101
|
Blay JY, Hindi N, Bollard J, Aguiar S, Angel M, Araya B, Badilla R, Bernabeu D, Campos F, Caro-Sánchez CHS, Carvajal B, Carvajal Montoya A, Casavilca-Zambrano S, Castro-Oliden V, Chacón M, Clara M, Collini P, Correa Genoroso R, Costa FD, Cuellar M, Dei Tos AP, Dominguez Malagon HR, Donati D, Dufresne A, Eriksson M, Farias-Loza M, Fernandez P, Frezza AM, Frisoni T, Garcia-Ortega DY, Gelderblom H, Gouin F, Gómez-Mateo MC, Gronchi A, Haro J, Huanca L, Jimenez N, Karanian M, Kasper B, Lopes David BB, Lopez-Pousa A, Lutter G, Martinez-Said H, Martinez-Tlahuel J, Mello CA, Morales Pérez JM, Moura David S, Nascimento AG, Ortiz-Cruz EJ, Palmerini E, Patel S, Pfluger Y, Provenzano S, Righi A, Rodriguez A, Salas R, Santos TTG, Scotlandi K, Soule T, Stacchiotti S, Valverde C, Waisberg F, Zamora Estrada E, Martin-Broto J. SELNET clinical practice guidelines for soft tissue sarcoma and GIST. Cancer Treat Rev 2022; 102:102312. [PMID: 34798363 DOI: 10.1016/j.ctrv.2021.102312] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 10/30/2021] [Indexed: 12/12/2022]
Affiliation(s)
- J Y Blay
- Léon Bérard Center, 28 rue Laennec 69373 Lyon Cedex 08, France.
| | - N Hindi
- Research Health Institute Fundacion Jimenez Diaz (IIS/FJD), 28015 Madrid, Spain; Hospital Fundación Jimenez Diaz University Hospital, 28040 Madrid, Spain; General de Villalba University Hospital, 28400 Madrid, Spain
| | - J Bollard
- Léon Bérard Center, 28 rue Laennec 69373 Lyon Cedex 08, France
| | - S Aguiar
- A.C.Camargo Cancer Center, Rua prof Antonio Prudente, 211 - Liberdade, São Paulo - SP 01509-010, Brazil
| | - M Angel
- Instituto Alexander Fleming. Av. Cramer 1180. CP C1426ANZ, Buenos Aires, Argentina
| | - B Araya
- Hospital Dr. R. A. Calderón Guardia, 7-9 Av, 15-17 St, Aranjuez, San José, Costa Rica
| | - R Badilla
- Hospital Dr. R. A. Calderón Guardia, 7-9 Av, 15-17 St, Aranjuez, San José, Costa Rica
| | - D Bernabeu
- Hospital Universitario La Paz, Paseo de la Castellana, 261, 28046 Madrid, Spain
| | - F Campos
- A.C.Camargo Cancer Center, Rua prof Antonio Prudente, 211 - Liberdade, São Paulo - SP 01509-010, Brazil
| | - C H S Caro-Sánchez
- Instituto Nacional de Cancerologia. Torre Nueva de Hospitalización, primer piso. Av. San Fernando 86, Colonia Niño Jesus. CP 14080, Tlalpan Mexico
| | - B Carvajal
- Fundación GIST México, Altadena 59, Nápoles, Benito Juárez, 03810 Ciudad de Mexico, CDMX, Mexico
| | - A Carvajal Montoya
- Hospital Dr. R. A. Calderón Guardia, 7-9 Av, 15-17 St, Aranjuez, San José, Costa Rica
| | - S Casavilca-Zambrano
- Instituto Nacional de Enfermedades Neoplásicas, Av. Angamos Este 2520, Lima 34, Peru
| | - V Castro-Oliden
- Instituto Nacional de Enfermedades Neoplásicas, Av. Angamos Este 2520, Lima 34, Peru
| | - M Chacón
- Instituto Alexander Fleming. Av. Cramer 1180. CP C1426ANZ, Buenos Aires, Argentina
| | - M Clara
- Instituto Nacional de Cancerologia. Torre Nueva de Hospitalización, primer piso. Av. San Fernando 86, Colonia Niño Jesus. CP 14080, Tlalpan Mexico
| | - P Collini
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milano, Italy
| | - R Correa Genoroso
- Hospital Clínico Universitario Virgen de la Victoria, Campus Universitario de Teatinos s/n, 29010 Malaga, Spain
| | - F D Costa
- A.C.Camargo Cancer Center, Rua prof Antonio Prudente, 211 - Liberdade, São Paulo - SP 01509-010, Brazil
| | - M Cuellar
- Fundación GIST México, Altadena 59, Nápoles, Benito Juárez, 03810 Ciudad de Mexico, CDMX, Mexico
| | - A P Dei Tos
- Treviso General Hospital Treviso, University of Padua, Padova, Italy
| | - H R Dominguez Malagon
- Instituto Nacional de Cancerologia. Torre Nueva de Hospitalización, primer piso. Av. San Fernando 86, Colonia Niño Jesus. CP 14080, Tlalpan Mexico
| | - D Donati
- IRCCS Istituto Ortopedico Rizzoli, University of Bologna, Via Pupilli, 1, 40136 Bologna, Italy
| | - A Dufresne
- Léon Bérard Center, 28 rue Laennec 69373 Lyon Cedex 08, France
| | - M Eriksson
- Skane University Hospital and Lund University, Lund, Sweden
| | - M Farias-Loza
- Instituto Nacional de Enfermedades Neoplásicas, Av. Angamos Este 2520, Lima 34, Peru
| | | | - A M Frezza
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milano, Italy
| | - T Frisoni
- IRCCS Istituto Ortopedico Rizzoli, University of Bologna, Via Pupilli, 1, 40136 Bologna, Italy
| | - D Y Garcia-Ortega
- Instituto Nacional de Cancerologia. Torre Nueva de Hospitalización, primer piso. Av. San Fernando 86, Colonia Niño Jesus. CP 14080, Tlalpan Mexico
| | - H Gelderblom
- Leiden University Medical Center, Leiden, the Netherlands
| | - F Gouin
- Léon Bérard Center, 28 rue Laennec 69373 Lyon Cedex 08, France
| | - M C Gómez-Mateo
- Hospital Universitario Miguel Servet, Paseo Isabel la Católica, 1-3, 50009 Zaragoza, Spain
| | - A Gronchi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milano, Italy
| | - J Haro
- Instituto Nacional de Enfermedades Neoplásicas, Av. Angamos Este 2520, Lima 34, Peru
| | - L Huanca
- Instituto Nacional de Enfermedades Neoplásicas, Av. Angamos Este 2520, Lima 34, Peru
| | - N Jimenez
- Hospital Dr. R. A. Calderón Guardia, 7-9 Av, 15-17 St, Aranjuez, San José, Costa Rica
| | - M Karanian
- Léon Bérard Center, 28 rue Laennec 69373 Lyon Cedex 08, France
| | - B Kasper
- University of Heidelberg, Mannheim Cancer Center, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - B B Lopes David
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milano, Italy
| | - A Lopez-Pousa
- Hospital de la Santa Creu i Sant Pau, Carrer de Sant Quintí, 89, 08041 Barcelona, Espagne
| | - G Lutter
- Instituto Alexander Fleming. Av. Cramer 1180. CP C1426ANZ, Buenos Aires, Argentina
| | - H Martinez-Said
- Centro Oncologico Integral, Hospital Medica Sur, Planta Baja Torre III - Cons. 305, Col. Toriello Guerra, Deleg. Tlalpan. C.P. 14050, Mexico, D.F
| | - J Martinez-Tlahuel
- Instituto Nacional de Cancerologia. Torre Nueva de Hospitalización, primer piso. Av. San Fernando 86, Colonia Niño Jesus. CP 14080, Tlalpan Mexico
| | - C A Mello
- A.C.Camargo Cancer Center, Rua prof Antonio Prudente, 211 - Liberdade, São Paulo - SP 01509-010, Brazil
| | - J M Morales Pérez
- Hospital Universitario Virgen del Rocio, Av Manuel Siurot s/n, 41013 Sevilla, Spain
| | - S Moura David
- Hospital Universitario Virgen del Rocio, Av Manuel Siurot s/n, 41013 Sevilla, Spain
| | - A G Nascimento
- A.C.Camargo Cancer Center, Rua prof Antonio Prudente, 211 - Liberdade, São Paulo - SP 01509-010, Brazil
| | - E J Ortiz-Cruz
- Hospital Universitario La Paz, MD Anderson Cancer Center, Calle de Arturo Soria, 270 28033 Madrid, Spain
| | - E Palmerini
- IRCCS Istituto Ortopedico Rizzoli, University of Bologna, Via Pupilli, 1, 40136 Bologna, Italy
| | - S Patel
- UT MD Anderson Cancer Center, Houston, TX, USA
| | - Y Pfluger
- Instituto Alexander Fleming. Av. Cramer 1180. CP C1426ANZ, Buenos Aires, Argentina
| | - S Provenzano
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milano, Italy
| | - A Righi
- IRCCS Istituto Ortopedico Rizzoli, University of Bologna, Via Pupilli, 1, 40136 Bologna, Italy
| | - A Rodriguez
- Instituto Alexander Fleming. Av. Cramer 1180. CP C1426ANZ, Buenos Aires, Argentina
| | - R Salas
- Fundación GIST México, Altadena 59, Nápoles, Benito Juárez, 03810 Ciudad de Mexico, CDMX, Mexico
| | - T T G Santos
- A.C.Camargo Cancer Center, Rua prof Antonio Prudente, 211 - Liberdade, São Paulo - SP 01509-010, Brazil
| | - K Scotlandi
- IRCCS Istituto Ortopedico Rizzoli, University of Bologna, Via Pupilli, 1, 40136 Bologna, Italy
| | - T Soule
- Instituto Alexander Fleming. Av. Cramer 1180. CP C1426ANZ, Buenos Aires, Argentina
| | - S Stacchiotti
- Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian, 1, 20133 Milano, Italy
| | - C Valverde
- Vall d́Hebrón University Hospital, Passeig de la Vall d'Hebron, 119, 08035 Barcelona, Spain
| | - F Waisberg
- Instituto Alexander Fleming. Av. Cramer 1180. CP C1426ANZ, Buenos Aires, Argentina
| | - E Zamora Estrada
- Hospital Dr. R. A. Calderón Guardia, 7-9 Av, 15-17 St, Aranjuez, San José, Costa Rica
| | - J Martin-Broto
- Research Health Institute Fundacion Jimenez Diaz (IIS/FJD), 28015 Madrid, Spain; Hospital Fundación Jimenez Diaz University Hospital, 28040 Madrid, Spain; General de Villalba University Hospital, 28400 Madrid, Spain
| |
Collapse
|
102
|
Van den Abbeele AD, Sakellis CG, George S. PET imaging of Gastrointestinal Stromal Tumors (GIST). Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00110-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
103
|
Shima T, Taniguchi K, Tokumaru Y, Inomata Y, Arima J, Lee SW, Takabe K, Yoshida K, Uchiyama K. Glucose transporter‑1 inhibition overcomes imatinib resistance in gastrointestinal stromal tumor cells. Oncol Rep 2021; 47:7. [PMID: 34738628 PMCID: PMC8600406 DOI: 10.3892/or.2021.8218] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/01/2021] [Indexed: 12/23/2022] Open
Abstract
Imatinib mesylate (imatinib) is the primary agent of choice used to treat gastrointestinal stromal tumors (GIST). However, drug resistance to imatinib poses a major obstacle to treatment efficacy. In addition, the relationship between imatinib resistance and glycolysis is poorly understood. Glucose transporter (GLUT)-1 is a key component of glycolysis. The present study aimed to assess the potential relationship between components in the glycolytic pathway and the acquisition of imatinib resistance by GIST cells, with particular focus on GLUT-1. An imatinib-resistant GIST cell line was established through the gradual and continuous imatinib treatment of the parental human GIST cell line GIST-T1. The expression of glycolysis-related molecules (GLUT-1, hexokinase 2, pyruvate kinase M2 and lactate dehydrogenase) was assessed in parental and imatinib-resistant cells by western blotting, reverse transcription-quantitative PCR and glucose and lactate measurement kits. In addition, clinical information and transcriptomic data obtained from the gene expression omnibus database (GSE15966) were used to confirm the in vitro results. The potential effects of GLUT-1 inhibition on the expression of proteins in the glycolysis (GLUT-1, hexokinase 2, pyruvate kinase M2 and lactate dehydrogenase) and apoptosis pathways (Bcl-2, cleaved PARP, caspase-3 and caspase-9) in imatinib-resistant cells were then investigated following gene silencing and treatment using the GLUT-1 inhibitor WZB117 by western blotting. For gene silencing, the mature siRNAs for SLC2A1 were used for cell transfection. Annexin V-FITC/PI double-staining followed by flow cytometry was used to measure apoptosis whereas three-dimensional culture experiments were used to create three-dimensional spheroid cells where cell viability and spheroid diameter were measured. Although imatinib treatment downregulated GLUT-1 expression and other glycolysis pathway components hexokinase 2, pyruvate kinase M2, and lactate dehydrogenase in parental GIST-T1 cells even at low concentrations. By contrast, expression of these glycolysis pathway components in imatinib-resistant cells were increased by imatinib treatment. WZB117 administration significantly downregulated AKT phosphorylation and Bcl-2 expression in imatinib-resistant cells, whereas the combined administration of imatinib and WZB117 conferred synergistic growth inhibition effects in apoptosis assay. WZB117 was found to exert additional inhibitory effects by inducing apoptosis in imatinib-resistant cells. Therefore, the present study suggests that GLUT-1 is involved in the acquisition of imatinib resistance by GIST cells, which can be overcome by combined treatment with WZB117 and imatinib.
Collapse
Affiliation(s)
- Takafumi Shima
- Department of General and Gastroenterological Surgery, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka 569‑8686, Japan
| | - Kohei Taniguchi
- Department of General and Gastroenterological Surgery, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka 569‑8686, Japan
| | - Yoshihisa Tokumaru
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Yosuke Inomata
- Department of General and Gastroenterological Surgery, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka 569‑8686, Japan
| | - Jun Arima
- Department of General and Gastroenterological Surgery, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka 569‑8686, Japan
| | - Sang-Woong Lee
- Department of General and Gastroenterological Surgery, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka 569‑8686, Japan
| | - Kazuaki Takabe
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Kazuhiro Yoshida
- Department of Surgical Oncology, Graduate School of Medicine, Gifu University, Gifu, Gifu 501‑1194, Japan
| | - Kazuhisa Uchiyama
- Department of General and Gastroenterological Surgery, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka 569‑8686, Japan
| |
Collapse
|
104
|
Tan BKJ, Teo CB, Tadeo X, Peng S, Soh HPL, Du SDX, Luo VWY, Bandla A, Sundar R, Ho D, Kee TW, Blasiak A. Personalised, Rational, Efficacy-Driven Cancer Drug Dosing via an Artificial Intelligence SystEm (PRECISE): A Protocol for the PRECISE CURATE.AI Pilot Clinical Trial. Front Digit Health 2021; 3:635524. [PMID: 34713106 PMCID: PMC8521832 DOI: 10.3389/fdgth.2021.635524] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/04/2021] [Indexed: 01/02/2023] Open
Abstract
Introduction: Oncologists have traditionally administered the maximum tolerated doses of drugs in chemotherapy. However, these toxicity-guided doses may lead to suboptimal efficacy. CURATE.AI is an indication-agnostic, mechanism-independent and efficacy-driven personalised dosing platform that may offer a more optimal solution. While CURATE.AI has already been applied in a variety of clinical settings, there are no prior randomised controlled trials (RCTs) on CURATE.AI-guided chemotherapy dosing for solid tumours. Therefore, we aim to assess the technical and logistical feasibility of a future RCT for CURATE.AI-guided solid tumour chemotherapy dosing. We will also collect exploratory data on efficacy and toxicity, which will inform RCT power calculations. Methods and analysis: This is an open-label, single-arm, two-centre, prospective pilot clinical trial, recruiting adults with metastatic solid tumours and raised baseline tumour marker levels who are planned for palliative-intent, capecitabine-based chemotherapy. As CURATE.AI is a small data platform, it will guide drug dosing for each participant based only on their own tumour marker levels and drug doses as input data. The primary outcome is the proportion of participants in whom CURATE.AI is successfully applied to provide efficacy-driven personalised dosing, as judged based on predefined considerations. Secondary outcomes include the timeliness of dose recommendations, participant and physician adherence to CURATE.AI-recommended doses, and the proportion of clinically significant dose changes. We aim to initially enrol 10 participants from two hospitals in Singapore, perform an interim analysis, and consider either cohort expansion or an RCT. Recruitment began in August 2020. This pilot clinical trial will provide key data for a future RCT of CURATE.AI-guided personalised dosing for precision oncology. Ethics and dissemination: The National Healthcare Group (NHG) Domain Specific Review Board has granted ethical approval for this study (DSRB 2020/00334). We will distribute our findings at scientific conferences and publish them in peer-reviewed journals. Trial registration number: NCT04522284
Collapse
Affiliation(s)
- Benjamin Kye Jyn Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chong Boon Teo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xavier Tadeo
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore
| | - Siyu Peng
- Department of Medicine, National University Health System, Singapore, Singapore
| | - Hazel Pei Lin Soh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sherry De Xuan Du
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vilianty Wen Ya Luo
- Haematology-Oncology Research Group, National University Cancer Institute, Singapore (NCIS), Singapore, Singapore
| | - Aishwarya Bandla
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
| | - Raghav Sundar
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Haematology-Oncology Research Group, National University Cancer Institute, Singapore (NCIS), Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore, Singapore
| | - Dean Ho
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Smart Systems Institute, National University of Singapore, Singapore, Singapore
| | - Theodore Wonpeum Kee
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore
| | - Agata Blasiak
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
105
|
Schmidt T, Ghadimi M, Fuchs HF, Bruns CJ. [Surgical and interdisciplinary treatment of gastrointestinal stromal tumors]. Chirurg 2021; 93:27-33. [PMID: 34709443 DOI: 10.1007/s00104-021-01527-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2021] [Indexed: 11/30/2022]
Abstract
Gastrointestinal stromal tumors (GISTs) are the most frequent potentially malignant mesenchymal tumors of the gastrointestinal tract. The treatment of GISTs has been revolutionized since imatinib and other tyrosine kinase inhibitors were introduced for the treatment of GISTs, which inhibit the tyrosine kinases c‑KIT and platelet-derived growth factor receptor (PDGFR) alpha. Even after the introduction of this targeted treatment GISTs can only be cured by surgical resection. With interdisciplinary multimodal treatment the prognosis of metastasized GIST can now be further improved by surgical resection of the primary tumor and the metastases, potentially leading to a cure. Neoadjuvant therapy can reduce the extent of surgical resection and hereby enable organ preservation and reduce surgical morbidity. To evaluate molecular and clinical predictors and to offer an optimal therapeutic plan, patients with GISTs and certainly patients with advanced GISTs should be evaluated by interdisciplinary sarcoma boards.
Collapse
Affiliation(s)
- Thomas Schmidt
- Klinik für Allgemein‑, Viszeral‑, Tumor und Transplantationschirurgie, Universitätsklinikum Köln, Kerpener Str. 62, 50937, Köln, Deutschland.
| | - Markus Ghadimi
- Klinik für Allgemein‑, Viszeral‑, Tumor und Transplantationschirurgie, Universitätsklinikum Köln, Kerpener Str. 62, 50937, Köln, Deutschland
| | - Hans F Fuchs
- Klinik für Allgemein‑, Viszeral‑, Tumor und Transplantationschirurgie, Universitätsklinikum Köln, Kerpener Str. 62, 50937, Köln, Deutschland
| | - Christiane J Bruns
- Klinik für Allgemein‑, Viszeral‑, Tumor und Transplantationschirurgie, Universitätsklinikum Köln, Kerpener Str. 62, 50937, Köln, Deutschland
| |
Collapse
|
106
|
Pestana RC, Roszik J, Groisberg R, Sen S, Van Tine BA, Conley AP, Subbiah V. Discovery of targeted expression data for novel antibody-based and chimeric antigen receptor-based therapeutics in soft tissue sarcomas using RNA-sequencing: clinical implications. Curr Probl Cancer 2021; 45:100794. [PMID: 34656365 DOI: 10.1016/j.currproblcancer.2021.100794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 08/11/2021] [Accepted: 09/02/2021] [Indexed: 11/18/2022]
Abstract
Recent failure of phase 3 trials and paucity of druggable oncogenic drivers hamper developmental therapeutics in sarcomas. Antibody-based therapeutics, like antibody-drug conjugates (ADCs) and chimeric antigen receptor (CAR)-based therapeutics, have emerged as promising strategies for anticancer drug delivery. The efficacy of these novel therapies is highly dependent on expression of the antibody target. We used RNA sequencing data from Cancer Genome Atlas (TCGA) to analyze expression of target antigens in sarcoma subtypes including dedifferentiated liposarcoma (DDLPS; n = 50), uterine leiomyosarcoma (ULMS; n = 27), leiomyosarcoma (STLMS; n = 53), undifferentiated pleomorphic sarcoma (UPS; n = 44), myxofibrosarcoma (MFS; n = 17), synovial sarcoma (SS; n = 10), and malignant peripheral nerve sheath tumor (MPNST; n = 5). We searched published literature and clinicaltrial.gov for ADC targets, bispecific antibodies, immunotoxins, radioimmunoconjugates, SPEAR T-cells, and CAR's that are in clinical trials. CD70 expression was significantly higher in DDLPS, UPS, and MFS than SS and STLMS. CDH3 expression was greater in LMS and ULMS than UPS (P < 0.001), MFS (P < 0.001), and DDLPS (P < 0.001). ERBB2 expression was low; however, it was overexpressed in MPNST when compared with UPS (P < 0.001), and MFS (P < 0.01). GPNMB was highly expressed in most sarcomas, with the exception of SS. LRRC15 also appeared to be a relevant target, especially in UPS. MSLN expression was relatively low except in SS and MPNST. PDGFRA was also highly expressed in most sarcomas with the exception of ULMS and STLMS. TNFRSF8 seems to be most appropriate in DDLPS, as well as MFS. AXL was expressed especially in MFS and STLMS. Sarcoma subtypes express multiple target genes relevant for ADCs, SPEAR T-cells and CAR's, warranting further clinical validation and evaluation.
Collapse
Affiliation(s)
- Roberto Carmagnani Pestana
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas; Centro de Oncologia e Hematologia Einstein Familia Dayan-Daycoval, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Jason Roszik
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roman Groisberg
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas; Rutgers Cancer Institute of New Jersey, New Jersey
| | - Shiraj Sen
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas; Sarah Cannon Research Institute at HealthONE, Denver, Colorado
| | - Brian A Van Tine
- Division of Medical Oncology, Washington University in St. Louis, St Louis, Missouri; Division of Pediatric Hematology and Oncology, St. Louis Children's Hospital, St Louis, Missouri; Siteman Cancer Center, St Louis, Missouri
| | - Anthony P Conley
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
107
|
Treatment of gastrointestinal stromal tumors: A single-center experience. North Clin Istanb 2021; 8:385-392. [PMID: 34585074 PMCID: PMC8430361 DOI: 10.14744/nci.2020.04468] [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: 10/21/2020] [Accepted: 11/24/2020] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE: Gastrointestinal stromal tumors are the most common mesenchymal tumors of the gastrointestinal tract. We aimed to examine the clinical characteristics and treatment outcomes of patients diagnosed with gastrointestinal stromal tumor (GIST) who were followed up and treated in our center. METHODS: This study retrospectively evaluated the clinical characteristics, disease stages, administered treatments, and treatment responses of 67 patients diagnosed with GIST who presented to our center between 2007 and 2015. RESULTS: Of the 67 patients included in our study, 24 (35.8%) were female and 43 (64.2%) were male. Median age at diagnosis was 54 years (23–86). Primary tumor localization was the stomach in 38.8% (n=26), small intestine in 46.2% (n=31), colorectal in 6% (n=4), and extra-gastrointestinal in 9% (n=6) of the patients. At diagnosis, 19 patients (28.4%) were at a metastatic stage. Fifty-seven patients (85.1%) underwent surgery. Thirty-three patients received one line, 20 patients received two lines, and 12 patients received three lines of treatment. The first-line treatment resulted in complete response in 12 patients (36.4%), partial response in 15 patients (45.5%), stable disease in 5 patients (15.2%), and progression in 1 patient (3%). Progression-free survival (PFS) was 36 months for the first-line treatment. The second-line treatment resulted in partial response in 7 patients (35%), stable disease in 12 patients (60%), and progression in 1 patient (5%). PFS was 12 months for the second-line treatment. The third-line treatment resulted in complete response in 1 patient (8.3%), partial response in 3 patients (25%), stable disease in 5 patients (41.7%), and progression in 3 patients (25%). PFS was 9 months for the third-line treatment. The fourth-line treatment resulted in stable disease in 4 patients (80%) and progression in 1 patient (20%). PFS was 4 months for the fourth-line treatment. Overall survival was 90 months for all patients. CONCLUSION: The use of tyrosine kinase inhibitors such as imatinib has a significant favorable effect on the prognosis in the treatment of GISTs, both in adjuvant therapy and in advanced stage disease.
Collapse
|
108
|
Roulleaux Dugage M, Jones RL, Trent J, Champiat S, Dumont S. Beyond the Driver Mutation: Immunotherapies in Gastrointestinal Stromal Tumors. Front Immunol 2021; 12:715727. [PMID: 34489967 PMCID: PMC8417712 DOI: 10.3389/fimmu.2021.715727] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are a subtype of soft tissue sarcoma (STS), and have become a concept of oncogenic addiction and targeted therapies.The large majority of these tumors develop after a mutation in KIT or platelet derived growth factor receptor α (PDGFRα), resulting in uncontrolled proliferation. GISTs are highly sensitive to imatinib. GISTs are immune infiltrated tumors with a predominance of tumor-associated macrophages (TAMs) and T-cells, including many CD8+ T-cells, whose numbers are prognostic. The genomic expression profile is that of an inhibited Th1 response and the presence of tertiary lymphoid structures and B cell signatures, which are known as predictive to response to ICI. However, the microtumoral environment has immunosuppressive attributes, with immunosuppressive M2 macrophages, overexpression of indoleamine 2,3-dioxygenase (IDO) or PD-L1, and loss of major histocompatibility complex type 1. In addition to inhibiting the KIT oncogene, imatinib appears to act by promoting cytotoxic T-cell activity, interacting with natural killer cells, and inhibiting the expression of PD-L1. Paradoxically, imatinib also appears to induce M2 polarization of macrophages. There have been few immunotherapy trials with anti-CTLA-4 or anti-PD-L1drugs and available clinical data are not very promising. Based on this comprehensive analysis of TME, we believe three immunotherapeutic strategies must be underlined in GIST. First, patients included in clinical trials must be better selected, based on the identified driver mutation (such as PDGFRα D842V mutation), the presence of tertiary lymphoid structures (TLS) or PD-L1 expression. Moreover, innovative immunotherapeutic agents also provide great interest in GIST, and there is a strong rationale for exploring IDO targeting after disease progression during imatinib therapy. Finally and most importantly, there is a strong rationale to combine of c-kit inhibition with immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Robin Lewis Jones
- Division of Clinical Studies, Institute of Cancer Research & Sarcoma Unit of the Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Jonathan Trent
- Department of Medicine, Division of Oncology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Stéphane Champiat
- Département d’Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Sarah Dumont
- Département d’Oncologie Médicale, Gustave Roussy, Université Paris Saclay, Villejuif, France
| |
Collapse
|
109
|
Singh AS, Hecht JR, Rosen L, Wainberg ZA, Wang X, Douek M, Hagopian A, Andes R, Sauer L, Brackert SR, Chow W, DeMatteo R, Eilber FC, Glaspy JA, Chmielowski B. A Randomized Phase 2 Study Of Nivolumab Monotherapy Or Nivolumab Combined with Ipilimumab In Patients with Advanced Gastrointestinal Stromal Tumors. Clin Cancer Res 2021; 28:84-94. [PMID: 34407970 DOI: 10.1158/1078-0432.ccr-21-0878] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/27/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Most GISTs are driven by KIT/PDGFRa mutations. TKI benefit is progressively less after imatinib failure. This phase II trial analyzed the efficacy of nivolumab (N) or nivolumab + ipilimumab (N + I) in refractory GIST patients. PATIENTS AND METHODS Advanced/metastatic GIST patients refractory to at least imatinib were randomized 1:1 in a noncomparative, parallel group, unblinded Phase 2 trial of N (240mg Q2wks) or N + I (240 mg Q2wks + 1mg/kg Q6wks). The primary endpoint was the ORR of N alone or N+I by RECIST 1.1 in the ITT population. RESULTS 36 patients with a median of 3 (1-6) prior lines of therapies were enrolled. 10/19(52.6%) patients had SD for a CBR of 52.6 % in the N arm and the median PFS was 11.7 wks (95% CI, 7.0, 17.4). In the N+I arm, 1/16(6.7%) patients had a CR and 4/16(25.0%) had SD for a CBR of 31.3% and a median PFS of 8.3wks (95% CI, 5.6, 22.2). The 4 and 6 month PFS were 42.1% and 26.3%, respectively for N and 31.3% and 18.8%, respectively for N+I. The most common adverse events (AEs) attributed to N and N+I were fatigue: 13.9% and 22.2%, respectively. There were 9 total attributable grade 3-4 AEs. CONCLUSIONS The primary endpoint of RR>15% was not observed for N or N + I. In a heavily pretreated GIST population, responses and long term disease control with both N and N+I were observed. No new safety signals have been observed.
Collapse
Affiliation(s)
- Arun S Singh
- Department of Hematology/Oncology, University of California, Los Angeles
| | - J Randolph Hecht
- Division of Hematology/Oncology, David Geffen School of Medicine at UCLA
| | - Lee Rosen
- Medicine, UCLA Santa Monica Hematology-Oncology
| | - Zev A Wainberg
- Division of Hematology/Oncology, David Geffen School of Medicine at UCLA
| | - Xiaoyan Wang
- Department of General Internal Medicine and Healthy Services Research, University of California, Los Angeles
| | | | - Anahis Hagopian
- Department of Hematology/Oncology, University of California, Los Angeles
| | - Rachel Andes
- Department of Hematology/Oncology, University of California, Los Angeles
| | - Lauren Sauer
- Department of Hematology/Oncology, University of California, Los Angeles
| | - Sandra R Brackert
- Department of Hematology/Oncology, University of California, Los Angeles
| | - Warren Chow
- Medical Oncology, City Of Hope National Medical Center
| | | | | | - John A Glaspy
- Department of Medicine, David Geffen School of Medicine at UCLA
| | | |
Collapse
|
110
|
Kim HJ, Park JY, Kim BJ, Kim JG, Kim HS, Park JM. Gastric Gastrointestinal Stromal Tumor with Repeated Recurrence at the Anastomosis Site in a Very Elderly Patient. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2021; 76:206-210. [PMID: 33100316 DOI: 10.4166/kjg.2020.76.4.206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/30/2020] [Accepted: 08/16/2020] [Indexed: 11/03/2022]
Abstract
Although high-risk gastrointestinal stromal tumors (GISTs) frequently recur, even after a complete resection and imatinib therapy, local recurrence at the suture line after complete resection is rare. The present case was an 88-year-old woman who was initially diagnosed with high-risk GIST without a distant metastasis. She underwent a complete surgical resection of the lesion and received adjuvant imatinib therapy for 18 months, which was discontinued due to severe drug-induced anemia. During the follow- up, an endoscopic examination performed 40 months after the initial surgery revealed local recurrence at the anastomosis site. Although a complete surgical resection was performed, repeated local recurrence was detected 18 months later, which progressed rapidly to metastatic disease. This paper reports a case of a completely resected gastric GIST with repeated local recurrence, despite the complete surgical resections and adjuvant imatinib therapy.
Collapse
Affiliation(s)
- Hye-Jin Kim
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Jae Yong Park
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Beom Jin Kim
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Jae Gyu Kim
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Hee Sung Kim
- Department of Pathology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Joong-Min Park
- Department of Surgery, Chung-Ang University College of Medicine, Seoul, Korea
| |
Collapse
|
111
|
Delayed adjuvant imatinib in patients with high risk of recurrence of gastrointestinal stromal tumor after radical surgery: a retrospective cohort study. J Cancer Res Clin Oncol 2021; 148:1493-1500. [PMID: 34319443 DOI: 10.1007/s00432-021-03749-6] [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: 05/01/2021] [Accepted: 07/26/2021] [Indexed: 02/05/2023]
Abstract
PURPOSE To investigate the impact of delayed adjuvant imatinib on GIST patients with high risk of recurrence. METHOD Adult GIST patients were retrospectively collected from our hospital between 2011 and 2018, and patients having high risk of recurrence were included for subsequent analyses. The primary endpoint was recurrence-free survival (RFS). RESULTS According to the interval between the radical surgery and the beginning of adjuvant imatinib, 222 patients were divided into three groups: group A (≤ 2 months, n = 41), group B (2-≤ 4 months, n = 113), and group C (4-≤ 6 months, n = 68). Univariate, multivariate, and survival analyses all showed that patients in group A had significantly more favorable RFS than those in group C but not group B, and patients taking adjuvant imatinib for over 12 months were also associated with longer RFS comparing to adjuvant imatinib of ≤ 12 months. When stratified by the duration of adjuvant imatinib, no significant differences were found in RFS among groups A, B, and C for adjuvant imatinib of ≤ 12 months. While for adjuvant imatinib of over 12 months, both groups A and B had significantly more favorable RFS than group C, and no significant difference in RFS was found between group A and B. CONCLUSION Delayed postoperative adjuvant imatinib for over 4 months in patients with high risk of recurrence of GIST may lead to worse RFS, and longer treatment with shorter delay has best results.
Collapse
|
112
|
Zalcberg JR, Heinrich MC, George S, Bauer S, Schöffski P, Serrano C, Gelderblom H, Jones RL, Attia S, D'Amato G, Chi P, Reichardt P, Somaiah N, Meade J, Reichert V, Shi K, Sherman ML, Ruiz-Soto R, von Mehren M, Blay JY. Clinical Benefit of Ripretinib Dose Escalation After Disease Progression in Advanced Gastrointestinal Stromal Tumor: An Analysis of the INVICTUS Study. Oncologist 2021; 26:e2053-e2060. [PMID: 34313371 PMCID: PMC8571742 DOI: 10.1002/onco.13917] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/20/2021] [Indexed: 12/16/2022] Open
Abstract
Background Ripretinib 150 mg once daily (QD) is indicated for advanced gastrointestinal stromal tumors (GISTs) as at least fourth‐line therapy. In INVICTUS, ripretinib intrapatient dose escalation (IPDE) to 150 mg b.i.d. was allowed after progressive disease (PD) on 150 mg QD by blinded independent central review using modified RECIST 1.1. We report the efficacy and safety of ripretinib IPDE to 150 mg b.i.d. after PD among patients randomized to ripretinib 150 mg QD in the INVICTUS study. Materials and Methods Tumor imaging was performed every 28‐day cycle for the first four cycles in the ripretinib 150 mg QD period and then every other cycle, including the 150 mg b.i.d. period. Among the ripretinib IPDE patients, progression‐free survival (PFS)1 was the time from randomization until PD; PFS2 was the time from the first dose of ripretinib 150 mg b.i.d. to PD or death. Results Among 43 ripretinib IPDE patients, median PFS1 was 4.6 months (95% confidence interval [CI], 2.7–6.4) and median PFS2 was 3.7 months (95% CI, 3.1–5.3). Median overall survival was 18.4 months (95% CI, 14.5–not estimable). Ripretinib 150 mg b.i.d. (median duration of treatment 3.7 months) was well tolerated with new or worsening grade 3–4 treatment‐emergent adverse events (TEAEs) of anemia in six (14%) and abdominal pain in three (7%) patients. Ripretinib 150 mg b.i.d. was discontinued because of TEAEs in seven (16%) patients. Conclusion Ripretinib 150 mg b.i.d. after PD on 150 mg QD may provide additional clinically meaningful benefit with an acceptable safety profile in patients with at least fourth‐line GISTs. Implications for Practice Of the 85 patients with advanced gastrointestinal stromal tumor having received at least three prior anticancer therapies randomized to ripretinib 150 mg once daily (QD) in the phase III INVICTUS study, 43 underwent ripretinib intrapatient dose escalation (IPDE) to 150 mg b.i.d. after progressive disease (PD). Median progression‐free survival was 4.6 months before and 3.7 months after ripretinib IPDE. The safety profile of ripretinib 150 mg b.i.d. was acceptable. These findings indicate ripretinib IPDE to 150 mg b.i.d. may provide additional clinical benefit in patients with PD on ripretinib 150 mg QD, for whom limited treatment options exist. This article presents further results from the INVICTUS study, focusing on patients who received ripretinib 150 mg QD who received intrapatient dose escalation to 150 mg b.i.d. after progressive disease.
Collapse
Affiliation(s)
- John R Zalcberg
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia.,Department of Medical Oncology, The Alfred Hospital, Melbourne, Australia
| | - Michael C Heinrich
- Portland VA Healthcare System and OHSU Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Suzanne George
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Sebastian Bauer
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, Essen, Germany
| | - Patrick Schöffski
- University Hospitals Leuven, Department of General Medical Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - César Serrano
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Robin L Jones
- Royal Marsden and Institute of Cancer Research, London, United Kingdom
| | | | - Gina D'Amato
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Ping Chi
- Memorial Sloan Kettering Cancer Center & Weill Cornell Medicine, New York, New York, USA
| | - Peter Reichardt
- Sarcoma Center, Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Neeta Somaiah
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Julie Meade
- Deciphera Pharmaceuticals, LLC, Waltham, Massachusetts, USA
| | | | - Kelvin Shi
- Deciphera Pharmaceuticals, LLC, Waltham, Massachusetts, USA
| | | | | | | | - Jean-Yves Blay
- Centre Léon Bérard & Université Claude Bernard, Lyon, France
| |
Collapse
|
113
|
Bauer S, George S, von Mehren M, Heinrich MC. Early and Next-Generation KIT/PDGFRA Kinase Inhibitors and the Future of Treatment for Advanced Gastrointestinal Stromal Tumor. Front Oncol 2021; 11:672500. [PMID: 34322383 PMCID: PMC8313277 DOI: 10.3389/fonc.2021.672500] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/22/2021] [Indexed: 12/21/2022] Open
Abstract
The majority of gastrointestinal stromal tumors (GIST) harbor an activating mutation in either the KIT or PDGFRA receptor tyrosine kinases. Approval of imatinib, a KIT/PDGFRA tyrosine kinase inhibitor (TKI), meaningfully improved the treatment of advanced GIST. Other TKIs subsequently gained approval: sunitinib as a second-line therapy and regorafenib as a third-line therapy. However, resistance to each agent occurs in almost all patients over time, typically due to secondary kinase mutations. A major limitation of these 3 approved therapies is that they target the inactive conformation of KIT/PDGFRA; thus, their efficacy is blunted against secondary mutations in the kinase activation loop. Neither sunitinib nor regorafenib inhibit the full spectrum of KIT resistance mutations, and resistance is further complicated by extensive clonal heterogeneity, even within single patients. To combat these limitations, next-generation TKIs were developed and clinically tested, leading to 2 new USA FDA drug approvals in 2020. Ripretinib, a broad-spectrum KIT/PDGFRA inhibitor, was recently approved for the treatment of adult patients with advanced GIST who have received prior treatment with 3 or more kinase inhibitors, including imatinib. Avapritinib, a type I kinase inhibitor that targets active conformation, was approved for the treatment of adults with unresectable or metastatic GIST harboring a PDGFRA exon 18 mutation, including PDGFRA D842V mutations. In this review, we will discuss how resistance mutations have driven the need for newer treatment options for GIST and compare the original GIST TKIs with the next-generation KIT/PDGFRA kinase inhibitors, ripretinib and avapritinib, with a focus on their mechanisms of action.
Collapse
Affiliation(s)
- Sebastian Bauer
- Department of Medical Oncology, West German Cancer Center, Essen University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Suzanne George
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Margaret von Mehren
- Department of Hematology and Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Michael C. Heinrich
- Department of Medicine, Portland VA Health Care System and OHSU Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
114
|
Battisti NML, Decoster L, Williams GR, Kanesvaran R, Wildiers H, Ring A. Targeted Therapies in Older Adults With Solid Tumors. J Clin Oncol 2021; 39:2128-2137. [PMID: 34043448 PMCID: PMC8260907 DOI: 10.1200/jco.21.00132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/07/2021] [Accepted: 02/23/2021] [Indexed: 12/23/2022] Open
Affiliation(s)
- Nicolò Matteo Luca Battisti
- Department of Medicine—Breast Unit, The Royal Marsden NHS Foundation Trust, Breast Cancer Research Division, The Institute of Cancer Research, Surrey, United Kingdom
| | - Lore Decoster
- Department of Medical Oncology, Universitair Ziekenhuis (UZ) Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Grant R. Williams
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | | | - Hans Wildiers
- Department of General Medical Oncology and Multidisciplinary Breast Centre, University Hospitals Leuven, Leuven Cancer Institute, Laboratory of Experimental Oncology, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Alistair Ring
- Department of Medicine—Breast Unit, The Royal Marsden NHS Foundation Trust, Breast Cancer Research Division, The Institute of Cancer Research, Surrey, United Kingdom
| |
Collapse
|
115
|
Imatinib-resistant gastrointestinal stromal tumors in the era of second- and third-line tyrosine kinase inhibitors: Does surgical resection have a role? Surgery 2021; 170:1481-1486. [PMID: 34090672 DOI: 10.1016/j.surg.2021.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Imatinib resistance is associated with a poor prognosis in patients with gastrointestinal stromal tumors. Although novel tyrosine kinase inhibitors have improved outcomes in imatinib-resistant gastrointestinal stromal tumors, the role of resection remains unclear. We sought to investigate factors predictive of overall and progression-free survival in patients with imatinib-resistant gastrointestinal stromal tumors. METHODS A query of our prospectively maintained Comprehensive Cancer Center registry was performed from 2003 to 2019 for patients with imatinib-resistant gastrointestinal stromal tumors. Clinicopathologic characteristics and medical and surgical treatments were collected; overall survival and progression-free survival after imatinib-resistance were analyzed with Kaplan-Meier and Cox proportional hazards modeling. RESULTS A total of 84 patients developed imatinib resistance at a median age of 59 years. Median time to imatinib resistance after diagnosis and overall survival after imatinib resistance was 50 and 51 months, respectively. After being diagnosed with imatinib resistance, 17 (20%) patients underwent resection. On multivariable analysis, resection after imatinib resistance was independently associated with improved progression-free survival (hazard ratio 0.50; P = .027) but not overall survival (hazard ratio 0.62; P = .215). Similar findings were found on subgroup analysis of patients treated with second-line sunitinib (n = 71). CONCLUSION Long-term survival can be achieved in patients who develop imatinib-resistant gastrointestinal stromal tumors. Surgical resection of imatinib-resistant gastrointestinal stromal tumors is associated with improved progression-free survival and should be considered in selected patients.
Collapse
|
116
|
Trullas-Jimeno A, Delgado J, Garcia-Ochoa B, Wang I, Sancho-Lopez A, Payares-Herrera C, Dalhus ML, Strøm BO, Egeland EJ, Enzmann H, Pignatti F. The EMA assessment of avapritinib in the treatment of gastrointestinal stromal tumours harbouring the PDGFRA D842V mutation. ESMO Open 2021; 6:100159. [PMID: 34023541 PMCID: PMC8165402 DOI: 10.1016/j.esmoop.2021.100159] [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: 03/09/2021] [Revised: 04/07/2021] [Accepted: 04/25/2021] [Indexed: 11/26/2022] Open
Abstract
Avapritinib is a protein kinase inhibitor designed to selectively inhibit oncogenic KIT and platelet-derived growth factor receptor alpha (PDGFRA) mutants by targeting the active conformation of the kinase. On 24 September 2020, a marketing authorisation valid through the European Union was issued for avapritinib as treatment of adult patients with unresectable or metastatic gastrointestinal stromal tumours (GIST) harbouring the PDGFRA D842V mutation. The drug was evaluated in an open-label, phase I, first-in-human, dose-escalation, open-label study to evaluate the safety, tolerability, pharmacokinetics, pharmacodynamics, and efficacy of avapritinib in adults with unresectable or metastatic GIST. The benefit of avapritinib was observed in patients with GIST harbouring the PDGFRA D842V mutation. The overall response rate was 95% (95% confidence interval 82.3%-99.4%), with a median duration of response of 22.1 months (95% confidence interval 14.1-not estimable months). The most common adverse events were nausea, fatigue, anaemia, periorbital and face oedema, hyperbilirubinaemia, diarrhoea, vomiting, increased lacrimation, and decreased appetite. Most of the reported cognitive effects were mild memory impairment. Rarer events were cases of severe encephalopathy and intracranial or gastrointestinal bleeding. The aim of this manuscript is to summarise the scientific review of the application leading to regulatory approval in the European Union. Avapritinib is a protein kinase inhibitor designed to inhibit oncogenic KIT and PDGFRA mutants. A marketing authorisation was issued for avapritinib as treatment of patients with GIST harbouring the PDGFRA D842V mutation. The overall response rate was 95% (95% CI 82.3-99.4), with a median duration of response of 22.1 months (95% CI 14.1-NE). The most common adverse events were nausea, fatigue, anaemia, periorbital and face oedema, hyperbilirubinaemia and diarrhoea.
Collapse
Affiliation(s)
- A Trullas-Jimeno
- Oncology and Haematology Office, European Medicines Agency, Amsterdam, The Netherlands
| | - J Delgado
- Oncology and Haematology Office, European Medicines Agency, Amsterdam, The Netherlands; Department of Haematology, Hospital Clinic, Barcelona, Spain.
| | - B Garcia-Ochoa
- Agencia Española de Medicamentos y Productos Sanitarios, Madrid, Spain; Committee for Medicinal Products for Human Use (CHMP), European Medicines Agency, Amsterdam, The Netherlands
| | - I Wang
- Committee for Medicinal Products for Human Use (CHMP), European Medicines Agency, Amsterdam, The Netherlands; Statens Legemiddelverk, Oslo, Norway
| | - A Sancho-Lopez
- Department of Clinical Pharmacology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - C Payares-Herrera
- Department of Clinical Pharmacology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | | | - B O Strøm
- Statens Legemiddelverk, Oslo, Norway
| | | | - H Enzmann
- Committee for Medicinal Products for Human Use (CHMP), European Medicines Agency, Amsterdam, The Netherlands; Bundesinstitut für Arzneimittel und Medizinprodukte, Bonn, Germany
| | - F Pignatti
- Oncology and Haematology Office, European Medicines Agency, Amsterdam, The Netherlands
| |
Collapse
|
117
|
Patel SR, Reichardt P. An updated review of the treatment landscape for advanced gastrointestinal stromal tumors. Cancer 2021; 127:2187-2195. [PMID: 33974733 PMCID: PMC8252111 DOI: 10.1002/cncr.33630] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 12/21/2022]
Abstract
Before the introduction of tyrosine kinase inhibitors (TKIs), the overall survival of patients with advanced or metastatic gastrointestinal stromal tumors (GISTs) was 10 to 20 months because of the lack of approved therapies. In the last 20 years, a treatment algorithm for patients with advanced GISTs, which includes imatinib, sunitinib, and regorafenib as first‐, second‐, and third‐line therapies, respectively, has been established. Recently, 2 new TKIs have been approved: ripretinib for fourth‐line therapy and avapritinib as first‐line therapy in patients harboring platelet‐derived growth factor receptor α (PDGFRA) exon 18 D842V mutations. Additionally, there are several experimental therapies under investigation that could advance individualized patient care. All of these therapies have varying efficacies and safety profiles that warrant an updated treatment landscape review. This review article summarizes the efficacy and safety data currently available for conventional TKIs along with recently approved and experimental therapies. With evolving treatment options and effective toxicity management, patients with advanced gastrointestinal stromal tumors are living longer than ever before. Recently approved targeted therapies and the investigation of experimental treatment options have the potential to alter the current treatment algorithm and encourage personalized patient care.
Collapse
Affiliation(s)
- Shreyaskumar R Patel
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Peter Reichardt
- Oncology and Palliative Care, Sarcoma Center, Helios Klinikum Berlin-Buch, Berlin, Germany
| |
Collapse
|
118
|
Dudzisz-Śledź M, Bylina E, Teterycz P, Rutkowski P. Treatment of Metastatic Gastrointestinal Stromal Tumors (GIST): A Focus on Older Patients. Drugs Aging 2021; 38:375-396. [PMID: 33651369 PMCID: PMC8096750 DOI: 10.1007/s40266-021-00841-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2021] [Indexed: 11/24/2022]
Abstract
Gastrointestinal stromal tumors (GIST) originating in the Cajal cells are the most common mesenchymal neoplasms of the gastrointestinal tract. The median age of patients with this diagnosis is 65 years, and over 20% of cases affect people over the age of 70 years. The effectiveness and tolerability of systemic treatment with tyrosine kinase inhibitors in older patients with GIST seem to be similar to that in younger patients, but some studies have shown that treatment of older patients is suboptimal. Disability, frailty, comorbidities, and concomitant medications may influence treatment decisions, and toxicities also more often lead to treatment discontinuation. The known safety profile and oral administration route of the tyrosine kinase inhibitors used in GIST may allow maximization of treatment and the best efficacy, especially in older patients. This review summarizes the efficacy data for the systemic treatment of GIST, including data for older patients and from real-world experiences, if available and significant. The reported safety data and general rules for toxicity management, including appropriate patient selection and the need for careful monitoring during treatment, are also discussed.
Collapse
Affiliation(s)
- Monika Dudzisz-Śledź
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781, Warsaw, Poland.
| | - Elżbieta Bylina
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781, Warsaw, Poland
| | - Paweł Teterycz
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781, Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781, Warsaw, Poland
| |
Collapse
|
119
|
Al-Share B, Alloghbi A, Al Hallak MN, Uddin H, Azmi A, Mohammad RM, Kim SH, Shields AF, Philip PA. Gastrointestinal stromal tumor: a review of current and emerging therapies. Cancer Metastasis Rev 2021; 40:625-641. [PMID: 33876372 DOI: 10.1007/s10555-021-09961-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023]
Abstract
Gastrointestinal stromal tumors (GIST) are rare neoplasms arising from the interstitial cell of Cajal in the gastrointestinal tract. Two thirds of GIST in adult patients have c-Kit mutation and smaller fractions have platelet derived growth factor receptor alpha (PDGFRA) mutation. Surgery is the only curative treatment for localized disease. Imatinib improves survival when used adjuvantly and in advanced disease. Several targeted therapies have also improved survival in GIST patients after progression on imatinib including sunitinib and regorafenib. Recently, United States Federal and Drug Administration (FDA) approved two new tyrosine kinase inhibitors for the treatment of heavily pretreated advanced/unresectable GIST including avapritinib (a selective inhibitor for PDGFRA exon 18 mutation including D842V mutations) and ripretinib (a broad-spectrum kinase inhibitor of c-Kit and PDGFRA). In this article, we will provide a comprehensive review of GIST including the current standard of care treatment and exploring future paradigm shifts in therapy.
Collapse
Affiliation(s)
- Bayan Al-Share
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Abdulrahman Alloghbi
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Mohammed Najeeb Al Hallak
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Hafiz Uddin
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Asfar Azmi
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Ramzi M Mohammad
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Steve H Kim
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Anthony F Shields
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Philip A Philip
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI, USA.
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
120
|
Abstract
The SMARCB1/INI1 gene was first discovered in the mid-1990s, and since then it has been revealed that loss of function mutations in this gene result in aggressive rhabdoid tumors. Recently, the term "rhabdoid tumor" has become synonymous with decreased SMARCB1/INI1 expression. When genetic aberrations in the SMARCB1/INI1 gene occur, the result can cause complete loss of expression, decreased expression, and mosaic expression. Although SMARCB1/INI1-deficient tumors are predominantly sarcomas, this is a diverse group of tumors with mixed phenotypes, which can often make the diagnosis challenging. Prognosis for these aggressive tumors is often poor. Moreover, refractory and relapsing progressive disease is common. As a result, accurate and timely diagnosis is imperative. Despite the SMARCB1/INI1 gene itself and its implications in tumorigenesis being discovered over two decades ago, there is a paucity of rhabdoid tumor cases reported in the literature that detail SMARCB1/INI1 expression. Much work remains if we hope to provide additional therapeutic strategies for patients with aggressive SMARCB1/INI1-deficient tumors.
Collapse
Affiliation(s)
- Nathaniel A Parker
- University of Kansas School of Medicine, 1010 N Kansas St, Wichita, KS, 67214, USA
| | - Ammar Al-Obaidi
- University of Kansas School of Medicine, 1010 N Kansas St, Wichita, KS, 67214, USA
| | - Jeremy M Deutsch
- Cancer Center of Kansas, 818 N. Emporia #403, Wichita, KS, 67214, USA
| |
Collapse
|
121
|
Zalcberg JR. Ripretinib for the treatment of advanced gastrointestinal stromal tumor. Therap Adv Gastroenterol 2021; 14:17562848211008177. [PMID: 33948116 PMCID: PMC8053826 DOI: 10.1177/17562848211008177] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Gastrointestinal stromal tumors (GISTs) are rare tumors of the gastrointestinal (GI) tract yet represent the most common GI sarcomas. Most GISTs are driven by activating mutations of the KIT and/or PDGFRA genes. Prior to the development of tyrosine kinase inhibitors (TKIs), GISTs were associated with a poor prognosis because conventional cytotoxic chemotherapy was relatively ineffective. However, TKIs that inhibit the most common driver mutations in KIT or PDGFRA have revolutionized the treatment of GISTs over the past two decades. Notwithstanding, ongoing management challenges relate to the development of secondary mutations in these genes, resulting in tumor progression. Due to both the intra- and inter-patient heterogeneity of these secondary mutations in GISTs, optimal treatment requires an agent that blocks as many mutant genes as possible. Ripretinib - a novel switch-control TKI - inhibits many of the most common primary and secondary activating KIT and PDGFRA mutants involved in GIST progression through a dual mechanism of action. In the pivotal INVICTUS phase III trial, patients with advanced GIST that had progressed on at least imatinib, sunitinib, and regorafenib and who received ripretinib experienced significantly longer progression-free survival (primary endpoint) as well as prolongation of overall survival, compared with those receiving placebo. Treatment with ripretinib was associated with durable improvements in quality-of-life indices and a manageable toxicity profile. The most frequent side effects were common to the class of TKIs used in the management of GIST. These results led to the approval of ripretinib for treatment of advanced GIST in adults who have received three or more TKIs, including imatinib. Ripretinib is also under investigation in the second-line treatment of advanced GIST in a phase III trial (INTRIGUE) comparing ripretinib with sunitinib in patients with advanced GIST after treatment with imatinib. PLAIN LANGUAGE SUMMARY Use of ripretinib for the treatment of gastrointestinal stromal tumors (GISTs) Gastrointestinal stromal tumors (GISTs) are a rare type of tumor most commonly located in the stomach and small intestine but can develop anywhere throughout the gastrointestinal tract. The symptoms of GISTs vary in extent depending on location of the primary tumor and include a feeling of fullness, abdominal pain, intestinal bleeding, and fatigue. Since these symptoms are nonspecific, making a diagnosis can be challenging. Most GISTs carry initial mutations in genes that control specific enzymes called tyrosine kinases. Historically, treatment of GISTs was limited because traditional chemotherapy is ineffective against these tumors. However, with the introduction of drugs that inhibit tyrosine kinases [i.e., tyrosine kinase inhibitors (TKIs)], survival has been extended substantially. However, many GISTs go on to develop secondary mutations that render them resistant to a given TKI. Prior to the approval of ripretinib, four TKIs were available for the treatment of GIST: imatinib; sunitinib; regorafenib; and, recently, avapritinib. Each drug is used until resistance develops or patients are unable to tolerate the side effects of treatment, after which the next drug is started. Ripretinib was recently approved by the FDA as the fourth drug in the usual treatment sequence recommended for patients with advanced GIST who have progressed (or are treatment intolerant) after receiving three or more TKIs, including imatinib. Approval of ripretinib was based on the results of the INVICTUS trial, which demonstrated that the drug significantly improves the time patients have without progression of the disease or death compared with placebo. The most common side effects related to ripretinib were hair loss, muscle pain, nausea, fatigue, hand-foot syndrome, and diarrhea, although most events were not very severe. Ripretinib is being further studied as the second TKI used in patients with GIST who have progressed on or could not tolerate first-line treatment with imatinib.
Collapse
|
122
|
Deo KB, Gautam S, Awale L, Yadav TN, Pradhan A, Pandit N. Cystic Ileal Gastrointestinal Stromal Tumor Masquerading as Metastatic Adnexal Carcinoma. J Gastrointest Cancer 2021; 51:1053-1056. [PMID: 32303996 DOI: 10.1007/s12029-020-00403-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Kunal Bikram Deo
- Department of Surgery, B. P. Koirala Institute of Health Sciences (BPKIHS), Dharan, Nepal.
| | - Sujan Gautam
- Department of Surgery, B. P. Koirala Institute of Health Sciences (BPKIHS), Dharan, Nepal
| | - Laligen Awale
- Department of Surgery, B. P. Koirala Institute of Health Sciences (BPKIHS), Dharan, Nepal
| | - Tek Narayan Yadav
- Department of Surgery, B. P. Koirala Institute of Health Sciences (BPKIHS), Dharan, Nepal
| | - Anju Pradhan
- Department of Pathology, B. P. Koirala Institute of Health Sciences (BPKIHS), Dharan, Nepal
| | - Narendra Pandit
- Department of Surgery, B. P. Koirala Institute of Health Sciences (BPKIHS), Dharan, Nepal
| |
Collapse
|
123
|
Klug LR, Corless CL, Heinrich MC. Inhibition of KIT Tyrosine Kinase Activity: Two Decades After the First Approval. J Clin Oncol 2021; 39:1674-1686. [PMID: 33797935 DOI: 10.1200/jco.20.03245] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Lillian R Klug
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR.,Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR.,VA Portland Health Care System, Portland, OR
| | - Christopher L Corless
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR.,Department of Pathology, Oregon Health & Science University, Portland, OR
| | - Michael C Heinrich
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR.,Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR.,VA Portland Health Care System, Portland, OR
| |
Collapse
|
124
|
Marqueen KE, Moshier E, Buckstein M, Ang C. Neoadjuvant therapy for gastrointestinal stromal tumors: A propensity score-weighted analysis. Int J Cancer 2021; 149:177-185. [PMID: 33634858 DOI: 10.1002/ijc.33536] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/22/2022]
Abstract
Retrospective and single-arm prospective studies have reported clinical benefit with neoadjuvant imatinib for GISTs. In the absence of randomized Phase III data, the impact of neoadjuvant systemic therapy (NAT) on survival compared to upfront resection (UR) remains unknown. We identified N = 16 308 patients within the National Cancer Database (2004-2016) who underwent resection of localized GIST of the stomach, esophagus, small bowel and colorectum, with or without ≥3 months of NAT. Inverse probability of treatment weighting adjusted for covariable imbalance among treatment groups. We estimated the effect of NAT on overall survival with a weighted time-dependent Cox proportional hazards model, and on 90-day postoperative mortality and R0 resection with weighted logistic regressions. Eight hundred sixty-five (5.3%) patients received NAT compared to 15 443 (94.7%) who underwent UR. Median NAT duration was 6.3 months. 53.7% of NAT patients were male vs 48.6% of UR patients, 67.3% vs 65.1% had primary gastric GIST and 72.8% vs 49.7% were at high risk. NAT patients had larger tumors and higher mitotic index. >3 months of NAT was associated with a significant survival benefit (weighted HR 0.85 [0.80-0.91]). 90-day postoperative mortality rate was 4/865 (0.5%) among NAT patients vs 346/15443 (2.2%). NAT was associated with lower odds of 90-day postoperative mortality. R0 resection rate was not significantly different between groups. In conclusion, despite higher risk features among NAT patients, this analysis suggests that NAT for localized GIST is associated with a modest survival benefit and lower risk of 90-day postoperative mortality, with no difference in likelihood of achieving an R0 resection.
Collapse
Affiliation(s)
- Kathryn E Marqueen
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Erin Moshier
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael Buckstein
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Celina Ang
- Department of Medicine, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
125
|
Abstract
Gastrointestinal stromal tumours (GIST) have an incidence of ~1.2 per 105 individuals per year in most countries. Around 80% of GIST have varying molecular changes, predominantly mutually exclusive activating KIT or PDGFRA mutations, but other, rare subtypes also exist. Localized GIST are curable, and surgery is their standard treatment. Risk factors for relapse are tumour size, mitotic index, non-gastric site and tumour rupture. Patients with GIST with KIT or PDGFRA mutations sensitive to the tyrosine kinase inhibitor (TKI) imatinib that are at high risk of relapse have improved survival with adjuvant imatinib treatment. In advanced disease, median overall survival has improved from 18 months to >70 months since the introduction of TKIs. The role of surgery in the advanced setting remains unclear. Resistance to TKIs arise mainly from subclonal selection of cells with resistance mutations in KIT or PDGFRA when they are the primary drivers. Advanced resistant GIST respond to second-line sunitinib and third-line regorafenib, as well as to the new broad-spectrum TKI ripretinib. Rare molecular forms of GIST with alterations involving NF1, SDH genes, BRAF or NTRK genes generally show primary resistance to standard TKIs, but some respond to specific inhibitors of the activated genes. Despite major advances, many questions in both advanced and localized disease remain unanswered.
Collapse
Affiliation(s)
- Jean-Yves Blay
- Department of Medicine, Centre Leon Berard, UNICANCER & University Lyon I, Lyon, France.
| | - Yoon-Koo Kang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Toshiroo Nishida
- Surgery Department, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | | |
Collapse
|
126
|
Pilco-Janeta DF, García-Valverde A, Gomez-Peregrina D, Serrano C. Emerging drugs for the treatment of gastrointestinal stromal tumors. Expert Opin Emerg Drugs 2021; 26:53-62. [PMID: 33645383 DOI: 10.1080/14728214.2021.1896704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Oncogenic activation of KIT or PDGFRA receptor tyrosine kinases is the crucial event in gastrointestinal stromal tumor (GIST) biology. Seminal works during the past two decades have underscored, first, the continuous relevance of KIT/PDGFRA oncogenic signaling after progression to targeted inhibition; second, the heterogeneity of KIT/PDGFRA acquired mutations, that cannot be efficiently suppressed by any given tyrosine kinase inhibitor (TKI); and third, the presence of specific mutants highly resistant to all approved therapies. AREAS COVERED This review discusses treatment options in advanced/metastatic GIST, including a detailed dissection of ripretinib and avapritinib, the two novel small molecule inhibitors approved by the Food and Drug Administration in 2020. EXPERT OPINION The three only therapeutic options since 2012 for metastatic GIST patients were imatinib, sunitinib, and regorafenib. Although imatinib was highly effective in treatment-naïve GIST, the benefit of second- and third-line sunitinib and regorafenib was modest, thus emphasizing the medical need for new treatment options. Ripretinib, a switch control inhibitor with broad anti-KIT/PDGFRA activity, has been approved as ≥4th line in GIST after progression to all standard therapies. Avapritinib, a type I TKI highly specific against the multi-resistant PDGFRA D842V mutation, is approved in this specific subset of GIST patients.
Collapse
Affiliation(s)
- Daniel F Pilco-Janeta
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Alfonso García-Valverde
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - David Gomez-Peregrina
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - César Serrano
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Hospital Campus, Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| |
Collapse
|
127
|
Jones RL, Serrano C, von Mehren M, George S, Heinrich MC, Kang YK, Schöffski P, Cassier PA, Mir O, Chawla SP, Eskens FALM, Rutkowski P, Tap WD, Zhou T, Roche M, Bauer S. Avapritinib in unresectable or metastatic PDGFRA D842V-mutant gastrointestinal stromal tumours: Long-term efficacy and safety data from the NAVIGATOR phase I trial. Eur J Cancer 2021; 145:132-142. [PMID: 33465704 PMCID: PMC9518931 DOI: 10.1016/j.ejca.2020.12.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND PDGFRA D842V mutations occur in 5-10% of gastrointestinal stromal tumours (GISTs), and previously approved tyrosine kinase inhibitors (TKIs) are inactive against this mutation. Consequently, patients have a poor prognosis. We present an updated analysis of avapritinib efficacy and long-term safety in this patient population. METHODS NAVIGATOR (NCT02508532), a two-part, open-label, dose-escalation/dose-expansion phase I study, enrolled adult patients with unresectable GISTs. Patients with PDGFRA D842V-mutant GIST were a prespecified subgroup within the overall safety population, which included patients who received ≥1 avapritinib dose. Primary end-points were overall response rate (ORR) and avapritinib safety profile. Secondary end-points were clinical benefit rate (CBR), duration of response (DOR) and progression-free survival (PFS). Overall survival (OS) was an exploratory end-point. RESULTS Between 7 October 2015 and 9 March 2020, 250 patients enrolled in the safety population; 56 patients were included in the PDGFRA D842V population, 11 were TKI-naïve. At data cut-off, median follow-up was 27.5 months. Safety profile was comparable between the overall safety and PDGFRA D842V populations. In the PDGFRA D842V population, the most frequent adverse events were nausea (38 [68%] patients) and diarrhoea (37 [66%]), and cognitive effects occurred in 32 (57%) patients. The ORR was 91% (51/56 patients). The CBR was 98% (55/56 patients). The median DOR was 27.6 months (95% confidence interval [CI]: 17.6-not reached [NR]); median PFS was 34.0 months (95% CI: 22.9-NR). Median OS was not reached. CONCLUSION Targeting PDGFRA D842V-mutant GIST with avapritinib resulted in an unprecedented, durable clinical benefit, with a manageable safety profile. Avapritinib should be considered as first-line therapy for these patients.
Collapse
Affiliation(s)
- Robin L Jones
- Royal Marsden Hospital and Institute of Cancer Research, London, UK.
| | - César Serrano
- Vall d' Hebron Institute of Oncology, Barcelona, Spain
| | | | | | - Michael C Heinrich
- Portland VA Health Care System and OHSU Knight Cancer Institute, Portland, OR, USA
| | - Yoon-Koo Kang
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Patrick Schöffski
- University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | | | | | | | | | - Piotr Rutkowski
- Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - William D Tap
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - Teresa Zhou
- Blueprint Medicines Corporation, Cambridge, MA, USA
| | - Maria Roche
- Blueprint Medicines Corporation, Cambridge, MA, USA
| | - Sebastian Bauer
- Department of Medical Oncology, University Hospital Essen, Sarcoma Center, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
128
|
Yoon H, Tang CM, Banerjee S, Yebra M, Noh S, Burgoyne AM, Torre JDL, Siena MD, Liu M, Klug LR, Choi YY, Hosseini M, Delgado AL, Wang Z, French RP, Lowy A, DeMatteo RP, Heinrich MC, Molinolo AA, Gutkind JS, Harismendy O, Sicklick JK. Cancer-associated fibroblast secretion of PDGFC promotes gastrointestinal stromal tumor growth and metastasis. Oncogene 2021; 40:1957-1973. [PMID: 33603171 PMCID: PMC7979540 DOI: 10.1038/s41388-021-01685-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/13/2021] [Accepted: 01/27/2021] [Indexed: 01/30/2023]
Abstract
Targeted therapies for gastrointestinal stromal tumor (GIST) are modestly effective, but GIST cannot be cured with single agent tyrosine kinase inhibitors. In this study, we sought to identify new therapeutic targets in GIST by investigating the tumor microenvironment. Here, we identified a paracrine signaling network by which cancer-associated fibroblasts (CAFs) drive GIST growth and metastasis. Specifically, CAFs isolated from human tumors were found to produce high levels of platelet-derived growth factor C (PDGFC), which activated PDGFC-PDGFRA signal transduction in GIST cells that regulated the expression of SLUG, an epithelial-mesenchymal transition (EMT) transcription factor and downstream target of PDGFRA signaling. Together, this paracrine induce signal transduction cascade promoted tumor growth and metastasis in vivo. Moreover, in metastatic GIST patients, SLUG expression positively correlated with tumor size and mitotic index. Given that CAF paracrine signaling modulated GIST biology, we directly targeted CAFs with a dual PI3K/mTOR inhibitor, which synergized with imatinib to increase tumor cell killing and in vivo disease response. Taken together, we identified a previously unappreciated cellular target for GIST therapy in order to improve disease control and cure rates.
Collapse
Affiliation(s)
- Hyunho Yoon
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Chih-Min Tang
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Sudeep Banerjee
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, CA, USA
- Department of Surgery, University of California, Los Angeles, CA, USA
| | - Mayra Yebra
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Sangkyu Noh
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Adam M Burgoyne
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Jorge De la Torre
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Martina De Siena
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, CA, USA
- Gastroenterology and Digestive Endoscopy, Fondazione Policlinico A.Gemelli Catholic University of Rome, Rome, Italy
| | - Mengyuan Liu
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Lillian R Klug
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR, USA
- Portland VA Health Care System, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Yoon Young Choi
- Division of Biomedical Informatics, Moores Cancer Center, University of California, San Diego, CA, USA
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Mojgan Hosseini
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Antonio L Delgado
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Zhiyong Wang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Randall P French
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Andrew Lowy
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Ronald P DeMatteo
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael C Heinrich
- Portland VA Health Care System, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Alfredo A Molinolo
- Department of Pathology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - J Silvio Gutkind
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Olivier Harismendy
- Division of Biomedical Informatics, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Jason K Sicklick
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, CA, USA.
| |
Collapse
|
129
|
Hedenström P, Andersson C, Sjövall H, Enlund F, Nilsson O, Nilsson B, Sadik R. Pretreatment Tumor DNA Sequencing of KIT and PDGFRA in Endosonography-Guided Biopsies Optimizes the Preoperative Management of Gastrointestinal Stromal Tumors. Mol Diagn Ther 2021; 24:201-214. [PMID: 32124386 PMCID: PMC7113213 DOI: 10.1007/s40291-020-00451-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Neoadjuvant tyrosine kinase inhibitor (TKI) therapy increases the chance of organ-preserving, radical resection in selected patients with gastrointestinal stromal tumors (GISTs). We aimed to evaluate systematic, immediate DNA sequencing of KIT and PDGFRA in pretreatment GIST tissue to guide neoadjuvant TKI therapy and optimize preoperative tumor response. METHODS All patients who were candidates for neoadjuvant therapy of a suspected GIST [the study cohort (SC)] were prospectively included from January 2014 to March 2018. Patients were subjected to pretreatment endosonography-guided fine-needle biopsy (EUS-FNB) or transabdominal ultrasound-guided needle biopsy (TUS-NB), followed by immediate tumor DNA sequencing (< 2 weeks). A historic (2006-2013) reference cohort (RC) underwent work-up without sequencing before neoadjuvant imatinib (n = 42). The rate of optimal neoadjuvant therapy (TherapyOPTIMAL) was calculated, and the induced tumor size reduction (Tumor RegressionMAX, %) was evaluated by computed tomography (CT) scan. RESULTS The success rate of pretreatment tumor DNA sequencing in the SC (n = 81) was 77/81 (95%) [EUS-FNB 71/74 (96%); TUS-NB 6/7 (86%)], with mutations localized in KIT (n = 58), PDGFRA (n = 18), or neither gene, wild type (n = 5). In patients with a final indication for neoadjuvant therapy, the TherapyOPTIMAL was higher in the SC compared with the RC [61/63 (97%) versus 33/42 (79%), p = 0.006], leading to a significantly higher Tumor RegressionMAX in patients treated with TKI (27% vs. 19%, p = 0.015). CONCLUSIONS Pretreatment endosonography-guided biopsy sampling followed by immediate tumor DNA sequencing of KIT and PDGFRA is highly accurate and valuable in guiding neoadjuvant TKI therapy in GIST. This approach minimizes maltreatment with inappropriate regimens and leads to improved tumor size reduction before surgery.
Collapse
Affiliation(s)
- Per Hedenström
- Division of Medical Gastroenterology, Department of Internal Medicine, Sahlgrenska University Hospital, Blå Stråket 3, 413 35, Gothenburg, Sweden. .,Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Carola Andersson
- Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Henrik Sjövall
- Division of Medical Gastroenterology, Department of Internal Medicine, Sahlgrenska University Hospital, Blå Stråket 3, 413 35, Gothenburg, Sweden.,Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Enlund
- Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ola Nilsson
- Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Bengt Nilsson
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Riadh Sadik
- Division of Medical Gastroenterology, Department of Internal Medicine, Sahlgrenska University Hospital, Blå Stråket 3, 413 35, Gothenburg, Sweden
| |
Collapse
|
130
|
Yoon H, Tang CM, Banerjee S, Delgado AL, Yebra M, Davis J, Sicklick JK. TGF-β1-mediated transition of resident fibroblasts to cancer-associated fibroblasts promotes cancer metastasis in gastrointestinal stromal tumor. Oncogenesis 2021; 10:13. [PMID: 33568624 PMCID: PMC7876107 DOI: 10.1038/s41389-021-00302-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/17/2020] [Accepted: 01/13/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are the most abundant cells in the tumor microenvironment. Crosstalk between tumor cells and CAFs contributes to tumor survival in most epithelial cancers. Recently, utilizing gastrointestinal stromal tumor (GIST) as a model for sarcomas, we identified paracrine networks by which CAFs promote tumor progression and metastasis. However, the mechanisms by which CAFs arise in sarcomas remain unclear. Here, RNA sequencing analysis revealed that transforming growth factor-β1 (TGF-β1) is highly expressed in both tumor cells and CAFs. To determine the functional role of TGF-β1, we treated normal gastric fibroblasts (GFs) with recombinant TGF-β1, which caused the GFs to adopt a more stellate morphology, as well as increased the mRNA expression of CAF-mediated genes (CCL2, RAB3B, and TNC) and genes encoding fibroblast growth factors (FGFs). Moreover, while either GIST or CAF conditioned media enhanced the transition from GFs to CAFs, a TGF-β1-blocking antibody attenuated this effect. Transwell migration assays revealed that the TGF-β1-mediated transition from GFs to CAFs enhanced tumor cell migration. This migratory effect was abrogated by an anti-TGF-β1 antibody, suggesting that TGF-β1 secreted from GIST cells or CAFs is associated with GIST migration via GF-to-CAF transition. In addition, the murine spleen-to-liver metastasis model showed that GF pre-treated with TGF-β1 promoted GIST metastasis. Collectively, these findings reveal unappreciated crosstalk among tumor cells, CAFs, and normal resident fibroblasts in the stroma of sarcomas, which enhances a GF-to-CAF transition associated with tumor migration and metastasis.
Collapse
Affiliation(s)
- Hyunho Yoon
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego, CA, USA
- Moores Cancer Center, University of California, San Diego, CA, USA
| | - Chih-Min Tang
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego, CA, USA
- Moores Cancer Center, University of California, San Diego, CA, USA
| | - Sudeep Banerjee
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego, CA, USA
- Moores Cancer Center, University of California, San Diego, CA, USA
- Department of Surgery, University of California, Los Angeles, CA, USA
| | - Antonio L Delgado
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego, CA, USA
- Moores Cancer Center, University of California, San Diego, CA, USA
| | - Mayra Yebra
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego, CA, USA
- Moores Cancer Center, University of California, San Diego, CA, USA
| | - Jacob Davis
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego, CA, USA
- Moores Cancer Center, University of California, San Diego, CA, USA
| | - Jason K Sicklick
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego, CA, USA.
- Moores Cancer Center, University of California, San Diego, CA, USA.
| |
Collapse
|
131
|
George S, Jones RL, Bauer S, Kang YK, Schöffski P, Eskens F, Mir O, Cassier PA, Serrano C, Tap WD, Trent J, Rutkowski P, Patel S, Chawla SP, Meiri E, Gordon M, Zhou T, Roche M, Heinrich MC, von Mehren M. Avapritinib in Patients With Advanced Gastrointestinal Stromal Tumors Following at Least Three Prior Lines of Therapy. Oncologist 2021; 26:e639-e649. [PMID: 33453089 PMCID: PMC8018324 DOI: 10.1002/onco.13674] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/05/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Most gastrointestinal stromal tumors (GIST) driven by KIT or platelet-derived growth factor receptor A (PDGFRA) mutations develop resistance to available tyrosine kinase inhibitor (TKI) treatments. NAVIGATOR is a two-part, single-arm, dose escalation and expansion study designed to evaluate safety and antineoplastic activity of avapritinib, a selective, potent inhibitor of KIT and PDGFRA, in patients with unresectable or metastatic GIST. MATERIALS AND METHODS Eligible patients were 18 years or older with histologically or cytologically confirmed unresectable GIST and Eastern Cooperative Oncology Group performance status ≤2 and initiated avapritinib at 300 mg or 400 mg once daily. Primary endpoints were safety in patients who initiated avapritinib at 300 mg or 400 mg once daily and overall response rate (ORR) in patients in the safety population with three or more previous lines of TKI therapy. RESULTS As of November 16, 2018, in the safety population (n = 204), the most common adverse events (AEs) were nausea (131 [64%]), fatigue (113 [55%]), anemia (102 [50%]), cognitive effects (84 [41%]), and periorbital edema (83 [41%]); 17 (8%) patients discontinued due to treatment-related AEs, most frequently confusion, encephalopathy, and fatigue. ORR in response-evaluable patients with GIST harboring KIT or non-D842V PDGFRA mutations and with at least three prior therapies (n = 103) was 17% (95% confidence interval [CI], 10-25). Median duration of response was 10.2 months (95% CI, 7.2-10.2), and median progression-free survival was 3.7 months (95% CI, 2.8-4.6). CONCLUSION Avapritinib has manageable toxicity with meaningful clinical activity as fourth-line or later treatment in some patients with GIST with KIT or PDGFRA mutations. IMPLICATIONS FOR PRACTICE In the NAVIGATOR trial, avapritinib, an inhibitor of KIT and platelet-derived growth factor receptor A tyrosine kinases, provided durable responses in a proportion of patients with advanced gastrointestinal stromal tumors (GIST) who had received three or more prior therapies. Avapritinib had a tolerable safety profile, with cognitive adverse events manageable with dose interruptions and modification in most cases. These findings indicate that avapritinib can elicit durable treatment responses in some patients with heavily pretreated GIST, for whom limited treatment options exist.
Collapse
Affiliation(s)
- Suzanne George
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Robin L Jones
- Royal Marsden Hospital and Institute of Cancer Research, London, UK
| | - Sebastian Bauer
- Department of Medical Oncology, West German Cancer Center, University of Duisburg-Essen, Essen, Germany
| | - Yoon-Koo Kang
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Patrick Schöffski
- University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Ferry Eskens
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | | | - Cesar Serrano
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - William D Tap
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| | - Jonathan Trent
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Piotr Rutkowski
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | - Sant P Chawla
- Sarcoma Oncology Center, Santa Monica, California, USA
| | - Eval Meiri
- Cancer Treatment Center of America, Atlanta, Georgia, USA
| | - Michael Gordon
- HonorHealth Research Institute, Scottsdale, Arizona, USA
| | - Teresa Zhou
- Blueprint Medicines Corporation, Cambridge, Massachusetts, USA
| | - Maria Roche
- Blueprint Medicines Corporation, Cambridge, Massachusetts, USA
| | - Micahel C Heinrich
- Portland VA Health Care System and OHSU Knight Cancer Institute, Portland, Oregon, USA
| | | |
Collapse
|
132
|
Anticancer effects of dietary administration of secoisolariciresinol
diglucoside in a patient of gastrointestinal stromal tumor: a case
report. INTERNATIONAL JOURNAL OF SURGERY: ONCOLOGY 2021. [DOI: 10.1097/ij9.0000000000000103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
133
|
Final analysis of the randomized trial on imatinib as an adjuvant in localized gastrointestinal stromal tumors (GIST) from the EORTC Soft Tissue and Bone Sarcoma Group (STBSG), the Australasian Gastro-Intestinal Trials Group (AGITG), UNICANCER, French Sarcoma Group (FSG), Italian Sarcoma Group (ISG), and Spanish Group for Research on Sarcomas (GEIS) ☆. Ann Oncol 2021; 32:533-541. [PMID: 33482247 DOI: 10.1016/j.annonc.2021.01.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 12/31/2020] [Accepted: 01/10/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND In 2004, we started an intergroup randomized trial of adjuvant imatinib versus no further therapy after R0-R1 surgery in localized, high/intermediate-risk gastrointestinal stromal tumors (GIST) patients. Interim analysis results were published in 2015 upon recommendation from an independent data review committee. We report the final outcome of the study. PATIENTS AND METHODS This was a randomized, open-label, multicenter phase III trial carried out at 112 hospitals in 12 countries. Patients were randomized to 2 years of imatinib, 400 mg daily, or no further therapy after surgery. The primary endpoint was imatinib failure-free survival (IFFS), while relapse-free survival (RFS), relapse-free interval (RFI), overall survival (OS) and toxicity were secondary endpoints. Adjusting for the interim analyses, results on IFFS were assessed on a 4.3% significance level; for the other endpoints, 5% was used. RESULTS Nine hundred and eight patients were randomized between January 2005 and October 2008: 454 to imatinib and 454 to observation; 835 patients were eligible. With a median follow-up of 9.1 years, 5 (10)-year IFFS was 87% (75%) in the imatinib arm versus 83% (74%) in the control arm [hazard ratio (HR) = 0.87, 95.7% confidence interval (CI) (0.65; 1.15), P = 0.31]; RFS was 70% versus 63% at 5 years and 63% versus 61% at 10 years, [HR = 0.71, 95% CI (0.57; 0.89), P = 0.002]; OS was 93% versus 92% at 5 years and 80% versus 78% at 10 years [HR = 0.88, 95% CI (0.65; 1.21), P = 0.43]. Among 526 patients with high-risk GIST by local pathology, 10-year IFFS and RFS were 69% versus 61%, and 48% versus 43%, respectively. CONCLUSIONS With 9.1 years of follow-up, a trend toward better long-term IFFS in imatinib-treated patients was observed in the high-risk subgroup. Although the difference was not statistically significant and the surrogacy value of such an endpoint is not validated, this may be seen as supporting the results reported by the Scandinavian/German trial, showing a sustained small but significant long-term OS benefit in high-risk GIST patients treated with 3 years of adjuvant imatinib.
Collapse
|
134
|
Song LJ, Ge HJ, Shi XQ, Shen WW. Prognostic and predictive values of the KIT11-mutated grading system in patients with gastrointestinal stromal tumors: a retrospective study. Hum Pathol 2021; 110:31-42. [PMID: 33476644 DOI: 10.1016/j.humpath.2021.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 01/10/2023]
Abstract
The KIT11 mutation is the most frequent mutation pattern in gastrointestinal stromal tumors (GISTs). However, few studies have investigated the correlation between the KIT11-mutated grading system and imatinib mesylate (IM) sensitivity (the first choice for adjuvant treatment of GISTs). Here, we elucidated the clinical value of the KIT11-mutated grading system for prognostic prediction in patients with GISTs treated with IM. A total of 106 patients with GIST were treated with IM (8: intermediate-risk, 98: high-risk; 10: KIT9-mutated, 86: KIT11-mutated, 5: wild-type, and 5: other mutations). KIT11-mutated patients were divided into 3 grades based on the KIT11-mutated site and type. Clinical backgrounds and prognostic outcomes were retrospectively compared between the 3 groups. Of 86 KIT11-mutated patients treated with IM, 32 (37.21%) had grade 1 tumors, 37 (43.02%) had grade 2 tumors, and 17 (19.77%) had grade 3 tumors. The 5-year disease-free survival (DFS) was significantly worse in patients with grade 3 KIT11-mutated GISTs (41.96%, p = 0.001) than in those with grade 1 (93%) and grade 2 (70.64%) cases. The multivariable analysis suggested that the KIT11-mutated grading system was an independent risk factor for DFS in patients treated with IM (hazard risk, 2.512; 95% confidence interval, 1.370-4.607; p = 0.003). In conclusion, the KIT11-mutated grading system provides good prognostic stratification for DFS in patients treated with IM. Grade 1 tumors predict a favorable response to IM.
Collapse
Affiliation(s)
- Ling-Jun Song
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080 China.
| | - Hui-Juan Ge
- Department of Pathology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 PR China
| | - Xiao-Qin Shi
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080 China
| | - Wei-Wei Shen
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080 China; Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
135
|
Kelly CM, Gutierrez Sainz L, Chi P. The management of metastatic GIST: current standard and investigational therapeutics. J Hematol Oncol 2021; 14:2. [PMID: 33402214 PMCID: PMC7786896 DOI: 10.1186/s13045-020-01026-6] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal stromal tumor (GIST) is the most common mesenchymal tumor of the gastrointestinal tract. The majority of GISTs harbor gain of function mutations in either KIT or PDGFRα. Determination of the GIST molecular subtype upon diagnosis is important because this information informs therapeutic decisions in both the adjuvant and metastatic setting. The management of GIST was revolutionized by the introduction of imatinib, a KIT inhibitor, which has become the standard first line treatment for metastatic GIST. However, despite a clinical benefit rate of 80%, the majority of patients with GIST experience disease progression after 2-3 years of imatinib therapy. Second and third line options include sunitinib and regorafenib, respectively, and yield low response rates and limited clinical benefit. There have been recent FDA approvals for GIST including ripretinib in the fourth-line setting and avapritinib for PDGFRA exon 18-mutant GIST. This article aims to review the optimal treatment approach for the management of patients with advanced GIST. It examines the standard treatment options available but also explores the novel treatment approaches in the setting of imatinib refractory GIST.
Collapse
Affiliation(s)
- Ciara M. Kelly
- grid.51462.340000 0001 2171 9952Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA ,grid.5386.8000000041936877XDepartment of Medicine, Weill Cornell Medical College, New York, USA
| | - Laura Gutierrez Sainz
- grid.81821.320000 0000 8970 9163Department of Medical Oncology, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Ping Chi
- grid.51462.340000 0001 2171 9952Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA ,grid.5386.8000000041936877XDepartment of Medicine, Weill Cornell Medical College, New York, USA ,grid.51462.340000 0001 2171 9952Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
136
|
Translating Knowledge About the Immune Microenvironment of Gastrointestinal Stromal Tumors into Effective Clinical Strategies. Curr Treat Options Oncol 2021; 22:9. [PMID: 33400014 DOI: 10.1007/s11864-020-00806-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2020] [Indexed: 12/25/2022]
Abstract
OPINION STATEMENT The role of targeted therapy is firmly established for gastrointestinal stromal tumors (GISTs); other modalities for targeting this disease are necessary for recurrent and refractory disease. There are several lines of evidence pointing to an active role of the immune system in GIST. Preclinical and clinical studies revealed that the most common type of immune cell infiltration in GISTs is tumor-associated macrophages (TAMs). The mechanism of how TAMs sculpt the tumor microenvironment in GIST is not clear, but it seems that the presence of immunosuppressive regulatory T cells (Tregs) is correlated with the number of TAMs, thus linking macrophages to immunosuppression. CD3+ T cells and NK infiltrates are found in the GIST microenvironment and carry some prognostic value. In early clinical trials, there is evidence for an active role for immunotherapy in treating GIST patients. Moreover, preclinical evidence has indicated that combining TKIs with checkpoint blockers may be synergistic in murine GIST models. Overall, there is substantial preclinical and clinical evidence to support a role for immunoregulation in GIST and further studies will be important for the development of immunotherapies for GIST.
Collapse
|
137
|
Zhang X, Liang Y, Li Y, Yin J. Comparative Efficacy and Safety of Different Regimens of Advanced Gastrointestinal Stromal Tumors After Failure Prior Tyrosine Kinase Inhibitors: A Network Meta-Analysis. Adv Ther 2021; 38:399-412. [PMID: 33131035 DOI: 10.1007/s12325-020-01545-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/20/2020] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The prospect of targeted therapies for advanced gastrointestinal stromal tumors (GISTs) has been dramatically transformed after encouraging results achieved in recent clinical trials. At present, the number of second- and third-line treatments are increasing, although the challenge is to take into account the differences between these interventions. Therefore, our goal is to evaluate the investigation of different regimens currently used in GISTs based on findings from phase II or phase III randomized controlled trials (RCTs), and then indirectly compare the effectiveness and safety of the available therapies. METHODS The qualified literatures in relevant sources were searched systematically. Studies to identify RCTs of which main endpoints were progression-free survival (PFS), overall survival (OS), and grade 3 or more adverse events (AEs) in patients with GISTs were considered for inclusion. RESULTS Eight RCTs met our inclusion criteria, which involved 2351 patients. For PFS, compared with placebo, imatinib, and sunitinib, regorafenib (HR = 0.12, 95% CI 0.07-0.23; HR = 0.27, 95% CI 0.19-0.39; HR = 0.36, 95% CI 0.19-0.72, respectively) and ripretinib (HR = 0.15, 95% CI 0.09-0.25; HR = 0.33, 95% CI 0.16-0.68; HR = 0.44, 95% CI 0.25-0.78, respectively) were significantly correlated with the improvement of PFS, and regorafenib may be the preferred option according to the analysis of treatment rankings. For OS, compared with placebo, imatinib, and sunitinib, masitinib (HR = 0.13, 95% CI 0.04-0.44; HR = 0.13, 95% CI 0.04-0.51; HR = 0.27, 95%CI 0.09-0.84) and ripretinib (HR = 0.36, 95% CI 0.21-0.62; HR = 0.36, 95% CI 0.16-0.80; HR = 0.18, 95% CI 0.09-0.36, respectively) were significantly more effective, and masitinib may be the best choice according to treatment ranking analysis. Statistically, regorafenib can be considered to be the highest in high-grade AEs, while the rate of severe AEs of ripretinib and masitinib was likely the lowest. CONCLUSION Our results show that ripretinib has the most favorable balance between effectiveness and tolerability among the different treatment regimens for GISTs.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Pharmacy, Kunming Yan'an Hospital, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, 650051, China
| | - Yueqin Liang
- Department of Pharmacy, Kunming Yan'an Hospital, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, 650051, China
| | - Yanhua Li
- Department of Pharmacy, Kunming Yan'an Hospital, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, 650051, China
| | - Jiafu Yin
- Department of Pharmacy, Tumor Hospital of Yunnan Province, Third Affiliated Hospital To Kunming Medical University, Kunming, 650118, China.
| |
Collapse
|
138
|
Steeghs EMP, Gelderblom H, Ho VKY, Voorham QJM, Willems SM, Grünberg K, Ligtenberg MJL. Nationwide evaluation of mutation-tailored treatment of gastrointestinal stromal tumors in daily clinical practice. Gastric Cancer 2021; 24:990-1002. [PMID: 33909171 PMCID: PMC8338807 DOI: 10.1007/s10120-021-01190-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Molecular analysis of KIT and PDGFRA is critical for tyrosine kinase inhibitor treatment selection of gastrointestinal stromal tumors (GISTs) and hence recommended by international guidelines. We performed a nationwide study into the application of predictive mutation testing in GIST patients and its impact on targeted treatment decisions in clinical practice. METHODS Real-world clinical and pathology information was obtained from GIST patients with initial diagnosis in 2017-2018 through database linkage between the Netherlands Cancer Registry and the nationwide Dutch Pathology Registry. RESULTS Predictive mutation analysis was performed in 89% of the patients with high risk or metastatic disease. Molecular testing rates were higher for patients treated in expertise centers (96%) compared to non-expertise centers (75%, P < 0.01). Imatinib therapy was applied in 81% of the patients with high risk or metastatic disease without patient's refusal or adverse characteristics, e.g., comorbidities or resistance mutations. Mutation analysis that was performed in 97% of these imatinib-treated cases, did not guarantee mutation-tailored treatment: 2% of these patients had the PDGFRA p.D842V resistance mutation and 7% initiated imatinib therapy at the normal instead of high dose despite of having a KIT exon 9 mutation. CONCLUSION In conclusion, nationwide real-world data show that over 81% of the eligible high risk or metastatic disease patients receive targeted therapy, which was tailored to the mutation status as recommended in guidelines in 88% of cases. Therefore, still 27% of these GIST patients misses out on mutation-tailored treatment. The reasons for suboptimal uptake of testing and treatment require further study.
Collapse
Affiliation(s)
- Elisabeth M. P. Steeghs
- grid.10417.330000 0004 0444 9382Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans Gelderblom
- grid.10419.3d0000000089452978Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Vincent K. Y. Ho
- Departments of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, The Netherlands
| | | | - Stefan M. Willems
- PALGA Foundation, Houten, The Netherlands ,grid.4494.d0000 0000 9558 4598Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Katrien Grünberg
- grid.10417.330000 0004 0444 9382Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marjolijn J. L. Ligtenberg
- grid.10417.330000 0004 0444 9382Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands ,grid.10417.330000 0004 0444 9382Laboratory of Tumor Genetics, Department of Pathology and Human Genetics, Radboud University Medical Center, Geert Grooteplein Zuid 10, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
139
|
Suresh Babu MC, Chaudhuri T, Babu KG, Lakshmaiah KC, Lokanatha D, Jacob LA, Rudresha AH, Lokesh KN, Rajeev LK. Metastatic gastrointestinal stromal tumor: A regional cancer center experience of 44 cases. South Asian J Cancer 2020; 6:118-121. [PMID: 28975120 PMCID: PMC5615881 DOI: 10.4103/sajc.sajc_290_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasms of the gastrointestinal tract. Historically, a poor prognosis for metastatic disease has been reported with systemic chemotherapy. Significant advances have been made in the last decade, since the introduction of different tyrosine kinase inhibitors (TKIs). Unfortunately, even though the TKIs have been used for a long time, there are very few published data of the experience of TKI therapy in metastatic GIST from India. MATERIALS AND METHODS Patients diagnosed with metastatic GIST from January 2005 to October 2016 at our center, who received first-line therapy with imatinib 400 mg/day, were reviewed retrospectively. Patients' profile, response to treatment, toxicity of TKI therapy, time to progression, and survival were evaluated. RESULTS Of the 44 metastatic GIST patients, 23 (52.2%) were males. Median age at diagnosis was 48 years. The most common presenting symptom was an abdominal pain (52%), followed by weight loss (23%). Most frequently affected metastatic site was liver (57%), followed by peritoneum (16%), and lungs (4.5%). Metastases to both liver and peritoneum were found in 10 patients (22.5%). All patients were initially treated with imatinib at a dose of 400 mg/day. Disease stabilization was documented in 21 cases (48%), and 13 patients (29%) achieved a partial response. TKI therapy was well-tolerated in most cases. Median progression-free survival (PFS) was 26 months, and estimated median survival was 48 months. Patients with lung metastases have a significantly inferior median PFS and overall survival, in comparison to patients with other metastatic sites (P < 0.05). CONCLUSIONS Imatinib therapy was well tolerated and induced a sustained clinical benefit in more than half of the patients with metastatic GIST. Lung metastases seemed to be a poor prognostic factor in this patient population.
Collapse
Affiliation(s)
- M C Suresh Babu
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - Tamojit Chaudhuri
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - K Govind Babu
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - K C Lakshmaiah
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - D Lokanatha
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - Linu Abraham Jacob
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - A H Rudresha
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - K N Lokesh
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - L K Rajeev
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| |
Collapse
|
140
|
Taghizadeh H, Mader RM, Müllauer L, Erhart F, Kautzky-Willer A, Prager GW. Precision Medicine for the Management of Therapy Refractory Colorectal Cancer. J Pers Med 2020; 10:jpm10040272. [PMID: 33322358 PMCID: PMC7768503 DOI: 10.3390/jpm10040272] [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: 10/14/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/30/2022] Open
Abstract
In this analysis, we examined the efficacy, feasibility, and limitations of molecular-based targeted therapies in heavily pretreated metastatic colorectal cancer (mCRC) patients after failure of all standard treatments. In this single-center, real-world retrospective analysis of our platform for precision medicine, we mapped the molecular profiles of 60 mCRC patients. Tumor samples of the patients were analyzed using next-generation sequencing panels of mutation hotspots, microsatellite instability testing, and immunohistochemistry. All profiles were reviewed by a multidisciplinary team to provide a targeted treatment recommendation after consensus discussion. In total, we detected 166 mutations in 53 patients. The five most frequently found mutations were TP53, KRAS, APC, PIK3CA, and PTEN. In 28 cases (47% of all patients), a molecularly targeted therapy could be recommended. Eventually, 12 patients (20%) received the recommended therapy. Six patients (10%) had a clinical benefit. The median time to treatment failure was 3.1 months. Our study demonstrates the feasibility and applicability of using targeted therapies in daily clinical practice for heavily pretreated mCRC patients. This could be used as a targeted treatment option in half of the patients.
Collapse
Affiliation(s)
- Hossein Taghizadeh
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, 1090 Vienna, Austria; (H.T.); (R.M.M.)
- Comprehensive Cancer Center Vienna, 1090 Vienna, Austria;
| | - Robert M. Mader
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, 1090 Vienna, Austria; (H.T.); (R.M.M.)
- Comprehensive Cancer Center Vienna, 1090 Vienna, Austria;
| | - Leonhard Müllauer
- Comprehensive Cancer Center Vienna, 1090 Vienna, Austria;
- Clinical Institute of Pathology, Medical University Vienna, 1090 Vienna, Austria
| | - Friedrich Erhart
- Department of Internal Medicine, Amstetten Region State Clinic, 3300 Amstetten, Austria;
| | - Alexandra Kautzky-Willer
- Department of Medicine III, Gender Medicine Unit, Medical University of Vienna, 1090 Vienna, Austria;
| | - Gerald W. Prager
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, 1090 Vienna, Austria; (H.T.); (R.M.M.)
- Comprehensive Cancer Center Vienna, 1090 Vienna, Austria;
- Correspondence: ; Tel.: +43-1-40400-44500
| |
Collapse
|
141
|
Mohammadi M, Gelderblom H. Systemic therapy of advanced/metastatic gastrointestinal stromal tumors: an update on progress beyond imatinib, sunitinib, and regorafenib. Expert Opin Investig Drugs 2020; 30:143-152. [PMID: 33252274 DOI: 10.1080/13543784.2021.1857363] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Introduction: Discovery of oncogenic mutations in the KIT and PDGFRA tyrosine kinase receptor was a crucial step for the development of tyrosine kinase inhibitors (TKIs). Since then, GIST became a model for the development of molecular-targeted therapy, which led to dramatically improved median overall survival of advanced GIST. Still, further progress is needed after third-line or for TKI resistant mutations. Areas covered: In this review, after a brief introduction on imatinib, sunitinib, and regorafenib, an overview of TKIs that was evaluated beyond these drugs is provided, with a main focus on the novel approved TKIs. Expert opinion: Combination therapies have thus far not fulfilled their promise in GIST, nor did immunotherapy. Increased understanding of GIST and advances in the development of molecular-targeted drugs led to the introduction of ripretinib and avapritinib. Furthermore, NTRK inhibitors became available for ultrarare NTRK fusions. Solutions for NF1 and BRAF mutated and SDH-deficient GIST are still to be awaited. This all underlines the need for adequate molecular profiling of high-risk GISTs before treatment is started. Possibly by using circulating tumor DNA in the future, targeting resistance mutations with specific drugs along the course of the disease would be easier, avoiding multiple tumor biopsies.
Collapse
Affiliation(s)
- Mahmoud Mohammadi
- Department of Medical Oncology, Leiden University Medical Center , Leiden, The Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center , Leiden, The Netherlands
| |
Collapse
|
142
|
Zhou G, Xiao K, Gong G, Wu J, Zhang Y, Liu X, Jiang Z, Ma C. A novel nomogram for predicting liver metastasis in patients with gastrointestinal stromal tumor: a SEER-based study. BMC Surg 2020; 20:298. [PMID: 33238982 PMCID: PMC7689971 DOI: 10.1186/s12893-020-00969-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Background Liver metastasis (LIM) of gastrointestinal stromal tumor (GIST) is associated with poor prognosis. The present study aimed at developing and validating nomogram to predict LIM in patients with GIST, thus helping clinical diagnosis and treatment. Methods The data of GIST patients derived from Surveillance, Epidemiology, and End Results (SEER) database from 2010 to 2016, which were then screened by univariate and multivariate logistic regression for the construction of LIM nomogram. The model discrimination of LIM nomogram was evaluated by concordance index (C-index) and calibration plots, while the predictive accuracy and clinical values were measured by decision curve analysis (DCA) and clinical impact plot. Furthermore, we validated predictive nomogram in the internal testing set. Results A total of 3797 patients were enrolled and divided randomly into training and validating groups in a 3-to-1 ratio. After logistic regression, the significant variables were sex, tumor location, tumor size, N stage and mitotic rate. The calibration curves showed the perfect agreement between nomogram predictions and actual observations, while the DCA and clinical impact plot showed the clinical utility of LIM nomogram. C-index of the nomogram was 0.812. What’s more, receiver operating characteristic curves (ROC) also showed good discrimination and calibration in the training set (AUC = 0.794, 95% CI 0.778–0.808) and the testing set (AUC = 0.775, 95% CI 0.748–0.802). Conclusion The nomogram for patients with GIST can effectively predict the individualized risk of liver metastasis and provide insightful information to clinicians to optimize therapeutic regimens.
Collapse
Affiliation(s)
- Guowei Zhou
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Keshuai Xiao
- Department of General Surgery, Xinyang Central Hospital, Xin Yang, 464000, Henan Province, China
| | - Guanwen Gong
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| | - Jiabao Wu
- Department of Pediatrics, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Ya Zhang
- Department of Gynecology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Xinxin Liu
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Zhiwei Jiang
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Chaoqun Ma
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
143
|
Taghizadeh H, Mader RM, Müllauer L, Fuereder T, Kautzky-Willer A, Prager GW. Outcome of Targeted Therapy Recommendations for Metastatic and Recurrent Head and Neck Cancers. Cancers (Basel) 2020; 12:cancers12113381. [PMID: 33203166 PMCID: PMC7696688 DOI: 10.3390/cancers12113381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/09/2020] [Accepted: 11/13/2020] [Indexed: 01/03/2023] Open
Abstract
Recurrent/metastatic (R/M) head and neck cancers bear a poor prognosis. In this analysis, we examined the efficacy and the outcome of targeted therapy recommendations based on the patients' molecular tumor portrait after failure of all standard therapy options. In this single-center, real-world retrospective analysis of our platform for precision medicine, we analyzed the molecular profile of 50 patients diagnosed with R/M head and neck cancer. Tumor samples of the patients were examined using next-generation sequencing panels of mutation hotspots, microsatellite instability (MSI) testing, and immunohistochemistry (IHC). In 31 cases (62.0% of all patients), a molecular-driven targeted therapy approach was recommended. Eventually, 14 patients (28%) received the suggested targeted therapy. Six of fourteen patients (43%) achieved stable disease conditions and four patients (29%) experienced a progressive disease. The median time to treatment failure was 2.8 months. Therapy recommendations were significantly more often issued for men (p = 0.037) than for women. This analysis demonstrated that precision medicine provided the basis for molecular-driven therapy recommendations in over half of the patients with advanced therapy refractory head and neck cancers, with significantly more therapy recommendations for men. Our analysis showed that although precision medicine approaches are implementable and feasible for the management of recurrent/metastatic head and neck cancers in daily clinical routine, there are major limitations and challenges that have to be overcome.
Collapse
Affiliation(s)
- Hossein Taghizadeh
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, 1090 Vienna, Austria; (H.T.); (R.M.M.); (T.F.)
- Comprehensive Cancer Center Vienna, 1090 Vienna, Austria;
| | - Robert M. Mader
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, 1090 Vienna, Austria; (H.T.); (R.M.M.); (T.F.)
- Comprehensive Cancer Center Vienna, 1090 Vienna, Austria;
| | - Leonhard Müllauer
- Comprehensive Cancer Center Vienna, 1090 Vienna, Austria;
- Clinical Institute of Pathology, Medical University Vienna, 1090 Vienna, Austria
| | - Thorsten Fuereder
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, 1090 Vienna, Austria; (H.T.); (R.M.M.); (T.F.)
- Comprehensive Cancer Center Vienna, 1090 Vienna, Austria;
| | - Alexandra Kautzky-Willer
- Department of Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, 1090 Vienna, Austria;
- Department of Medicine III, Gender Medicine Unit, Medical University of Vienna, 1090 Vienna, Austria
| | - Gerald W. Prager
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, 1090 Vienna, Austria; (H.T.); (R.M.M.); (T.F.)
- Comprehensive Cancer Center Vienna, 1090 Vienna, Austria;
- Correspondence: ; Tel.: +43-1-40400-44500
| |
Collapse
|
144
|
Esteban-Villarrubia J, Soto-Castillo JJ, Pozas J, San Román-Gil M, Orejana-Martín I, Torres-Jiménez J, Carrato A, Alonso-Gordoa T, Molina-Cerrillo J. Tyrosine Kinase Receptors in Oncology. Int J Mol Sci 2020; 21:E8529. [PMID: 33198314 PMCID: PMC7696731 DOI: 10.3390/ijms21228529] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/07/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
Tyrosine kinase receptors (TKR) comprise more than 60 molecules that play an essential role in the molecular pathways, leading to cell survival and differentiation. Consequently, genetic alterations of TKRs may lead to tumorigenesis and, therefore, cancer development. The discovery and improvement of tyrosine kinase inhibitors (TKI) against TKRs have entailed an important step in the knowledge-expansion of tumor physiopathology as well as an improvement in the cancer treatment based on molecular alterations over many tumor types. The purpose of this review is to provide a comprehensive review of the different families of TKRs and their role in the expansion of tumor cells and how TKIs can stop these pathways to tumorigenesis, in combination or not with other therapies. The increasing growth of this landscape is driving us to strengthen the development of precision oncology with clinical trials based on molecular-based therapy over a histology-based one, with promising preliminary results.
Collapse
Affiliation(s)
- Jorge Esteban-Villarrubia
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (J.J.S.-C.); (J.P.); (M.S.R.-G.); (I.O.-M.); (J.T.-J.)
| | - Juan José Soto-Castillo
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (J.J.S.-C.); (J.P.); (M.S.R.-G.); (I.O.-M.); (J.T.-J.)
| | - Javier Pozas
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (J.J.S.-C.); (J.P.); (M.S.R.-G.); (I.O.-M.); (J.T.-J.)
| | - María San Román-Gil
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (J.J.S.-C.); (J.P.); (M.S.R.-G.); (I.O.-M.); (J.T.-J.)
| | - Inmaculada Orejana-Martín
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (J.J.S.-C.); (J.P.); (M.S.R.-G.); (I.O.-M.); (J.T.-J.)
| | - Javier Torres-Jiménez
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (J.J.S.-C.); (J.P.); (M.S.R.-G.); (I.O.-M.); (J.T.-J.)
| | - Alfredo Carrato
- Medical Oncology Department, Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, Alcalá University, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (A.C.); (J.M.-C.)
| | - Teresa Alonso-Gordoa
- Medical Oncology Department, Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, Alcalá University, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (A.C.); (J.M.-C.)
| | - Javier Molina-Cerrillo
- Medical Oncology Department, Ramón y Cajal Health Research Institute (IRYCIS), CIBERONC, Alcalá University, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (A.C.); (J.M.-C.)
| |
Collapse
|
145
|
Riedel RF, Ballman KV, Lu Y, Attia S, Loggers ET, Ganjoo KN, Livingston MB, Chow W, Wright J, Ward JH, Rushing D, Okuno SH, Reed DR, Liebner DA, Keedy VL, Mascarenhas L, Davis LE, Ryan C, Reinke DK, Maki RG. A Randomized, Double-Blind, Placebo-Controlled, Phase II Study of Regorafenib Versus Placebo in Advanced/Metastatic, Treatment-Refractory Liposarcoma: Results from the SARC024 Study. Oncologist 2020; 25:e1655-e1662. [PMID: 32701199 PMCID: PMC7648334 DOI: 10.1634/theoncologist.2020-0679] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 07/09/2020] [Indexed: 01/11/2023] Open
Abstract
LESSONS LEARNED The results from the liposarcoma cohort of SARC024 confirm previously published data and do not support the routine use of regorafenib in this patient population. Continued exploration of novel therapies, including combination approaches, is warranted for a patient population in whom limited treatment options exist. BACKGROUND Regorafenib is a multitargeted kinase inhibitor with a kinase profile overlapping, but distinct from, pazopanib, an agent approved for recurrent and metastatic non-gastrointestinal stromal tumor (GIST), non-adipocytic soft tissue sarcoma. We conducted a randomized, phase II study of regorafenib versus placebo in refractory liposarcoma patients. METHODS Patients with advanced or metastatic, treatment-refractory liposarcoma were randomized 1:1 to receive regorafenib 160 mg or placebo once daily (3 weeks on, 1 week off). Patients with well-differentiated liposarcoma only were excluded. Crossover for placebo was allowed upon progression. The primary endpoint was progression-free survival (PFS), according to RECIST version 1.1. RESULTS Forty-eight subjects with liposarcoma (34 dedifferentiated, 12 myxoid/round cell, 2 pleomorphic) were enrolled. Median PFS was 1.87 (95% confidence interval [CI], 0.92-3.67) months for regorafenib versus 2.07 (95% CI, 1.64-3.44) months for placebo; stratified hazard ratio [HR], 0.85 (95% CI, 0.46, 1.58), p = .62. No responses were seen on regorafenib. One PR was observed on placebo. Median overall survival was 6.46 (95% CI, 4.16-23.48) months for regorafenib and 4.89 (95% CI, 3.02-9.77) months for placebo, stratified HR, 0.66 (95% CI, 0.31-1.40), p = .28). Treatment-related adverse events were similar to the known safety profile of regorafenib. CONCLUSION Regorafenib did not appear to improve PFS in treatment-refractory liposarcoma. No new significant safety signals were observed.
Collapse
Affiliation(s)
- Richard F. Riedel
- Duke Cancer Institute, Duke University Medical CenterDurhamNorth CarolinaUSA
| | | | - Yao Lu
- Weill Cornell MedicineNew YorkNew YorkUSA
| | | | | | | | | | - Warren Chow
- City of Hope Cancer CenterDuarteCaliforniaUSA
| | | | - John H. Ward
- Huntsman Cancer Institute, University of UtahSalt Lake CityUtahUSA
| | - Daniel Rushing
- Melvin and Bren Simon Cancer Center, Indiana UniversityIndianapolisIndianaUSA
| | | | - Damon R. Reed
- H. Lee Moffitt Cancer Center and Research InstituteTampaFloridaUSA
| | | | - Vicki L. Keedy
- Vanderbilt‐Ingram Cancer Center, Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Leo Mascarenhas
- Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Lara E. Davis
- Knight Cancer Institute, Oregon Health & Science UniversityPortlandOregonUSA
| | - Christopher Ryan
- Knight Cancer Institute, Oregon Health & Science UniversityPortlandOregonUSA
| | - Denise K. Reinke
- Sarcoma Alliance for Research through Collaboration (SARC)Ann ArborMichiganUSA
| | - Robert G. Maki
- Abramson Cancer Center, University of Pennsylvania School of MedicinePhiladelphiaPennsylvaniaUSA
| |
Collapse
|
146
|
Development of synthetic lethality in cancer: molecular and cellular classification. Signal Transduct Target Ther 2020; 5:241. [PMID: 33077733 PMCID: PMC7573576 DOI: 10.1038/s41392-020-00358-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/27/2022] Open
Abstract
Recently, genetically targeted cancer therapies have been a topic of great interest. Synthetic lethality provides a new approach for the treatment of mutated genes that were previously considered unable to be targeted in traditional genotype-targeted treatments. The increasing researches and applications in the clinical setting made synthetic lethality a promising anticancer treatment option. However, the current understandings on different conditions of synthetic lethality have not been systematically assessed and the application of synthetic lethality in clinical practice still faces many challenges. Here, we propose a novel and systematic classification of synthetic lethality divided into gene level, pathway level, organelle level, and conditional synthetic lethality, according to the degree of specificity into its biological mechanism. Multiple preclinical findings of synthetic lethality in recent years will be reviewed and classified under these different categories. Moreover, synthetic lethality targeted drugs in clinical practice will be briefly discussed. Finally, we will explore the essential implications of this classification as well as its prospects in eliminating existing challenges and the future directions of synthetic lethality.
Collapse
|
147
|
The V654A second-site KIT mutation increases tumor oncogenesis and STAT activation in a mouse model of gastrointestinal stromal tumor. Oncogene 2020; 39:7153-7165. [PMID: 33024275 PMCID: PMC7718339 DOI: 10.1038/s41388-020-01489-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/19/2020] [Accepted: 09/24/2020] [Indexed: 01/02/2023]
Abstract
Gastrointestinal stromal tumor (GIST) is the most common human sarcoma and arises in the gastrointestinal tract. Most GISTs are caused by activating mutations in the KIT receptor tyrosine kinase, such as the exon 11 KIT V559Δ mutation. The small molecule imatinib inhibits KIT and has been a mainstay of therapy in GIST. Unfortunately, imatinib-treated patients typically relapse, most often due to clonal emergence of the resistance-associated KIT V654A mutation. To determine the biologic impact of this second-site mutation in vivo, we created a mouse model with the corresponding V558Δ;V653A Kit double mutation restricted (a) spatially to ETV1+ cells, which include the interstitial cells of Cajal (ICCs) from which GISTs presumably originate, and (b) temporally through tamoxifen treatment after birth. This resulted in the first in vivo model of the most common second-site mutation associated with imatinib resistance in GIST and the first in vivo demonstration that cell-autonomous expression of mutant KIT in the ICC lineage leads to GIST. GISTs driven by the V558Δ;V653A Kit double mutation were resistant to imatinib, while cabozantinib was more effective in overcoming resistance than sunitinib. Compared to control mice with a single V558Δ Kit mutation, mice with a double V558Δ; V653A Kit mutation had increased tumor oncogenesis and associated KIT-dependent STAT activation. Our findings demonstrate that the biologic consequences of a second-site mutation in an oncogenic driver may include not only a mechanism for drug resistance, but changes in tumor oncogenic potential and differential activation of signaling pathways.
Collapse
|
148
|
Liu P, Tan F, Liu H, Ge J, Liu S, Lei T, Zhao X. Skin Metastasis of Gastrointestinal Stromal Tumors: A Case Series and Literature Review. Cancer Manag Res 2020; 12:7681-7690. [PMID: 32904396 PMCID: PMC7455533 DOI: 10.2147/cmar.s261823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/04/2020] [Indexed: 12/12/2022] Open
Abstract
Background Gastrointestinal stromal tumors (GISTs) extremely and rarely metastasize to the skin, and such metastases have not been well characterized. Methods Retrospective analysis of clinicopathological data of patients with skin metastasis of a GIST (SM-GIST) admitted to Xiangya Hospital (Changsha, Hunan, China) and literature review were conducted. Results Including our 4 cases, a total of 17 cases have been reported to date. The mean age of the patients was 55.4 years (29~70 years) and there was not sex predominance (male 10 and female 7). Primary tumors were often located in the stomach (n=9), duodenum (n=2) and small bowel (n=2). Meanwhile, SM-GIST mainly occurred in head and face (n=6), extremities (n=6), followed by abdomen wall (n=5), back (n=3) and chest (n=2). Mutation analysis revealed that the frequency of wild-type GIST (WT-GIST), exon 9, 11 and 13 mutations was 6, 1, 4 and 1, respectively. The average time to SM-GIST was 4.22 years, specifically 4.59 years in gastric and 3.8 years in non-gastric. Moreover, for the resection only group (including chemotherapy), such average time was 3.63 years, while for the combined group (resection and tyrosine kinase inhibitors (TKIs)), it was about 4.74 years. The mean survival was approximately 6.2 years. However, after the diagnosis of SM-GIST, survival was only about 1.69 years. Conclusion SM-GIST is a rare malignant condition. Non-gastric GIST, surgery without TKIs, high invasiveness and tumor burden, and molecular subtype (mutation in exon 9, 11 and wild-type) may be conducive to the development of SM-GIST. Additionally, it is also a sign of poor prognosis.
Collapse
Affiliation(s)
- Peng Liu
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, People's Republic of China
| | - Fengbo Tan
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, People's Republic of China
| | - Heli Liu
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, People's Republic of China
| | - Jie Ge
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, People's Republic of China
| | - Sheng Liu
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, People's Republic of China
| | - Tianxiang Lei
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, People's Republic of China
| | - Xianhui Zhao
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, People's Republic of China
| |
Collapse
|
149
|
Feasibility and first reports of the MATCH-R repeated biopsy trial at Gustave Roussy. NPJ Precis Oncol 2020; 4:27. [PMID: 32964129 PMCID: PMC7478969 DOI: 10.1038/s41698-020-00130-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 07/24/2020] [Indexed: 12/17/2022] Open
Abstract
Unravelling the biological processes driving tumour resistance is necessary to support the development of innovative treatment strategies. We report the design and feasibility of the MATCH-R prospective trial led by Gustave Roussy with the primary objective of characterizing the molecular mechanisms of resistance to cancer treatments. The primary clinical endpoints consist of analyzing the type and frequency of molecular alterations in resistant tumours and compare these to samples prior to treatment. Patients experiencing disease progression after an initial partial response or stable disease for at least 24 weeks underwent a tumour biopsy guided by CT or ultrasound. Molecular profiling of tumours was performed using whole exome sequencing, RNA sequencing and panel sequencing. At data cut-off for feasibility analysis, out of 333 inclusions, tumour biopsies were obtained in 303 cases (91%). From these biopsies, 278 (83%) had sufficient quality for analysis by high-throughput next generation sequencing (NGS). All 278 samples underwent targeted NGS, 215 (70.9%) RNA sequencing and 222 (73.2%) whole exome sequencing. In total, 163 tumours were implanted in NOD scid gamma (NSG) or nude mice and 54 patient-derived xenograft (PDX) models were established, with a success rate of 33%. Adverse events secondary to invasive tumour sampling occurred in 24 patients (7.6%). Study recruitment is still ongoing. Systematic molecular profiling of tumours and the development of patient-derived models of acquired resistance to targeted agents and immunotherapy is feasible and can drive the selection of the next therapeutic strategy.
Collapse
|
150
|
Li HL, Wang LH, Hu YL, Feng Y, Li XH, Liu YF, Li P, Mao QS, Xue WJ. Clinical and prognostic significance of CC chemokine receptor type 8 protein expression in gastrointestinal stromal tumors. World J Gastroenterol 2020; 26:4656-4668. [PMID: 32884223 PMCID: PMC7445867 DOI: 10.3748/wjg.v26.i31.4656] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/07/2020] [Accepted: 07/18/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasms of the gastrointestinal tract. Surgical resection and tyrosine kinase inhibitors are defined as the main treatments but cannot cure patients with advanced GIST, which eventually develops into recurrence and acquired drug resistance. Therefore, it is necessary to identify prognostic biomarkers and new therapeutic targets for GISTs. CC chemokine receptor type 8 (CCR8) protein participates in regulation of immune responses. Recent studies on CCR8 in non-small cell lung cancer and colorectal cancer showed that it was highly expressed in tumor-infiltrating regulatory T cells and correlated with a poor prognosis.
AIM To detect CCR8 expression in GIST tissues and analyze its relationships with clinicopathological features and prognosis in patients with GISTs.
METHODS Tissue samples were used for the tissue microarrays construction. The microarrays were then subjected to immunohistochemical analyses to detect CCR8 expression. Next, Kaplan–Meier analysis was utilized to calculate the survival rate of patients with complete follow-up data, and the potential prognostic value of CCR8 was evaluated by Cox regression analysis. Finally, a Gene Ontology/Kyoto Encyclopedia of Genes and Genomes single-gene enrichment chart of CCR8 was constructed using the STRING database.
RESULTS CCR8-positive signals were detected as brown or brown-yellow particles by immunohistochemistry located in the cytoplasm. Among 125 tissue samples, 74 had CCR8 high expression and 51 had low or negative expression. Statistical analyses suggested CCR8 was significantly correlated with tumor size, mitotic index, AFIP-Miettinen risk classification and tumor location. Kaplan–Meier and multivariate analyses showed that patients with low or negative CCR8 expression, mitotic index < 5/high-power fields (HPF) and tumor diameter < 5 cm had a better prognosis. Based on the STRING database, CCR8 was significantly enriched in biological processes such as tumor immunity, T lymphocyte chemotaxis, migration and pathways like the nuclear factor-κB and tumor necrosis factor pathways as well as intestinal immune regulation networks.
CONCLUSION CCR8 is a prognostic biomarker for malignant potential of GISTs, with high expression correlated with malignancy and poor prognosis.
Collapse
Affiliation(s)
- Huai-Liang Li
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu Province, China
| | - Lin-Hua Wang
- Department of Intensive Care Unit, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu Province, China
| | - Yi-Lin Hu
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu Province, China
| | - Ying Feng
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu Province, China
| | - Xiao-Hong Li
- Department of Surgical Comprehensive Laboratory, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu Province, China
| | - Yi-Fei Liu
- Department of Pathology, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu Province, China
| | - Peng Li
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu Province, China
| | - Qin-Sheng Mao
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu Province, China
| | - Wan-Jiang Xue
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong 226001, Jiangsu Province, China
| |
Collapse
|