451
|
Sarvizadeh M, Ghasemi F, Tavakoli F, Sadat Khatami S, Razi E, Sharifi H, Biouki NM, Taghizadeh M. Vaccines for colorectal cancer: an update. J Cell Biochem 2018; 120:8815-8828. [PMID: 30536960 DOI: 10.1002/jcb.28179] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 11/12/2018] [Indexed: 12/29/2022]
Abstract
Colorectal cancer (CRC) is known as the third most common and fourth leading cancer associated death worldwide. The occurrence of metastasis has remained as a critical challenge in CRC, so that distant metastasis (mostly to the liver) has been manifested in about 20%-25% of patients. Several screening approaches have introduced for detecting CRC in different stages particularly in early stages. The standard treatments for CRC are surgery, chemotherapy and radiotherapy, in alone or combination. Immunotherapy is a set of novel approaches with the aim of remodeling the immune system battle with metastatic cancer cells, such as immunomodulatory monoclonal antibodies (immune checkpoint inhibitors), adoptive cell transfer (ACT) and cancer vaccine. Cancer vaccines are designed to trigger the intense response of immune system to tumor-specific antigens. In two last decades, introduction of new cancer vaccines and designing several clinical trials with vaccine therapy, have been taken into consideration in colon cancer patients. This review will describe the treatment approaches with the special attention to vaccines applied to treat colorectal cancer.
Collapse
Affiliation(s)
- Mostafa Sarvizadeh
- The Advocate Center for Clinical Research, Ayatollah Yasrebi Hospital, Kashan, Iran
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Fatemeh Tavakoli
- Department of Biotechnology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Sadat Khatami
- Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Ebrahim Razi
- The Advocate Center for Clinical Research, Ayatollah Yasrebi Hospital, Kashan, Iran
| | - Hossein Sharifi
- The Advocate Center for Clinical Research, Ayatollah Yasrebi Hospital, Kashan, Iran
| | - Nousin Moussavi Biouki
- Department of Surgery, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
452
|
Vacante M, Borzì AM, Basile F, Biondi A. Biomarkers in colorectal cancer: Current clinical utility and future perspectives. World J Clin Cases 2018; 6:869-881. [PMID: 30568941 PMCID: PMC6288499 DOI: 10.12998/wjcc.v6.i15.869] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/30/2018] [Accepted: 11/07/2018] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer death worldwide. CRC has poor prognosis and there is a crucial need for new diagnostic and prognostic biomarkers to avoid CRC-related deaths. CRC can be considered a sporadic disease in most cases (75%-80%), but it has been suggested that crosstalk between gene mutations (i.e., mutations of BRAF, KRAS, and p53 as well as microsatellite instability) and epigenetic alterations (i.e., DNA methylation of CpG island promoter regions) could play a pivotal role in cancer development. A number of studies have focused on molecular testing to guide targeted and conventional treatments for patients with CRC, sometimes with contrasting results. Some of the most useful innovations in the management of CRC include the possibility to detect the absence of KRAS, BRAF, NRAS and PIK3CA gene mutations with the subsequent choice to administer targeted adjuvant therapy with anti-epidermal growth factor receptor antibodies. Moreover, CRC patients can benefit from tests for microsatellite instability and for the detection of loss of heterozygosity of chromosome 18q that can be helpful in guiding therapeutic decisions as regards the administration of 5-FU. The aim of this review was to summarize the most recent evidence on the possible use of genetic or epigenetic biomarkers for diagnosis, prognosis and response to therapy in CRC patients.
Collapse
Affiliation(s)
- Marco Vacante
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania 95123, Italy
| | - Antonio Maria Borzì
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania 95123, Italy
| | - Francesco Basile
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania 95123, Italy
| | - Antonio Biondi
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania 95123, Italy
| |
Collapse
|
453
|
Liao X, Li H, Liu Z, Liao S, Li Q, Liang C, Huang Y, Xie M, Wei J, Li Y. Clinical efficacy and safety of apatinib in patients with advanced colorectal cancer as the late-line treatment. Medicine (Baltimore) 2018; 97:e13635. [PMID: 30558053 PMCID: PMC6320137 DOI: 10.1097/md.0000000000013635] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
There is currently no standard therapeutic regimen available for patients with advanced colorectal cancer in whom the disease continues to progress after 2 or more lines of chemotherapy. The purpose of this study is to investigate the efficacy and safety of apatinib in patients with advanced colorectal cancer for whom at least two lines of prior chemotherapy had failed.Twenty seven patients with advanced colorectal cancer who had failed at least 2 lines chemotherapy were treated with apatinib (500 mg/day). As a comparison control, 26 advanced colorectal cancer patients with comparable clinical baseline characteristics including age, sex, Eastern Cooperative Oncology Group (ECOG) score, pathological type, carcinoembryonic antigen (CEA) level, tumor location, number and location(s) of metastasis, and previous chemotherapies were subject to observation. Survival analyses were performed via the Kaplan-Meier method. The toxicity were evaluated in all patients this study according to the National Cancer Institute Common Toxicity Criteria 4 (NCI CTC version 4.0).A total of 53 well-matched patients with advanced colorectal cancer were retrospectively analyzed. The median follow-up time was 6.0 months (2.0-16.0 months). The median PFS was significantly longer for apatinib group than for observation group (2.0 vs. 1.1 months; HR = 3.88; 95% confidence interval [CI], 1.91-7.88; P < .001). However, there was no significant difference between the 2 groups for median OS (5.0 vs. 4.0 months; HR = 1.03; 95% CI, 0.56-1.90; P = .914). The disease control rate of the apatinib group was significantly better than that of the observation group (70.4% vs 26.9%, P = .002). There was no significant difference in the overall remission rate between the 2 groups (3.7% vs 0%, P = .322). Advanced colorectal cancer patients with 2 or fewer metastatic sites experienced longer PFS than those with more than 2 sites. High ECOG scores, cancer localization to the right side of colon and lymph node metastasis were associated with increased risk of death and all remained independent factors affecting OS. The most common grade 3/4 treatment-related adverse events were hypertension and hand-foot skin syndrome.Apatinib treatment for patients with advanced colorectal cancer who had failed chemotherapy achieved better disease control and prolonged PFS relative to untreated controls. The toxicity was manageable.
Collapse
Affiliation(s)
| | | | | | | | - Qian Li
- Department of First Chemotherapy
| | | | - Yu Huang
- Department of First Chemotherapy
| | | | - Junbao Wei
- Department of Therapeutic Radiology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | | |
Collapse
|
454
|
What Is the Best Systemic Therapy for Left-sided RAS Wild-type Metastatic Colorectal Cancer? CURRENT COLORECTAL CANCER REPORTS 2018. [DOI: 10.1007/s11888-018-0414-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
455
|
Corsini LR, Fanale D, Passiglia F, Incorvaia L, Gennusa V, Bazan V, Russo A. Monoclonal antibodies for the treatment of non-hematological tumors: a safety review. Expert Opin Drug Saf 2018; 17:1197-1209. [PMID: 30457416 DOI: 10.1080/14740338.2018.1550068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: The introduction of monoclonal antibodies (moAbs) into clinical practice revolutionized the treatment strategies in several solid tumors. These agents differ from cytotoxic chemotherapy for their mechanism of action and toxicity. By targeting specific antigens present on healthy cells and modulating immune system activity, these biological drugs are able to generate a wide spectrum of peculiar adverse events that can negatively impact on patients' quality of life. Areas covered: In this review, the main side effects associated with the use of moAbs have been described to show their incidence and current management strategies, which may drive clinicians in their daily practice. Expert opinion: The majority of these drugs represents an example of successful innovation, since they are able to induce a significant improvement of patients' survival and quality of life without any increase in related side effects as compared to standard cancer treatments. For this reason, they have become new milestones in personalized therapy for different non-hematological malignancies. With the increasing use of moAbs in treatment regimens, it is strongly recommended that clinicians are knowledgeable about the side effects associated with these agents, their management and monitoring, to optimize the clinical treatment of cancer patients.
Collapse
Affiliation(s)
- Lidia Rita Corsini
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| | - Daniele Fanale
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| | - Francesco Passiglia
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| | - Lorena Incorvaia
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| | - Vincenzo Gennusa
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| | - Viviana Bazan
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| | - Antonio Russo
- a Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology , University of Palermo , Palermo , Italy
| |
Collapse
|
456
|
Holch JW, Ricard I, Stintzing S, Fischer von Weikersthal L, Decker T, Kiani A, Vehling-Kaiser U, Heintges T, Kahl C, Kullmann F, Scheithauer W, Moehler M, Jelas I, Modest DP, Westphalen CB, von Einem JC, Michl M, Heinemann V. Relevance of baseline carcinoembryonic antigen for first-line treatment against metastatic colorectal cancer with FOLFIRI plus cetuximab or bevacizumab (FIRE-3 trial). Eur J Cancer 2018; 106:115-125. [PMID: 30496943 DOI: 10.1016/j.ejca.2018.10.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022]
Abstract
PURPOSE Increased baseline carcinoembryonic antigen (CEA) serum level is associated with inferior overall survival (OS) in metastatic colorectal cancer (mCRC). However, limited data exist on its predictive relevance for targeted therapies. Therefore, we analysed its relevance in FIRE-3, a randomised phase III study. EXPERIMENTAL DESIGN FIRE-3 evaluated first-line FOLFIRI plus cetuximab (FOLFIRI/Cet) versus FOLFIRI plus bevacizumab (FOLFIRI/Bev) in mCRC patients with RAS-WT tumour (i.e. wild-type in KRAS and NRAS exons 2-4). Herein, the impact of CEA on patient outcome was investigated. RESULTS Of 400 patients, 356 (89.0%) were evaluable for CEA. High CEA (>10 ng/ml; N = 237) compared to low CEA (≤10 ng/ml; N = 119) was associated with shorter OS in the FOLFIRI/Bev arm (hazard ratio [HR] = 1.50; P = 0.036), while no significant OS difference was observed in the FOLFIRI/Cet arm (HR = 1.07; P = 0.74). In patients with high CEA, FOLFIRI/Cet compared to FOLFIRI/Bev showed a greater OS benefit (HR = 0.56; P < 0.001) than in patients with low CEA (HR = 0.78; P = 0.30). Furthermore, FOLFIRI/Cet exhibited significantly superior objective response rate in patients with high CEA (odds ratio = 2.21; P = 0.006) in contrast to patients with low CEA (odds ratio = 0.90; P = 0.85). CONCLUSION In patients with RAS-WT mCRC receiving first-line chemotherapy with FOLFIRI/Cet versus FOLFIRI/Bev, elevated CEA was associated with inferior survival in the bevacizumab arm, while this was not the case when cetuximab was applied. Comparison of OS and objective response rate according to treatment arms indicated that cetuximab was greatly superior to bevacizumab in patients with elevated CEA, while this effect was markedly lower and lost statistical significance in patients with low CEA.
Collapse
Affiliation(s)
- J W Holch
- Department of Internal Medicine III, Comprehensive Cancer Center Munich, University Hospital Grosshadern, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany.
| | - I Ricard
- Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - S Stintzing
- Department of Internal Medicine III, Comprehensive Cancer Center Munich, University Hospital Grosshadern, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - L Fischer von Weikersthal
- Praxis für Onkologie/Haematologie, Gesundheitszentrum St. Marien GmbH, Mariahilfbergweg 7, 92224 Amberg, Germany
| | - T Decker
- Onkologie Ravensburg, Elisabethenstrasse 19, 88212 Ravensburg, Germany
| | - A Kiani
- Department of Medicine IV, Klinikum Bayreuth GmbH, Preuschwitzer Strasse 101, 95445 Bayreuth, Germany
| | - U Vehling-Kaiser
- Hämato-onkologische Tagesklinik, Dr. Med. Ursula Vehling-Kaiser, Ländgasse 132-135, 84028 Landshut, Germany
| | - T Heintges
- Department of Medicine II, Lukaskrankenhaus, Preußenstrasse 84, 41462 Neuss, Germany
| | - C Kahl
- Department of Hematology, Oncology and Palliative Care, Klinikum Magdeburg gGmbH, Birkenallee 34, 39130 Magdeburg, Germany
| | - F Kullmann
- Department of Internal Medicine I, Klinikum Weiden, Söllnerstrasse 16, 92637 Weiden, Germany
| | - W Scheithauer
- Department of Internal Medicine I & CCC, Medical University Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - M Moehler
- University Medical Center Mainz, I. Dept. of Internal Medicine, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - I Jelas
- Department of Internal Medicine III, Comprehensive Cancer Center Munich, University Hospital Grosshadern, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - D P Modest
- Department of Internal Medicine III, Comprehensive Cancer Center Munich, University Hospital Grosshadern, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - C B Westphalen
- Department of Internal Medicine III, Comprehensive Cancer Center Munich, University Hospital Grosshadern, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - J C von Einem
- Department of Internal Medicine III, Comprehensive Cancer Center Munich, University Hospital Grosshadern, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - M Michl
- Department of Internal Medicine III, Comprehensive Cancer Center Munich, University Hospital Grosshadern, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - V Heinemann
- Department of Internal Medicine III, Comprehensive Cancer Center Munich, University Hospital Grosshadern, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| |
Collapse
|
457
|
How the BRAF V600E Mutation Defines a Distinct Subgroup of Colorectal Cancer: Molecular and Clinical Implications. Gastroenterol Res Pract 2018; 2018:9250757. [PMID: 30598662 PMCID: PMC6287148 DOI: 10.1155/2018/9250757] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/18/2018] [Indexed: 12/13/2022] Open
Abstract
The BRAF oncogene is an integral component of the MAP kinase pathway, and an activating V600E mutation occurs in 15% of sporadic colorectal cancer. This is an early event in serrated pathway tumourigenesis, and the BRAF V600E has been commonly associated with the CpG island methylator phenotype, microsatellite instability (MSI), and a consistent clinical presentation including a proximal location and predilection for elderly females. A proportion of the BRAF mutant lesions remain as microsatellite stable (MSS), and in contrast to the MSI cancers, they have an aggressive phenotype and correlate with poor patient outcomes. Recent studies have found that they have clinical and molecular features of both the BRAF mutant/MSI and the conventional BRAF wild-type cancers and comprise a distinct colorectal cancer subgroup. This review highlights the importance of the BRAF mutation occurring in colorectal cancer stratified for molecular background and discusses its prognostic and clinical significance.
Collapse
|
458
|
Claassen YHM, van der Valk MJM, Breugom AJ, Frouws MA, Bastiaannet E, Liefers G, van de Velde CJH, Kapiteijn E, Cochrane Colorectal Cancer Group. Survival differences with immediate versus delayed chemotherapy for asymptomatic incurable metastatic colorectal cancer. Cochrane Database Syst Rev 2018; 11:CD012326. [PMID: 30480771 PMCID: PMC6517244 DOI: 10.1002/14651858.cd012326.pub2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND For patients with asymptomatic, incurable, metastatic colorectal cancer, palliative, systemic treatment can be started immediately, or can be delayed until disease-related symptoms occur. How the potential survival benefit of starting palliative, systemic treatment immediately after diagnosis weighs up against the potential side effects is currently under debate, and was investigated in this review. OBJECTIVES To assess the effects of immediate versus delayed chemotherapy, with or without targeted therapy, on overall survival, toxicity, quality of life, progression-free survival, and compliance with chemotherapy for individuals with asymptomatic, metastatic, incurable colorectal cancer. SEARCH METHODS We searched CENTRAL; 2018, Issue 8, MEDLINE Ovid, Embase Ovid, PsycINFO, the World Health Organization International Clinical Trials Registry Platform, and Clinicaltrials.gov, from inception to 23 August 2018. We did not apply limitations based on language or date of publication. We searched the reference lists of all included studies to identify trials that may not have been identified from the electronic searches. SELECTION CRITERIA Randomised controlled trials evaluating immediate versus delayed chemotherapy in persons with asymptomatic, metastatic, incurable colorectal cancer. DATA COLLECTION AND ANALYSIS We applied standard methodological procedures, according to the recommendations of Cochrane and Cochrane Colorectal Cancer. Two review authors independently reviewed the studies identified by literature searches, selected relevant trials, extracted data, and assessed risk of bias of the included studies. We used the Cochrane tool to assess risk of bias, Review Manager 5 software for meta-analysis, GRADE methods to evaluate the quality of the evidence, and GRADEpro GDT software to develop a 'Summary of findings' table. MAIN RESULTS We included three randomised controlled trials (351 participants) investigating immediate versus delayed chemotherapy in people diagnosed with asymptomatic, metastatic, incurable colorectal cancer. Giving immediate versus delayed chemotherapy may make little or no difference to overall survival (hazard ratio (HR) 1.17, 95% confidence interval (CI) 0.93 to 1.46; 3 studies, 351 persons; low-quality evidence). For toxicity, giving immediate versus delayed chemotherapy may make little or no difference to the risk of grade 3 or 4 nausea and vomiting (risk ratio (RR) 0.84, 95% CI 0.31 to 2.25; 2 studies, 140 persons; very low-quality evidence), stomatitis (RR 1.10, 95% CI 0.47 to 2.55; 2 studies, 140 persons; very low-quality evidence), or diarrhoea (RR 0.69, 95% CI 0.34 to 1.40; 2 studies, 140 persons, very low-quality evidence). We are uncertain whether delayed chemotherapy made a difference to quality of life (very low-quality evidence), progression-free survival (low-quality evidence), or compliance with chemotherapy (low-quality evidence), as we had insufficient data to pool for these outcomes. AUTHORS' CONCLUSIONS Based on a limited number of trials, very sparse data, and uncertainty of the evidence, this review was unable to establish whether there was a difference in overall survival or other clinically relevant outcomes, between immediate or delayed chemotherapy in persons with metastatic, incurable, colorectal cancer. The results should be interpreted with caution.
Collapse
Affiliation(s)
| | | | - Anne J Breugom
- Leiden University Medical CentreDepartment of SurgeryLeidenNetherlands
| | - Martine A. Frouws
- Leiden University Medical CentreDepartment of SurgeryLeidenNetherlands
| | | | | | | | - Ellen Kapiteijn
- Leiden University Medical CentreDepartment of Medical OncologyLeidenNetherlands
| | | |
Collapse
|
459
|
Goel G. Molecular characterization and biomarker identification in colorectal cancer: Toward realization of the precision medicine dream. Cancer Manag Res 2018; 10:5895-5908. [PMID: 30510457 PMCID: PMC6250110 DOI: 10.2147/cmar.s162967] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Colorectal cancer (CRC) is a major public health problem, both in the USA and globally. Over the past 20 years, significant advances have been made in the treatment of patients with metastatic CRC (mCRC). Recent efforts in the field of biomarkers have focused on the development of molecular diagnostics to define the subset of patients with mCRC that is likely to derive most benefit from anti-EGFR therapy. Herein, we review the recent advancements in molecular stratification of CRC and the role of current as well as emerging biomarkers in this disease. It is now clear that the presence of activating mutations in the KRAS and NRAS genes serves as reliable predictive markers for resistance to anti-EGFR therapy in mCRC. It is also clear that further improvements in the survival of mCRC patients will probably be made possible only with identification of new predictive molecular biomarkers and their evaluation using rational and innovative clinical trials. The recent advances in DNA sequencing technology and "omics"-based approaches have provided promising new strategies for the development of novel molecular biomarkers in this disease.
Collapse
Affiliation(s)
- Gaurav Goel
- Division of Hematology-Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,
| |
Collapse
|
460
|
Iemoto T, Nishiumi S, Kobayashi T, Fujigaki S, Hamaguchi T, Kato K, Shoji H, Matsumura Y, Honda K, Yoshida M. Serum level of octanoic acid predicts the efficacy of chemotherapy for colorectal cancer. Oncol Lett 2018; 17:831-842. [PMID: 30655836 PMCID: PMC6312949 DOI: 10.3892/ol.2018.9731] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/19/2018] [Indexed: 12/18/2022] Open
Abstract
The survival times of patients with advanced colorectal cancer (CRC) have increased due to the introduction of chemotherapy involving irinotecan and cetuximab. However, further studies are required on the effective pretreatment methods for identifying patients with CRC who would respond to particular treatments. The aim of the present study was to identify biomarkers for predicting the efficacy of chemotherapy for CRC. A total of 123 serum samples were collected from 31 patients with CRC just prior to each of the first four rounds of chemotherapy. Serum metabolome analysis was performed using a multiplatform metabolomics system, and univariate Cox regression hazards analysis of the time to disease progression was conducted. Octanoic acid and 1,5-anhydro-D-glucitol were identified as biomarker candidates. In addition, the serum level of octanoic acid was indicated to be significantly associated with the time to disease progression (hazard ratio, 3.3; 95% confidence interval, 1.099–11.840; P=0.033). The serum levels of fatty acids, in particular polyunsaturated fatty acids, tended to be downregulated in the partial response group. The findings of the present study suggest that the serum level of octanoic acid may serve as a useful predictor for the prognosis of CRC.
Collapse
Affiliation(s)
- Takao Iemoto
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Shin Nishiumi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Takashi Kobayashi
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Seiji Fujigaki
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | - Tetsuya Hamaguchi
- Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Ken Kato
- Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Hirokazu Shoji
- Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yasuhiro Matsumura
- Division of Developmental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba 277-8577, Japan
| | - Kazufumi Honda
- Division of Chemotherapy and Clinical Research, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Masaru Yoshida
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan.,Division of Metabolomics Research, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan.,AMED-CREST, AMED, Kobe, Hyogo 650-0017, Japan
| |
Collapse
|
461
|
Preoperative radiomic signature based on multiparametric magnetic resonance imaging for noninvasive evaluation of biological characteristics in rectal cancer. Eur Radiol 2018; 29:3200-3209. [PMID: 30413959 DOI: 10.1007/s00330-018-5763-x] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/21/2018] [Accepted: 09/14/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To develop and validate radiomic models in evaluating biological characteristics of rectal cancer based on multiparametric magnetic resonance imaging (MP-MRI). METHODS This study consisted of 345 patients with rectal cancer who underwent MP-MRI. We focused on evaluating five postoperative confirmed characteristics: lymph node (LN) metastasis, tumor differentiation, fraction of Ki-67-positive tumor cells, human epidermal growth factor receptor 2 (HER-2), and KRAS-2 gene mutation status. Data from 197 patients were used to develop the biological characteristics evaluation models. Radiomic features were extracted from MP-MRI and then refined for reproducibility and redundancy. The refined features were investigated for usefulness in building radiomic signatures by using two feature-ranking methods (MRMR and WLCX) and three classifiers (RF, SVM, and LASSO). Multivariable logistic regression was used to build an integrated evaluation model combining radiomic signatures and clinical characteristics. The performance was evaluated using an independent validation dataset comprising 148 patients. RESULTS The MRMR and LASSO regression produced the best-performing radiomic signatures for evaluating HER-2, LN metastasis, tumor differentiation, and KRAS-2 gene status, with AUC values of 0.696 (95% CI, 0.610-0.782), 0.677 (95% CI, 0.591-0.763), 0.720 (95% CI, 0.621-0.819), and 0.651 (95% CI, 0.539-0.763), respectively. The best-performing signatures for evaluating Ki-67 produced an AUC value of 0.699 (95% CI, 0.611-0.786), and it was developed by WLCX and RF algorithm. The integrated evaluation model incorporating radiomic signature and MRI-reported LN status had improved AUC of 0.697 (95% CI, 0.612-0.781). CONCLUSION Radiomic signatures based on MP-MRI have potential to noninvasively evaluate the biological characteristics of rectal cancer. KEY POINTS • Radiomic features were extracted from MP-MRI images of the rectal tumor. • The proposed radiomic signatures demonstrated discrimination ability in identifying the histopathological, immunohistochemical, and genetic characteristics of rectal cancer. • All MRI sequences were important and could provide complementary information in radiomic analysis.
Collapse
|
462
|
Yu IS, Cheung WY. Metastatic Colorectal Cancer in the Era of Personalized Medicine: A More Tailored Approach to Systemic Therapy. Can J Gastroenterol Hepatol 2018; 2018:9450754. [PMID: 30519549 PMCID: PMC6241232 DOI: 10.1155/2018/9450754] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 10/30/2018] [Indexed: 12/21/2022] Open
Abstract
Colorectal cancer is the second most common malignancy diagnosed in Canada. Despite declining incidence and mortality rates in recent years, there is still a significant number of cases that are metastatic at presentation. Fluoropyrimidine-based chemotherapy was the backbone of colorectal cancer treatment, but the addition of irinotecan and oxaliplatin to form combination regimens has significantly improved overall survival. In the past decade, the development of novel biologic agents including therapies directed against vascular endothelial growth factor and epidermal growth factor receptor has further altered the landscape of metastatic colorectal cancer treatment. However, clinical trials have demonstrated that not all patients respond to these therapies similarly and consideration must be given to individual patient- and tumor-related factors. A more tailored and biomarker driven approach to treatment selection can optimize outcomes and avoid unnecessary adverse effects. In this review article, we offer a comprehensive overview of the panel of clinical- and tumor-associated characteristics that influence treatment decisions in metastatic colorectal cancer and how this sets the foundation for a more personalized treatment strategy in oncology.
Collapse
Affiliation(s)
- Irene S. Yu
- University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
463
|
Qin S, Li J, Wang L, Xu J, Cheng Y, Bai Y, Li W, Xu N, Lin LZ, Wu Q, Li Y, Yang J, Pan H, Ouyang X, Qiu W, Wu K, Xiong J, Dai G, Liang H, Hu C, Zhang J, Tao M, Yao Q, Wang J, Chen J, Eggleton SP, Liu T. Efficacy and Tolerability of First-Line Cetuximab Plus Leucovorin, Fluorouracil, and Oxaliplatin (FOLFOX-4) Versus FOLFOX-4 in Patients With RAS Wild-Type Metastatic Colorectal Cancer: The Open-Label, Randomized, Phase III TAILOR Trial. J Clin Oncol 2018; 36:3031-3039. [PMID: 30199311 PMCID: PMC6324088 DOI: 10.1200/jco.2018.78.3183] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Cetuximab in combination with chemotherapy is a standard-of-care first-line treatment regimen for patients with RAS wild-type (wt) metastatic colorectal cancer (mCRC); however, the efficacy of cetuximab plus leucovorin, fluorouracil, and oxaliplatin (FOLFOX) has never before been proven in a controlled and randomized phase III trial. To our knowledge, the TAILOR trial ( ClinicalTrials.gov identifier: NCT01228734) is the first randomized, multicenter, phase III study of the addition of cetuximab to first-line FOLFOX prospectively choosing a RAS wt population and thus providing confirmative data for the efficacy and safety of cetuximab plus FOLFOX versus FOLFOX alone. PATIENTS AND METHODS TAILOR is an open-label, randomized (1:1), multicenter, phase III trial in patients from China comparing FOLFOX-4 with or without cetuximab in RAS wt (KRAS/NRAS, exons 2 to 4) mCRC. The primary end point of TAILOR was progression-free survival time; secondary end points included overall survival time, overall response rate, and safety and tolerability. RESULTS In the modified intent-to-treat population of 393 patients with RAS wt mCRC, adding cetuximab to FOLFOX-4 significantly improved the primary end point of progression-free survival time compared with FOLFOX-4 alone (hazard ratio, 0.69; 95% CI, 0.54 to 0.89; P = .004; median, 9.2 v 7.4 months, respectively), as well as the secondary end points of overall survival time (current assessment after 300 events: hazard ratio, 0.76; 95% CI, 0.61 to 0.96; P = .02; median, 20.7 v 17.8 months, respectively) and overall response rate (odds ratio, 2.41; 95% CI, 1.61 to 3.61; P < .001; 61.1% v 39.5%, respectively). Treatment was well tolerated, and there were no new or unexpected safety findings. CONCLUSION The TAILOR study met all of its objectives and relevant clinical end points, confirming cetuximab in combination with FOLFOX as an effective standard-of-care first-line treatment regimen for patients with RAS wt mCRC.
Collapse
Affiliation(s)
- Shukui Qin
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Jin Li
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Liwei Wang
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Jianming Xu
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Ying Cheng
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Yuxian Bai
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Wei Li
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Nong Xu
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Li-zhu Lin
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Qiong Wu
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Yunfeng Li
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Jianwei Yang
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Hongming Pan
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Xuenong Ouyang
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Wensheng Qiu
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Kaichun Wu
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Jianping Xiong
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Guanghai Dai
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Houjie Liang
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Chunhong Hu
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Jun Zhang
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Min Tao
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Qiang Yao
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Junyuan Wang
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Jiongjie Chen
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - S. Peter Eggleton
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| | - Tianshu Liu
- Shukui Qin, Bayi Hospital, Nanjing University of Chinese Medicine, Nanjing; Jin Li, Fudan University Cancer Hospital and Tongji University East Hospital; Liwei Wang, Shanghai First People’s Hospital; Jun Zhang, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Tianshu Liu, Affiliated Zhongshan Hospital of Fudan University, Shanghai; Jianming Xu, 307 Hospital of the Chinese People’s Liberation Army; Guanghai Dai, Chinese People’s Liberation Army General Hospital; Junyuan Wang and Jiongjie Chen, Merck Serono, Beijing; Ying Cheng, Jilin Cancer Hospital; Wei Li, First Affiliated Hospital of Jilin University, Jilin; Yuxian Bai, Affiliated Hospital of Harbin Medical University, Harbin; Nong Xu, First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou; Li-zhu Lin, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou; Qiong Wu, First Affiliated Hospital of Bengbu Medical College, Bengbu; Yunfeng Li, Yunnan Province Cancer Hospital, Kunming; Jianwei Yang, Fujian Province Cancer Hospital; Xuenong Ouyang, Fuzhou General Hospital, Fuzhou; Hongming Pan, Sir Run Run Shaw Hospital Affiliated With School of Medicine, Zhejiang University, Zhejiang; Wensheng Qiu, Affiliated Hospital of Qingdao University, Qingdao; Kaichun Wu, Xijing Hospital, Fourth Military Medical University, Shaanxi; Jianping Xiong, First Affiliated Hospital of Nanchang University, Nanchang; Houjie Liang, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing; Chunhong Hu, Second Xiangya Hospital of Central South University, Changsha; Min Tao, First Affiliated Hospital of Suzhou University, Suzhou; Qiang Yao, Tianjin People’s Hospital, Tianjin, People’s Republic of China; and S. Peter Eggleton, Merck KGaA, Darmstadt, Germany
| |
Collapse
|
464
|
Survival outcome and prognostic model of patients with colorectal cancer on phase 1 trials. Invest New Drugs 2018; 37:490-497. [PMID: 30315379 DOI: 10.1007/s10637-018-0675-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/02/2018] [Indexed: 12/22/2022]
Abstract
Background Patients with metastatic colorectal cancer (mCRC) who progress on standard therapies may be eligible for phase I trials. To better delineate the risk-benefit ratio, we assessed toxicities, clinical outcomes and prognostic factors. Methods Records of mCRC patients on phase I trials at our institution over 18 years were reviewed. Univariable (UVA) and multivariable analyses (MVA) were undertaken and a prognostic model developed. Results There were 187 enrollments on 37 phase I trials. Median age was: 59 (29-83) years and number of prior therapies: 3 (0-8). The clinical benefit rate (CBR): response (5.6%) + stable disease, was 43.1%. Median progression free survival (PFS) and overall survival (OS) was 7.7 weeks and 43.7 weeks, respectively. The MVA identified age > 60 years (HR 1.63, p < 0.004), albumin<3.5 g/dL (HR 3.69, p < 0.001), direct bilirubin>ULN (HR1.69, p < 0.01), and WBC ≥ 5.2 k/uL (HR 1.97, p < 0.001) as negative prognostic factors. A risk score based on the MVA revealed that patients with a score of 0-1 had an improved OS (58.7 weeks) compared to a score of 2 (49.9 weeks, p < 0.01) and 3 (14.1 weeks, p < 0.001). Conclusions Phase 1 trials may offer similar or better clinical outcome for mCRC patients than standard third line therapies; the prognostic model could assist in selecting appropriate patients.
Collapse
|
465
|
Sreekumar R, Harris S, Moutasim K, DeMateos R, Patel A, Emo K, White S, Yagci T, Tulchinsky E, Thomas G, Primrose JN, Sayan AE, Mirnezami AH. Assessment of Nuclear ZEB2 as a Biomarker for Colorectal Cancer Outcome and TNM Risk Stratification. JAMA Netw Open 2018; 1:e183115. [PMID: 30646224 PMCID: PMC6324431 DOI: 10.1001/jamanetworkopen.2018.3115] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
IMPORTANCE At present, patients with colorectal cancer (CRC) are risk stratified using TNM histologic features. More recently, an association between a mesenchymal phenotype and a high risk of disease recurrence and micrometastases has been recognized. OBJECTIVE To investigate the association of the epithelial to mesenchymal transition (EMT)-inducing transcription factor ZEB2 (zinc finger E box-binding homeobox 2), survival outcomes, and the efficacy of ZEB2 as a biomarker when added as refinement to TNM staging after curative intent surgery for CRC. DESIGN, SETTING, AND PARTICIPANTS ZEB2 expression was assessed using a previously validated scoring system as part of a prospective, observational, masked diagnostic study from January 1, 2008, to December 31, 2013. Data were prospectively collected and analyzed for association with oncologic outcomes from January 1, 2017, to December 31, 2018. An initial test cohort from an academic university medical center of 126 consecutive patients with CRC and, subsequently, an independent validation cohort of 210 patients were examined. ZEB2 positivity was scored by 2 independent, masked pathologists. External validity was tested using an open access gene expression portal. Nomograms were developed with or without ZEB2. MAIN OUTCOMES AND MEASURES Systemic and local recurrence of CRC. RESULTS The test cohort consisted of 126 consecutive patients (mean [SD] age, 72.7 [11.7] years; 61 [48.4%] male) and the validation cohort of 210 patients (mean [SD] age, 72.0 [10.6] years; 111 [52.9%] male). A total of 52 tumors (41.3%) in the test cohort and 104 (49.5%) in the validation cohort were scored nuclear ZEB2 positive. Survival analysis by the log-rank test found that ZEB2 expression was associated with a significant reduction in overall survival and disease-free survival in both cohorts. Cox proportional hazards regression analysis highlighted ZEB2 as an independent biomarker of shorter overall survival and disease-free survival. Analysis of node-negative disease (n = 222) identified ZEB2 as an independent biomarker of early recurrence and reduced survival. External validation confirmed these findings. Addition of ZEB2 expression to nomograms composed of conventional TNM risk factors improved the ability to identify patients at high risk of recurrence demonstrated by the improvement in concordance index in both test (0.73 to 0.77) and validation (0.82 to 0.87) cohorts. CONCLUSIONS AND RELEVANCE The findings suggest that expression of ZEB2 is associated with poor oncologic outcome and distant recurrence. The study also found that the addition of ZEB2 to existing TNM classification improved the ability to stratify patients for risk of recurrence. The results of this study suggest that addition of ZEB2 expression status to the TNM staging system improves the ability to stratify patients at high risk of recurrence.
Collapse
Affiliation(s)
- Rahul Sreekumar
- Cancer Sciences Division, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Department of Surgery, Southampton University Hospital National Health Service Trust, Southampton, United Kingdom
| | - Scott Harris
- Medical Statistics and Mathematics Department, University of Southampton, Southampton, United Kingdom
| | - Karwan Moutasim
- Cancer Sciences Division, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ricardo DeMateos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ashish Patel
- Cancer Sciences Division, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Katherine Emo
- Cancer Sciences Division, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Sophie White
- Department of Surgery, Southampton University Hospital National Health Service Trust, Southampton, United Kingdom
| | - Tamer Yagci
- Department of Molecular Biology and Genetics, Gebze Technical University, Gebze, Turkey
| | - Eugene Tulchinsky
- Department of Cancer Sciences, University of Leicester, Leicester, United Kingdom
| | - Gareth Thomas
- Cancer Sciences Division, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - John N. Primrose
- Cancer Sciences Division, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Department of Surgery, Southampton University Hospital National Health Service Trust, Southampton, United Kingdom
| | - A. Emre Sayan
- Cancer Sciences Division, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Alex H. Mirnezami
- Cancer Sciences Division, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Department of Surgery, Southampton University Hospital National Health Service Trust, Southampton, United Kingdom
| |
Collapse
|
466
|
An Update of Efficacy and Safety of Cetuximab in Metastatic Colorectal Cancer: A Narrative Review. Adv Ther 2018; 35:1497-1509. [PMID: 30218345 DOI: 10.1007/s12325-018-0791-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Indexed: 02/06/2023]
Abstract
Colorectal cancer is the second most common cancer, representing 13% of all diagnosed cancers. Cetuximab is a recombinant chimeric monoclonal IgG1 antibody and epidermal growth factor receptor (EGFR) inhibitor. Cetuximab is approved for the first-line treatment in combination with chemotherapy or as a single agent in patients who have failed or are intolerant to chemotherapy in patients with EGFR-expressing, RAS wild-type metastatic colorectal cancer. Cetuximab efficacy emerged from studies that were conducted to approve the drug. Cetuximab is well tolerated; its toxicities are caused by its mechanism of action and the most common adverse reaction is skin toxicity. The main purpose of this manuscript is to present an update on the evidence-based summary of efficacy and safety and on the cost-effectiveness of cetuximab. Furthermore, it suggests a management of adverse drug reactions to improve the tolerability of the drug.
Collapse
|
467
|
Moati E, Taly V, Didelot A, Perkins G, Blons H, Taieb J, Laurent-Puig P, Zaanan A. Role of circulating tumor DNA in the management of patients with colorectal cancer. Clin Res Hepatol Gastroenterol 2018; 42:396-402. [PMID: 29627453 DOI: 10.1016/j.clinre.2018.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/13/2018] [Accepted: 03/06/2018] [Indexed: 02/04/2023]
Abstract
Colorectal cancer is a major health burden with a prognosis that has been improved with the progresses in diagnosis and the advance of chemotherapy and personalized medicine. However, because of intra-tumor heterogeneity, clonal evolution and selection, tumors often develop resistance to treatments. "Liquid biopsy" is a minimally invasive method, based on analysis of tumor-specific material in peripheral blood samples of patients. Analysis of tumor specific genetic or epigenetic alterations in cell-free circulating nucleic acids may reflect the molecular heterogeneity of the underlying disease process and serial testing could allow to monitor its temporal genomic changing without using re-biopsy. In this review, we focused on the role of circulating tumor DNA (ctDNA) as a biomarker in the management of patients with colorectal cancer at early and advanced stages. Through recent studies, we described its promising clinical applications for diagnosis, detection of recurrence after surgery and monitoring for tumor response or therapeutic resistance in metastatic setting. Such recent developments offer new perspectives for personalized medicine in colorectal cancer but still needs some standardized detection methods and further studies to validate its use in clinical routine.
Collapse
Affiliation(s)
- Emilie Moati
- INSERM UMR-S1147, CNRS SNC5014, Paris Descartes University, Equipe labellisée Ligue Nationale contre le cancer, Paris, France
| | - Valérie Taly
- INSERM UMR-S1147, CNRS SNC5014, Paris Descartes University, Equipe labellisée Ligue Nationale contre le cancer, Paris, France
| | - Audrey Didelot
- INSERM UMR-S1147, CNRS SNC5014, Paris Descartes University, Equipe labellisée Ligue Nationale contre le cancer, Paris, France
| | - Géraldine Perkins
- INSERM UMR-S1147, CNRS SNC5014, Paris Descartes University, Equipe labellisée Ligue Nationale contre le cancer, Paris, France; Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, AP-HP, Paris Descartes University, Paris, France
| | - Hélène Blons
- INSERM UMR-S1147, CNRS SNC5014, Paris Descartes University, Equipe labellisée Ligue Nationale contre le cancer, Paris, France; Department of Biology, European Georges Pompidou Hospital, AP-HP, Paris, France
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, AP-HP, Paris Descartes University, Paris, France
| | - Pierre Laurent-Puig
- INSERM UMR-S1147, CNRS SNC5014, Paris Descartes University, Equipe labellisée Ligue Nationale contre le cancer, Paris, France; Department of Biology, European Georges Pompidou Hospital, AP-HP, Paris, France
| | - Aziz Zaanan
- INSERM UMR-S1147, CNRS SNC5014, Paris Descartes University, Equipe labellisée Ligue Nationale contre le cancer, Paris, France; Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, AP-HP, Paris Descartes University, Paris, France.
| |
Collapse
|
468
|
Jensen CE, Villanueva JY, Loaiza-Bonilla A. Differences in overall survival and mutation prevalence between right- and left-sided colorectal adenocarcinoma. J Gastrointest Oncol 2018; 9:778-784. [PMID: 30505575 DOI: 10.21037/jgo.2018.06.10] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Prior reports have demonstrated inferior outcomes for patients with right-sided colorectal cancer (CRC) compared to patients with left-sided disease, as well as differences in treatment response based on disease sidedness. Differences in prognosis remain even among patients with metastatic disease, indicating that anatomy or stage at diagnosis alone cannot explain all of these findings. While genetic differences between right- and left-sided CRC have long been described, the genetic and molecular drivers underlying differences in prognosis and treatment response remain incompletely understood. Methods We compared mutation prevalence between right- (cecum to splenic flexure) and left-sided (descending colon to rectum) CRC among 38 genes in a retrospective review of next-generation sequencing data of CRC samples obtained in routine clinical practice at a single academic medical center. Results Among 288 cases (167 left-sided, 103 right-sided, 18 synchronous or without clear primary), patients with left-sided primaries had a longer overall survival from pathologic diagnosis (median 1,823 days vs. 1,006 days for right-sided cases, P=0.004). Among the assessed genes, BRAF and CTNNB1 mutations were more prevalent in right-sided CRC. BRAF was mutated in 15.5% of right-sided CRC (95% CI: 8.5-22.5%) compared to 4.8% (95% CI: 1.6-8.0%) (P=0.003). CTNNB1 was mutated in 3.9% of right-sided CRC (95% CI: 0.2-7.6%) compared to no instances of CTNNB1 mutations in left-sided disease (P=0.01). Conclusions This difference in mutation prevalence may implicate these genetic pathways in the mechanisms underlying the discrepant outcomes and treatment responses between right- and left-sided CRC described in this and prior studies.
Collapse
Affiliation(s)
- Christopher E Jensen
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | | | | |
Collapse
|
469
|
Xue WS, Men SY, Liu W, Liu RH. A meta-analysis of safety and efficacy of regorafenib for refractory metastatic colorectal cancer. Medicine (Baltimore) 2018; 97:e12635. [PMID: 30290640 PMCID: PMC6200445 DOI: 10.1097/md.0000000000012635] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Patients with metastatic colorectal cancer (mCRC) often suffer from progressive disease despite previous therapy. It has been a great challenge for those patients. In 2012, regorafenib was approved for mCRC. In this meta-analysis, we aimed to collect and present existing data to explorethe clinical use of regorafenib. METHODS The online electronic databases, such as PubMed, Embase, and the Cochrane library, updated to November 2017 were systematically searched. Trials on the effectiveness of regorafenib in patients who suffer from treatment-refractory metastatic colorectal cancer were included, of which the main outcomes included 3 parameters: overall survival (OS), progression-free survival (PFS), and grade 3/4 AE. RESULTS Totally, 4 trials were included in this meta-analysis. The OD was significantly better with the use of regorafenib (OR = 0.78, 95%CI = 0.65-0.94, I = 69%, P = .008), and PFS (OR = 0.52, 95%CI = 0.34-0.79, I = 97%, P = .002). However, the most common toxicities occurred more frequently in the regorafenib group than the control group (OR = 3.73, 95%CI = 1.68-8.28, I = 79%, P = .001). CONCLUSION Regorafenib demonstrates better efficacy and has manageable adverse-event profile for treatment-refractory mCRC. Considering the safety feature of regorafenib, further studies and clinical trials are warranted to investigate the dosing of regorafenib and alternative approaches are needed to explore predictive biomarker fortherapy selection.
Collapse
Affiliation(s)
| | - Si-Ye Men
- Department of General Surgery, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Liu
- Department of Anorectal Surgery
| | | |
Collapse
|
470
|
Del Carmen S, Sayagués JM, Bengoechea O, Anduaga MF, Alcazar JA, Gervas R, García J, Orfao A, Bellvis LM, Sarasquete ME, Del Mar Abad M. Spatio-temporal tumor heterogeneity in metastatic CRC tumors: a mutational-based approach. Oncotarget 2018; 9:34279-34288. [PMID: 30344942 PMCID: PMC6188146 DOI: 10.18632/oncotarget.26081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 08/10/2018] [Indexed: 12/12/2022] Open
Abstract
It is well known that activating mutations in the KRAS and NRAS genes are associated with poor response to anti-EGFR therapies in patients with metastatic colorectal cancer (mCRC). Approximately half of the patients with wild-type (WT) KRAS colorectal carcinoma do not respond to these therapies. This could be because the treatment decision is determined by the mutational profile of the primary tumor, regardless of the presence of small tumor subclones harboring RAS mutations in lymph nodes or liver metastases. We analyzed the mutational profile of the KRAS, NRAS, BRAF and PI3KCA genes using low-density microarray technology in samples of 26 paired primary tumors, 16 lymph nodes and 34 liver metastases from 26 untreated mCRC patients (n=76 samples). The most frequent mutations found in primary tumors were KRAS (15%) and PI3KCA (15%), followed by NRAS (8%) and BRAF (4%). The distribution of the mutations in the 16 lymph node metastases analyzed was as follows: 4 (25%) in KRAS gene, 3 (19%) in NRAS gene and 1 mutation each in PI3KCA and BRAF genes (6%). As expected, the most prevalent mutation in liver metastasis was in the KRAS gene (35%), followed by PI3KCA (9%) and BRAF (6%). Of the 26 cases studied, 15 (58%) displayed an overall concordance in the mutation status detected in the lymph node metastases and liver metastases compared with primary tumor, suggesting no clonal evolution. In contrast, the mutation profiles differed in the primary tumor and lymph node/metastases samples of the remaining 11 patients (48%), suggesting a spatial and temporal clonal evolution. We confirm the presence of different mutational profiles among primary tumors, lymph node metastases and liver metastases. Our results suggest the need to perform mutational analysis in all available tumor samples of patients before deciding to commence anti-EGFR treatment.
Collapse
Affiliation(s)
- Sofía Del Carmen
- Department of Pathology and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - José María Sayagués
- Cytometry Service-NUCLEUS, Department of Medicine, Cancer Research Center (IBMCC-CSIC/USAL) and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - Oscar Bengoechea
- Department of Pathology and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - María Fernanda Anduaga
- General and Gastrointestinal Surgery Service and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - Jose Antonio Alcazar
- General and Gastrointestinal Surgery Service and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - Ruth Gervas
- Department of Pathology and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - Jacinto García
- General and Gastrointestinal Surgery Service and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - Alberto Orfao
- Cytometry Service-NUCLEUS, Department of Medicine, Cancer Research Center (IBMCC-CSIC/USAL) and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - Luis Muñoz Bellvis
- General and Gastrointestinal Surgery Service and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | | | - María Del Mar Abad
- Department of Pathology and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| |
Collapse
|
471
|
Iwahashi N, Sakai K, Noguchi T, Yahata T, Toujima S, Nishio K, Ino K. A comprehensive gene mutation analysis of liquid biopsy samples from patients with metastatic colorectal cancer to the ovary: A case report. Oncol Lett 2018; 16:6431-6436. [PMID: 30405780 PMCID: PMC6202479 DOI: 10.3892/ol.2018.9467] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 09/17/2018] [Indexed: 01/20/2023] Open
Abstract
Liquid biopsies of circulating tumor DNA (ctDNA) can detect molecular alterations, including tumor-specific mutations, and have recently been used as a non-invasive diagnostic, prognostic, and predictive tool. However, this technique is not commonly used in the gynecological field. Gene mutation profiling of liquid biopsy samples was performed using CAncer Personalized Profiling by deep Sequencing (CAPP-Seq), a novel next-generation sequencing-based approach to ultrasensitive ctDNA detection, in order to make it possible to molecularly diagnose metastatic colorectal cancer to the ovary. Liquid biopsy (plasma) samples and formalin-fixed paraffin-embedded tumor samples were obtained from two patients with ovarian tumors, who had a history of surgery for colorectal cancer, and comprehensive gene mutation profiling was conducted using CAPP-Seq. In patient 1, mutations were identified in the same three regions in both the ovarian tumor and preoperative plasma sample (in the KRAS G13D, APC E1306*, and TP53 H193Y genes). In patient 2, mutation was identified in the same one region in all the primary colorectal tumor, the ovarian tumor, and preoperative plasma sample (in APC R216* gene). These mutations are well-known genetic signatures of colorectal cancer, suggesting that the ovarian tumor was metastatic. Tthe gene mutation patterns of colorectal cancer were examined by subjecting liquid biopsy samples from patients with suspected metastatic ovarian tumors to CAPP-Seq. Gene mutation profiling of liquid biopsy samples can contribute to the preoperative differential diagnosis of metastatic ovarian cancer and its subsequent personalized treatment.
Collapse
Affiliation(s)
- Naoyuki Iwahashi
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama 641-0012, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
| | - Tomoko Noguchi
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama 641-0012, Japan
| | - Tamaki Yahata
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama 641-0012, Japan
| | - Saori Toujima
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama 641-0012, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan
| | - Kazuhiko Ino
- Department of Obstetrics and Gynecology, Wakayama Medical University, Wakayama 641-0012, Japan
| |
Collapse
|
472
|
Additional Biomarkers beyond RAS That Impact the Efficacy of Cetuximab plus Chemotherapy in mCRC: A Retrospective Biomarker Analysis. JOURNAL OF ONCOLOGY 2018; 2018:5072987. [PMID: 30305811 PMCID: PMC6165607 DOI: 10.1155/2018/5072987] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 06/17/2018] [Accepted: 06/27/2018] [Indexed: 12/22/2022]
Abstract
Purpose We aimed to identify new predictive biomarkers for cetuximab in first-line treatment for patients with RAS wild-type metastatic colorectal cancer (mCRC). Methods The study included patients with KRAS wild-type unresectable liver-limited mCRC treated with chemotherapy with or without cetuximab. Next-generation sequencing was done for single nucleotide polymorphism according to custom panel. Potential predictive biomarkers were identified and integrated into a predictive model within a training cohort. The model was validated in a validation cohort. Results Thirty-one of 247(12.6%) patients harbored RAS mutations. In training cohort (N=93), six potential predictive genes, namely, ATP6V1B1, CUL9, ERBB2, LY6G6D, PTCH1, and RBMXL3, were identified. According to predictive model, patients were divided into responsive group (n=66) or refractory group (n=27). In responsive group, efficacy outcomes were significantly improved by addition of cetuximab to chemotherapy. In refractory group, no benefit was observed. Interaction test was significant across all endpoints. In validation cohort (N=123), similar results were also observed. Conclusions In the first-line treatment of mCRC, the predictive model integrating six new predictive mutations divided patients well, indicating a promising approach to further refine patient selection for cetuximab on the basis of RAS mutations.
Collapse
|
473
|
Van der Jeught K, Xu HC, Li YJ, Lu XB, Ji G. Drug resistance and new therapies in colorectal cancer. World J Gastroenterol 2018; 24:3834-3848. [PMID: 30228778 PMCID: PMC6141340 DOI: 10.3748/wjg.v24.i34.3834] [Citation(s) in RCA: 407] [Impact Index Per Article: 58.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/25/2018] [Accepted: 07/16/2018] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is often diagnosed at an advanced stage when tumor cell dissemination has taken place. Chemo- and targeted therapies provide only a limited increase of overall survival for these patients. The major reason for clinical outcome finds its origin in therapy resistance. Escape mechanisms to both chemo- and targeted therapy remain the main culprits. Here, we evaluate major resistant mechanisms and elaborate on potential new therapies. Amongst promising therapies is α-amanitin antibody-drug conjugate targeting hemizygous p53 loss. It becomes clear that a dynamic interaction with the tumor microenvironment exists and that this dictates therapeutic outcome. In addition, CRC displays a limited response to checkpoint inhibitors, as only a minority of patients with microsatellite instable high tumors is susceptible. In this review, we highlight new developments with clinical potentials to augment responses to checkpoint inhibitors.
Collapse
Affiliation(s)
- Kevin Van der Jeught
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Han-Chen Xu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Yu-Jing Li
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Xiong-Bin Lu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
474
|
Kim SL, Min IS, Park YR, Lee ST, Kim SW. Lipocalin 2 inversely regulates TRAIL sensitivity through p38 MAPK-mediated DR5 regulation in colorectal cancer. Int J Oncol 2018; 53:2789-2799. [PMID: 30221676 DOI: 10.3892/ijo.2018.4562] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 08/28/2018] [Indexed: 11/05/2022] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) induces apoptosis through death receptors (DRs)4 and/or 5 expressed on the cell surface. Multiple clinical trials are underway to evaluate the antitumor activity of recombinant human TRAIL and agonistic antibodies to DR4 or DR5. However, their therapeutic potential is limited by the high frequency of cancer resistance. In this study, we provide evidence demonstrating the role of lipocalin 2 (LCN2) in the TRAIL-mediated apoptosis of human colorectal cancer (CRC). By analyzing the mRNA expression data of 71 CRC tissues from patients, we found that DR5 was preferentially expressed in CRC tissues with a low LCN2 expression level compared to tissues with a high LCN2 expression level. Moreover, we analyzed the association between DR5 and LCN2 expression and this analysis revealed that DR5 expression in CRC tended to be inversely associated with LCN2 expression. By contrast, no association was found between the DR4 and LCN2 expression levels. The expression patterns of LCN2 in human CRC cell lines also exhibited an inverse association with DR5 expression. The knockdown of LCN2 by siRNA in the TRAIL‑resistant CRC cells expressing high levels of LCN2 led to a significant increase in TRAIL-induced apoptosis through the upregulation of DR5 protein and mRNA expression. The mechanism through which LCN2 silencing sensitized the CRC cells to TRAIL was dependent on the extrinsic pathway of apoptosis. In addition, we identified that the knockdown of LCN2 enhanced the sensitivity of the cells to TRAIL through the p38 MAPK/CHOP-dependent upregulation of DR5. Taken together, the findings of this study suggest that LCN2 is responsible for TRAIL sensitivity and LCN2 may thus prove to be a promising target protein in DR-targeted CRC therapy.
Collapse
Affiliation(s)
- Se-Lim Kim
- Department of Internal Medicine and Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju 561-712, Korea
| | - In Suk Min
- Department of Internal Medicine and Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju 561-712, Korea
| | - Young Ran Park
- Department of Internal Medicine and Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju 561-712, Korea
| | - Soo Teik Lee
- Department of Internal Medicine and Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju 561-712, Korea
| | - Sang-Wook Kim
- Department of Internal Medicine and Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju 561-712, Korea
| |
Collapse
|
475
|
Weng WH, Leung WH, Pang YJ, Kuo LW, Hsu HH. EPA significantly improves anti-EGFR targeted therapy by regulating miR-378 expression in colorectal cancer. Oncol Lett 2018; 16:6188-6194. [PMID: 30333883 DOI: 10.3892/ol.2018.9408] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 06/26/2018] [Indexed: 01/05/2023] Open
Abstract
It is known that colorectal cancer (CRC) cells containing mutations of the genes KRAS and BRAF are predominate mechanisms causing resistance to epidermal growth factor receptor (EGFR) inhibitors, and commonly exhibit a lower expression of microRNA-378 (miR-378) when compared with the wild type. In the present study, the aim was to determine the possible mechanism which associates miR-378 with the mitogen-activated protein kinase pathway, and to determine the efficiency of eicosapentaenoic acid ethyl ester (EPA) in its ability to restore sensitivity towards cetuximab, an EGFR inhibitor. The results demonstrated that a combined treatment of 40 µM EPA with 0.2 µM cetuximab can significantly suppress the cell growth in KRAS-mutant and control wild-type cells. Furthermore, the higher phosphorylated protein level of extracellular-signal-regulated kinase 1/2 was notable in KRAS EPA-treated cells (P=0.006-0.047) and resulted in significantly increased cell death; however, inconsistent results were indicated in EPA-treated BRAF-mutant cells, compared with the original cells (without treatment). KRAS-mutant and wild-type Caco-2 cells treated with EPA exhibited increased cetuximab response rates, but these response rates were reduced in the BRAF-mutant cells. In conclusion, upregulation of miR-378 induced by EPA may result in the significant restoration of sensitivity to cetuximab in the KRAS-mutant cells. The present data will contribute to a notable potential therapeutic solution for future clinical CRC treatments.
Collapse
Affiliation(s)
- Wen-Hui Weng
- Department of Chemical Engineering and Biotechnology, Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology, Taipei 10608, Taiwan, R.O.C
| | - Wai-Hung Leung
- Department of Chemical Engineering and Biotechnology, Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology, Taipei 10608, Taiwan, R.O.C.,Division of Colorectal Surgery, Department of Surgery, Mackay Memorial Hospital, Taipei 10491, Taiwan, R.O.C
| | - Yeu Jye Pang
- Department of Internal Medicine, Queen Elizabeth The Queen Mother Hospital, Margate CT9 4AN, UK
| | - Li-Wei Kuo
- Department of Chemical Engineering and Biotechnology, Graduate Institute of Biochemical and Biomedical Engineering, National Taipei University of Technology, Taipei 10608, Taiwan, R.O.C
| | - Hsi-Hsien Hsu
- Division of Colorectal Surgery, Department of Surgery, Mackay Memorial Hospital, Taipei 10491, Taiwan, R.O.C
| |
Collapse
|
476
|
Bolaños-Díaz R, Sanabria-Montañez C, Farfán-Tello C, Calderón-Cahua M. Cost-effectiveness of Cetuximab as a treatment strategy for metastatic colon cancer in Peru: chemotherapy/Cetuximab versus chemotherapy alone. JOURNAL OF PHARMACEUTICAL HEALTH SERVICES RESEARCH 2018. [DOI: 10.1111/jphs.12262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
477
|
Musculoskeletal Metastasis from Primary Rectal Cancer: Series of Two Cases of a Very Rare Occurrence with a Short Literature Review. J Gastrointest Cancer 2018; 50:991-996. [PMID: 30175394 DOI: 10.1007/s12029-018-0165-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
478
|
Iwamoto S, Ooki A, Morita S, Hara H, Tanioka H, Satake H, Kataoka M, Kotaka M, Kagawa Y, Nakamura M, Shingai T, Ishikawa M, Miyake Y, Sudo T, Hashiguchi Y, Yabuno T, Sakamoto J, Tsuji A, Ando M, Yamaguchi K. A prospective Phase II study to examine the relationship between quality of life and adverse events of first-line chemotherapy plus cetuximab in patients with KRAS wild-type unresectable metastatic colorectal cancer: QUACK trial. Cancer Med 2018; 7:4217-4227. [PMID: 30051609 PMCID: PMC6144158 DOI: 10.1002/cam4.1623] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/24/2018] [Accepted: 05/28/2018] [Indexed: 12/21/2022] Open
Abstract
A prospective trial has not been performed to investigate associations between quality of life (QOL), adverse events (AEs), and overall survival (OS) in the first-line treatment with cetuximab plus standard chemotherapy for advanced/metastatic colorectal cancer (mCRC). Associations between patient outcome and health-related QOL (HRQOL) together with skin toxicity-related QOL were prospectively evaluated using EORTC QLQ-C30 and DLQI questionnaires. One hundred and forty mCRC patients were analyzed in this study, and 87.8% received pre-emptive skin treatment. Skin toxicity had no clinical impact on HRQOL or skin-related QOL during the first 8 weeks and throughout the study period. An early skin reaction with a grade ≥2 at 8 weeks was significantly associated with a favorable OS compared with a grade of ≤1 (HR, 0.50; 95% CI, 0.24-0.95; P = .035) and was confirmed to be an independent predictor of OS (HR, 0.48; 95% CI, 0.21-0.97; P = .040). Patients symptomatic at baseline who responded to treatment had improved HRQOL compared to nonresponding patients. Severe mucositis/stomatitis had a statistically significant and clinically meaningful negative impact on HRQOL (mean changes from baseline throughout the study period in global health status were -12.64 for a grade of ≥2 vs -0.35 for a grade of 0 or 1 (P = .005)). In conclusion, severe early skin reactions predict favorable OS for patients treated with cetuximab plus chemotherapy without impairing QOL. In addition, mucositis/stomatitis was the most substantial AE compromising both QOL and treatment compliance.
Collapse
Affiliation(s)
| | - Akira Ooki
- Department of GastroenterologySaitama Cancer CenterSaitamaJapan
- Department of Gastroenterological ChemotherapyCancer Institute Hospital of Japanese Foundation for Cancer ResearchTokyoJapan
| | - Satoshi Morita
- Department of Biomedical Statistics and BioinformaticsKyoto UniversityKyotoJapan
| | - Hiroki Hara
- Department of GastroenterologySaitama Cancer CenterSaitamaJapan
| | - Hiroaki Tanioka
- Department of Medical OncologyOkayama Rosai HospitalOkayamaJapan
| | - Hironaga Satake
- Department of Medical OncologyKobe City Medical Center General HospitalKobeJapan
| | - Masato Kataoka
- Department of SurgeryNational Hospital Organization Nagoya Medical CenterNagoyaJapan
| | | | | | | | | | | | - Yasuhiro Miyake
- Department of SurgeryOsaka Minato Central HospitalOsakaJapan
| | - Takeshi Sudo
- Department of SurgeryYamagata Prefectural Central HospitalYamagataJapan
| | | | - Taichi Yabuno
- Department of SurgeryYokohama Municipal Citizen's HospitalYokohamaJapan
| | | | - Akihito Tsuji
- Department of Medical OncologyKagawa UniversityTakamatsuJapan
| | - Masahiko Ando
- Center for Advanced Medicine and Clinical ResearchNagoya UniversityNagoyaJapan
| | - Kensei Yamaguchi
- Department of GastroenterologySaitama Cancer CenterSaitamaJapan
- Department of Gastroenterological ChemotherapyCancer Institute Hospital of Japanese Foundation for Cancer ResearchTokyoJapan
| |
Collapse
|
479
|
Russo G, Pennisi M, Boscarino R, Pappalardo F. Continuous Petri Nets and microRNA Analysis in Melanoma. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 15:1492-1499. [PMID: 28767374 DOI: 10.1109/tcbb.2017.2733529] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Personalized target therapies represent one of the possible treatment strategies to fight the ongoing battle against cancer. New treatment interventions are still needed for an effective and successful cancer therapy. In this scenario, we simulated and analyzed the dynamics of BRAF V600E melanoma patients treated with BRAF inhibitors in order to find potentially interesting targets that may make standard treatments more effective in particularly aggressive tumors that may not respond to selective inhibitor drugs. To this aim, we developed a continuous Petri Net model that simulates fundamental signalling cascades involved in melanoma development, such as MAPK and PI3K/AKT, in order to deeply analyze these complex kinase cascades and predict new crucial nodes involved in melanomagenesis. The model pointed out that some microRNAs, like hsa-mir-132, downregulates expression levels of p120RasGAP: under high concentrations of p120RasGAP, MAPK pathway activation is significantly decreased and consequently also PI3K/PDK1/AKT activation. Furthermore, our analysis carried out through the Genomic Data Commons (GDC) Data Portal shows the evidence that hsa-mir-132 is significantly associated with clinical outcome in melanoma cancer genomic data sets of BRAF-mutated patients. In conclusion, targeting miRNAs through antisense oligonucleotides technology may suggest the way to enhance the action of BRAF-inhibitors.
Collapse
|
480
|
The influence of tumour site on prognosis in metastatic colorectal carcinomas with primary tumour resection. Int J Colorectal Dis 2018; 33:1215-1223. [PMID: 29915904 DOI: 10.1007/s00384-018-3098-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/08/2018] [Indexed: 02/04/2023]
Abstract
PURPOSE The aim of our study was to compare the characteristics and prognosis between right- and left-sided metastatic colorectal carcinomas. METHODS Data from 937 patients with stage IV colorectal carcinomas (synchronous distant metastasis) who had a resection of the primary tumour between 1985 and 2014 were analysed. Carcinomas in the caecum to transverse colon were defined as right-sided (n = 250; 26.7%). They were compared to tumours located from the splenic flexure to the rectum categorised as left-sided (n = 687; 73.3%). RESULTS In right-sided carcinomas, we observed significantly more female patients (50.8 vs 36.2%; p < 0.001), more unfavourable histological types (24.0 vs 8.6%; p < 0.001), more M1c carcinomas (metastases to the peritoneum ± others; 32.0 vs 14.4%; p < 0.001) and more emergencies (11.6 vs 7.1%; p = 0.029), while multimodal treatment was utilised in fewer patients (51.6 vs 63.8%; p = 0.001) and curative resections were less frequently (24.1 vs 35.4%; p = 0.002). Prognosis was significantly worse in patients with right-sided carcinomas (2-year-survival 27.2 vs 44.6%, p < 0.01). This difference was more pronounced after R2 resection (15.3 vs 29.7%; p < 0.001), than after macroscopic curative resection (2-year-survival 63.9 vs 71.9%; p = 0.106). In multivariate Cox regression analysis, tumour site was found to be an independent prognostic factor for overall survival (HR 1.2; 95% CI 1.0-1.5; p = 0.012). During the three 10-year periods, the prognosis improved equally in patients with right- and left-sided carcinomas, while the differences in survival remained identical. CONCLUSIONS In a surgical patient cohort undergoing primary tumour resection, significant differences in prognosis were observed between patients with metastatic right- and left-sided colorectal carcinomas.
Collapse
|
481
|
Ghasabi M, Mansoori B, Mohammadi A, Duijf PH, Shomali N, Shirafkan N, Mokhtarzadeh A, Baradaran B. MicroRNAs in cancer drug resistance: Basic evidence and clinical applications. J Cell Physiol 2018; 234:2152-2168. [PMID: 30146724 DOI: 10.1002/jcp.26810] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/30/2018] [Indexed: 12/19/2022]
Abstract
Development of drug resistance has considerably limited the efficacy of cancer treatments, including chemotherapy and targeted therapies. Hence, understanding the molecular mechanisms underpinning the innate or the acquired resistance to these therapies is critical to improve drug efficiency and clinical outcomes. Several studies have implicated microRNAs (miRNA) in this process. MiRNAs repress gene expression by specific binding to complementary sequences in the 3' region of target messenger RNAs (mRNAs), followed by target mRNA degradation or blocked translation. By targeting molecules specific to a particular pathway within tumor cells, the new generation of cancer treatment strategies has shown significant advantages over conventional chemotherapy. However, the long-term efficacy of targeted therapies often remains poor, because tumor cells develop resistance to such therapeutics. Targeted therapies often involve monoclonal antibodies (mAbs), such as those blocking the ErB/HER tyrosine kinases, epidermal growth factor receptor (cetuximab) and HER2 (trastuzumab), and those inhibiting vascular endothelial growth factor receptor signaling (e.g., bevacizumab). Even though these are among the most used agents in tumor medicine, clinical response to these drugs is reduced due to the emergence of drug resistance as a result of toxic effects in the tumor microenvironment. Research on different types of human cancers has revealed that aberrant expression of miRNAs promotes resistance to the aforementioned drugs. In this study, we review the mechanisms of tumor cell resistance to mAb therapies and the role of miRNAs therein. Emerging treatment strategies combine therapies using innovative miRNA mimics or antagonizers with conventional approaches to maximize outcomes of patients with cancer.
Collapse
Affiliation(s)
- Mehri Ghasabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pascal Hg Duijf
- University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naghmeh Shirafkan
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
482
|
Ahlborn LB, Tuxen IV, Mouliere F, Kinalis S, Schmidt AY, Rohrberg KS, Santoni-Rugiu E, Nielsen FC, Lassen U, Yde CW, Oestrup O, Mau-Sorensen M. Circulating tumor DNA as a marker of treatment response in BRAF V600E mutated non-melanoma solid tumors. Oncotarget 2018; 9:32570-32579. [PMID: 30220966 PMCID: PMC6135692 DOI: 10.18632/oncotarget.25948] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/29/2018] [Indexed: 01/31/2023] Open
Abstract
Purpose We evaluated longitudinal tracking of BRAF V600E in circulating cell-free DNA (cfDNA) as a marker of treatment response to BRAF inhibitor (BRAFi) combination therapies in non-melanoma solid tumors included in the Copenhagen Prospective Personalized Oncology (CoPPO) program. Experimental design Patients with BRAF V600E-mutated tumors were treated with combination therapies including BRAFi. Quantification of mutant cfDNA from plasma was determined and correlated to clinical outcomes. Exome sequencing was performed to identify possible resistance mutations. Results Twenty-three patients had BRAF-mutated tumors out of 455 patients included in CoPPO and 17 started BRAFi combination (EGFRi/MEKi) therapy. Tumor responses were achieved in 8 out of 16 evaluable patients and the median overall- and progression-free survival (OS and PFS) was 15 and 4.8 months, respectively. Longitudinal measurements of BRAF V600E-mutant cfDNA indicated disease progression prior to radiological evaluation and a reduction in the mutant fraction of more than 50% after 4 and 12 weeks of therapy was associated with a significantly longer PFS (p=0.003 and p=0.029) and OS (p=0.029 and p=0.017). Furthermore, the baseline mutant fraction and total level of cfDNA positively correlated with tumor burden (p=0.026 and p=0.024). Finally, analysis of cfDNA at progression revealed novel mutations potentially affecting the MAPK pathway. Conclusion BRAFi combination therapies showed a response rate of 50% in BRAF V600E-mutated non-melanoma tumors. The fraction of BRAF-mutant cfDNA represent a sensitive indicator for clinical outcomes with plasma collected at week 4 and 12 as crucial time points for monitoring response and disease progression.
Collapse
Affiliation(s)
- Lise Barlebo Ahlborn
- The Phase I Unit, Department of Oncology, Rigshospitalet, Copenhagen University, Copenhagen, Denmark.,Center for Genomic Medicine, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Ida Viller Tuxen
- The Phase I Unit, Department of Oncology, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Florent Mouliere
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Savvas Kinalis
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Ane Y Schmidt
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Kristoffer Staal Rohrberg
- The Phase I Unit, Department of Oncology, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Eric Santoni-Rugiu
- Department of Pathology, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Finn Cilius Nielsen
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Ulrik Lassen
- The Phase I Unit, Department of Oncology, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | | | - Olga Oestrup
- Center for Genomic Medicine, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Morten Mau-Sorensen
- The Phase I Unit, Department of Oncology, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
483
|
Kim CA, Ahmed S, Ahmed S, Brunet B, Chalchal H, Deobald R, Doll C, Dupre MP, Gordon V, Lee-Ying RM, Lim H, Liu D, Loree JM, McGhie JP, Mulder K, Park J, Yip B, Wong RP, Zaidi A. Report from the 19th annual Western Canadian Gastrointestinal Cancer Consensus Conference; Winnipeg, Manitoba; 29-30 September 2017. ACTA ACUST UNITED AC 2018; 25:275-284. [PMID: 30111968 DOI: 10.3747/co.25.4109] [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: 11/15/2022]
Abstract
The 19th annual Western Canadian Gastrointestinal Cancer Consensus Conference (wcgccc) was held in Winnipeg, Manitoba, 29-30 September 2017. The wcgccc is an interactive multidisciplinary conference attended by health care professionals from across Western Canada (British Columbia, Alberta, Saskatchewan, and Manitoba) who are involved in the care of patients with gastrointestinal cancer. Surgical, medical, and radiation oncologists; pathologists; radiologists; and allied health care professionals participated in presentation and discussion sessions for the purpose of developing the recommendations presented here. This consensus statement addresses current issues in the management of colorectal cancer.
Collapse
Affiliation(s)
- C A Kim
- Manitoba-Medical Oncology (Kim, Gordon, Wong) and Radiation Oncology (Shahida Ahmed), CancerCare Manitoba, University of Manitoba, Winnipeg; Surgery (Park, Yip) and Pathology (Dupre), University of Manitoba, Winnipeg
| | - S Ahmed
- Saskatchewan- Medical Oncology (Shahid Ahmed, Zaidi), Radiation Oncology (Brunet), and Surgery (Deobald), Saskatchewan Cancer Agency, University of Saskatchewan, Saskatoon; Medical Oncology (Chalchal), Allan Blair Cancer Centre, Regina
| | - S Ahmed
- Manitoba-Medical Oncology (Kim, Gordon, Wong) and Radiation Oncology (Shahida Ahmed), CancerCare Manitoba, University of Manitoba, Winnipeg; Surgery (Park, Yip) and Pathology (Dupre), University of Manitoba, Winnipeg
| | - B Brunet
- Saskatchewan- Medical Oncology (Shahid Ahmed, Zaidi), Radiation Oncology (Brunet), and Surgery (Deobald), Saskatchewan Cancer Agency, University of Saskatchewan, Saskatoon; Medical Oncology (Chalchal), Allan Blair Cancer Centre, Regina
| | - H Chalchal
- Saskatchewan- Medical Oncology (Shahid Ahmed, Zaidi), Radiation Oncology (Brunet), and Surgery (Deobald), Saskatchewan Cancer Agency, University of Saskatchewan, Saskatoon; Medical Oncology (Chalchal), Allan Blair Cancer Centre, Regina
| | - R Deobald
- Saskatchewan- Medical Oncology (Shahid Ahmed, Zaidi), Radiation Oncology (Brunet), and Surgery (Deobald), Saskatchewan Cancer Agency, University of Saskatchewan, Saskatoon; Medical Oncology (Chalchal), Allan Blair Cancer Centre, Regina
| | - C Doll
- Alberta-Medical Oncology (Mulder), Cross Cancer Institute, University of Alberta, Edmonton; Medical Oncology (Lee-Ying) and Radiation Oncology (Doll), Tom Baker Cancer Centre, University of Calgary, Calgary
| | - M P Dupre
- Manitoba-Medical Oncology (Kim, Gordon, Wong) and Radiation Oncology (Shahida Ahmed), CancerCare Manitoba, University of Manitoba, Winnipeg; Surgery (Park, Yip) and Pathology (Dupre), University of Manitoba, Winnipeg
| | - V Gordon
- Manitoba-Medical Oncology (Kim, Gordon, Wong) and Radiation Oncology (Shahida Ahmed), CancerCare Manitoba, University of Manitoba, Winnipeg; Surgery (Park, Yip) and Pathology (Dupre), University of Manitoba, Winnipeg
| | - R M Lee-Ying
- Alberta-Medical Oncology (Mulder), Cross Cancer Institute, University of Alberta, Edmonton; Medical Oncology (Lee-Ying) and Radiation Oncology (Doll), Tom Baker Cancer Centre, University of Calgary, Calgary
| | - H Lim
- British Columbia-Medical Oncology (Lim, Loree), BC Cancer, University of British Columbia, Vancouver; Medical Oncology (McGhie), BC Cancer, University of British Columbia, Victoria; Radiology (Liu), University of British Columbia, Vancouver
| | - D Liu
- British Columbia-Medical Oncology (Lim, Loree), BC Cancer, University of British Columbia, Vancouver; Medical Oncology (McGhie), BC Cancer, University of British Columbia, Victoria; Radiology (Liu), University of British Columbia, Vancouver
| | - J M Loree
- British Columbia-Medical Oncology (Lim, Loree), BC Cancer, University of British Columbia, Vancouver; Medical Oncology (McGhie), BC Cancer, University of British Columbia, Victoria; Radiology (Liu), University of British Columbia, Vancouver
| | - J P McGhie
- British Columbia-Medical Oncology (Lim, Loree), BC Cancer, University of British Columbia, Vancouver; Medical Oncology (McGhie), BC Cancer, University of British Columbia, Victoria; Radiology (Liu), University of British Columbia, Vancouver
| | - K Mulder
- Alberta-Medical Oncology (Mulder), Cross Cancer Institute, University of Alberta, Edmonton; Medical Oncology (Lee-Ying) and Radiation Oncology (Doll), Tom Baker Cancer Centre, University of Calgary, Calgary
| | - J Park
- Manitoba-Medical Oncology (Kim, Gordon, Wong) and Radiation Oncology (Shahida Ahmed), CancerCare Manitoba, University of Manitoba, Winnipeg; Surgery (Park, Yip) and Pathology (Dupre), University of Manitoba, Winnipeg
| | - B Yip
- Manitoba-Medical Oncology (Kim, Gordon, Wong) and Radiation Oncology (Shahida Ahmed), CancerCare Manitoba, University of Manitoba, Winnipeg; Surgery (Park, Yip) and Pathology (Dupre), University of Manitoba, Winnipeg
| | - R P Wong
- Manitoba-Medical Oncology (Kim, Gordon, Wong) and Radiation Oncology (Shahida Ahmed), CancerCare Manitoba, University of Manitoba, Winnipeg; Surgery (Park, Yip) and Pathology (Dupre), University of Manitoba, Winnipeg
| | - A Zaidi
- Saskatchewan- Medical Oncology (Shahid Ahmed, Zaidi), Radiation Oncology (Brunet), and Surgery (Deobald), Saskatchewan Cancer Agency, University of Saskatchewan, Saskatoon; Medical Oncology (Chalchal), Allan Blair Cancer Centre, Regina
| | | |
Collapse
|
484
|
Li S, Fang XD, Wang XY, Fei BY. Fos-like antigen 2 (FOSL2) promotes metastasis in colon cancer. Exp Cell Res 2018; 373:57-61. [PMID: 30114390 DOI: 10.1016/j.yexcr.2018.08.016] [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: 04/28/2018] [Revised: 08/10/2018] [Accepted: 08/11/2018] [Indexed: 01/02/2023]
Abstract
Among different cancers, incidence and mortality of colorectal cancer (CRC) is one of the highest. KRAS mutation is one of the underlying features in the pathogenesis of CRC with CRC tumors harboring mutant KRAS exhibiting a more aggressive behavior compared to CRC tumors with wild type KRAS. We had earlier shown that the microRNA-143 (miR-143) replenishment not only chemosensitizers CRC cell line with mutant KRAS instead of wild-type KRAS gene, to paclitaxel-mediated cytotoxicity, but also inhibits cell migration and invasion ability. Hence, the study aimed to determine how miR-143 replenishment is inhibiting pre-metastatic behavior in CRC cells with mutant KRAS. Top ten mRNA targets of miR-143 as predicted by TargetScan were evaluated by qRT-PCR in LoVo cells which were performed mock transfection or miR-143 mimic transfection. Evaluation of the changes in cognate mRNA target(s) was done in 30 paired CRC tissue and tumor adjacent normal tissue specimens and in LoVo cells by western blot. Effect of the mRNA target on pro-metastatic behavior was assayed by gain- and loss-of-function studies using a combination of western blotting and in vitro cell proliferation and transwell migration/invasion assay in LoVo cells and in the normal colonic epithelium cell line FHC. In vivo effect of the cognate mRNA target on CRC metastasis was assayed by xenograft assay. Of the 10 predicted mRNA targets, FOSL2 (P < 0.05) and IGFBP5 (P > 0.05) was down regulated in LoVo cells transfected with the miR-143 mimic. FOSL2 mRNA levels were significantly downregulated in CRC tissue specimens compared with adjacent normal tissue (P < 0.05). Immunoblot analysis showed that FOSL2, but not IGFBP5, protein expression is down regulated in LoVo cells after the miR-143 mimic transfection. FOSL2 overexpression in the normal colonic epithelial cell line FHC or siRNA-mediated silencing in LoVo cells induced and repressed, respectively, pro-mesenchymal cell features. Whereas manipulation of FOSL2 expression did not have any effect on cell proliferation rates, silencing its expression inhibited cell migration and invasion ability in vitro. In addition, silencing of FOSL2 expression in the LoVo cells can significantly inhibited invasion of hepatic, while no effect was found for tumorigenic potential. Our results suggest that FOSL2 is a critical regulator of CRC metastasis and might be an important marker for prognostic in CRC patients.
Collapse
Affiliation(s)
- Shuo Li
- Department of Hepatobiliary & Pancreatic Surgery, The China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Xue-Dong Fang
- Department of Gastrointestinal Colorectal and anal Surgery, The China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| | - Xiu-Ying Wang
- Medical Record Department, The China Japan Union Hospital of Jilin University, Changchun 130021, China
| | - Bing-Yuan Fei
- Department of Gastrointestinal Colorectal and anal Surgery, The China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
485
|
Sánchez-Botet A, Gasa L, Quandt E, Hernández-Ortega S, Jiménez J, Mezquita P, Carrasco-García MÀ, Kron SJ, Vidal A, Villanueva A, Ribeiro MPC, Clotet J. The atypical cyclin CNTD2 promotes colon cancer cell proliferation and migration. Sci Rep 2018; 8:11797. [PMID: 30087414 PMCID: PMC6081371 DOI: 10.1038/s41598-018-30307-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 07/27/2018] [Indexed: 01/20/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide, with 8–10% of these tumours presenting a BRAF (V600E) mutation. Cyclins are known oncogenes deregulated in many cancers, but the role of the new subfamily of atypical cyclins remains elusive. Here we have performed a systematic analysis of the protein expression levels of eight atypical cyclins in human CRC tumours and several cell lines, and found that CNTD2 is significantly upregulated in CRC tissue compared to the adjacent normal one. CNTD2 overexpression in CRC cell lines increases their proliferation capacity and migration, as well as spheroid formation capacity and anchorage-independent growth. Moreover, CNTD2 increases tumour growth in vivo on xenograft models of CRC with wild-type BRAF. Accordingly, CNTD2 downregulation significantly diminished the proliferation of wild-type BRAF CRC cells, suggesting that CNTD2 may represent a new prognostic factor and a promising drug target in the management of CRC.
Collapse
Affiliation(s)
- Abril Sánchez-Botet
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Laura Gasa
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Eva Quandt
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Sara Hernández-Ortega
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Javier Jiménez
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Pau Mezquita
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Miquel Àngel Carrasco-García
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain.,Pathology Department, Hospital Universitari General de Catalunya, Sant Cugat del Vallès, Barcelona, Spain
| | - Stephen J Kron
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, USA
| | - August Vidal
- Department of Pathology, University Hospital of Bellvitge, Bellvitge Biomedical Research Institute (IDIBELL), CIBERONC, L'Hospitalet del Llobregat, Barcelona, Spain.,Xenopat S.L., Business Bioincubator, Bellvitge Health Science Campus, Barcelona, Spain
| | - Alberto Villanueva
- Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO) Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - Mariana P C Ribeiro
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain.
| | - Josep Clotet
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain.
| |
Collapse
|
486
|
Wu Z, Guo HF, Xu H, Cheung NKV. Development of a Tetravalent Anti-GPA33/Anti-CD3 Bispecific Antibody for Colorectal Cancers. Mol Cancer Ther 2018; 17:2164-2175. [PMID: 30082472 DOI: 10.1158/1535-7163.mct-18-0026] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 06/17/2018] [Accepted: 07/31/2018] [Indexed: 01/09/2023]
Abstract
Despite progress in the treatment of colorectal cancer, curing metastatic colorectal cancer remains a major unmet medical need worldwide. Here, we describe a T-cell-engaging bispecific antibody (T-BsAb) to redirect polyclonal cytotoxic T cells to eradicate colorectal cancer. A33, a murine antibody specific for GPA33, was humanized to huA33 and reformatted to huA33-BsAb, based on a novel IgG(L)-scFv platform by linking the anti-CD3 huOKT3 scFv to the carboxyl end of the light chain. This T-BsAb was stably expressed in CHO cells and purified as a stable monomer by HPLC, retaining immunoreactivity by FACS through 30 days of incubation at 37°C. In vitro, it induced activation and expansion of unstimulated T cells and elicited potent T-cell-dependent cell-mediated cytotoxicity against colon and gastric cancer cells in an antigen-specific manner. In vivo, huA33-BsAb inhibited the colon and gastric cancer xenografts, in both subcutaneous and intraperitoneal tumor models. More importantly, both microsatellite instable and microsatellite stable colorectal cancer were effectively eliminated by huA33-BsAb. These preclinical results provide further support for the use of IgG(L)-scFv platform to build BsAb, and especially one targeting GPA33 for colorectal cancer. These preclinical results also support further development of huA33-BsAb as a potential immunotherapeutic. Mol Cancer Ther; 17(10); 2164-75. ©2018 AACR.
Collapse
Affiliation(s)
- Zhihao Wu
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hong-Fen Guo
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hong Xu
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | |
Collapse
|
487
|
Price TJ, Tang M, Gibbs P, Haller DG, Peeters M, Arnold D, Segelov E, Roy A, Tebbutt N, Pavlakis N, Karapetis C, Burge M, Shapiro J. Targeted therapy for metastatic colorectal cancer. Expert Rev Anticancer Ther 2018; 18:991-1006. [PMID: 30019590 DOI: 10.1080/14737140.2018.1502664] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Outcomes in metastatic colorectal cancer are improving, with better understanding and use of targeted therapies. Areas covered: A review of the literature and recent conference presentations was undertaken on the topic of systemic treatment of metastatic colorectal cancer. This article reviews the current evidence for targeted therapies in advanced colorectal cancer, including up-to-date data regarding anti-epidermal growth factor receptor (EGFR) and anti-vascular endothelial growth factor (VEGF) agents, the relevance of primary tumor location and novel subgroups such as BRAF mutated, HER2 amplified, and mismatch-repair-deficient cancers. Expert commentary: EGFR-targeted and VEGF-targeted antibodies are now routinely incorporated into treatment strategies for metastatic colorectal cancer (mCRC). The use of EGFR-targeted antibodies should be restricted to patients with extended RAS wild-type profiles, and there is evidence that they should be further restricted to patients with left-sided tumors. Clinically, mCRC can be divided into subgroups based on RAS, BRAF, HER2, and MMR status, each of which have distinct treatment pathways.
Collapse
Affiliation(s)
- Timothy J Price
- a Medical Oncology , The Queen Elizabeth Hospital, Woodville, and University of Adelaide , Adelaide , Australia
| | - Monica Tang
- b Medical Oncology , NHMRC Clinical Trials Centre, University of Sydney , Sydney , Australia
| | - Peter Gibbs
- c Medical Oncology , Western Hospital , Melbourne , Australia.,d Medical Oncology , Walter and Eliza Hall Institute , Melbourne , Australia
| | - Daniel G Haller
- e Medical Oncology , Abrahamson Cancer Centre at the Perelman School of Medicine, University of Pennsylvania , Philadelphia , USA
| | - Marc Peeters
- f Medical Oncology , University Hospital Antwerp, Edegem, Belgiumg Asklepios Tumorzentrum Hamburg , Hamburg , Germany
| | - Dirk Arnold
- g Medical Oncology , Asklepios Tumorzentrum Hamburg , Germany
| | - Eva Segelov
- h Medical Oncology , Monash University School of Clinical Sciences at Monash Health, Monash Medical Centre , Clayton , Australia
| | - Amitesh Roy
- i Medical Oncology , Flinders Centre for Innovation in Cancer , Bedford Park , Australia.,j Medical Oncology , Flinders University , Bedford Park , Australia
| | - Niall Tebbutt
- k Medical Oncology , Austin Health , Heidelberg , Australia
| | - Nick Pavlakis
- l Medical Oncology , Royal North Shore Hospital , St Leonards , Australia
| | - Chris Karapetis
- i Medical Oncology , Flinders Centre for Innovation in Cancer , Bedford Park , Australia
| | - Matthew Burge
- m Medical Oncology , Royal Brisbane Hospital , Brisbane , Australia
| | - Jeremy Shapiro
- n Medical Oncology , Cabrini Hospital and Monash University , Melbourne , Australia
| |
Collapse
|
488
|
Hu P, Zhang S, Wu T, Ni D, Fan W, Zhu Y, Qian R, Shi J. Fe-Au Nanoparticle-Coupling for Ultrasensitive Detections of Circulating Tumor DNA. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1801690. [PMID: 29931715 DOI: 10.1002/adma.201801690] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/02/2018] [Indexed: 05/15/2023]
Abstract
Effectiveness of cancer therapy relies heavily on the efficient early diagnosis. Circulating tumor DNA (ctDNA) detection is one of the most clinically meaningful liquid biopsy approaches for the noninvasive cancer early diagnosis, which, unfortunately, cannot be applied as a routine diagnostic tool till a number of obstacles, for example, unsatisfactory specificity and sensitivity, and extremely high costs, are overcome. Here, the first paradigm of nanomaterial's application in the extremely specific, ultrasensitive, and yet economical ctDNA detections is reported based on a facile nanoparticle-coupling strategy without amplification, with which polymerase chain reaction (PCR)-introduced bias and other shortcomings are successfully circumvented. Aiming at seven Kirsten rat sarcoma-2 virus (KRAS) point mutations, the present strategy exhibits high specificity and an ultrahigh sensitivity of detecting as low as 0.1 pg mL-1 of KRAS point mutation without prior PCR amplification. Discriminating KRAS gene mutations in lung adenocarcinoma patients at an extremely low detection limit equivalent to 0.12% mutation relative to wild-type gene is successful. It is envisioned that this nanoparticle-coupling approach could be routinely applied clinically for ultra-early diagnosis and monitoring of diverse malignant tumors, thus facilitating the fight against cancer.
Collapse
Affiliation(s)
- Ping Hu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Shengjian Zhang
- Department of Radiology, Cancer Hospital/Institute and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P. R. China
| | - Tong Wu
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Dalong Ni
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Wenpei Fan
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Yan Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Rong Qian
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
489
|
Lindsay CR, Jamal-Hanjani M, Forster M, Blackhall F. KRAS: Reasons for optimism in lung cancer. Eur J Cancer 2018; 99:20-27. [PMID: 29894909 DOI: 10.1016/j.ejca.2018.05.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/21/2018] [Accepted: 05/13/2018] [Indexed: 01/07/2023]
Abstract
Despite being the most frequent gain-of-function genetic alteration in human cancer, KRAS mutation has to date offered only limited potential as a prognostic and predictive biomarker. Results from the phase III SELECT-1 trial in non-small cell lung cancer (NSCLC) recently added to a number of historical and more contemporary disappointments in targeting KRAS mutant disease, including farnesyl transferase inhibition and synthetic lethality partners such as STK33. This narrative review uses the context of these previous failures to demonstrate how the knowledge gained from these experiences can be used as a platform for exciting advances in NSCLC on the horizon. It now seems clear that mutational subtype (most commonly G12C) of individual mutations is of greater relevance than the categorical evaluation of KRAS mutation presence or otherwise. A number of direct small molecules targeted to these subtypes are in development and have shown promising biological activity, with some in the late stages of preclinical validation.
Collapse
Affiliation(s)
- C R Lindsay
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, UK; Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, London and Manchester, UK.
| | - M Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, London and Manchester, UK; Department of Oncology, University College of London Hospital and UCL Cancer Institute, London, UK
| | - M Forster
- Cancer Research UK Lung Cancer Centre of Excellence, London and Manchester, UK; Department of Oncology, University College of London Hospital and UCL Cancer Institute, London, UK
| | - F Blackhall
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester, UK; Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, London and Manchester, UK
| |
Collapse
|
490
|
Harty G, Jarrett J, Jofre-Bonet M. Consequences of Biomarker Analysis on the Cost-Effectiveness of Cetuximab in Combination with FOLFIRI as a First-Line Treatment of Metastatic Colorectal Cancer: Personalised Medicine at Work. APPLIED HEALTH ECONOMICS AND HEALTH POLICY 2018; 16:515-525. [PMID: 29948926 PMCID: PMC6028886 DOI: 10.1007/s40258-018-0395-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
BACKGROUND Therapies may be more efficacious when targeting a patient subpopulation with specific attributes, thereby enhancing the cost-effectiveness of treatment. In the CRYSTAL study, patients with metastatic colorectal cancer (mCRC) were treated with cetuximab plus FOLFIRI or FOLFIRI alone until disease progression, unacceptable toxic effects or withdrawal of consent. OBJECTIVE To determine if stratified use of cetuximab based on genetic biomarker detection improves cost-effectiveness. METHODS We used individual patient data from CRYSTAL to compare the cost-effectiveness, cost per life-year (LY) and cost per quality-adjusted LY (QALY) gained of cetuximab plus FOLFIRI versus FOLFIRI alone in three cohorts of patients with mCRC: all randomised patients (intent-to-treat; ITT), tumours with no detectable mutations in codons 12 and 13 of exon 2 of the KRAS protein ('KRAS wt') and no detectable mutations in exons 2, 3 and 4 of KRAS and exons 2, 3 and 4 of NRAS ('RAS wt'). Survival analysis was conducted using RStudio, and a cost-utility model was modified to allow comparison of the three cohorts. RESULTS The deterministic base-case ICER (cost per QALY gained) was £130,929 in the ITT, £72,053 in the KRAS wt and £44,185 in the RAS wt cohorts for cetuximab plus FOLFIRI compared with FOLFIRI alone. At a £50,000 willingness-to-pay threshold, cetuximab plus FOLFIRI has a 2.8, 20 and 63% probability of being cost-effective for the ITT, KRAS wt and RAS wt cohorts, respectively, versus FOLFIRI alone. CONCLUSION Screening for mutations in both KRAS and NRAS may provide the most cost-effective approach to patient selection.
Collapse
Affiliation(s)
- Gerard Harty
- Merck Serono Ltd, Bedfont Cross, Stanwell Road, Feltham, Middlesex, TW14 8NX, UK
| | - James Jarrett
- Mapi Group, Ltd, 73 Collier Street, London, N1 9BE, UK
| | | |
Collapse
|
491
|
Monoclonal antibodies as immunomodulatory therapy against cancer and autoimmune diseases. Curr Opin Pharmacol 2018; 41:114-121. [DOI: 10.1016/j.coph.2018.05.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/22/2022]
|
492
|
Margonis GA, Buettner S, Andreatos N, Kim Y, Wagner D, Sasaki K, Beer A, Schwarz C, Løes IM, Smolle M, Kamphues C, He J, Pawlik TM, Kaczirek K, Poultsides G, Lønning PE, Cameron JL, Burkhart RA, Gerger A, Aucejo FN, Kreis ME, Wolfgang CL, Weiss MJ. Association of BRAF Mutations With Survival and Recurrence in Surgically Treated Patients With Metastatic Colorectal Liver Cancer. JAMA Surg 2018; 153:e180996. [PMID: 29799910 DOI: 10.1001/jamasurg.2018.0996] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Importance BRAF mutations are reportedly associated with aggressive tumor biology. However, in contrast with primary colorectal cancer, the association of V600E and non-V600E BRAF mutations with survival and recurrence after resection of colorectal liver metastases (CRLM) has not been well studied. Objective To investigate the prognostic association of BRAF mutations with survival and recurrence independently and compared with other prognostic determinants, such as KRAS mutations. Design, Setting, and Participants In this cohort study, all patients who underwent resection for CRLM with curative intent from January 1, 2000, through December 31, 2016, at the institutions participating in the International Genetic Consortium for Colorectal Liver Metastasis and had data on BRAF and KRAS mutational status were retrospectively identified. Multivariate Cox proportional hazards regression models were used to assess long-term outcomes. Interventions Hepatectomy in patients with CRLM. Main Outcomes and Measures The association of V600E and non-V600E BRAF mutations with disease-free survival (DFS) and overall survival (OS). Results Of 853 patients who met inclusion criteria (510 men [59.8%] and 343 women [40.2%]; mean [SD] age, 60.2 [12.4] years), 849 were included in the study analyses. Forty-three (5.1%) had a mutated (mut) BRAF/wild-type (wt) KRAS (V600E and non-V600E) genotype; 480 (56.5%), a wtBRAF/wtKRAS genotype; and 326 (38.4%), a wtBRAF/mutKRAS genotype. Compared with the wtBRAF/wtKRAS genotype group, patients with a mutBRAF/wtKRAS genotype more frequently were female (27 [62.8%] vs 169 [35.2%]) and 65 years or older (22 [51.2%] vs 176 [36.9%]), had right-sided primary tumors (27 [62.8%] vs 83 [17.4%]), and presented with a metachronous liver metastasis (28 [64.3%] vs 229 [46.8%]). On multivariable analysis, V600E but not non-V600E BRAF mutation was associated with worse OS (hazard ratio [HR], 2.76; 95% CI, 1.74-4.37; P < .001) and DFS (HR, 2.04; 95% CI, 1.30-3.20; P = .002). The V600E BRAF mutation had a stronger association with OS and DFS than the KRAS mutations (β for OS, 10.15 vs 2.94; β for DFS, 7.14 vs 2.27). Conclusions and Relevance The presence of the V600E BRAF mutation was associated with worse prognosis and increased risk of recurrence. The V600E mutation was not only a stronger prognostic factor than KRAS but also was the strongest prognostic determinant in the overall cohort.
Collapse
Affiliation(s)
| | - Stefan Buettner
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Surgery, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Nikolaos Andreatos
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yuhree Kim
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Doris Wagner
- Department of General Surgery, Medical University of Graz, Graz, Austria
| | - Kazunari Sasaki
- Department of General Surgery, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio
| | - Andrea Beer
- Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Christoph Schwarz
- Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Inger Marie Løes
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Maria Smolle
- Division for Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Carsten Kamphues
- Department of General, Visceral and Vascular Surgery, Charité Campus Benjamin Franklin, University of Berlin-Charité, Berlin, Germany
| | - Jin He
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus.,Deputy Editor
| | - Klaus Kaczirek
- Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - George Poultsides
- Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Per Eystein Lønning
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - John L Cameron
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Richard A Burkhart
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Armin Gerger
- Division for Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Federico N Aucejo
- Department of General Surgery, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio
| | - Martin E Kreis
- Department of General, Visceral and Vascular Surgery, Charité Campus Benjamin Franklin, University of Berlin-Charité, Berlin, Germany
| | | | - Matthew J Weiss
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
493
|
Shulman K, Barnett-Griness O, Friedman V, Greenson JK, Gruber SB, Lejbkowicz F, Rennert G. Outcomes of Chemotherapy for Microsatellite Instable-High Metastatic Colorectal Cancers. JCO Precis Oncol 2018; 2:PO.17.00253. [PMID: 32913995 PMCID: PMC7446482 DOI: 10.1200/po.17.00253] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
PURPOSE Microsatellite instable-high (MSI-H) colorectal cancers (CRCs) are known to carry better survival in the local disease stage even without treatment. The influence of types of treatment on survival of MSI-H metastatic CRCs (mCRCs) is still unclear and is evaluated in this study. MATERIALS AND METHODS Patients with MSI-H mCRC treated with first-line chemotherapy, with or without bevacizumab, identified in the Israeli population-based Molecular Epidemiology of Colorectal Cancer (MECC) study, were diagnosed between 1998 and 2013 and followed up until May 2017; MSI status was determined by comparing 10 markers in tumor and normal tissue. Dates of metastases and death and treatment details were extracted from oncology records. RESULTS Among 590 patients treated for mCRC, 106 (18%) had MSI-H tumors. Patients with MSI-H had a median overall survival (OS, from start of first-line treatment) of 1.6 years. The presence of a somatic B-Raf proto-oncogene (BRAF) mutation was a significant adverse prognostic factor in the MSI-H group (hazard ratio [HR], 1.8; 95% CI, 1.1 to 3.0; P = .026). MSI-H tumors without BRAF mutation (n = 87) had similar OS benefit from fluorouracil (FU) only as from any combination protocols (HR, 0.93; P = .78), whereas microsatellite-stable (MSS) tumors without BRAF mutation (n = 456) showed improved OS over FU-only regimens when combination chemotherapy with or without bevacizumab was used (HR, 0.58; P < .01; P value for interaction = .07). Patients with MSI-H/BRAF wild type (WT) had survival advantage over patients with MSS disease (adjusted HR, 0.58; 95% CI, 0.35 to 0.98) when treated with FU-only protocols. CONCLUSION Clinical outcomes differ substantially between patients with MSS/BRAF-WT mCRC and MSI-H/BRAF-WT mCRC, with measurable differences between chemotherapy regimens. MSI-H mCRCs are a clinically distinct subset of colorectal cancers. Their current poor outcome suggests that new clinical trials are needed to identify therapeutic options, potentially taking advantage of the new developments in the field of immunotherapy.
Collapse
Affiliation(s)
- Katerina Shulman
- Katerina Shulman, Hillel Yaffe Medical Center, Hadera; Katerina Shulman, Ofra Barnett-Griness, Vered Friedman, Flavio Lejbkowicz, and Gad Rennert, Carmel Medical Center, and Technion, Haifa, Israel; Joel K. Greenson, University of Michigan, Ann Arbor, MI; and Stephen B. Gruber, University of Southern California, Los Angeles, CA
| | - Ofra Barnett-Griness
- Katerina Shulman, Hillel Yaffe Medical Center, Hadera; Katerina Shulman, Ofra Barnett-Griness, Vered Friedman, Flavio Lejbkowicz, and Gad Rennert, Carmel Medical Center, and Technion, Haifa, Israel; Joel K. Greenson, University of Michigan, Ann Arbor, MI; and Stephen B. Gruber, University of Southern California, Los Angeles, CA
| | - Vered Friedman
- Katerina Shulman, Hillel Yaffe Medical Center, Hadera; Katerina Shulman, Ofra Barnett-Griness, Vered Friedman, Flavio Lejbkowicz, and Gad Rennert, Carmel Medical Center, and Technion, Haifa, Israel; Joel K. Greenson, University of Michigan, Ann Arbor, MI; and Stephen B. Gruber, University of Southern California, Los Angeles, CA
| | - Joel K. Greenson
- Katerina Shulman, Hillel Yaffe Medical Center, Hadera; Katerina Shulman, Ofra Barnett-Griness, Vered Friedman, Flavio Lejbkowicz, and Gad Rennert, Carmel Medical Center, and Technion, Haifa, Israel; Joel K. Greenson, University of Michigan, Ann Arbor, MI; and Stephen B. Gruber, University of Southern California, Los Angeles, CA
| | - Stephen B. Gruber
- Katerina Shulman, Hillel Yaffe Medical Center, Hadera; Katerina Shulman, Ofra Barnett-Griness, Vered Friedman, Flavio Lejbkowicz, and Gad Rennert, Carmel Medical Center, and Technion, Haifa, Israel; Joel K. Greenson, University of Michigan, Ann Arbor, MI; and Stephen B. Gruber, University of Southern California, Los Angeles, CA
| | - Flavio Lejbkowicz
- Katerina Shulman, Hillel Yaffe Medical Center, Hadera; Katerina Shulman, Ofra Barnett-Griness, Vered Friedman, Flavio Lejbkowicz, and Gad Rennert, Carmel Medical Center, and Technion, Haifa, Israel; Joel K. Greenson, University of Michigan, Ann Arbor, MI; and Stephen B. Gruber, University of Southern California, Los Angeles, CA
| | - Gad Rennert
- Katerina Shulman, Hillel Yaffe Medical Center, Hadera; Katerina Shulman, Ofra Barnett-Griness, Vered Friedman, Flavio Lejbkowicz, and Gad Rennert, Carmel Medical Center, and Technion, Haifa, Israel; Joel K. Greenson, University of Michigan, Ann Arbor, MI; and Stephen B. Gruber, University of Southern California, Los Angeles, CA
| |
Collapse
|
494
|
Skin toxicity with anti-EGFR monoclonal antibody in cancer patients: a meta-analysis of 65 randomized controlled trials. Cancer Chemother Pharmacol 2018; 82:571-583. [PMID: 30006755 DOI: 10.1007/s00280-018-3644-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 07/04/2018] [Indexed: 12/30/2022]
Abstract
We performed a meta-analysis to fully investigate the skin toxicities of anti-EGFR monoclonal antibody (EGFR-MoAbs) in cancer patients. The relevant studies of the randomized controlled trials (RCTs) in cancer patients treated with EGFR-MoAbs were retrieved and the systematic evaluation was conducted. EMBASE, MEDLINE, and PubMed were searched for articles published till November 2017. The relevant RCTs in cancer patients treated with EGFR-MoAbs were retrieved and the systematic evaluation was conducted. 65 RCTs and 25994 patients were included. The current meta-analysis suggests that the use of EGFR-MoAbs significantly increases the risk of developing all-grade and high-grade skin toxicity, such as rash, hand-foot syndrome, dry skin and oral mucositis. Rash was the most common skin toxicity. Patients receiving nimotuzumab were associated with the least risk of skin toxicity. The risk of high-grade skin toxicity tended to be higher in the study in which the EGFR-MoAbs treatment duration was longer. The available data suggested that the use of EGFR-MoAbs significantly increases the risk of developing skin toxicity. Physicians should be aware of skin toxicity and should monitor cancer patients when receiving EGFR-MoAbs.
Collapse
|
495
|
Shimada Y, Tajima Y, Nagahashi M, Ichikawa H, Oyanagi H, Okuda S, Takabe K, Wakai T. Clinical Significance of BRAF Non-V600E Mutations in Colorectal Cancer: A Retrospective Study of Two Institutions. J Surg Res 2018; 232:72-81. [PMID: 30463788 DOI: 10.1016/j.jss.2018.06.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/22/2018] [Accepted: 06/06/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Recent advances in next-generation sequencing have enabled the detection of BRAF V600E mutations as well as BRAF non-V600E mutations in a single assay. The present work aimed to describe the clinicopathological characteristics and clinical outcome of the BRAF non-V600E mutant-type in colorectal cancer (CRC). PATIENTS AND METHODS CRC samples from 111 Stage IV patients were analyzed for somatic mutations using a 415-gene comprehensive genomic sequencing panel. Patients were classified according to BRAF status as wild-type, V600E mutant-type, or non-V600E mutant-type. Differences between clinicopathological characteristics and genetic alterations were analyzed among the three groups. Overall survival (OS) and the response to anti-EGFR therapy were also analyzed. RESULTS Comprehensive genomic sequencing revealed that 98 patients (88%), 7 patients (6%), and 6 patients (6%) were wild-type, V600E mutant-type, and non-V600E mutant-type, respectively. Non-V600E mutant-type tumors were frequently left-sided (83%), while V600E mutant-type tumors were frequently right-sided (86%; P = 0.025). Non-V600E mutant-type showed better OS than V600E mutant-type (P = 0.038), with no significant difference compared with wild-type tumors. The two patients with non-V600E mutations who underwent repeated metastasectomies showed no evidence of disease at final follow-up. Regarding the efficacy of anti-EGFR therapy, the patient with an I326V mutation had progressive disease (+115%) despite no genetic alterations detected in the EGFR pathway that could drive resistance, suggesting an alternate resistance mechanism. CONCLUSIONS Non-V600E mutant-type is more likely to be left-sided and demonstrates better OS than V600E mutant-type. Further preclinical and clinical investigations are needed to clarify the role of non-V600E mutations in CRC.
Collapse
Affiliation(s)
- Yoshifumi Shimada
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Niigata, Japan.
| | - Yosuke Tajima
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Niigata, Japan
| | - Masayuki Nagahashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Niigata, Japan
| | - Hiroshi Ichikawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Niigata, Japan
| | - Hidehito Oyanagi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Niigata, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Niigata, Japan
| | - Kazuaki Takabe
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Niigata, Japan; Division of Breast Surgery, Roswell Park Cancer Institute, Buffalo, New York; Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, New York; Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan; Department of Surgery, Yokohama City University, Yokohama, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata City, Niigata, Japan
| |
Collapse
|
496
|
Naito Y, Takahashi H, Shitara K, Okamoto W, Bando H, Kuwata T, Kuboki Y, Matsumoto S, Miki I, Yamanaka T, Watanabe A, Kojima M. Feasibility study of cancer genome alterations identified by next generation sequencing: ABC study. Jpn J Clin Oncol 2018; 48:559-564. [PMID: 29659903 PMCID: PMC5974784 DOI: 10.1093/jjco/hyy052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/05/2018] [Indexed: 01/08/2023] Open
Abstract
Background To confirm the feasibility and explore the clinical applicability of amplicon sequencing by next generation sequencing (NGS) of biopsy samples from patients with advanced solid tumors, we conducted a prospective study. Methods Patients with unresectable, advanced, or recurrent solid tumors were included. Key eligibility criteria were as follows: 20 years or older, any planned systemic therapy, adequate lesion for biopsy, and written informed consent. Samples were fixed in 10% buffered formalin and embedded in paraffin. Cancer-derived DNA was extracted, and amplicon sequencing was performed using Ion AmpliseqTM Cancer Hotspot Panel version 1.0 or version 2.0 by central vendor. We evaluated the success rate of sequencing, and the proportion of the patients with actionable mutations. We organized an expert panel to share the results of targeted sequence, make annotations and reports, and discuss concomitant ethical/legal/social issues. Results A total of 232 patients were included, and 208 were successfully analyzed (success rate of 89.7%). The biopsy procedures were safe, with only one case of Grade 3 vasovagal reaction. The proportion of actionable/druggable mutations was 38.9% (81/208), which was not significantly different between the cancer panel version 1.0 and version 2.0 (P = 0.476). Expert panel could discuss the findings and make sufficient reports. Conclusions We confirmed the feasibility of NGS-based amplicon sequencing using biopsy samples, making the basis for nationwide genome screening for cancer patients using biopsy samples. Our results suggest that focused panel may be sufficient to detect major mutations.
Collapse
Affiliation(s)
- Yoichi Naito
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Hideaki Takahashi
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Kohei Shitara
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Wataru Okamoto
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Hideaki Bando
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Takeshi Kuwata
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Yasutoshi Kuboki
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Shingo Matsumoto
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Izumi Miki
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | | | | | - Motohiro Kojima
- National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| |
Collapse
|
497
|
Basso M, Dadduzio V, Rossi S, D'Argento E, Strippoli A, Di Salvatore M, Orlandi A, Zurlo V, Di Noia V, Barone C. The interference between oxaliplatin & anti-EGFR therapies: a different hypothesis to explain the 'unexplainable'. Per Med 2018; 15:319-327. [PMID: 29927360 DOI: 10.2217/pme-2017-0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This paper has been written because we have a differing idea concerning the suspected negative interference between oxaliplatin and anti-EGFR therapies in cancer patients. Several multicenter, randomized, controlled clinical trials investigated whether the efficacy of oxaliplatin-based chemotherapy is improved by the addition of anti-EGFR therapies in patients affected by KRAS wild-type advanced colorectal cancer. Results of these trials have produced puzzling findings, with some studies demonstrating improved survival and other studies showing no differences in overall survival between experimental and control arms. Moreover, a detrimental effect has been demonstrated in some settings. Nevertheless, the extent of this interaction remains uncertain. Some physicians proposed personal interpretations. This paper describes our hypothesis.
Collapse
Affiliation(s)
- Michele Basso
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Vincenzo Dadduzio
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy.,Unit of Oncology 1, Department of Clinical & Experimental Oncology, Istituto Oncologico Veneto IRCCS, Padua, Italy
| | - Sabrina Rossi
- Department of Medical Oncology, Humanitas Clinical & Research Center, Rozzano, Milan, Italy
| | - Ettore D'Argento
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonia Strippoli
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mariantonietta Di Salvatore
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Armando Orlandi
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valeria Zurlo
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Vincenzo Di Noia
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carlo Barone
- Department of Medical Oncology, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
498
|
Villéger R, Lopès A, Veziant J, Gagnière J, Barnich N, Billard E, Boucher D, Bonnet M. Microbial markers in colorectal cancer detection and/or prognosis. World J Gastroenterol 2018; 24:2327-2347. [PMID: 29904241 PMCID: PMC6000297 DOI: 10.3748/wjg.v24.i22.2327] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/03/2018] [Accepted: 05/18/2018] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer worldwide. CRC is still associated with a poor prognosis among patients with advanced disease. On the contrary, due to its slow progression from detectable precancerous lesions, the prognosis for patients with early stages of CRC is encouraging. While most robust methods are invasive and costly, actual patient-friendly screening methods for CRC suffer of lack of sensitivity and specificity. Therefore, the development of sensitive, non-invasive and cost-effective methods for CRC detection and prognosis are necessary for increasing the chances of a cure. Beyond its beneficial functions for the host, increasing evidence suggests that the intestinal microbiota is a key factor associated with carcinogenesis. Many clinical studies have reported a disruption in the gut microbiota balance and an alteration in the faecal metabolome of CRC patients, suggesting the potential use of a microbial-based test as a non-invasive diagnostic and/or prognostic tool for CRC screening. This review aims to discuss the microbial signatures associated with CRC known to date, including dysbiosis and faecal metabolome alterations, and the potential use of microbial variation markers for non-invasive early diagnosis and/or prognostic assessment of CRC and advanced adenomas. We will finally discuss the possible use of these markers as predicators for treatment response and their limitations.
Collapse
Affiliation(s)
- Romain Villéger
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
| | - Amélie Lopès
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Research Biologics, Sanofi R&D, Vitry-Sur-Seine 94400, France
| | - Julie Veziant
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Chirurgie digestive, Centre Hospitalier Universitaire, Clermont-Ferrand 63000, France
| | - Johan Gagnière
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Chirurgie digestive, Centre Hospitalier Universitaire, Clermont-Ferrand 63000, France
| | - Nicolas Barnich
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Université Clermont Auvergne, Institut Universitaire de Technologie de Clermont-Ferrand, Clermont-Ferrand 63000, France
| | - Elisabeth Billard
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Université Clermont Auvergne, Institut Universitaire de Technologie de Clermont-Ferrand, Clermont-Ferrand 63000, France
| | - Delphine Boucher
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Université Clermont Auvergne, Institut Universitaire de Technologie de Clermont-Ferrand, Clermont-Ferrand 63000, France
| | - Mathilde Bonnet
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, Clermont-Ferrand 63000, France
- Université Clermont Auvergne, Institut Universitaire de Technologie de Clermont-Ferrand, Clermont-Ferrand 63000, France
| |
Collapse
|
499
|
Shin W, Lee SK, Hwang JH, Park JC, Cho YH, Ro EJ, Song Y, Seo HR, Choi KY. Identification of Ras-degrading small molecules that inhibit the transformation of colorectal cancer cells independent of β-catenin signaling. Exp Mol Med 2018; 50:1-10. [PMID: 29884842 PMCID: PMC5994827 DOI: 10.1038/s12276-018-0102-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 03/06/2018] [Indexed: 12/17/2022] Open
Abstract
Although the development of drugs that control Ras is an emerging topic in cancer therapy, no clinically applicable drug is currently available. We have previously utilized knowledge of the Wnt/β-catenin signaling-dependent mechanism of Ras protein stability regulation to identify small molecules that inhibit the proliferation and transformation of various colorectal cancer (CRC) cells via degradation of both β-catenin and Ras. Due to the absence of Ras degradation in cells expressing a nondegradable mutant form of β-catenin and the need to determine an alternative mechanism of Ras degradation, we designed a cell-based system to screen compounds that degrade Ras independent of the Wnt/β-catenin signaling pathway. A cell-based high-content screening (HCS) system that monitors the levels of EGFP-K-RasG12V was established using HCT-116 cells harboring a nondegradable mutant CTNNB1 (ΔS45). Through HCS of a chemical library composed of 10,000 compounds and subsequent characterization of hits, we identified several compounds that degrade Ras without affecting the β-catenin levels. KY7749, one of the most effective compounds, inhibited the proliferation and transformation of CRC cells, especially KRAS-mutant cells that are resistant to the EGFR monoclonal antibody cetuximab. Small molecules that degrade Ras independent of β-catenin may able to be used in treatments for cancers caused by aberrant EGFR and Ras. Mutations in KRAS, a gene regulating cell proliferation, occur in 40–50% of colorectal cancer (CRC) patients. These cancers are usually insensitive to antibody drugs targeting the epidermal growth factor receptor (EGFR) on cancer cells. Kang-Yell Choi at Yonsei University, Seoul, South Korea and co-workers had previously identified small molecules that inhibited CRC by degrading the signaling proteins β-catenin and Ras. However, CRC cells with β-catenin mutations are resistant to Ras-degrading compounds. In this study, they screened 10,000 small molecules that could degrade Ras in CRC cells expressing nondegradable mutant β-catenin. They identified small molecules that inhibited growth of CRC cells resistant to EGFR-mediated antibody therapy. Further investigation of these molecules may help develop new drugs for treating CRC.
Collapse
Affiliation(s)
- Wookjin Shin
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Sang-Kyu Lee
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jeong-Ha Hwang
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jong-Chan Park
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yong-Hee Cho
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Eun Ji Ro
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yeonhwa Song
- Cancer Biology Research Laboratory, Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Haeng Ran Seo
- Cancer Biology Research Laboratory, Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kang-Yell Choi
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, Republic of Korea. .,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
500
|
Brägelmann J, Dammert MA, Dietlein F, Heuckmann JM, Choidas A, Böhm S, Richters A, Basu D, Tischler V, Lorenz C, Habenberger P, Fang Z, Ortiz-Cuaran S, Leenders F, Eickhoff J, Koch U, Getlik M, Termathe M, Sallouh M, Greff Z, Varga Z, Balke-Want H, French CA, Peifer M, Reinhardt HC, Örfi L, Kéri G, Ansén S, Heukamp LC, Büttner R, Rauh D, Klebl BM, Thomas RK, Sos ML. Systematic Kinase Inhibitor Profiling Identifies CDK9 as a Synthetic Lethal Target in NUT Midline Carcinoma. Cell Rep 2018; 20:2833-2845. [PMID: 28930680 PMCID: PMC5622049 DOI: 10.1016/j.celrep.2017.08.082] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 06/27/2017] [Accepted: 08/24/2017] [Indexed: 12/27/2022] Open
Abstract
Kinase inhibitors represent the backbone of targeted cancer therapy, yet only a limited number of oncogenic drivers are directly druggable. By interrogating the activity of 1,505 kinase inhibitors, we found that BRD4-NUT-rearranged NUT midline carcinoma (NMC) cells are specifically killed by CDK9 inhibition (CDK9i) and depend on CDK9 and Cyclin-T1 expression. We show that CDK9i leads to robust induction of apoptosis and of markers of DNA damage response in NMC cells. While both CDK9i and bromodomain inhibition over time result in reduced Myc protein expression, only bromodomain inhibition induces cell differentiation and a p21-induced cell-cycle arrest in these cells. Finally, RNA-seq and ChIP-based analyses reveal a BRD4-NUT-specific CDK9i-induced perturbation of transcriptional elongation. Thus, our data provide a mechanistic basis for the genotype-dependent vulnerability of NMC cells to CDK9i that may be of relevance for the development of targeted therapies for NMC patients.
Collapse
Affiliation(s)
- Johannes Brägelmann
- Molecular Pathology, Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - Marcel A Dammert
- Molecular Pathology, Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - Felix Dietlein
- Department I of Internal Medicine and Center for Integrated Oncology, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | | | - Axel Choidas
- Lead Discovery Center (LDC) GmbH, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - Stefanie Böhm
- Molecular Pathology, Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - André Richters
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Str. 4a, 44221 Dortmund, Germany
| | - Debjit Basu
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Str. 4a, 44221 Dortmund, Germany
| | - Verena Tischler
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - Carina Lorenz
- Molecular Pathology, Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - Peter Habenberger
- Lead Discovery Center (LDC) GmbH, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - Zhizhou Fang
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Str. 4a, 44221 Dortmund, Germany
| | - Sandra Ortiz-Cuaran
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - Frauke Leenders
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - Jan Eickhoff
- Lead Discovery Center (LDC) GmbH, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - Uwe Koch
- Lead Discovery Center (LDC) GmbH, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - Matthäus Getlik
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Str. 4a, 44221 Dortmund, Germany
| | - Martin Termathe
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Str. 4a, 44221 Dortmund, Germany
| | - Muhammad Sallouh
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Str. 4a, 44221 Dortmund, Germany
| | - Zoltán Greff
- Vichem Chemie Research Ltd., Herman Ottó u. 15, Budapest, Hungary
| | - Zoltán Varga
- Vichem Chemie Research Ltd., Herman Ottó u. 15, Budapest, Hungary
| | - Hyatt Balke-Want
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany; Department I of Internal Medicine and Center for Integrated Oncology, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Christopher A French
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Martin Peifer
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany
| | - H Christian Reinhardt
- Department I of Internal Medicine and Center for Integrated Oncology, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - László Örfi
- Vichem Chemie Research Ltd., Herman Ottó u. 15, Budapest, Hungary; Department of Pharmaceutical Chemistry, Semmelweis University, Hőgyes E. U.9, Budapest, Hungary
| | - György Kéri
- Vichem Chemie Research Ltd., Herman Ottó u. 15, Budapest, Hungary
| | - Sascha Ansén
- Department I of Internal Medicine and Center for Integrated Oncology, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Lukas C Heukamp
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany; Institute of Pathology, Medical Faculty, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Reinhard Büttner
- Institute of Pathology, Medical Faculty, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Daniel Rauh
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Str. 4a, 44221 Dortmund, Germany
| | - Bert M Klebl
- Lead Discovery Center (LDC) GmbH, Otto-Hahn-Str. 15, 44227 Dortmund, Germany
| | - Roman K Thomas
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany; Institute of Pathology, Medical Faculty, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany.
| | - Martin L Sos
- Molecular Pathology, Institute of Pathology, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 115b, 50931 Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|