1
|
Bleckman RF, Haag CMSC, Rifaela N, Beukema G, Mathijssen RHJ, Steeghs N, Gelderblom H, Desar IME, Cleven A, ter Elst A, Schuuring E, Reyners AKL. Levels of circulating tumor DNA correlate with tumor volume in gastro-intestinal stromal tumors: an exploratory long-term follow-up study. Mol Oncol 2024; 18:2658-2667. [PMID: 38790141 PMCID: PMC11547224 DOI: 10.1002/1878-0261.13644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/26/2024] [Accepted: 03/15/2024] [Indexed: 05/26/2024] Open
Abstract
Patients with gastro-intestinal stromal tumors (GISTs) undergoing tyrosine kinase inhibitor therapy are monitored with regular computed tomography (CT) scans, exposing patients to cumulative radiation. This exploratory study aimed to evaluate circulating tumor DNA (ctDNA) testing to monitor treatment response and compare changes in ctDNA levels with RECIST 1.1 and total tumor volume measurements. Between 2014 and 2021, six patients with KIT proto-oncogene, receptor tyrosine kinase (KIT) exon-11-mutated GIST from whom long-term plasma samples were collected prospectively were included in the study. ctDNA levels of relevant plasma samples were determined using the KIT exon 11 digital droplet PCR drop-off assay. Tumor volume measurements were performed using a semi-automated approach. In total, 94 of 130 clinically relevant ctDNA samples were analyzed. Upon successful treatment response, ctDNA became undetectable in all patients. At progressive disease, ctDNA was detectable in five out of six patients. Higher levels of ctDNA correlated with larger tumor volumes. Undetectable ctDNA at the time of progressive disease on imaging was consistent with lower tumor volumes compared to those with detectable ctDNA. In summary, ctDNA levels seem to correlate with total tumor volume at the time of progressive disease. Our exploratory study shows promise for including ctDNA testing in treatment follow-up.
Collapse
Affiliation(s)
- Roos F. Bleckman
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Charlotte M. S. C. Haag
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Naomi Rifaela
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Gerrieke Beukema
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Ron H. J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer InstituteErasmus University Medical CenterRotterdamThe Netherlands
| | - Neeltje Steeghs
- Department of Medical OncologyThe Netherlands Cancer Institute Antoni van LeeuwenhoekAmsterdamThe Netherlands
| | - Hans Gelderblom
- Department of Medical OncologyLeiden University Medical CenterThe Netherlands
| | - Ingrid M. E. Desar
- Department of Medical OncologyRadboud University Medical CenterNijmegenThe Netherlands
| | - Arjen Cleven
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
- Department of Medical OncologyLeiden University Medical CenterThe Netherlands
| | - Arja ter Elst
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Ed Schuuring
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Anna K. L. Reyners
- Department of Medical Oncology and Pathology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| |
Collapse
|
2
|
Heinrich MC, Jones RL, George S, Gelderblom H, Schöffski P, von Mehren M, Zalcberg JR, Kang YK, Razak AA, Trent J, Attia S, Le Cesne A, Siontis BL, Goldstein D, Boye K, Sanchez C, Steeghs N, Rutkowski P, Druta M, Serrano C, Somaiah N, Chi P, Reichmann W, Sprott K, Achour H, Sherman ML, Ruiz-Soto R, Blay JY, Bauer S. Ripretinib versus sunitinib in gastrointestinal stromal tumor: ctDNA biomarker analysis of the phase 3 INTRIGUE trial. Nat Med 2024; 30:498-506. [PMID: 38182785 PMCID: PMC10878977 DOI: 10.1038/s41591-023-02734-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 11/22/2023] [Indexed: 01/07/2024]
Abstract
INTRIGUE was an open-label, phase 3 study in adult patients with advanced gastrointestinal stromal tumor who had disease progression on or intolerance to imatinib and who were randomized to once-daily ripretinib 150 mg or sunitinib 50 mg. In the primary analysis, progression-free survival (PFS) with ripretinib was not superior to sunitinib. In clinical and nonclinical studies, ripretinib and sunitinib have demonstrated differential activity based on the exon location of KIT mutations. Therefore, we hypothesized that mutational analysis using circulating tumor DNA (ctDNA) might provide further insight. In this exploratory analysis (N = 362), baseline peripheral whole blood was analyzed by a 74-gene ctDNA next-generation sequencing-based assay. ctDNA was detected in 280/362 (77%) samples with KIT mutations in 213/362 patients (59%). Imatinib-resistant mutations were found in the KIT ATP-binding pocket (exons 13/14) and activation loop (exons 17/18). Mutational subgroup assessment showed 2 mutually exclusive populations with differential treatment effects. Patients with only KIT exon 11 + 13/14 mutations (ripretinib, n = 21; sunitinib, n = 20) had better PFS with sunitinib versus ripretinib (median, 15.0 versus 4.0 months). Patients with only KIT exon 11 + 17/18 mutations (ripretinib, n = 27; sunitinib, n = 25) had better PFS with ripretinib versus sunitinib (median, 14.2 versus 1.5 months). The results of this exploratory analysis suggest ctDNA sequencing may improve the prediction of the efficacy of single-drug therapies and support further evaluation of ripretinib in patients with KIT exon 11 + 17/18 mutations. ClinicalTrials.gov identifier: NCT03673501.
Collapse
Affiliation(s)
- Michael C Heinrich
- Division of Hematology/Oncology, Portland VA Health Care System, Portland, OR, USA
- Department of Medicine, OHSU Knight Cancer Institute, Portland, OR, USA
| | - Robin L Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | - Suzanne George
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Patrick Schöffski
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Margaret von Mehren
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, USA
| | - John R Zalcberg
- Department of Medical Oncology, Monash University School of Public Health and Preventive Medicine, Alfred Health, Melbourne, Victoria, Australia
| | - Yoon-Koo Kang
- Department of Oncology, Asan Medical Center, University of Ulsan, Seoul, Korea
| | - Albiruni Abdul Razak
- Division of Medical Oncology, Toronto Sarcoma Program, Princess Margaret Cancer Center, Toronto, ON, Canada
| | - Jonathan Trent
- Department of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL, USA
| | - Steven Attia
- Department of Medical Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Axel Le Cesne
- Medical Oncology Department, Gustave Roussy, Villejuif, France
| | | | - David Goldstein
- Department of Medical Oncology, Prince of Wales Hospital, Randwick, New South Wales, Australia
| | - Kjetil Boye
- Department of Tumor Biology, Oslo University Hospital, Oslo, Norway
| | - Cesar Sanchez
- Department of Hematology-Oncology, Centro de Cáncer, Hospital Clínico Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warszawa, Poland
| | - Mihaela Druta
- Sarcoma Program, Moffitt Cancer Center, Tampa, FL, USA
| | - César Serrano
- Sarcoma Translational Research Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ping Chi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Kam Sprott
- Biometrics, Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
- Translational Medicine, Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
| | - Haroun Achour
- Biometrics, Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
- Clinical Development, Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
| | - Matthew L Sherman
- Clinical Development, Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
| | - Rodrigo Ruiz-Soto
- Clinical Development, Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Sebastian Bauer
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.
| |
Collapse
|
3
|
Ko TK, Lee E, Ng CCY, Yang VS, Farid M, Teh BT, Chan JY, Somasundaram N. Circulating Tumor DNA Mutations in Progressive Gastrointestinal Stromal Tumors Identify Biomarkers of Treatment Resistance and Uncover Potential Therapeutic Strategies. Front Oncol 2022; 12:840843. [PMID: 35273917 PMCID: PMC8904145 DOI: 10.3389/fonc.2022.840843] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/28/2022] [Indexed: 12/27/2022] Open
Abstract
Liquid biopsy circulating tumor DNA (ctDNA)-based approaches may represent a non-invasive means for molecular interrogation of gastrointestinal stromal tumors (GISTs). We deployed a customized 29-gene Archer® LiquidPlex™ targeted panel on 64 plasma samples from 46 patients. The majority were known to harbor KIT mutations (n = 41, 89.1%), while 3 were PDGFRA exon 18 D842V mutants and the rest (n = 2) were wild type for KIT and PDGFRA. In terms of disease stage, 14 (30.4%) were localized GISTs that had undergone complete surgical resection while the rest (n = 32) were metastatic. Among ten patients, including 7 on tyrosine kinase inhibitors, with evidence of disease progression at study inclusion, mutations in ctDNA were detected in 7 cases (70%). Known somatic mutations in KIT (n = 5) or PDGFRA (n = 1) in ctDNA were identified only among 6 of the 10 patients. These KIT mutants included duplication, indels, and single-nucleotide variants. The median mutant AF in ctDNA was 11.0% (range, 0.38%–45.0%). In patients with metastatic progressive KIT-mutant GIST, tumor burden was higher with detectable KIT ctDNA mutation than in those without (median, 5.97 cm vs. 2.40 cm, p = 0.0195). None of the known tumor mutations were detected in ctDNA for localized cases (n = 14) or metastatic cases without evidence of disease progression (n = 22). In patients with serial samples along progression of disease, secondary acquired mutations, including a potentially actionable PIK3CA exon 9 c.1633G>A mutation, were detected. ctDNA mutations were not detectable when patients responded to a switch in TKI therapy. In conclusion, detection of GIST-related mutations in ctDNA using a customized targeted NGS panel represents an attractive non-invasive means to obtain clinically tractable information at the time of disease progression.
Collapse
Affiliation(s)
- Tun Kiat Ko
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore.,Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Elizabeth Lee
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore.,Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Cedric Chuan-Young Ng
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore.,Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore
| | - Valerie Shiwen Yang
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore.,Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Mohamad Farid
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Bin Tean Teh
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore, Singapore.,Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore.,Institute of Molecular and Cell Biology, Singapore, Singapore.,Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Jason Yongsheng Chan
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore.,Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Nagavalli Somasundaram
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
4
|
Moati E, Taly V, Garinet S, Didelot A, Taieb J, Laurent-Puig P, Zaanan A. Role of Circulating Tumor DNA in Gastrointestinal Cancers: Current Knowledge and Perspectives. Cancers (Basel) 2021; 13:4743. [PMID: 34638228 PMCID: PMC8507552 DOI: 10.3390/cancers13194743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/06/2021] [Accepted: 09/18/2021] [Indexed: 12/21/2022] Open
Abstract
Gastrointestinal (GI) cancers are major health burdens worldwide and biomarkers are needed to improve the management of these diseases along their evolution. Circulating tumor DNA (ctDNA) is a promising non-invasive blood and other bodily-fluid-based biomarker in cancer management that can help clinicians in various cases for the detection, diagnosis, prognosis, monitoring and personalization of treatment in digestive oncology. In addition to the well-studied prognostic role of ctDNA, the main real-world applications appear to be the assessment of minimal residual disease to further guide adjuvant therapy and predict relapse, but also the monitoring of clonal evolution to tailor treatments in metastatic setting. Other challenges such as predicting response to treatment including immune checkpoint inhibitors could also be among the potential applications of ctDNA. Although the level of advancement of ctDNA development in the different tumor localizations is still inhomogeneous, it might be now reliable enough to be soon used in clinical routine for colorectal cancers and shows promising results in other GI cancers.
Collapse
Affiliation(s)
- Emilie Moati
- Department of Gastroenterology and Digestive Oncology, Institut du Cancer Paris Carpem, Assistance Publique des Hôpitaux de Paris, European Georges Pompidou Hospital, 75015 Paris, France; (E.M.); (J.T.)
| | - Valerie Taly
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Centre National de la Recherche Scientifique, Sorbonne Université, USPC, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 75006 Paris, France; (V.T.); (S.G.); (A.D.); (P.L.-P.)
| | - Simon Garinet
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Centre National de la Recherche Scientifique, Sorbonne Université, USPC, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 75006 Paris, France; (V.T.); (S.G.); (A.D.); (P.L.-P.)
- Department of Biochemistry, Institut du Cancer Paris Carpem, Assistance Publique des Hôpitaux de Paris, European Georges Pompidou Hospital, 75015 Paris, France
| | - Audrey Didelot
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Centre National de la Recherche Scientifique, Sorbonne Université, USPC, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 75006 Paris, France; (V.T.); (S.G.); (A.D.); (P.L.-P.)
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, Institut du Cancer Paris Carpem, Assistance Publique des Hôpitaux de Paris, European Georges Pompidou Hospital, 75015 Paris, France; (E.M.); (J.T.)
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Centre National de la Recherche Scientifique, Sorbonne Université, USPC, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 75006 Paris, France; (V.T.); (S.G.); (A.D.); (P.L.-P.)
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Centre National de la Recherche Scientifique, Sorbonne Université, USPC, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 75006 Paris, France; (V.T.); (S.G.); (A.D.); (P.L.-P.)
- Department of Biochemistry, Institut du Cancer Paris Carpem, Assistance Publique des Hôpitaux de Paris, European Georges Pompidou Hospital, 75015 Paris, France
| | - Aziz Zaanan
- Department of Gastroenterology and Digestive Oncology, Institut du Cancer Paris Carpem, Assistance Publique des Hôpitaux de Paris, European Georges Pompidou Hospital, 75015 Paris, France; (E.M.); (J.T.)
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Centre National de la Recherche Scientifique, Sorbonne Université, USPC, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 75006 Paris, France; (V.T.); (S.G.); (A.D.); (P.L.-P.)
| |
Collapse
|
5
|
Gómez-Peregrina D, García-Valverde A, Pilco-Janeta D, Serrano C. Liquid Biopsy in Gastrointestinal Stromal Tumors: Ready for Prime Time? Curr Treat Options Oncol 2021; 22:32. [PMID: 33641024 DOI: 10.1007/s11864-021-00832-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
OPINION STATEMENT Gastrointestinal stromal tumor (GIST) constitutes a paradigm for clinically effective targeted inhibition of oncogenic driver mutations. Therefore, GIST has emerged as a compelling clinical and biological model to study oncogene addiction and to validate preclinical concepts for drug response and drug resistance. Oncogenic activation of KIT or PDGFRA receptor tyrosine kinases is the essential drivers of GIST progression throughout all stages of the disease. Interestingly, KIT/PDGFRA genotype predicts the response to first-line imatinib and to all tyrosine kinase inhibitors (TKIs) approved or in investigation after imatinib failure. Considering that TKIs are effective only against a subset of KIT or PDGFRA resistance mutations, close monitoring of tumor dynamics with non-invasive methods such as liquid biopsy emerges as a necessary step forward in the field. Liquid biopsy, in contrast to solid tumor biopsy, aims to characterize tumors irrespective of heterogeneity. Although there are several components in the peripheral blood, most recent studies have been focused on circulating tumor (ct)DNA, due to the technological feasibility, the stability of DNA itself and DNA alterations, and the therapeutic development in precision oncology largely based on the identification of genetic driver mutations. In the present review, we systematically dissect the current wealth of data of ctDNA in GIST. To do so, a critical understanding of the promises and limitations of the current technologies will be followed by an exposition of the knowledge gathered with such studies in GIST. Collectively, our goal is to establish clear premises that can be used as the foundations to build future studies towards the clinical implementation of ctDNA evaluation in GIST patients.
Collapse
Affiliation(s)
- David Gómez-Peregrina
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Hospital Campus, C/ Natzaret 115-117, 08035, Barcelona, Spain
| | - Alfonso García-Valverde
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Hospital Campus, C/ Natzaret 115-117, 08035, Barcelona, Spain
| | - Daniel Pilco-Janeta
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Hospital Campus, C/ Natzaret 115-117, 08035, Barcelona, Spain
| | - César Serrano
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Hospital Campus, C/ Natzaret 115-117, 08035, Barcelona, Spain. .,Department of Medical Oncology, Vall d'Hebron University Hospital, P/Vall d'Hebron 119, 08035, Barcelona, Spain.
| |
Collapse
|
6
|
Teufel M, Seidel H, Köchert K, Meinhardt G, Finn RS, Llovet JM, Bruix J. Biomarkers Associated With Response to Regorafenib in Patients With Hepatocellular Carcinoma. Gastroenterology 2019; 156:1731-1741. [PMID: 30738047 DOI: 10.1053/j.gastro.2019.01.261] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS In a phase 3 trial (RESORCE), regorafenib increased overall survival compared with placebo in patients with hepatocellular carcinoma (HCC) previously treated with sorafenib. In an exploratory study, we analyzed plasma and tumor samples from study participants to identify genetic, microRNA (miRNA), and protein biomarkers associated with response to regorafenib. METHODS We obtained archived tumor tissues and baseline plasma samples from patients with HCC given regorafenib in the RESORCE trial. Baseline plasma samples from 499 patients were analyzed for expression of 294 proteins (DiscoveryMAP) and plasma samples from 349 patients were analyzed for levels of 750 miRNAs (miRCURY miRNA PCR). Tumor tissues from 7 responders and 10 patients who did not respond (progressors) were analyzed by next-generation sequencing (FoundationOne). Forty-six tumor tissues were analyzed for expression patterns of 770 genes involved in oncogenic and inflammatory pathways (PanCancer Immune Profiling). Associations between plasma levels of proteins and miRNAs and response to treatment (overall survival and time to progression) were evaluated using a Cox proportional hazards model. RESULTS Decreased baseline plasma concentrations of 5 of 266 evaluable proteins (angiopoietin 1, cystatin B, the latency-associated peptide of transforming growth factor beta 1, oxidized low-density lipoprotein receptor 1, and C-C motif chemokine ligand 3; adjusted P ≤ .05) were significantly associated with increased overall survival time after regorafenib treatment. Levels of these 5 proteins, which have roles in inflammation and/or HCC pathogenesis, were not associated with survival independently of treatment. Only 20 of 499 patients had high levels and a reduced survival time. Plasma levels of α-fetoprotein and c-MET were associated with poor outcome (overall survival) independently of regorafenib treatment only. We identified 9 plasma miRNAs (MIR30A, MIR122, MIR125B, MIR200A, MIR374B, MIR15B, MIR107, MIR320, and MIR645) whose levels significantly associated with overall survival time with regorafenib (adjusted P ≤ .05). Functional analyses of these miRNAs indicated that their expression level associated with increased overall survival of patients with tumors of the Hoshida S3 subtype. Next-generation sequencing analyses of tumor tissues revealed 49 variants in 27 oncogenes or tumor suppressor genes. Mutations in CTNNB1 were detected in 3 of 10 progressors and VEGFA amplification in 1 of 7 responders. CONCLUSION We identified expression patterns of plasma proteins and miRNAs that associated with increased overall survival times of patients with HCC following treatment with regorafenib in the RESORCE trial. Levels of these circulating biomarkers and genetic features of tumors might be used to identify patients with HCC most likely to respond to regorafenib. ClinicalTrials.gov number NCT01774344. NCBI GEO accession numbers: mRNA data (NanoString): GSE119220; miRNA data (Exiqon): GSE119221.
Collapse
Affiliation(s)
| | | | | | | | - Richard S Finn
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Josep M Llovet
- BCLC Group, Liver Unit, Hospital Clinic, University of Barcelona, IDIBAPS, CIBEREHD, Barcelona, Spain; Liver Cancer Program, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Jordi Bruix
- BCLC Group, Liver Unit, Hospital Clinic, University of Barcelona, IDIBAPS, CIBEREHD, Barcelona, Spain
| |
Collapse
|
7
|
Ravegnini G, Sammarini G, Serrano C, Nannini M, Pantaleo MA, Hrelia P, Angelini S. Clinical relevance of circulating molecules in cancer: focus on gastrointestinal stromal tumors. Ther Adv Med Oncol 2019; 11:1758835919831902. [PMID: 30854029 PMCID: PMC6399766 DOI: 10.1177/1758835919831902] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/30/2018] [Indexed: 12/12/2022] Open
Abstract
In recent years, growing research interest has focused on the so-called liquid biopsy. A simple blood test offers access to a plethora of information, which might be extremely helpful in understanding or characterizing specific diseases. Blood contains different molecules, of which circulating free DNA (cfDNA), circulating tumor DNA (ctDNA), circulating tumor cells (CTCs) and extracellular vesicles (EVs) are the most relevant. Conceivably, these molecules have the potential for tumor diagnosis, monitoring tumor evolution, and evaluating treatment response and pharmacological resistance. This review aims to present a state-of-the-art of recent advances in circulating DNA and circulating RNA in gastrointestinal stromal tumors (GISTs). To date, progress in liquid biopsy has been scarce in GISTs due to several issues correlated with the nature of the pathology. Namely, heterogeneity in primary and secondary mutations in key driver genes has greatly slowed the development and application in GISTs, unlike in other tumor types in which liquid biopsy has already been translated into clinical practice. However, meaningful novel data have shown in recent years a significant clinical potential of ctDNA, CTCs, EVs and circulating RNA in GISTs.
Collapse
Affiliation(s)
- Gloria Ravegnini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giulia Sammarini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - César Serrano
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Margherita Nannini
- Department of Specialized, Experimental and Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Maria A Pantaleo
- Department of Specialized, Experimental and Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy and Biotechnology, Via Irnerio 48, 40126 Bologna, Italy
| |
Collapse
|
8
|
Montemurro M, Cioffi A, Dômont J, Rutkowski P, Roth AD, von Moos R, Inauen R, Toulmonde M, Burkhard RO, Knuesli C, Bauer S, Cassier P, Schwarb H, Le Cesne A, Koeberle D, Bärtschi D, Dietrich D, Biaggi C, Prior J, Leyvraz S. Long-term outcome of dasatinib first-line treatment in gastrointestinal stromal tumor: A multicenter, 2-stage phase 2 trial (Swiss Group for Clinical Cancer Research 56/07). Cancer 2018; 124:1449-1454. [DOI: 10.1002/cncr.31234] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 12/06/2017] [Accepted: 12/15/2017] [Indexed: 11/09/2022]
Affiliation(s)
| | - Angela Cioffi
- Medical Oncology-Sarcoma; Gustave Roussy Institute; Villejuif France
| | - Julien Dômont
- Medical Oncology-Sarcoma; Gustave Roussy Institute; Villejuif France
| | - Piotr Rutkowski
- Soft Tissue/Bone Sarcoma and Melanoma; Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology; Warsaw Poland
| | - Arnaud D. Roth
- Division of Oncology; Geneva University Hospital; Geneva Switzerland
| | - Roger von Moos
- Medical Oncology and Hematology; Cantonal Hospital Graubunden; Chur Switzerland
| | - Roman Inauen
- Department of Oncology; Cantonal Hospital St. Gallen; St. Gallen Switzerland
| | | | - Roger O. Burkhard
- Oncology Center; Hirslanden Hospital and Health Care; Zurich Switzerland
| | - Claudio Knuesli
- Medical Oncology; Hospital St. Claraspital; Basel Switzerland
| | - Sebastian Bauer
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center; University of Duisburg-Essen; Essen Germany
| | | | - Heike Schwarb
- Oncology/Internal Medicine; Cantonal Hospital Baden; Baden Switzerland
| | - Axel Le Cesne
- Medical Oncology-Sarcoma; Gustave Roussy Institute; Villejuif France
| | - Dieter Koeberle
- Department of Oncology/Hematology; Cantonal Hospital St. Gallen; St. Gallen Switzerland
| | - Daniela Bärtschi
- Coordinating Center; Swiss Group for Clinical Cancer Research; Bern Switzerland
| | - Daniel Dietrich
- Coordinating Center; Swiss Group for Clinical Cancer Research; Bern Switzerland
| | - Christine Biaggi
- Coordinating Center; Swiss Group for Clinical Cancer Research; Bern Switzerland
| | - John Prior
- Nuclear Medicine and Molecular Imaging; University Hospital of Lausanne; Lausanne Switzerland
| | - Serge Leyvraz
- Medical Oncology; University Hospital of Lausanne; Lausanne Switzerland
| |
Collapse
|
9
|
Abstract
Circulating tumor DNA (ctDNA) analysis is currently gaining momentum as an innovative methodology for characterizing the tumor genome and monitoring therapeutic efficacy in the multifocal, genetically and spatially heterogeneous plasma cell malignancy, multiple myeloma (MM). Circulating cell-free DNA (cfDNA), which consists of a combination of DNA derived from both tumor and normal cells, is present in extracellular bodily fluids. The presence of ctDNA within this admixture has been demonstrated recently in MM. In this chapter, we describe the routinely utilized methodology for the extraction and longitudinal analysis of specific mutations present in ctDNA derived from peripheral blood plasma of MM patients.
Collapse
Affiliation(s)
- Sridurga Mithraprabhu
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital-Monash University, Melbourne, VIC, Australia
- Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia
| | - Andrew Spencer
- Myeloma Research Group, Australian Centre for Blood Diseases, Alfred Hospital-Monash University, Melbourne, VIC, Australia.
- Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia.
- Department of Clinical Haematology, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
10
|
Florou V, Wilky BA, Trent JC. Latest advances in adult gastrointestinal stromal tumors. Future Oncol 2017; 13:2183-2193. [DOI: 10.2217/fon-2017-0245] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common GI tract mesenchymal tumors. GIST patients are optimally managed by a precision medicine approach. Herein, we discuss the latest advances in precision medicine and ongoing clinical trials relevant to GIST. Circulating tumor DNA for detection of mutational changes could replace tissue biopsies and radiographic imaging once validated. Most GISTs are KIT/PDGFRα mutated, and despite the good clinical response to imatinib, treatment is generally not curative, more often due to secondary mutations. New mechanisms to bypass this resistance by inhibiting KIT downstream pathways and by targeting multiple KIT or PDGFRα mutations are being investigated. Immunotherapy for GIST patients is in its infancy. These approaches may lead to more effective, less toxic therapies.
Collapse
Affiliation(s)
- Vaia Florou
- Division of Hematology/Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
- Sylvester Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Breelyn A Wilky
- Division of Hematology/Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
- Sylvester Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Jonathan C Trent
- Division of Hematology/Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
- Sylvester Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| |
Collapse
|
11
|
D'Ambrosio L, Palesandro E, Boccone P, Tolomeo F, Miano S, Galizia D, Manca A, Chiara G, Bertotto I, Russo F, Campanella D, Venesio T, Sangiolo D, Pignochino Y, Siatis D, De Simone M, Ferrero A, Pisacane A, Dei Tos AP, Aliberti S, Aglietta M, Grignani G. Impact of a risk-based follow-up in patients affected by gastrointestinal stromal tumour. Eur J Cancer 2017; 78:122-132. [PMID: 28448856 DOI: 10.1016/j.ejca.2017.03.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/16/2017] [Accepted: 03/24/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Follow-up aims to precociously identify recurrences, metastases or treatment-related adverse events so as to undertake the appropriate therapy. Guidelines admit lack of knowledge on optimal surveillance schedule, but suggest follow-up based on experts' opinion and risk stratification. To identify the impact, if any, of regular follow-up, we interrogated our prospectively collected database whether early detection of recurrences affected both clinical management and, likely, the outcome. PATIENTS AND METHODS We required information to be available on primary surgery and ≥3°years of follow-up for non-recurring patients. We analysed recurrence characteristics (asymptomatic versus symptomatic, low- versus high tumour burden) and computed tomography (CT) scan counts to detect one recurrence. Kaplan-Meier method estimated recurrence-free survival (RFS), post-recurrence progression-free survival (PR-PFS), and disease-specific overall survival (OS). Comparisons used Hazard ratios (HR) with 95% confidence intervals (CIs). Multivariate analyses employed the Cox proportional hazards model. All tests were two-sided. RESULTS Between 01/2001 and 12/2012 we found 233 study-eligible patients. Estimated 5- and 10-year RFS were 61.8% and 50.4%, respectively. After a 68-month median follow-up, we observed 94 (40.3%) recurrences [73/94 (77.7%) asymptomatic versus 21/94 (22.3%) symptomatic and 45/94 (47.9%) low- versus 49/94 (52.1%) high tumour burden]. Multivariate analysis revealed that symptomatic and high tumour burden recurrences were highly predictive of both worse PR-PFS (HR:3.19, P < 0.001; HR:2.80, P = 0.003, respectively) and OS (HR:3.65, P < 0.001; HR:2.38, P = 0.026, respectively). Finally, 29 second (primary) cancers were detected during follow-up. CONCLUSIONS Regular follow-up detects recurrences at an earlier stage and may be associated with a better PR-PFS and OS for these patients. In the absence of randomised trials, these evidences support follow-up effort and cost.
Collapse
Affiliation(s)
- Lorenzo D'Ambrosio
- Sarcoma Unit, Division of Medical Oncology Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy; University of Torino, Department of Oncology, Regione Gonzole, 10, 10043 Orbassano, TO, Italy
| | - Erica Palesandro
- Sarcoma Unit, Division of Medical Oncology Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy; University of Torino, Department of Oncology, Regione Gonzole, 10, 10043 Orbassano, TO, Italy
| | - Paola Boccone
- Sarcoma Unit, Division of Medical Oncology Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy; University of Torino, Department of Oncology, Regione Gonzole, 10, 10043 Orbassano, TO, Italy
| | - Francesco Tolomeo
- Sarcoma Unit, Division of Medical Oncology Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy; University of Torino, Department of Oncology, Regione Gonzole, 10, 10043 Orbassano, TO, Italy
| | - Sara Miano
- Sarcoma Unit, Division of Medical Oncology Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy; University of Torino, Department of Oncology, Regione Gonzole, 10, 10043 Orbassano, TO, Italy
| | - Danilo Galizia
- Sarcoma Unit, Division of Medical Oncology Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy; University of Torino, Department of Oncology, Regione Gonzole, 10, 10043 Orbassano, TO, Italy
| | - Antonio Manca
- Radiology Unit, Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy
| | - Gabriele Chiara
- Radiology Unit, Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy
| | - Ilaria Bertotto
- Radiology Unit, Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy
| | - Filippo Russo
- Radiology Unit, Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy
| | - Delia Campanella
- Radiology Unit, Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy
| | - Tiziana Venesio
- Pathology Unit, Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy
| | - Dario Sangiolo
- Sarcoma Unit, Division of Medical Oncology Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy; University of Torino, Department of Oncology, Regione Gonzole, 10, 10043 Orbassano, TO, Italy
| | - Ymera Pignochino
- Sarcoma Unit, Division of Medical Oncology Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy; University of Torino, Department of Oncology, Regione Gonzole, 10, 10043 Orbassano, TO, Italy
| | - Dimitrios Siatis
- Surgical Oncology Unit, Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy
| | - Michele De Simone
- Surgical Oncology Unit, Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy
| | - Alessandro Ferrero
- Department of General and Oncological Surgery, Ospedale Mauriziano "Umberto I", Via Magellano 1, 10128 Torino, TO, Italy
| | - Alberto Pisacane
- Pathology Unit, Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy
| | - Angelo Paolo Dei Tos
- Department of Pathology and Molecular Genetics, Treviso General Hospital, Piazza Ospedale 23, 31100 Treviso, TV, Italy; University of Padova, Department of Medicine, Via 8 febbraio 2, 35122 Padova, PD, Italy
| | - Sandra Aliberti
- Sarcoma Unit, Division of Medical Oncology Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy
| | - Massimo Aglietta
- Sarcoma Unit, Division of Medical Oncology Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy; University of Torino, Department of Oncology, Regione Gonzole, 10, 10043 Orbassano, TO, Italy
| | - Giovanni Grignani
- Sarcoma Unit, Division of Medical Oncology Candiolo Cancer Institute - FPO, IRCCS, Strada Provinciale 142, Km 3.95, 10060 Candiolo, TO, Italy; University of Torino, Department of Oncology, Regione Gonzole, 10, 10043 Orbassano, TO, Italy.
| |
Collapse
|
12
|
Abstract
Gastrointestinal stromal tumors had the reputation for poor outcomes because of their lack of response to nonsurgical interventions. The discovery of gain-of-function mutations involving receptor tyrosine kinase growth factor receptors altered the biological understanding and management. Beginning in 2000, management of these tumors has changed dramatically because of the availability of tyrosine kinase inhibitors. The role of surgery continues to be refined. This article reviews how surgery and systemic therapy are being used, incorporating definitions of risk. Decisions on how to treat a patient is based on the risk of progression, pathologic characteristics, and tumor location.
Collapse
|
13
|
Szucs Z, Thway K, Fisher C, Bulusu R, Constantinidou A, Benson C, van der Graaf WT, Jones RL. Promising novel therapeutic approaches in the management of gastrointestinal stromal tumors. Future Oncol 2016; 13:185-194. [PMID: 27600625 DOI: 10.2217/fon-2016-0194] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Primary and secondary resistance to currently available licensed tyrosine kinase inhibitors poses a real clinical challenge in the management of advanced gastrointestinal stromal tumors. Within the frame of early phase clinical trials novel systemic treatments are currently being evaluated to target both the well explored and novel emerging downstream effectors of KIT and PDGFRA signaling. Alternative therapeutic approaches also include exploring novel inhibitors of the KIT/PDGFRA receptors, immune checkpoint and cyclin-dependent kinase inhibitors. The final clinical trial outcome data for these agents are highly anticipated. Integration of new diagnostic techniques into routine clinical practice can potentially guide tailored delivery of agents in the treatment of a highly polyclonal, heterogeneous disease such as heavily pretreated advanced gastrointestinal stromal tumor.
Collapse
Affiliation(s)
- Zoltan Szucs
- The Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
| | - Khin Thway
- The Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
| | - Cyril Fisher
- The Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
| | - Ramesh Bulusu
- Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| | | | - Charlotte Benson
- The Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
| | - Winette Ta van der Graaf
- The Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK.,The Institute of Cancer Research, Cotswold Road, Sutton, SM2 5NG, UK
| | - Robin L Jones
- The Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
| |
Collapse
|
14
|
Detecting Primary KIT Mutations in Presurgical Plasma of Patients with Gastrointestinal Stromal Tumor. Mol Diagn Ther 2016; 20:347-51. [DOI: 10.1007/s40291-016-0203-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
15
|
Lyseng-Williamson KA. Regorafenib: a guide to its use in advanced gastrointestinal stromal tumor (GIST) after failure of imatinib and sunitinib. BioDrugs 2014; 27:525-31. [PMID: 23975637 DOI: 10.1007/s40259-013-0061-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Regorafenib (Stivarga(®)), a new inhibitor of multiple kinases, is indicated as third-line treatment in patients with locally advanced, unresectable or metastatic gastrointestinal stromal tumor (GIST) who have been previously treated with imatinib and sunitinib in the USA. In a phase III trial in patients with progressive GIST after failure of standard therapies, regorafenib plus best supportive care increased median progression-free survival by >5-fold relative to best supportive care alone. Although regorafenib is associated with several specific drug-related adverse events, it is reasonably well tolerated if recommendations for dose modifications (i.e. treatment interruption, dose reductions and/or permanent treatment discontinuation based on tolerability) and other precautions are followed.
Collapse
|
16
|
Patel JD, Krilov L, Adams S, Aghajanian C, Basch E, Brose MS, Carroll WL, de Lima M, Gilbert MR, Kris MG, Marshall JL, Masters GA, O'Day SJ, Polite B, Schwartz GK, Sharma S, Thompson I, Vogelzang NJ, Roth BJ. Clinical Cancer Advances 2013: Annual Report on Progress Against Cancer From the American Society of Clinical Oncology. J Clin Oncol 2014; 32:129-60. [DOI: 10.1200/jco.2013.53.7076] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A MESSAGE FROM ASCO'S PRESIDENTSince its founding in 1964, the American Society of Clinical Oncology (ASCO) has been committed to improving cancer outcomes through research and the delivery of quality care. Research is the bedrock of discovering better treatments—providing hope to the millions of individuals who face a cancer diagnosis each year.The studies featured in “Clinical Cancer Advances 2013: Annual Report on Progress Against Cancer From the American Society of Clinical Oncology” represent the invaluable contributions of thousands of patients who participate in clinical trials and the scientists who conduct basic and clinical research. The insights described in this report, such as how cancers hide from the immune system and why cancers may become resistant to targeted drugs, enable us to envision a future in which cancer will be even more controllable and preventable.The scientific process is thoughtful, deliberate, and sometimes slow, but each advance, while helping patients, now also points toward new research questions and unexplored opportunities. Both dramatic and subtle breakthroughs occur so that progress against cancer typically builds over many years. Success requires vision, persistence, and a long-term commitment to supporting cancer research and training.Our nation's longstanding investment in federally funded cancer research has contributed significantly to a growing array of effective new treatments and a much deeper understanding of the drivers of cancer. But despite this progress, our position as a world leader in advancing medical knowledge and our ability to attract the most promising and talented investigators are now threatened by an acute problem: Federal funding for cancer research has steadily eroded over the past decade, and only 15% of the ever-shrinking budget is actually spent on clinical trials. This dismal reality threatens the pace of progress against cancer and undermines our ability to address the continuing needs of our patients.Despite this extremely challenging economic environment, we continue to make progress. Maintaining and accelerating that progress require that we keep our eyes on the future and pursue a path that builds on the stunning successes of the past. We must continue to show our policymakers the successes in cancer survival and quality of life (QOL) they have enabled, emphasizing the need to sustain our national investment in the remarkably productive US cancer research enterprise.We must also look to innovative methods for transforming how we care for—and learn from—patients with cancer. Consider, for example, that fewer than 5% of adult patients with cancer currently participate in clinical trials. What if we were able to draw lessons from the other 95%? This possibility led ASCO this year to launch CancerLinQ, a groundbreaking health information technology initiative that will provide physicians with access to vast quantities of clinical data about real-world patients and help achieve higher quality, higher value cancer care.As you read the following pages, I hope our collective progress against cancer over the past year inspires you. More importantly, I hope the pride you feel motivates you to help us accelerate the pace of scientific advancement.Clifford A. Hudis, MD, FACPPresidentAmerican Society of Clinical Oncology
Collapse
Affiliation(s)
- Jyoti D. Patel
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Lada Krilov
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Sylvia Adams
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Carol Aghajanian
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Ethan Basch
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Marcia S. Brose
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - William L. Carroll
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Marcos de Lima
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Mark R. Gilbert
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Mark G. Kris
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - John L. Marshall
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Gregory A. Masters
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Steven J. O'Day
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Blasé Polite
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Gary K. Schwartz
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Sunil Sharma
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Ian Thompson
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Nicholas J. Vogelzang
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| | - Bruce J. Roth
- Jyoti D. Patel, Northwestern University; Blasé Polite, University of Chicago Medicine, Chicago, IL; Lada Krilov, American Society of Clinical Oncology, Alexandria, VA; Sylvia Adams and William L. Carroll, New York University Cancer Institute; Carol Aghajanian, Mark G. Kris, and Gary K. Schwartz, Memorial Sloan-Kettering Cancer Center, New York, NY; Ethan Basch, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Marcia S. Brose, University of Pennsylvania Abramson Cancer
| |
Collapse
|
17
|
Actualités 2013 : le point de vue du comité de rédaction du Bulletin du Cancer. Bull Cancer 2014; 101:75-92. [DOI: 10.1684/bdc.2013.1874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|