Center of Neurofibromatosis, Assistance Publique-Hôpital Paris (AP-HP), Hôpital Henri-Mondor, F-94010 Créteil, France
| | - Salah Ferkal
- INSERM, Centre d’Investigation Clinique 006, Referral Center of Neurofibromatosis, Assistance Publique-Hôpital Paris (AP-HP), Hôpital Henri-Mondor, F-94010 Créteil, France
| | - Patrick Combemale
- Rhône-Alpes Auvergne Competence Center for the treatment of Neurofibromatosis type 1, Léon Bérard Comprehensive Cancer Center, Hôpitaux Universitaires de Lyon, Université de Lyon, F-69008 Lyon, France
| | - Pierre Wolkenstein
- Faculty of medicine, Université Paris-Est Creteil (UPEC), F-94010 Créteil Cedex, France
- Assistance Publique-Hôpital Paris (AP-HP), Hôpital Henri-Mondor, Service de Dermatologie, F-94010 Créteil, France
- INSERM, Centre d’Investigation Clinique 006, Referral Center of Neurofibromatosis, Assistance Publique-Hôpital Paris (AP-HP), Hôpital Henri-Mondor, F-94010 Créteil, France
| |
Collapse
|
28
|
Alessandrino F, Smith DA, Tirumani SH, Ramaiya NH. Cancer genome landscape: a radiologist's guide to cancer genome medicine with imaging correlates. Insights Imaging 2019; 10:111. [PMID: 31781977 PMCID: PMC6883020 DOI: 10.1186/s13244-019-0800-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022] Open
Abstract
The introduction of high throughput sequence analysis in the past decade and the decrease in sequencing costs has made available an enormous amount of genomic data. These data have shaped the landscape of cancer genome, which encompasses mutations determining tumorigenesis, the signaling pathways involved in cancer growth, the tumor heterogeneity, and its role in development of metastases. Tumors develop acquiring a series of driver mutations over time. Of the many mutated genes present in cancer, only few specific mutations are responsible for invasiveness and metastatic potential, which, in many cases, have characteristic imaging appearance. Ten signaling pathways, each with targetable components, have been identified as responsible for cancer growth. Blockage of any of these pathways form the basis for molecular targeted therapies, which are associated with specific pattern of response and toxicities. Tumor heterogeneity, responsible for the different mutation pattern of metastases and primary tumor, has been classified in intratumoral, intermetastatic, intrametastatic, and interpatient heterogeneity, each with specific imaging correlates. The purpose of this article is to introduce the key components of the landscapes of cancer genome and their imaging counterparts, describing the types of mutations associated with tumorigenesis, the pathways of cancer growth, the genetic heterogeneity involved in metastatic disease, as well as the current challenges and opportunities for cancer genomics research.
Collapse
Affiliation(s)
- Francesco Alessandrino
- Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA. .,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| | - Daniel A Smith
- Department of Radiology, UH Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Sree Harsha Tirumani
- Department of Radiology, UH Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Nikhil H Ramaiya
- Department of Radiology, UH Cleveland Medical Center, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| |
Collapse
|
29
|
Turnquist C, Watson RA, Protheroe A, Verrill C, Sivakumar S. Tumor heterogeneity: does it matter? Expert Rev Anticancer Ther 2019; 19:857-867. [PMID: 31510810 DOI: 10.1080/14737140.2019.1667236] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: It has long been recognized that tumors are composed of a mosaic of cells and numerous methods have been developed to detect tumor heterogeneity, including in situ hybridization, multi-regional sampling, cytological assays, and whole genome and single cell sequencing. Using these methods, heterogeneity has been observed at the genetic, epigenetic, and phenotypic level in numerous cancers. With the advent of deep sequencing technology, we now appreciate a greater complexity of distinct genotypes and phenotypes that drive the biological behavior of cancer. Despite decades of progress in detecting tumor heterogeneity, the question remains: to what extent does it matter? Areas covered: This review explores the evidence for and against the importance of tumor heterogeneity in three main areas: prognostication, development of targeted therapeutics and tumor resistance; summarizing current understanding before evaluating ongoing experimental and clinical developments. Expert opinion: Theoretical understanding and in vitro detection of intratumour heterogeneity promises much but is yet to translate into meaningful clinical benefit. However, the recent emergence of a host of technological innovations and upcoming clinical trials may soon change the landscape of this field.
Collapse
Affiliation(s)
- Casmir Turnquist
- University of Oxford Medical School, John Radcliffe Hospital , Oxford , UK
| | | | | | - Clare Verrill
- Nuffield Department of Surgical Sciences, Oxford NIHR Biomedical Research Centre , Oxford , UK
| | - Shivan Sivakumar
- Department of Oncology, University of Oxford , Oxford , UK.,Kennedy Institute of Rheumatology, University of Oxford , Oxford , UK
| |
Collapse
|
30
|
Wang X, Wu K, Xiao LK, Wu XY. ShRNA-mediated BMI-1 gene silencing inhibits gastrointestinal stromal tumor cell telomerase activity and enhances apoptosis. Kaohsiung J Med Sci 2018; 34:606-615. [PMID: 30392567 DOI: 10.1016/j.kjms.2018.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/30/2018] [Accepted: 06/05/2018] [Indexed: 11/15/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most frequently occurring mesenchymal tumors of the gastrointestinal tract. Telomerase activity is well acknowledged as a critical factor in oncogenesis. The objective of the present study is to evaluate the effect of BMI gene silencing on proliferation, apoptosis and telomerase activity in human GIST882 cells. GIST882 cells were transfected with a eukaryotic expression vector of an shRNA fragment. The silencing efficiency in the GIST882 cells was determined by RT-qPCR and a western blot analysis. After the shRNA-BMI-1 plasmid was transfected into the GIST882 cells and nude mice, a cell counting kit-8 (CCK-8) assay and flow cytometry were utilized to detect the GIST882 cell proliferation, the apoptosis rate and the cell cycle. Tumor growth was observed by tumor xenograft in nude mice. Telomerase activity and telomere length were detected by a Southern blot and a target region amplified polymorphism. The shRNA-BMI-1 recombinant plasmid was successfully constructed. The mRNA and protein expression of the BMI-1 gene in GIST882 cells was suppressed by the shRNA-BMI-1 recombinant plasmid. Meanwhile, BMI-1 gene silencing inhibited the cell proliferation, tumor growth, and cell cycle in the GIST882 cells. However, cell apoptosis was increased and telomerase activity was decreased with the silencing of the BMI-1 gene. Collectively, the results of this study suggest that silencing the BMI-1 gene may provide a new target for the treatment of GISTs.
Collapse
Affiliation(s)
- Xin Wang
- Department of General Surgery, Chongqing General Hospital (Zhongshan Branch), Chongqing, PR China.
| | - Kun Wu
- Department of General Surgery, Chongqing General Hospital (Zhongshan Branch), Chongqing, PR China
| | - Lin-Kang Xiao
- Department of General Surgery, Chongqing General Hospital (Zhongshan Branch), Chongqing, PR China
| | - Xing-Ye Wu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| |
Collapse
|
31
|
Kettle JG, Anjum R, Barry E, Bhavsar D, Brown C, Boyd S, Campbell A, Goldberg K, Grondine M, Guichard S, Hardy CJ, Hunt T, Jones RDO, Li X, Moleva O, Ogg D, Overman RC, Packer MJ, Pearson S, Schimpl M, Shao W, Smith A, Smith JM, Stead D, Stokes S, Tucker M, Ye Y. Discovery of N-(4-{[5-Fluoro-7-(2-methoxyethoxy)quinazolin-4-yl]amino}phenyl)-2-[4-(propan-2-yl)-1 H-1,2,3-triazol-1-yl]acetamide (AZD3229), a Potent Pan-KIT Mutant Inhibitor for the Treatment of Gastrointestinal Stromal Tumors. J Med Chem 2018; 61:8797-8810. [PMID: 30204441 DOI: 10.1021/acs.jmedchem.8b00938] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
While the treatment of gastrointestinal stromal tumors (GISTs) has been revolutionized by the application of targeted tyrosine kinase inhibitors capable of inhibiting KIT-driven proliferation, diverse mutations to this kinase drive resistance to established therapies. Here we describe the identification of potent pan-KIT mutant kinase inhibitors that can be dosed without being limited by the tolerability issues seen with multitargeted agents. This effort focused on identification and optimization of an existing kinase scaffold through the use of structure-based design. Starting from a series of previously reported phenoxyquinazoline and quinoline based inhibitors of the tyrosine kinase PDGFRα, potency against a diverse panel of mutant KIT driven Ba/F3 cell lines was optimized, with a particular focus on reducing activity against a KDR driven cell model in order to limit the potential for hypertension commonly seen in second and third line GIST therapies. AZD3229 demonstrates potent single digit nM growth inhibition across a broad cell panel, with good margin to KDR-driven effects. Selectivity over KDR can be rationalized predominantly by the interaction of water molecules with the protein and ligand in the active site, and its kinome selectivity is similar to the best of the approved GIST agents. This compound demonstrates excellent cross-species pharmacokinetics, shows strong pharmacodynamic inhibition of target, and is active in several in vivo models of GIST.
Collapse
Affiliation(s)
- Jason G Kettle
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Rana Anjum
- Oncology, IMED Biotech Unit , AstraZeneca , 35 Gatehouse Park , Waltham , Massachusetts 02451 , United States
| | - Evan Barry
- Oncology, IMED Biotech Unit , AstraZeneca , 35 Gatehouse Park , Waltham , Massachusetts 02451 , United States
| | - Deepa Bhavsar
- Oncology, IMED Biotech Unit , AstraZeneca , 35 Gatehouse Park , Waltham , Massachusetts 02451 , United States
| | - Crystal Brown
- Oncology, IMED Biotech Unit , AstraZeneca , 35 Gatehouse Park , Waltham , Massachusetts 02451 , United States
| | - Scott Boyd
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Andrew Campbell
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Kristin Goldberg
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Michael Grondine
- Oncology, IMED Biotech Unit , AstraZeneca , 35 Gatehouse Park , Waltham , Massachusetts 02451 , United States
| | - Sylvie Guichard
- Oncology, IMED Biotech Unit , AstraZeneca , 35 Gatehouse Park , Waltham , Massachusetts 02451 , United States
| | - Christopher J Hardy
- Discovery Sciences, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Tom Hunt
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Rhys D O Jones
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Xiuwei Li
- Pharmaron Beijing Co., Ltd. , 6 Taihe Road BDA , Beijing 100176 , P. R. China
| | - Olga Moleva
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Derek Ogg
- Discovery Sciences, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Ross C Overman
- Discovery Sciences, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Martin J Packer
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Stuart Pearson
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Marianne Schimpl
- Discovery Sciences, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Wenlin Shao
- Oncology, IMED Biotech Unit , AstraZeneca , 35 Gatehouse Park , Waltham , Massachusetts 02451 , United States
| | - Aaron Smith
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - James M Smith
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Darren Stead
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Steve Stokes
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Michael Tucker
- Oncology, IMED Biotech Unit , AstraZeneca , Unit 310, Darwin Building, Cambridge Science Park, Milton Road , Cambridge CB4 0WG , United Kingdom
| | - Yang Ye
- Pharmaron Beijing Co., Ltd. , 6 Taihe Road BDA , Beijing 100176 , P. R. China
| |
Collapse
|
32
|
Gunaratnam M, Collie GW, Reszka AP, Todd AK, Parkinson GN, Neidle S. A naphthalene diimide G-quadruplex ligand inhibits cell growth and down-regulates BCL-2 expression in an imatinib-resistant gastrointestinal cancer cell line. Bioorg Med Chem 2018; 26:2958-2964. [DOI: 10.1016/j.bmc.2018.04.050] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/23/2018] [Accepted: 04/25/2018] [Indexed: 12/24/2022]
|
33
|
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: 142] [Impact Index Per Article: 23.7] [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
|