301
|
van der Kaaij RT, Wassenaar ECE, Koemans WJ, Sikorska K, Grootscholten C, Los M, Huitema A, Schellens JHM, Veenhof AAFA, Hartemink KJ, Aalbers AGJ, van Ramshorst B, Boerma D, Boot H, van Sandick JW. Treatment of PERItoneal disease in Stomach Cancer with cytOreductive surgery and hyperthermic intraPEritoneal chemotherapy: PERISCOPE I initial results. Br J Surg 2020; 107:1520-1528. [PMID: 32277764 DOI: 10.1002/bjs.11588] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/04/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The role of cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy (HIPEC) in gastric cancer is unknown. This non-randomized dose-finding phase I-II study was designed to assess the safety and feasibility of HIPEC, following systemic chemotherapy, in patients with gastric cancer and limited peritoneal dissemination. The maximum tolerated dose of normothermic intraperitoneal docetaxel in combination with a fixed dose of intraperitoneal oxaliplatin was also explored. METHODS Patients with resectable cT3-cT4a gastric adenocarcinoma with limited peritoneal metastases and/or tumour-positive peritoneal cytology were included. An open HIPEC technique was used with 460 mg/m2 hyperthermic oxaliplatin for 30 min followed by normothermic docetaxel for 90 min in escalating doses (0, 50, 75 mg/m2 ). RESULTS Between 2014 and 2017, 37 patients were included. Of 25 patients who completed the full study protocol, four were treated at dose level 1 (0 mg/m2 docetaxel), six at dose level 2 (50 mg/m2 ) and four at dose level 3 (75 mg/m2 ). At dose level 3, two dose-limiting toxicities occurred, both associated with postoperative ileus. Thereafter, another 11 patients were treated at dose level 2, with no more dose-limiting toxicities. Based on this, the maximum tolerated dose was 50 mg/m2 intraperitoneal docetaxel. Serious adverse events were scored in 17 of 25 patients. The reoperation rate was 16 per cent (4 of 25) and the treatment-related mortality rate was 8 per cent (2 patients, both in dose level 3). CONCLUSION Gastrectomy combined with cytoreductive surgery and HIPEC was feasible using 460 mg/m2 oxaliplatin and 50 mg/m2 normothermic docetaxel.
Collapse
Affiliation(s)
| | | | - W J Koemans
- Department of Surgical Oncology, Amsterdam, the Netherlands
| | - K Sikorska
- Department of Biometrics, Amsterdam, the Netherlands
| | - C Grootscholten
- Department of Gastrointestinal Oncology, Amsterdam, the Netherlands
| | - M Los
- Department of Medical Oncology, St Antonius Hospital, Nieuwegein, the Netherlands
| | - A Huitema
- Department of Pharmacy, Amsterdam, the Netherlands
| | - J H M Schellens
- >Department of Clinical Pharmacology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | | | - K J Hartemink
- Department of Surgical Oncology, Amsterdam, the Netherlands
| | - A G J Aalbers
- Department of Surgical Oncology, Amsterdam, the Netherlands
| | | | - D Boerma
- Department of Surgery, Nieuwegein, the Netherlands
| | - H Boot
- Department of Gastrointestinal Oncology, Amsterdam, the Netherlands
| | | |
Collapse
|
302
|
van Kleef J, van den Boorn H, Verhoeven R, Vanschoenbeek K, Abu-Hanna A, Zwinderman A, Sprangers M, van Oijen M, De Schutter H, van Laarhoven H. External Validation of the Dutch SOURCE Survival Prediction Model in Belgian Metastatic Oesophageal and Gastric Cancer Patients. Cancers (Basel) 2020; 12:E834. [PMID: 32244310 PMCID: PMC7225946 DOI: 10.3390/cancers12040834] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022] Open
Abstract
The SOURCE prediction model predicts individualised survival conditional on various treatments for patients with metastatic oesophageal or gastric cancer. The aim of this study was to validate SOURCE in an external cohort from the Belgian Cancer Registry. Data of Belgian patients diagnosed with metastatic disease between 2004 and 2014 were extracted (n = 4097). Model calibration and discrimination (c-indices) were determined. A total of 2514 patients with oesophageal cancer and 1583 patients with gastric cancer with a median survival of 7.7 and 5.4 months, respectively, were included. The oesophageal cancer model showed poor calibration (intercept: 0.30, slope: 0.42) with an absolute mean prediction error of 14.6%. The mean difference between predicted and observed survival was -2.6%. The concordance index (c-index) of the oesophageal model was 0.64. The gastric cancer model showed good calibration (intercept: 0.02, slope: 0.91) with an absolute mean prediction error of 2.5%. The mean difference between predicted and observed survival was 2.0%. The c-index of the gastric cancer model was 0.66. The SOURCE gastric cancer model was well calibrated and had a similar performance in the Belgian cohort compared with the Dutch internal validation. However, the oesophageal cancer model had not. Our findings underscore the importance of evaluating the performance of prediction models in other populations.
Collapse
Affiliation(s)
- J.J. van Kleef
- Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Department of Medical Oncology, 1105 AZ Amsterdam, The Netherlands; (J.J.v.K.); (H.G.v.d.B.); (M.G.H.v.O.)
| | - H.G. van den Boorn
- Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Department of Medical Oncology, 1105 AZ Amsterdam, The Netherlands; (J.J.v.K.); (H.G.v.d.B.); (M.G.H.v.O.)
| | - R.H.A. Verhoeven
- Department of Research & Development, Netherlands Comprehensive Cancer Organisation (IKNL), 3511 DT Utrecht, The Netherlands;
| | - K. Vanschoenbeek
- Belgian Cancer Registry, 1210 Brussels, Belgium; (K.V.); (H.D.S.)
| | - A. Abu-Hanna
- Department of Medical Informatics, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands;
| | - A.H. Zwinderman
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - M.A.G. Sprangers
- Department of Medical Psychology, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam Public Health Research Institute, 1105 AZ Amsterdam, The Netherlands;
| | - M.G.H. van Oijen
- Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Department of Medical Oncology, 1105 AZ Amsterdam, The Netherlands; (J.J.v.K.); (H.G.v.d.B.); (M.G.H.v.O.)
| | - H. De Schutter
- Belgian Cancer Registry, 1210 Brussels, Belgium; (K.V.); (H.D.S.)
| | - H.W.M. van Laarhoven
- Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, Department of Medical Oncology, 1105 AZ Amsterdam, The Netherlands; (J.J.v.K.); (H.G.v.d.B.); (M.G.H.v.O.)
| |
Collapse
|
303
|
Li Y, Wang P, Wu LL, Yan J, Pang XY, Liu SJ. miR-26a-5p Inhibit Gastric Cancer Cell Proliferation and Invasion Through Mediated Wnt5a. Onco Targets Ther 2020; 13:2537-2550. [PMID: 32273724 PMCID: PMC7108881 DOI: 10.2147/ott.s241199] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/04/2020] [Indexed: 01/26/2023] Open
Abstract
Purpose Gastric cancer (GC) is a malignant disease of digestive tract. Clinically, radiation therapy is widely applied in treating GC, while with undesirable outcome due to tumor re-proliferation and recurrence and metastasis after radiation. Therefore, it is crucial to explore potential molecular mechanisms to develop therapeutic strategies. The present study found that miR-26a-5p has low expression in GC patients and could regulate Wnt5a to inhibit tumor growth, which was a potential therapeutic target for GC. To explore the expression and related mechanism of miR-26a-5p and Wnt5a in GC. Patients and Methods MiR-26a-5p and Wnt5a were extracted from the transcriptome data of GC downloaded from TCGA database for analysis. The expression levels of miR-26a-5p and Wnt5a in patients’ tissues and serum were detected by qRT-PCR, and their correlation with patients’ pathological data and survival was analyzed. In addition, miR-26a-5p and Wnt5a overexpression and inhibition vectors were transfected into cells to observe the effects on the proliferation, invasion and apoptosis of GC cells. The relationship between miR-26a-5p and Wnt5a was analyzed by dual luciferase report. Results The database and clinical samples showed that miR-26a-5p level was low while Wnt5a was high in GC. MiR-26a-5p level decreased in patients with stage III+IV, lymphatic metastasis and tumor ≥3cm, and Wnt5a was contrary to that of the miR-26a-5p, with diagnostic value. Overexpressed miR-26a-5p and inhibited Wnt5a enhanced apoptosis, decreased proliferation and invasion, reduced Bcl-2 and β-catenin proteins, and elevated Caspase 3, E-cadherin and Bax proteins, while inhibited miR-26a-5p and over-expressed Wnt5a showed the opposite results. Dual luciferase report confirmed that miR-26a-5p targeted to regulate Wnt5a, and rescue experiments found that these effects could be counteracted by reducing miR-26a-5p level. Conclusion Overexpressed miR-26a-5p can inhibit Wnt5a expression, promote cell apoptosis, and suppress cell proliferation and invasion in GC.
Collapse
Affiliation(s)
- Yu Li
- Department of Oncology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang Province, People's Republic of China
| | - Peng Wang
- Department of Oncology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang Province, People's Republic of China
| | - Lei-Lei Wu
- School of Pharmaceutical Sciences, Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang Province, People's Republic of China
| | - Jun Yan
- Department of Oncology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang Province, People's Republic of China
| | - Xue-Ying Pang
- Department of Oncology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang Province, People's Republic of China
| | - Song-Jiang Liu
- Department of Oncology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang Province, People's Republic of China
| |
Collapse
|
304
|
Catenacci DVT, Rasco D, Lee J, Rha SY, Lee KW, Bang YJ, Bendell J, Enzinger P, Marina N, Xiang H, Deng W, Powers J, Wainberg ZA. Phase I Escalation and Expansion Study of Bemarituzumab (FPA144) in Patients With Advanced Solid Tumors and FGFR2b-Selected Gastroesophageal Adenocarcinoma. J Clin Oncol 2020; 38:2418-2426. [PMID: 32167861 PMCID: PMC7367551 DOI: 10.1200/jco.19.01834] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To evaluate the safety, pharmacokinetics, and preliminary activity of bemarituzumab in patients with FGFR2b-overexpressing gastric and gastroesophageal junction adenocarcinoma (GEA). PATIENTS AND METHODS FPA144-001 was a phase I, open-label, multicenter trial consisting of the following 3 parts: part 1a involved dose escalation in patients with recurrent solid tumors at doses ranging from 0.3 to 15 mg/kg; part 1b involved dose escalation in patients with advanced-stage GEA; and part 2 involved dose expansion in patients with advanced-stage GEA that overexpressed FGFR2b at various levels (4 cohorts; high, medium, low, and no FGFR2b overexpression) and 1 cohort of patients with FGFR2b-overexpressing advanced-stage bladder cancer. RESULTS Seventy-nine patients were enrolled; 19 were enrolled in part 1a, 8 in part 1b, and 52 in part 2. No dose-limiting toxicities were reported, and the recommended dose was identified as 15 mg/kg every 2 weeks based on safety, tolerability, pharmacokinetic parameters, and clinical activity. The most frequent treatment-related adverse events (TRAEs) were fatigue (17.7%), nausea (11.4%), and dry eye (10.1%). Grade 3 TRAEs included nausea (2 patients) and anemia, neutropenia, increased AST, increased alkaline phosphatase, vomiting, and an infusion reaction (1 patient each). Three (10.7%) of 28 patients assigned to a cohort receiving a dose of ≥ 10 mg/kg every 2 weeks for ≥ 70 days reported reversible grade 2 corneal TRAEs. No TRAEs of grade ≥ 4 were reported. Five (17.9%; 95% CI, 6.1% to 36.9%) of 28 patients with high FGFR2b-overexpressing GEA had a confirmed partial response. CONCLUSION Overall, bemarituzumab seems to be well tolerated and demonstrated single-agent activity as late-line therapy in patients with advanced-stage GEA. Bemarituzumab is currently being evaluated in combination with chemotherapy in a phase III trial as front-line therapy for patients with high FGFR2b-overexpressing advanced-stage GEA.
Collapse
Affiliation(s)
| | - Drew Rasco
- The START Center for Cancer Care, San Antonio, TX
| | - Jeeyun Lee
- Samsung Medical Center, Seoul, South Korea
| | - Sun Young Rha
- Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seoul, South Korea
| | - Keun-Wook Lee
- Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seoul, South Korea
| | - Yung Jue Bang
- Seoul National University College of Medicine, Seoul, South Korea
| | - Johanna Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN
| | | | | | - Hong Xiang
- Five Prime Therapeutics, South San Francisco, CA
| | - Wei Deng
- Five Prime Therapeutics, South San Francisco, CA
| | | | | |
Collapse
|
305
|
Choi JH, Choi YW, Kang SY, Jeong GS, Lee HW, Jeong SH, Park JS, Ahn MS, Sheen SS. Combination versus single-agent as palliative chemotherapy for gastric cancer. BMC Cancer 2020; 20:167. [PMID: 32122320 PMCID: PMC7052983 DOI: 10.1186/s12885-020-6666-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 02/21/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Although combination chemotherapy (CC) is generally recommended in recurrent or primary metastatic gastric cancer (RPMGC), the results of randomized trials are conflicting. METHODS A retrospective review was conducted on 687 RPMGC patients who received palliative chemotherapy. We compared the overall survival (OS) between CC and single-agent chemotherapy (SC) among these patients, and we analyzed the clinicopathological characteristics affecting outcome including neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR). RESULTS Although 521 patients (75.8%) underwent CC, SC was more frequently performed in elderly patients (57.6%) and ECOG performance status (PS) 2 or 3 (65.8%) patients (p < 0.0001, in each case). The median OS of patients who received CC was significantly longer than that of patients who received SC (11 vs. 8 months, p < 0.0001). No difference in OS between CC and SC was observed in elderly patients (p = 0.583), poor PS (p = 0.810), signet ring cell (p = 0.347), palliative surgical resection (p = 0.307), and high PLR (p = 0.120), with a significant interaction between age and type of regimen (p = 0.012). Moreover, there was no difference in OS between CC and SC after propensity score matching (p = 0.322). Multivariate analysis revealed that palliative resection and ≥ second-line chemotherapy were independently associated with favorable OS (p < 0.0001, in each case), whereas poor PS (p = 0.004), signet ring cell (p < 0.0001), peritoneal metastasis (p = 0.04), high NLR (p = 0.001), and high PLR (p = 0.033) were independent prognostic factors of poor OS. CONCLUSIONS Although CC is the standard of care in RPMGC, SC can be considered a reasonable option in certain subgroups, such as elderly patients.
Collapse
Affiliation(s)
- Jin-Hyuk Choi
- Department of Hematology-Oncology, Ajou University School of Medicine, 164 World Cup-ro, Suwon, Yeongtong-gu, 16499, South Korea
| | - Yong Won Choi
- Department of Hematology-Oncology, Ajou University School of Medicine, 164 World Cup-ro, Suwon, Yeongtong-gu, 16499, South Korea
| | - Seok Yun Kang
- Department of Hematology-Oncology, Ajou University School of Medicine, 164 World Cup-ro, Suwon, Yeongtong-gu, 16499, South Korea
| | - Geum Sook Jeong
- Department of Hematology-Oncology, Ajou University School of Medicine, 164 World Cup-ro, Suwon, Yeongtong-gu, 16499, South Korea
| | - Hyun Woo Lee
- Department of Hematology-Oncology, Ajou University School of Medicine, 164 World Cup-ro, Suwon, Yeongtong-gu, 16499, South Korea
| | - Seong Hyun Jeong
- Department of Hematology-Oncology, Ajou University School of Medicine, 164 World Cup-ro, Suwon, Yeongtong-gu, 16499, South Korea
| | - Joon Seong Park
- Department of Hematology-Oncology, Ajou University School of Medicine, 164 World Cup-ro, Suwon, Yeongtong-gu, 16499, South Korea
| | - Mi Sun Ahn
- Department of Hematology-Oncology, Ajou University School of Medicine, 164 World Cup-ro, Suwon, Yeongtong-gu, 16499, South Korea.
| | - Seung Soo Sheen
- Department of Pulmonary and Critical Care Medicine, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
306
|
Li Y, Sui X, Su Z, Yu C, Shi X, Johnson NL, Chu F, Li Y, Li K, Ding X. Meta-Analysis of Paclitaxel-Based Chemotherapy Combined With Traditional Chinese Medicines for Gastric Cancer Treatment. Front Pharmacol 2020; 11:132. [PMID: 32174834 PMCID: PMC7056897 DOI: 10.3389/fphar.2020.00132] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/31/2020] [Indexed: 12/22/2022] Open
Abstract
This study aimed to compare the efficacy and safety of traditional Chinese medicines (TCMs) combined with paclitaxel-based chemotherapy and paclitaxel-based chemotherapy alone for gastric cancer treatment. Literature searches (up to September 25, 2019) were performed using the Cochrane Library, EMBASE, PubMed, Chinese Science and Technology Journals (CQVIP), Wanfang, and China Academic Journals (CNKI) databases. Data from 14 randomized controlled trials (RCTs), with 1,109 participants, were included. The results indicated that, compared with paclitaxel-based chemotherapy alone, the combination of TCMs and paclitaxel-based chemotherapy significantly improved the tumor response rate (TRR; RR: 1.39; 95% CI: 1.24–1.57; p < 0.001, I2 = 12%), increased the quality of life based on the Karnofsky Performance Scale score (RR: 1.53; 95% CI: 1.19–1.96; p < 0.001, I2 = 0%), and reduced the side effects, such as neutropenia (RR: 0.68; 95% CI: 0.55–0.84; p < 0.001, I2 = 44%), leukopenia (RR: 0.69; 95% CI: 0.54–0.90; p < 0.01, I2 = 40%), thrombocytopenia (RR: 0.66; 95% CI: 0.46–0.96; p < 0.05, I2 = 32%), and nausea and vomiting (RR: 0.50; 95% CI: 0.32–0.80; p < 0.01, I2 = 85%). Hepatic dysfunction (RR: 0.63; 95% CI: 0.33–1.20; p = 0.16, I2 = 0%), neurotoxicity (RR: 0.64; 95% CI: 0.26–1.55; p = 0.32, I2 = 0%), and anemia (RR: 0.65; 95% CI: 0.40–1.04; p = 0.07, I2 = 0%) were similar between the two groups. Evidence from the meta-analysis suggested that compared with paclitaxel-based chemotherapy alone, the combination of TCMs and paclitaxel-based chemotherapy may increase the TRR, improve quality of life, and reduce multiple chemotherapy-related side effects in gastric cancer patients. Additional rigorously designed large RCTs are required to confirm the efficacy and safety of this treatment.
Collapse
Affiliation(s)
- Yicong Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xinbing Sui
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,Department of Cancer Pharmacology, Holistic Integrative Pharmacy Institutes, College of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Zeqi Su
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chunyue Yu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoguang Shi
- Department of Surgery, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Nadia L Johnson
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fuhao Chu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Kexin Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xia Ding
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
307
|
Wang X, Xu Z, Sun J, Lv H, Wang Y, Ni Y, Chen S, Hu C, Wang L, Chen W, Cheng X. Cisplatin resistance in gastric cancer cells is involved with GPR30-mediated epithelial-mesenchymal transition. J Cell Mol Med 2020; 24:3625-3633. [PMID: 32052561 PMCID: PMC7131920 DOI: 10.1111/jcmm.15055] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Cisplatin is the major chemotherapeutic drug in gastric cancer, particularly in treating advanced gastric cancer. Tumour cells often develop resistance to chemotherapeutic drugs, which seriously affects the efficacy of chemotherapy. GPR30 is a novel oestrogen receptor that is involved in the invasion, metastasis and drug resistance of many tumours. Targeting GPR30 has been shown to increase the drug sensitivity of breast cancer cells. However, few studies have investigated the role of GPR30 in gastric cancer. Epithelial‐mesenchymal transition (EMT) has been shown to be associated with the development of chemotherapeutic drug resistance. In this study, we demonstrated that GPR30 is involved in cisplatin resistance by promoting EMT in gastric cancer. GPR30 knockdown resulted in increased sensitivity of different gastric cancer (GC) cells to cisplatin and alterations in the epithelial/mesenchymal markers. Furthermore, G15 significantly enhanced the cisplatin sensitivity of GC cells while G1 inhibited this phenomenon. In addition, EMT occurred when AGS and BGC‐823 were treated with cisplatin. Down‐regulation of GPR30 with G15 inhibited this transformation, while G1 promoted it. Taken together, these results revealed the role of GPR30 in the formation of cisplatin resistance, suggesting that targeting GPR30 signalling may be a potential strategy for improving the efficacy of chemotherapy in gastric cancer.
Collapse
Affiliation(s)
- Xiaofeng Wang
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhiyuan Xu
- Department of Abdominal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jiancheng Sun
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hang Lv
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diagnosis and Treatment of Digestive System Tumor, Hangzhou, China
| | - Yiping Wang
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diagnosis and Treatment of Digestive System Tumor, Hangzhou, China
| | - Yixiu Ni
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shangqi Chen
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Can Hu
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lijing Wang
- Department of Ultrasonics, Zhejiang Cancer Hospital, Hangzhou, China
| | - Wei Chen
- Cancer Institute of Integrated traditional Chinese and Western Medicine, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Xiangdong Cheng
- Department of Abdominal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
308
|
Ke X, Qin Q, Deng T, Liao Y, Gao SJ. Heterogeneous Responses of Gastric Cancer Cell Lines to Tenovin-6 and Synergistic Effect with Chloroquine. Cancers (Basel) 2020; 12:cancers12020365. [PMID: 32033497 PMCID: PMC7072542 DOI: 10.3390/cancers12020365] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/14/2020] [Accepted: 02/02/2020] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is the fifth most frequently diagnosed cancer and the third leading cause of cancer death. Approximately 15% of GC is associated with Epstein-Barr virus (EBV). GC is largely incurable with a dismal five-year survival rate. There is an urgent need to identify new therapeutic agents for the treatment of GC. Tenovin-6 was initially identified as a p53 activator, but it was later found to inhibit autophagy flux, and the protein deacetylase activity of sirtuins. Tenovin-6 shows promising therapeutic effect in various malignancies. However, it remains unknown whether Tenovin-6 is effective for GC. In this study, we found that EBV-positive and -negative GC cell lines were sensitive to Tenovin-6 but with different response times and doses. Tenovin-6 suppressed anchorage-independent growth of GC cells. Tenovin-6 induced different levels of apoptosis and phases of cell-cycle arrest depending on the cell lines with some manifesting gap 1 (G1) and others showing synthesis (S) phase cell-cycle arrest. Mechanistically, Tenovin-6 induced autophagy or p53 activation in GC cells depending on the status of TP53 gene. However, initiation of autophagy following treatment with Tenovin-6 conferred some protective effect on numerous cells. Combined treatment with Tenovin-6 and autophagy inhibitor chloroquine increased the cytotoxic effect by inducing microtubule-associated protein 1 light chain 3B (LC3B)-II accumulation, and by enhancing apoptosis and cell-cycle arrest. These results indicated that Tenovin-6 can be used as a potential therapeutic agent for GC, but the genetic background of the cancer cells might determine the response and mechanism of action. Treatment with Tenovin-6 alone or in combination with chloroquine could be a promising therapeutic approach for GC.
Collapse
Affiliation(s)
- Xiangyu Ke
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou 515000, China; (X.K.); (T.D.); (Y.L.)
| | - Qingsong Qin
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou 515000, China; (X.K.); (T.D.); (Y.L.)
- Correspondence: (Q.Q.); (S.-J.G.); Tel.: +86-0754-88900474 (Q.Q.); +1-412-339-9484 (S.-J.G.)
| | - Tianyi Deng
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou 515000, China; (X.K.); (T.D.); (Y.L.)
| | - Yueyan Liao
- Laboratory of Human Virology and Oncology, Shantou University Medical College, Shantou 515000, China; (X.K.); (T.D.); (Y.L.)
| | - Shou-Jiang Gao
- UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Correspondence: (Q.Q.); (S.-J.G.); Tel.: +86-0754-88900474 (Q.Q.); +1-412-339-9484 (S.-J.G.)
| |
Collapse
|
309
|
Sethi NS, Kikuchi O, Duronio GN, Stachler MD, McFarland JM, Ferrer-Luna R, Zhang Y, Bao C, Bronson R, Patil D, Sanchez-Vega F, Liu JB, Sicinska E, Lazaro JB, Ligon KL, Beroukhim R, Bass AJ. Early TP53 alterations engage environmental exposures to promote gastric premalignancy in an integrative mouse model. Nat Genet 2020; 52:219-230. [PMID: 32025000 PMCID: PMC7031028 DOI: 10.1038/s41588-019-0574-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 12/18/2019] [Indexed: 12/30/2022]
Abstract
Somatic alterations in cancer genes are being detected in normal and premalignant tissue, thus placing greater emphasis on gene-environment interactions that enable disease phenotypes. By combining early genetic alterations with disease-relevant exposures, we developed an integrative mouse model to study gastric premalignancy. Deletion of Trp53 in gastric cells confers a selective advantage and promotes the development of dysplasia in the setting of dietary carcinogens. Organoid derivation from dysplastic lesions facilitated genomic, transcriptional and functional evaluation of gastric premalignancy. Cell cycle regulators, most notably Cdkn2a, were upregulated by p53 inactivation in gastric premalignancy, serving as a barrier to disease progression. Co-deletion of Cdkn2a and Trp53 in dysplastic gastric organoids promoted cancer phenotypes but also induced replication stress, exposing a susceptibility to DNA damage response inhibitors. These findings demonstrate the utility of mouse models that integrate genomic alterations with relevant exposures and highlight the importance of gene-environment interactions in shaping the premalignant state.
Collapse
Affiliation(s)
- Nilay S Sethi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Gastrointestinal Cancer Treatment Center, Dana-Farber Cancer Institute, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| | - Osamu Kikuchi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gina N Duronio
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Matthew D Stachler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - James M McFarland
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Ruben Ferrer-Luna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Yanxi Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Chunyang Bao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Roderick Bronson
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Deepa Patil
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Francisco Sanchez-Vega
- Department of Surgery and Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jie-Bin Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ewa Sicinska
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jean-Bernard Lazaro
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Keith L Ligon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rameen Beroukhim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Adam J Bass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Gastrointestinal Cancer Treatment Center, Dana-Farber Cancer Institute, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| |
Collapse
|
310
|
Martin SP, Drake JA, Hernandez JM, Davis JL. Bidirectional chemotherapy in patients with gastric cancer and peritoneal metastasis. J Gastrointest Oncol 2020; 11:108-111. [PMID: 32175112 DOI: 10.21037/jgo.2019.11.05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is a lethal malignancy due to the combination of late-stage presentation, propensity for early metastasis, and lack of effective systemic therapies. Given the high rates of gastric peritoneal metastasis, both macro- and microscopic, regional therapy represents both an attractive and rational treatment option for patients given its success in other peritoneal surface malignancies. Bidirectional chemotherapy (intraperitoneal and intravenous) for treatment of metastatic gastric cancer has not been evaluated prospectively in a contemporary North American cohort. Here we present the rationale and design of a phase II clinical trial of intraperitoneal paclitaxel in combination with intravenous paclitaxel and oral capecitabine. We hypothesize that the combination of systemic and regional chemotherapy may result in improved progression free survival (PFS) for patients with gastric adenocarcinoma and peritoneal-only metastasis. In addition to studying clinical outcomes associated with this treatment regimen, both basic and translational science efforts are planned to better understand this complex malignancy.
Collapse
Affiliation(s)
- Sean P Martin
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Justin A Drake
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan M Hernandez
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeremy L Davis
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
311
|
Arbabi Moghadam S, Rezania V, Tuszynski JA. Cell death and survival due to cytotoxic exposure modelled as a two-state Ising system. ROYAL SOCIETY OPEN SCIENCE 2020; 7:191578. [PMID: 32257323 PMCID: PMC7062046 DOI: 10.1098/rsos.191578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/08/2020] [Indexed: 06/11/2023]
Abstract
Cancer chemotherapy agents are assessed for their therapeutic utility primarily by their ability to cause apoptosis of cancer cells and their potency is given by an IC50 value. Chemotherapy uses both target-specific and systemic-action drugs and drug combinations to treat cancer. It is important to judiciously choose a drug type, its dosage and schedule for optimized drug selection and administration. Consequently, the precise mathematical formulation of cancer cells' response to chemotherapy may assist in the selection process. In this paper, we propose a mathematical description of the cancer cell response to chemotherapeutic agent exposure based on a time-tested physical model of two-state multiple-component systems near criticality. We describe the Ising model methodology and apply it to a diverse panel of cytotoxic drugs administered against numerous cancer cell lines in a dose-response manner. The analysed dataset was generated by the Netherlands Translational Research Center B.V. (Oncolines). This approach allows for an accurate and consistent analysis of cytotoxic agents' effects on cancer cell lines and reveals the presence or absence of the bystander effect through the interaction constant. By calculating the susceptibility function, we see the value of IC50 coinciding with the peak of this measure of the system's sensitivity to external perturbations.
Collapse
Affiliation(s)
- S. Arbabi Moghadam
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
| | - V. Rezania
- Department of Physical Sciences, MacEwan University, Edmonton, Alberta, Canada T5 J 4S2
| | - J. A. Tuszynski
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada T6G 1Z2
- DIMEAS, Politecnico di Torino, Turin, Italy
| |
Collapse
|
312
|
Mehta R, Kommalapati A, Kim RD. The Impact of Ramucirumab Treatment on Survival and Quality of Life in Patients with Gastric Cancer. Cancer Manag Res 2020; 12:51-57. [PMID: 32021422 PMCID: PMC6954857 DOI: 10.2147/cmar.s199827] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/17/2019] [Indexed: 01/26/2023] Open
Abstract
Gastric cancer is the sixth most common cancer and is known to be the fifth-leading cause of cancer-related deaths globally in 2018. Systemic therapy remains the only curative option in advanced gastric carcinoma with the primary goal of improving the Health-related Quality of Life (HRQoL) (including palliation of symptoms such as dysphagia) and prolonging overall survival. Recently, ramucirumab is approved by the United States Food and Drug Administration (US-FDA) as a second-line agent either as monotherapy or in combination with paclitaxel in advanced or metastatic gastric and gastro-esophageal junction adenocarcinoma patients who have progressed on prior treatment with fluoropyrimidine or platinum containing chemotherapy. HRQoL is a subjective term that typically constitutes four components - psychological, social, occupational and physical well being. This has been evaluated as secondary endpoint in the pivotal Phase III trials with ramucirumab. HRQoL measurement can potentially provide additional information for clinical decision making beyond that of traditional medical outcomes. The present work is primarily focused on discussing HRQoL in gastric cancer patients and the impact of ramucirumab on the HRQoL in the patients with advanced gastric cancer. We also summarized the studies that evaluated the benefits of systemic therapies on HRQoL in advanced gastric cancer.
Collapse
Affiliation(s)
- Rutika Mehta
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Anuhya Kommalapati
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Richard D Kim
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
313
|
Wang H, Chen B, Lin Y, Zhou Y, Li X. Legumain Promotes Gastric Cancer Progression Through Tumor-associated Macrophages In vitro and In vivo. Int J Biol Sci 2020; 16:172-180. [PMID: 31892854 PMCID: PMC6930372 DOI: 10.7150/ijbs.36467] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/14/2019] [Indexed: 12/14/2022] Open
Abstract
Tumor-associated macrophages (TAMs) play a crucial role in the tumor microenvironment. Legumain (LGMN) has been shown to be a tumor-promoting protein, but the effect of LGMN on TAMs in the progression of gastric cancer (GC) is under exploration. Our studies included the construction of LGMN-knockdown and LGMN-overexpressing TAMs induced from the human cell line THP-1 (PMA/IL-4/IL-13) and murine cell line Raw264.7 (IL-4/IL-13). A CCK-8 assay and transwell migration assay indicated that upregulation of LGMN expression in TAMs stimulated cell proliferation, migration and invasion in vitro, while downregulation of LGMN expression reduced cell proliferation, migration and invasion. In vivo experiments revealed slower growth, less angiogenesis, and less Ki67 expression in LGMN-knockdown TAMs injected with gastric cancer cells compared to control TAMs injected with GC cells. Together, these study results suggested that LGMN+ TAMs, which may serve as a potential target for GC treatment, promoted gastric cancer cell proliferation and angiogenesis in vitro and in vivo.
Collapse
Affiliation(s)
- Hongbin Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Gastroenterology, Punan Hospital, Pudong New Area, Shanghai, China
| | - Binghong Chen
- Department of Neurosurgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingying Lin
- Department of Neurosurgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Zhou
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaobo Li
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
314
|
Pan T, Jin Z, Yu Z, Wu X, Chang X, Fan Z, Li F, Wang X, Li Z, Zhou Q, Li J, Liu B, Su L. Cathepsin L promotes angiogenesis by regulating the CDP/Cux/VEGF-D pathway in human gastric cancer. Gastric Cancer 2020; 23:974-987. [PMID: 32388635 PMCID: PMC7567730 DOI: 10.1007/s10120-020-01080-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Increasing evidence indicates that angiogenesis plays an important role in tumor progression. The function of cathepsin L (CTSL), an endosomal proteolytic enzyme, in promoting tumor metastasis is well recognized. The mechanisms by which CTSL has promoted the angiogenesis of gastric cancer (GC), however, remains unclear. METHODS The nuclear expression levels of CTSL were assessed in GC samples. The effects of CTSL on GC angiogenesis were determined by endothelial tube formation analysis, HUVEC migration assay, and chick embryo chorioallantoic membrane (CAM) assay. The involvement of the CDP/Cux/VEGF-D pathway was analyzed by angiogenesis antibody array, Western blot, co-immunoprecipitation (Co-IP) and dual-luciferase reporter assay. RESULTS In this study, we found that the nuclear CTSL expression level in GC was significantly higher than that in adjacent nontumor gastric tissues and was a potential important clinical prognostic factor. Loss- and gain-of-function assays indicated that CTSL promotes the tubular formation and migration of HUVEC cells in vitro. The CAM assay also showed that CTSL promotes angiogenesis of GC in vivo. Mechanistic analysis demonstrated that CTSL can proteolytically process CDP/Cux and produce the physiologically relevant p110 isoform, which stably binds to VEGF-D and promotes the transcription of VEGF-D, thus contributing to the angiogenesis of GC. CONCLUSION The findings of the present study suggested that CTSL plays a constructive role in the regulation of angiogenesis in human GC and could be a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Tao Pan
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhijian Jin
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenjia Yu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiongyan Wu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyu Chang
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyuan Fan
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangyuan Li
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofeng Wang
- Department of General Surgery, First People's Hospital, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Li
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Quan Zhou
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianfang Li
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingya Liu
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Liping Su
- Department of Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
315
|
Lei Z, Wang J, Li Z, Li B, Luo J, Wang X, Wang J, Ba M, Tang H, He Q, Liao Q, Yang X, Guan T, Liang H, Cui S, On Behalf Of The Chinese Peritoneal Oncology Study Group. Hyperthermic intraperitoneal chemotherapy for gastric cancer with peritoneal metastasis: A multicenter propensity score-matched cohort study. Chin J Cancer Res 2020; 32:794-803. [PMID: 33447001 PMCID: PMC7797229 DOI: 10.21147/j.issn.1000-9604.2020.06.12] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective Systemic chemotherapy has limited efficacy in the treatment of peritoneal metastasis (PM) in gastric cancer (GC). Hyperthermic intraperitoneal chemotherapy (HIPEC) combined with complete cytoreductive surgery (CRS) has shown promising outcomes but remains controversial. The present study aimed to evaluate the safety and efficacy of HIPEC without CRS in GC patients with PM. Methods This retrospective propensity score-matched multicenter cohort study included GC patients with PM treated with either chemotherapy alone (Cx group) or with HIPEC combined with chemotherapy (HIPEC-Cx group) in four Chinese high-volume gastric medical centers between 2010 and 2017. The primary outcomes were median survival time (MST) and 3-year overall survival (OS). Propensity score matching was performed to compensate for controlling potential confounding effects and selection bias. Results Of 663 eligible patients, 498 were matched. The MST in the Cx and HIPEC-Cx groups was 10.8 and 15.9 months, respectively [hazard ratio (HR)=0.71, 95% confidence interval (95% CI), 0.58−0.88; P=0.002]. The 3-year OS rate was 10.1% (95% CI, 5.4%−14.8%) and 18.4% (95% CI, 12.3%−24.5%) in the Cx and HIPEC-Cx groups, respectively (P=0.017). The complication rates were comparable. The time to first flatus and length of hospital stay for patients undergoing HIPEC combined with chemotherapy was longer than that of chemotherapy alone (4.6±2.4 dvs. 2.7±1.8 d, P<0.001; 14.2±5.8 dvs. 11.4±7.7 d, P<0.001), respectively. The median follow-up period was 33.2 months.
Conclusions Compared with standard systemic chemotherapy, HIPEC combined with chemotherapy revealed a statistically significant survival benefit for GC patients with PM, without compromising patient safety.
Collapse
Affiliation(s)
- Ziying Lei
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Jiahong Wang
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Zhi Li
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Tumor Hospital of Henan Province, Zhengzhou 450008, China
| | - Baozhong Li
- Department of Surgery, Anyang Tumor Hospital, Anyang 455000, China
| | - Jiali Luo
- Department of Oncology, Guangzhou Medical University, Guangzhou 510095, China
| | - Xuejun Wang
- Department of Gastrointestinal Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jin Wang
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Mingchen Ba
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Hongsheng Tang
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Qingjun He
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Quanxing Liao
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Xiansheng Yang
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Tianpei Guan
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Han Liang
- Department of Gastrointestinal Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Shuzhong Cui
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - On Behalf Of The Chinese Peritoneal Oncology Study Group
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China.,Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Tumor Hospital of Henan Province, Zhengzhou 450008, China.,Department of Surgery, Anyang Tumor Hospital, Anyang 455000, China.,Department of Oncology, Guangzhou Medical University, Guangzhou 510095, China.,Department of Gastrointestinal Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
316
|
Dijksterhuis WPM, Verhoeven RHA, Meijer SL, Slingerland M, Haj Mohammad N, de Vos-Geelen J, Beerepoot LV, van Voorthuizen T, Creemers GJ, van Oijen MGH, van Laarhoven HWM. Increased assessment of HER2 in metastatic gastroesophageal cancer patients: a nationwide population-based cohort study. Gastric Cancer 2020; 23:579-590. [PMID: 31927675 PMCID: PMC7305095 DOI: 10.1007/s10120-020-01039-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Addition of trastuzumab to first-line palliative chemotherapy in gastroesophageal cancer patients with HER2 overexpression has shown to improve survival. Real-world data on HER2 assessment and administration of trastuzumab are lacking. The aim of this study was to assess HER2 testing, trastuzumab administration, and overall survival (OS) in a nationwide cohort of metastatic gastroesophageal cancer patients. METHODS Data of patients with synchronous metastatic gastroesophageal adenocarcinoma diagnosed in 2010-2016 that received palliative systemic treatment (n = 2846) were collected from the Netherlands Cancer Registry and Dutch Pathology Registry. The ToGA trial criteria were used to determine HER2 overexpression. Proportions of HER2 tested patients were analyzed between hospital volume categories using Chi-square tests, and over time using trend analysis. OS was tested using the Kaplan Meier method with log rank test. RESULTS HER2 assessment increased annually, from 18% in 2010 to 88% in 2016 (P < 0.01). Median OS increased from 6.9 (2010-2013) to 7.9 months (2014-2016; P < 0.05). Between the hospitals, the proportion of tested patients varied between 29-100%, and was higher in high-volume hospitals (P < 0.01). Overall, 77% of the HER2 positive patients received trastuzumab. Median OS was higher in patients with positive (8.8 months) and negative (7.4 months) HER2 status, compared to non-tested patients (5.6 months; P < 0.05). CONCLUSION Increased determination of HER2 and administration of trastuzumab have changed daily practice management of metastatic gastroesophageal cancer patients receiving palliative systemic therapy, and possibly contributed to their improved survival. Further increase in awareness of HER2 testing and trastuzumab administration may improve quality of care and patient outcomes.
Collapse
Affiliation(s)
- Willemieke P. M. Dijksterhuis
- grid.7177.60000000084992262Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands ,grid.470266.10000 0004 0501 9982Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - Rob H. A. Verhoeven
- grid.470266.10000 0004 0501 9982Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - Sybren L. Meijer
- grid.7177.60000000084992262Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marije Slingerland
- grid.10419.3d0000000089452978Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nadia Haj Mohammad
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Judith de Vos-Geelen
- grid.412966.e0000 0004 0480 1382Department of Internal Medicine, Division of Medical Oncology, GROW-School for Oncology and Developmental Biology, Maastricht UMC+, Maastricht, The Netherlands
| | - Laurens V. Beerepoot
- grid.416373.4Department of Medical Oncology, Elisabeth-TweeSteden Hospital, Tilburg, The Netherlands
| | - Theo van Voorthuizen
- grid.415930.aDepartment of Medical Oncology, Rijnstate Hospital, Arnhem, The Netherlands
| | - Geert-Jan Creemers
- grid.413532.20000 0004 0398 8384Department of Medical Oncology, Catharina Hospital, Eindhoven, The Netherlands
| | - Martijn G. H. van Oijen
- grid.7177.60000000084992262Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands ,grid.470266.10000 0004 0501 9982Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - Hanneke W. M. van Laarhoven
- grid.7177.60000000084992262Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
317
|
Zhou X, Liu M, Ren Q, Zhu W, Wang Y, Chen H, Chen J. Oral and injectable Marsdenia tenacissima extract (MTE) as adjuvant therapy to chemotherapy for gastric cancer: a systematic review. Altern Ther Health Med 2019; 19:366. [PMID: 31830977 PMCID: PMC6909592 DOI: 10.1186/s12906-019-2779-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 11/28/2019] [Indexed: 12/14/2022]
Abstract
Background Marsdenia tenacissima extract (MTE) is a phytochemical widely used as complementary therapy in cancer care. This systematic review was conducted to investigate the anticancer and detoxification effects of MTE, as an adjuvant therapy to chemotherapy, for treating gastric cancer. Methods Ten databases were searched to identify randomized controlled trials (RCTs) comparing oral or injectable MTE plus chemotherapy versus chemotherapy alone for treating gastric cancer up to May 1, 2019. In meta-analyses, proportional odds ratios (PORs) with 95% confidence intervals (CIs) were pooled for the ordinal outcomes using the generalized linear model, and risk ratios (RRs) with 95% CIs were pooled for dichotomous outcomes using the Mantel-Haenszel method. Results Seventeen RCTs with 1329 individuals were included, with a moderate to high risk of selection and performance bias. Compared to chemotherapy alone, MTE adjuvant therapy significantly improved the response to anticancer treatment (POR 2.01, 95% CI 1.60–2.53) and patients’ performance status (POR 3.15, 95% CI 2.22–4.48) and reduce the incidences of chemotherapy-induced leukopenia (RR 0.66, 95% CI 0.56–0.78), thrombocytopenia (RR 0.64, 95% CI 0.48–0.86), anemia (RR 0.89, 95% CI 0.72–1.10), nausea/vomiting (RR 0.79, 95% CI 0.69–0.91), hepatic injury (RR 0.77, 95% CI 0.61–0.96), and peripheral neurotoxicity (RR 0.77, 95% CI 0.59–1.01). However, MTE did not significantly alleviate anemia, diarrhea, constipation, kidney injury, and oral mucosal lesions after chemotherapy. Incidence of nausea/vomiting was lower in patients receiving oral MTE than those receiving injectable MTE (RR 0.47 vs. 0.82, interaction P = 0.04). Heterogeneity was generally low among these outcomes. Three out of five RCTs that reported survival data supported the effects of MTE for prolonging progression-free and/or overall survival. No studies reported safety outcomes of MTE. Conclusions The current evidence with limitations of risk of selection and performance bias suggests that MTE, as an adjuvant therapy to chemotherapy, is effective for inhibiting cancer growth and reducing incidences of multiple chemotherapy side effects. Oral MTE may be a better choice. Uncertainty remains regarding the effects of MTE on survival endpoints and the subgroup differences between acute and chronic use of MTE and between different chemotherapy regimens.
Collapse
|
318
|
Bang YJ, Ruiz EY, Van Cutsem E, Lee KW, Wyrwicz L, Schenker M, Alsina M, Ryu MH, Chung HC, Evesque L, Al-Batran SE, Park SH, Lichinitser M, Boku N, Moehler MH, Hong J, Xiong H, Hallwachs R, Conti I, Taieb J. Phase III, randomised trial of avelumab versus physician's choice of chemotherapy as third-line treatment of patients with advanced gastric or gastro-oesophageal junction cancer: primary analysis of JAVELIN Gastric 300. Ann Oncol 2019; 29:2052-2060. [PMID: 30052729 PMCID: PMC6225815 DOI: 10.1093/annonc/mdy264] [Citation(s) in RCA: 398] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background There currently are no internationally recognised treatment guidelines for patients with advanced gastric cancer/gastro-oesophageal junction cancer (GC/GEJC) in whom two prior lines of therapy have failed. The randomised, phase III JAVELIN Gastric 300 trial compared avelumab versus physician’s choice of chemotherapy as third-line therapy in patients with advanced GC/GEJC. Patients and methods Patients with unresectable, recurrent, locally advanced, or metastatic GC/GEJC were recruited at 147 sites globally. All patients were randomised to receive either avelumab 10 mg/kg by intravenous infusion every 2 weeks or physician’s choice of chemotherapy (paclitaxel 80 mg/m2 on days 1, 8, and 15 or irinotecan 150 mg/m2 on days 1 and 15, each of a 4-week treatment cycle); patients ineligible for chemotherapy received best supportive care. The primary end point was overall survival (OS). Secondary end points included progression-free survival (PFS), objective response rate (ORR), and safety. Results A total of 371 patients were randomised. The trial did not meet its primary end point of improving OS {median, 4.6 versus 5.0 months; hazard ratio (HR)=1.1 [95% confidence interval (CI) 0.9–1.4]; P = 0.81} or the secondary end points of PFS [median, 1.4 versus 2.7 months; HR=1.73 (95% CI 1.4–2.2); P > 0.99] or ORR (2.2% versus 4.3%) in the avelumab versus chemotherapy arms, respectively. Treatment-related adverse events (TRAEs) of any grade occurred in 90 patients (48.9%) and 131 patients (74.0%) in the avelumab and chemotherapy arms, respectively. Grade ≥3 TRAEs occurred in 17 patients (9.2%) in the avelumab arm and in 56 patients (31.6%) in the chemotherapy arm. Conclusions Treatment of patients with GC/GEJC with single-agent avelumab in the third-line setting did not result in an improvement in OS or PFS compared with chemotherapy. Avelumab showed a more manageable safety profile than chemotherapy. Trial registration ClinicalTrials.gov: NCT02625623.
Collapse
Affiliation(s)
- Y-J Bang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea.
| | | | - E Van Cutsem
- Department of Digestive Oncology, University Hospitals Gasthuisberg/Leuven, KU Leuven, Leuven, Belgium
| | - K-W Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - L Wyrwicz
- Department of Gastrointestinal Cancer, M. Sklodowska-Curie Memorial Cancer Center, Warsaw, Poland
| | - M Schenker
- Centrul de Oncologie Sf. Nectarie, Craiova, Romania
| | - M Alsina
- Department of Medical Oncology, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - M-H Ryu
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - H-C Chung
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Yonsei University Health System, Seoul, South Korea
| | - L Evesque
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France
| | - S-E Al-Batran
- Department of Clinical Oncology and Hematology, Krankenhaus Nordwest, Frankfurt, Germany
| | - S H Park
- Department of Medicine, Samsung Medical Center, Seoul, South Korea
| | - M Lichinitser
- Department of Chemotherapy and Combined Therapy, N. N. Blokhin Russian Oncological Scientific Center, Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - N Boku
- Department of Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - M H Moehler
- Department of Internal Medicine, Johannes Gutenberg University Mainz, Mainz, Germany
| | - J Hong
- Global Clinical Development Immuno-Oncology, EMD Serono, Billerica, USA
| | - H Xiong
- Global Clinical Development Immuno-Oncology, EMD Serono, Billerica, USA
| | - R Hallwachs
- Global Research & Development, Merck KGaA, Darmstadt, Germany
| | - I Conti
- Global Clinical Development Immuno-Oncology, EMD Serono, Billerica, USA
| | - J Taieb
- Department of Gastroenterology and Digestive Oncology, Université Sorbonne Paris Cité, Paris Descartes University, Georges Pompidou European Hospital, Paris, France
| |
Collapse
|
319
|
Guo X, Zhao F, Ma X, Shen G, Ren D, Zheng F, Du F, Wang Z, Ahmad R, Yuan X, Zhao J, Zhao J. A comparison between triplet and doublet chemotherapy in improving the survival of patients with advanced gastric cancer: a systematic review and meta-analysis. BMC Cancer 2019; 19:1125. [PMID: 31747911 PMCID: PMC6865072 DOI: 10.1186/s12885-019-6294-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 10/25/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Chemotherapy can improve the survival of patients with advanced gastric cancer. However, whether triplet chemotherapy can further improve the survival of patients with advanced gastric cancer compared with doublet chemotherapy remains controversial. This study reviewed and updated all published and eligible randomized controlled trials (RCTs) to compare the efficacy, prognosis, and toxicity of triplet chemotherapy with doublet chemotherapy in patients with advanced gastric cancer. METHODS RCTs on first-line chemotherapy in advanced gastric cancer on PubMed, Embase, and the Cochrane Register of Controlled Trials and all abstracts from the annual meetings of the European Society for Medical Oncology (ESMO) and the American Society of Clinical Oncology conferences up to October 2018 were searched. The primary outcome was overall survival, while the secondary outcomes were progression-free survival (PFS), time to progress (TTP), objective response rate (ORR), and toxicity. RESULTS Our analysis included 23 RCTs involving 4540 patients and 8 types of triplet and doublet chemotherapy regimens, and systematic review and meta-analysis revealed that triplet chemotherapy was superior compared with doublet chemotherapy in terms of improving median OS (HR = 0.92; 95% CI, 0.86-0.98; P = 0.02) and PFS (HR = 0.82; 95% CI, 0.69-0.97; P = 0.02) and TTP (HR = 0.92; 95% CI, 0.86-0.98; P = 0.02) and ORR (OR = 1.21; 95% CI, 1.12-1.31; P < 0.0001) among overall populations. Compared with doublet chemotherapy, subgroup analysis indicated that OS improved with fluoropyrimidine-based (HR = 0.80; 95% CI, 0.66-0.96; P = 0.02), platinum-based (HR = 0.75; 95% CI, 0.57-0.99; P = 0.04), and other drug-based triplet (HR = 0.79; 95% CI, 0.69-0.90; P = 0.0006) chemotherapies while not with anthracycline-based (HR = 0.70; 95% CI, 0.42-1.15; P = 0.16), mitomycin-based (HR = 0.81; 95% CI, 0.47-1.39; P = 0.44), taxane-based (HR = 0.91; 95% CI, 0.81-1.01; P = 0.07), and irinotecan-based triplet (HR = 1.01; 95% CI, 0.82-1.24; P = 0.94) chemotherapies. For different patients, compared with doublet chemotherapy, triplet chemotherapy improved OS (HR = 0.89; 95% CI, 0.81-0.99; P = 0.03) among Western patients but did not improve (HR = 0.96; 95% CI, 0.86-1.07; P = 0.47) that among Asian patients. CONCLUSIONS Compared with doublet chemotherapy, triplet chemotherapy improved OS, PFS, TTP, and ORR in patients with advanced gastric cancer in the population overall, and improved OS in Western but not in Asian patients.
Collapse
Affiliation(s)
- Xinjian Guo
- Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Fuxing Zhao
- Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Xinfu Ma
- Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Guoshuang Shen
- Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Dengfeng Ren
- Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Fangchao Zheng
- Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
- Shouguang Hospital of Traditional Chinese Medicine, Weifang, 262700, China
- Department of Medical Oncology, Cancer hospital, Chinese academy of medical sciences, Peking Union Medical College, Beijing, 100021, China
| | - Feng Du
- Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Ziyi Wang
- Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Raees Ahmad
- Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Xinyue Yuan
- Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China
| | - Junhui Zhao
- Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China.
| | - Jiuda Zhao
- Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai University, Xining, 810000, China.
| |
Collapse
|
320
|
de Man FM, Goey AKL, van Schaik RHN, Mathijssen RHJ, Bins S. Individualization of Irinotecan Treatment: A Review of Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics. Clin Pharmacokinet 2019. [PMID: 29520731 PMCID: PMC6132501 DOI: 10.1007/s40262-018-0644-7] [Citation(s) in RCA: 252] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Since its clinical introduction in 1998, the topoisomerase I inhibitor irinotecan has been widely used in the treatment of solid tumors, including colorectal, pancreatic, and lung cancer. Irinotecan therapy is characterized by several dose-limiting toxicities and large interindividual pharmacokinetic variability. Irinotecan has a highly complex metabolism, including hydrolyzation by carboxylesterases to its active metabolite SN-38, which is 100- to 1000-fold more active compared with irinotecan itself. Several phase I and II enzymes, including cytochrome P450 (CYP) 3A4 and uridine diphosphate glucuronosyltransferase (UGT) 1A, are involved in the formation of inactive metabolites, making its metabolism prone to environmental and genetic influences. Genetic variants in the DNA of these enzymes and transporters could predict a part of the drug-related toxicity and efficacy of treatment, which has been shown in retrospective and prospective trials and meta-analyses. Patient characteristics, lifestyle and comedication also influence irinotecan pharmacokinetics. Other factors, including dietary restriction, are currently being studied. Meanwhile, a more tailored approach to prevent excessive toxicity and optimize efficacy is warranted. This review provides an updated overview on today’s literature on irinotecan pharmacokinetics, pharmacodynamics, and pharmacogenetics.
Collapse
Affiliation(s)
- Femke M de Man
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015, Rotterdam, The Netherlands
| | - Andrew K L Goey
- Department of Hospital Pharmacy, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015, Rotterdam, The Netherlands
| | - Sander Bins
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015, Rotterdam, The Netherlands.
| |
Collapse
|
321
|
Zhao L, Li J, Bai C, Nie Y, Lin G. Multi-Modality Treatment for Patients With Metastatic Gastric Cancer: A Real-World Study in China. Front Oncol 2019; 9:1155. [PMID: 31737573 PMCID: PMC6839024 DOI: 10.3389/fonc.2019.01155] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/16/2019] [Indexed: 12/26/2022] Open
Abstract
Introduction: People with metastatic gastric cancer (GC) have a poor prognosis. The study aims to investigate the efficacy of multi-modality treatment for patients with metastatic GC. Methods: We retrospectively identified 267 patients with stage IV gastric cancer who were treated with systemic chemotherapy: 114 received multi-modality treatments, 153 received systematic chemotherapy alone. The survival of these two groups was compared by log rank test, the independent prognostic factors were investigated using univariate and multivariate analyses. Results: The median survival of metastatic GC patients who received multi-modality treatment was significantly longer than those who received systematic chemotherapy alone (18.4 vs. 11.4 months, P < 0.001). Multivariate analysis identified tumor histologic differentiation, CA19–9 level, previous curative resection, palliative gastrectomy, and metastasectomy as independent prognostic factors for overall survival. In the multimodality treatment group, patients who received palliative gastrectomy or metastasectomy had a longer survival than those who only received intraperitoneal chemotherapy or radiotherapy (21.6 vs. 15.2 months, P = 0.014). Conclusion: Multi-modality treatments offer a survival benefit for patients with metastatic GC. Future prospective studies are needed to confirm the result.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiarui Li
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongdu Nie
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guole Lin
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
322
|
Tjulandin SA, Tryakin AA, Besova NS, Sholokhova E, Ivanova JI, Cheng WY, Schmerold LM, Thompson-Leduc P, Novick D. Real-world treatment patterns among patients with advanced gastric cancer in Russia: a chart review study. J Drug Assess 2019; 8:150-158. [PMID: 31656688 PMCID: PMC6792042 DOI: 10.1080/21556660.2019.1669610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 09/16/2019] [Indexed: 12/24/2022] Open
Abstract
Objective: Little evidence is available on the management of patients with metastatic and/or unresectable gastric cancer (mGC) after the failure of first-line treatment. This study presents real-world data on characteristics and treatment patterns of patients with mGC in Russia. Methods: Eligible patients were ≥18 years old, diagnosed with mGC ≥ January 1, 2012, received first-line chemotherapy followed by second-line chemotherapy or best supportive care (BSC), had ≥3 months of follow-up after the start of second-line chemotherapy or BSC (except in cases of death), and had not participated in a clinical trial. Data were summarized using descriptive statistics. Results: A total of 88 physicians provided data from 202 charts. Mean age at mGC diagnosis was 53.7 (standard deviation: 11.2) years; 70.8% of patients were male. Reasons for first-line treatment discontinuation included disease progression (50.5%) and adverse events/toxicity (39.1%). There were 52 unique treatment regimens prescribed in second-line; capecitabine (14.5%), paclitaxel (9.3%), and capecitabine + oxaliplatin (8.7%) were the most frequent. Reasons for second-line treatment discontinuation included disease progression (39.8%) and patient refusal to continue (37.5%). During 2nd-line treatment, the most common treatment-related symptoms were nausea/vomiting (75.0%), while pain (73.8%) was the most common disease-related symptom. Antiemetics (63.4%), chemotherapy (61.6%), non-narcotic analgesics (48.3%), endoscopy (45.9%), and nutritional support (35.5%) were most frequently used as supportive care. Conclusions: Second-line treatment patterns for patients with mGC in Russia are heterogeneous. Results of this study indicate the need for more intensive implementation of the most active regimens in second-line treatment of mGC according to international and national guidelines.
Collapse
Affiliation(s)
- Sergei A Tjulandin
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexey A Tryakin
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Natalia S Besova
- N. N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | | | | | | | | | | |
Collapse
|
323
|
Song J, Li Z, Chen P, Yu J, Wang F, Yang Z, Wang X. A 18FDG PET/CT-based volume parameter is a predictor of overall survival in patients with local advanced gastric cancer. Chin J Cancer Res 2019; 31:632-640. [PMID: 31564806 PMCID: PMC6736658 DOI: 10.21147/j.issn.1000-9604.2019.04.07] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Objective The present study investigated the prognosis value of preoperative fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) in patients with local advanced gastric cancer (LAGC). Methods In total, 144 patients [median age 63 (range: 48−80) years old] with LAGC underwent18F-FDG PET/CT prior to any treatment. The maximum standardized uptake values (SUVmax), mean standardized uptake values (SUVmean), metabolic tumor volume (MTV) and total lesion glycolysis (TLG) of the primary lesion were measured on PET/CT and correlated with clinicopathological features and survival.
Results Significant differences in SUVmean, SUVmax, MTV and TLG were found according to Lauren’s classification, histologic grade and T category (P<0.05). During the 26.5-month follow-up, 51 (35.4%) patients died and 70 (48.6%) exhibited disease progression. The optimal thresholds of MTV and TLG were 15.1 cm3 and 47.3 cm3, respectively. The 3-year progression-free survival (PFS) and overall survival (OS) for patients with high TLG values were 30% and 38% compared to 38% and 47% for low TLG values, respectively (P<0.05). Univariate and multifactor analyses demonstrated that lymph node metastasis and T stage were independent prognostic factors for PFS; T stage, histologic grade and TLG were independent prognostic factors for OS (P<0.05). Molecular markers had no relationship with patient’s outcomes.
Conclusions Metabolic activity of primary gastric tumors from 18F-FDG PET/CT is a prognostic factor in patients with LAGC.
Collapse
Affiliation(s)
| | - Zhongwu Li
- Department of Pathology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | | | | | | | | | | |
Collapse
|
324
|
Cheng J, Cai M, Shuai X, Gao J, Wang G, Tao K. First-line systemic therapy for advanced gastric cancer: a systematic review and network meta-analysis. Ther Adv Med Oncol 2019; 11:1758835919877726. [PMID: 31632469 PMCID: PMC6767753 DOI: 10.1177/1758835919877726] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/30/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Systemic therapy is the standard treatment against advanced gastric cancer. Fluoropyrimidine plus platinum doublet has been recommended as the preferred first-line strategy. However, there is still a lack of a comprehensive and hierarchical evidence that compares all eligible literature simultaneously. METHODS Record retrieval was conducted in PubMed, Web of Science, Cochrane Central Register of Controlled Trials, Embase, ASCO, and ESMO meeting library from inception to October 2018. Randomized controlled trials featuring comparisons between different first-line systemic treatments against advanced gastric cancer were eligible. Overall survival was utilized as the primary endpoint. Pairwise and network calculations were based on a random-effects model and the hierarchical ranking was numerically indicated by P-score. All procedures were conducted according to Cochrane Handbook 5.1 and PRISMA for Network Meta-analysis (Registration identifier: CRD42018084951). RESULTS A total of 119 studies were eligible for our pooled analysis. Concerning general analysis, 'fluoropyrimidine plus platinum-based triplet' topped the overall survival hierarchy (HR 0.91 [0.83-0.99], P-score = 0.903, p = 0.04) while it ranked in second place for progression-free survival and objective response rate. However, it displayed worse tolerability against 'fluoropyrimidine plus platinum doublet'. More specifically, 'Capecitabine plus cisplatin-based triplet plus targeted medication' topped the ranking among all fluoropyrimidine plus platinum-based regimens in additional analysis. Nevertheless, it did not reach statistical advantage against fluoropyrimidine plus oxaliplatin doublet in terms of survival benefits, while still displaying significantly worse safety profile. CONCLUSIONS Taken together, fluoropyrimidine plus oxaliplatin doublet (especially capecitabine or S-1) should still be considered as the preferred first-line regimen owing to its comparable survival benefits and lower toxicity.
Collapse
Affiliation(s)
- Ji Cheng
- Department of Gastrointestinal Surgery, Union
Hospital, Tongji
- Medical College, Huazhong University of Science
and Technology, No.1277 Jiefang Avenue, Wuhan 430022, China
- Department of Pathology, Beth Israel Deaconess
Medical Center, Harvard Medical School, Boston, MA 02115
| | - Ming Cai
- Department of Gastrointestinal Surgery, Union
Hospital, Tongji Medical College, Huazhong
- University of Science and Technology, Wuhan,
China
| | - Xiaoming Shuai
- Department of Gastrointestinal Surgery, Union
Hospital, Tongji Medical College, Huazhong
- University of Science and Technology, Wuhan,
China
| | - Jinbo Gao
- Department of Gastrointestinal Surgery, Union
Hospital, Tongji Medical College, Huazhong
- University of Science and Technology, Wuhan,
China
| | - Guobin Wang
- Department of Gastrointestinal Surgery, Union
Hospital, Tongji Medical College, Huazhong
- University of Science and Technology, Wuhan,
China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union
Hospital, Tongji Medical College, Huazhong
- University of Science and Technology, No.1277
Jiefang Avenue, Wuhan 430022, China
| |
Collapse
|
325
|
Yang JC, Chang N, Wu DC, Cheng WC, Chung WM, Chang WC, Lei FJ, Liu CJ, Wu IC, Lai HC, Ma WL. Preclinical evaluation of exemestane as a novel chemotherapy for gastric cancer. J Cell Mol Med 2019; 23:7417-7426. [PMID: 31557413 PMCID: PMC6815818 DOI: 10.1111/jcmm.14605] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/18/2019] [Accepted: 07/23/2019] [Indexed: 12/14/2022] Open
Abstract
CYP19A1/aromatase (Ar) is a prognostic biomarker of gastric cancer (GCa). Ar is a critical enzyme for converting androstenedione to oestradiol in the steroidogenesis cascade. For decades, Ar has been targeted with Ar inhibitors (ARIs) in gynaecologic malignancies; however, it is unexplored in GCa. A single‐cohort tissue microarray examination was conducted to study the association between Ar expression and disease outcome in Asian patients with GCa. The results revealed that Ar was a prognostic promoter. Bioinformatics analyses conducted on a Caucasian‐based cDNA microarray databank showed Ar to be positively associated with GCa prognosis for multiple clinical modalities, including surgery, 5‐Fluorouracil (5‐FU) for adjuvant chemotherapy, or HER2 positivity. These findings imply that targeting Ar expression exhibits a potential for fulfilling unmet medical needs. Hence, Ar‐targeting compounds were tested, and the results showed that exemestane exhibited superior cancer‐suppressing efficacy to other ARIs. In addition, exemestane down‐regulated Ar expression. Ablating Ar abundance with short hairpin (sh)Ar could also suppress GCa cell growth, and adding 5‐FU could facilitate this effect. Notably, adding oestradiol could not prevent exemestane or shAr effects, implicating a nonenzymatic mechanism of Ar in cancer growth. Regarding translational research, treatment with exemestane alone exhibited tumour suppression efficacy in a dose‐dependent manner. Combining subminimal doses of 5‐FU and exemestane exerted an excellent tumour suppression effect without influencing bodyweight. This study validated the therapeutic potentials of exemestane in GCa. Combination of metronomic 5‐FU and exemestane for GCa therapy is recommended.
Collapse
Affiliation(s)
- Juan-Cheng Yang
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of OBS & GYN, China Medical University Hospital, Taichung, Taiwan
| | - Ning Chang
- Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan
| | - Deng-Chyang Wu
- Department of Medicine, Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Wei-Chung Cheng
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of OBS & GYN, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Min Chung
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of OBS & GYN, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Chun Chang
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of OBS & GYN, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan
| | - Fu-Ju Lei
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan
| | - Chung-Jung Liu
- Department of Medicine, Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - I-Chen Wu
- Department of Medicine, Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Hsueh-Chou Lai
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of OBS & GYN, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan
| | - Wen-Lung Ma
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of OBS & GYN, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan.,Department of Nursing, Asia University, Taichung, Taiwan
| |
Collapse
|
326
|
Khayamian MA, Shalileh S, Vanaei S, Salemizadeh Parizi M, Ansaryan S, Saghafi M, Abbasvandi F, Ebadi A, Soltan Khamsi P, Abdolahad M. Electrochemical generation of microbubbles by carbon nanotube interdigital electrodes to increase the permeability and material uptakes of cancer cells. Drug Deliv 2019; 26:928-934. [PMID: 31526074 PMCID: PMC6758649 DOI: 10.1080/10717544.2019.1662514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Artificial cavitation as a prerequisite of sonoporation, plays an important role on the ultrasound (US) assisted drug delivery systems. In this study, we have proposed a new method of microbubble (MB) generation by local electrolysis of the medium. An integrated interdigital array of three-electrode system was designed and patterned on a nickel-coated quartz substrate and then, a short DC electrical pulse was applied that consequently resulted in distributed generation of microbubbles at the periphery of the electrodes. Growth of the carbon nanotube (CNT) nanostructures on the surface of the electrodes approximately increased the number of generated microbubbles up to 7-fold and decreased their average size from ∼20 µm for bare to ∼7 µm for CNT electrodes. After optimizing the three-electrode system, biocompatibility assays of the CNT electrodes stimulated by DC electrical micropulses were conducted. Finally, the effect of the proposed method on the sonoporation efficiency and drug uptake of breast cells were assessed using cell cycle and Annexin V/PI flow cytometry analysis. These results show the potential of electrochemical generation of MBs by CNT electrodes as an easy, available and promising technique for artificial cavitation and ultrasound assisted drug delivery.
Collapse
Affiliation(s)
- Mohammad Ali Khayamian
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,School of Mechanical Engineering, College of Engineering, University of Tehran , Tehran , Iran
| | - Shahriar Shalileh
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran
| | - Shohreh Vanaei
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,School of Biology, College of Science, University of Tehran , Tehran , Iran
| | - Mohammad Salemizadeh Parizi
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran
| | - Saeid Ansaryan
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran
| | - Mohammad Saghafi
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran
| | - Fereshteh Abbasvandi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR , Tehran , Iran
| | - Amirali Ebadi
- MEMS and NEMS Laboratory, Department of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran , Tehran , Iran
| | - Pouya Soltan Khamsi
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran
| | - Mohammad Abdolahad
- Nano Electronic Center of Excellence, Nano Bio Electronic Devices Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran.,Nano Electronic Center of Excellence, Thin Film and Nanoelectronic Lab, School of Electrical and Computer Engineering, University of Tehran , Tehran , Iran
| |
Collapse
|
327
|
Qiu JG, Wang L, Liu WJ, Wang JF, Zhao EJ, Zhou FM, Ji XB, Wang LH, Xia ZK, Wang W, Lin MCM, Liu LZ, Huang YX, Jiang BH. Apigenin Inhibits IL-6 Transcription and Suppresses Esophageal Carcinogenesis. Front Pharmacol 2019; 10:1002. [PMID: 31572184 PMCID: PMC6749068 DOI: 10.3389/fphar.2019.01002] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/06/2019] [Indexed: 01/05/2023] Open
Abstract
Esophagus cancer is the seventh cause of cancer-related deaths globally. In this study, we analyzed interleukin 6 (IL-6) gene expression in human esophagus cancer patients and showed that IL-6 mRNA levels are significantly higher in tumor tissues and negatively correlated with overall survival, suggesting that IL-6 is a potential therapeutic target for esophagus cancer. We further demonstrated that apigenin, a nature flavone product of green plants, inhibited IL-6 transcription and gene expression in human esophagus cancer Eca-109 and Kyse-30 cells. Apigenin significantly and dose-dependently inhibited cell proliferation and promoted apoptosis while stimulating the cleaved PARP (poly ADP-ribose polymerase) (C-PARP) and caspase-8 expression. It suppressed VEGF (Vascular endothelial growth Factor) expression and tumor-induced angiogenesis. Pretreatment of cells with IL-6 could completely reverse apigenin-induced cellular changes. Finally, using a preclinical nude mice model subcutaneously xenografted with Eca-109 cells, we demonstrated the in vivo antitumor activity and mechanisms of apigenin. Taken together, this study revealed for the first time that apigenin is a new IL-6 transcription inhibitor and that inhibiting IL-6 transcription is one of the mechanisms by which apigenin exhibits its anticancer effects. The potential clinical applications of apigenin in treating esophagus cancer warrant further investigations.
Collapse
Affiliation(s)
- Jian-Ge Qiu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Wen-Jing Liu
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ju-Feng Wang
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Er-Jiang Zhao
- Department of Biostatistics, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng-Mei Zhou
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiang-Bo Ji
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Li-Hong Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhong-Kun Xia
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Wei Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Marie Chia-mi Lin
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Ling-Zhi Liu
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
| | - Ying-Xue Huang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Ying-Xue Huang, ; Bing-Hua Jiang,
| | - Bing-Hua Jiang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
- *Correspondence: Ying-Xue Huang, ; Bing-Hua Jiang,
| |
Collapse
|
328
|
Zhao K, Wang Z, Li X, Liu JL, Tian L, Chen JQ. Exosome-mediated transfer of CLIC1 contributes to the vincristine-resistance in gastric cancer. Mol Cell Biochem 2019; 462:97-105. [PMID: 31473882 DOI: 10.1007/s11010-019-03613-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 08/17/2019] [Indexed: 12/26/2022]
Abstract
Our previous study shows that high Chloride intracellular channel 1 (CLIC1) expression can efficiently enhance invasion and migration of gastric cancer (GC) cells in vitro. Growing evidences have found that exosomes are involved in chemotherapy resistance in several cancers including GC. We aimed to evaluate the effect of the exosome-mediated transfer of CLIC1 in the vincristine-resistance of GC. The effect of exosome-mediated transfer of CLIC1 on the development of resistance to vincristine in GC cell line SGC-7901 and the potential underlying mechanisms were investigated by Cell Counting Kit-8 (CCK8), RT-PCR, and Western blotting. Exosomes were isolated from cell supernatants by differential ultracentrifugation. Comparing with SGC-7901, the expression level of CLIC1 is higher in vincristine‑resistant cell line SGC-7901/VCR (P < 0.05). After silencing the expression of CLIC1 by RNA interference, the half inhibition concentration (IC50) to vincristine decreased significantly in SGC-7901/VCR, and the expression of CLIC1 decreased significantly in exosomes from SGC-7901/VCR. After 48 h co-culturing with exosomes from SGC-7901/VCR, the IC50 to vincristine in SGC-7901 increased significantly, and the expression of CLIC1, P-gp, and Bcl-2 were significantly up-regulated. CLIC1 was closely associated with the resistance to vincristine in GC, and exosome-mediated transfer of CLIC1 could induce the development of resistance to vincristine in vitro. The possible mechanism was related to up-regulated P-gp and Bcl-2. However, in vivo study was needed to confirm the results in future.
Collapse
Affiliation(s)
- Kun Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Zhen Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Xin Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Jin-Lu Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Lei Tian
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| | - Jun-Qiang Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
329
|
Ter Veer E, van Oijen MGH, van Laarhoven HWM. The Use of (Network) Meta-Analysis in Clinical Oncology. Front Oncol 2019; 9:822. [PMID: 31508373 PMCID: PMC6718703 DOI: 10.3389/fonc.2019.00822] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022] Open
Abstract
Meta-analysis is important in oncological research to provide a more reliable answer to a clinical research question that was assessed in multiple studies but with inconsistent results. Pair-wise meta-analysis can be applied when comparing two treatments at once, whereas it is possible to compare multiple treatments at once with network meta-analysis (NMA). After careful systematic review of the literature and quality assessment of the identified studies, there are several assumptions in the use of meta-analysis. First, the added value of meta-analysis should be evaluated by examining the comparability of study populations. Second, the appropriate comparator in meta-analysis should be chosen according to the types of comparisons made in individual studies: (1) Experimental and comparator arms are different treatments (A vs. B); (2) Substitution of a conventional treatment by an experimental treatment (A+B vs. A+C); or (3) Addition of an experimental treatment (A+B vs. B). Ideally there is one common comparator treatment, but when there are multiple common comparators, the most efficacious comparator is preferable. Third, treatments can only be adequately pooled in meta-analysis or merged into one treatment node in NMA when considering likewise mechanism of action and similar setting in which treatment is indicated. Fourth, for both pair-wise meta-analysis and NMA, adequate assessment of heterogeneity should be performed and sub-analysis and sensitivity analysis can be applied to objectify a possible confounding factor. Network inconsistency, as statistical manifestation of violating the transitivity assumption, can best be evaluated by node-split modeling. NMA has advantages over pair-wise meta-analysis, such as clarification of inconsistent outcomes from multiple studies including multiple common comparators and indirect effect calculation of missing direct comparisons between important treatments. Also, NMA can provide increased statistical power and cross-validation of the observed treatment effect of weak connections with reasonable network connectivity and sufficient sample-sizes. However, inappropriate use of NMA can cause misleading results, and may emerge when there is low network connectivity, and therefore low statistical power. Furthermore, indirect evidence is still observational and should be interpreted with caution. NMA should therefore preferably be conducted and interpreted by both expert clinicians in the field and an experienced statistician. Finally, the use of meta-analysis can be extended to other areas, for example the identification of prognostic and predictive factors. Also, the integration of evidence from both meta-analysis and expert opinion can improve the construction of prognostic models in real-world databases.
Collapse
Affiliation(s)
- Emil Ter Veer
- Department of Medical Oncology, Cancer Centre Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Martijn G H van Oijen
- Department of Medical Oncology, Cancer Centre Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Cancer Centre Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
330
|
Dijksterhuis WPM, Verhoeven RHA, Slingerland M, Haj Mohammad N, de Vos-Geelen J, Beerepoot LV, van Voorthuizen T, Creemers GJ, van Oijen MGH, van Laarhoven HWM. Heterogeneity of first-line palliative systemic treatment in synchronous metastatic esophagogastric cancer patients: A real-world evidence study. Int J Cancer 2019; 146:1889-1901. [PMID: 31340065 PMCID: PMC7027521 DOI: 10.1002/ijc.32580] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/26/2019] [Accepted: 06/13/2019] [Indexed: 12/16/2022]
Abstract
The optimal first-line palliative systemic treatment strategy for metastatic esophagogastric cancer is not well defined. The aim of our study was to explore real-world use of first-line systemic treatment in esophagogastric cancer and assess the effect of treatment strategy on overall survival (OS), time to failure (TTF) of first-line treatment and toxicity. We selected synchronous metastatic esophagogastric cancer patients treated with systemic therapy (2010-2016) from the nationwide Netherlands Cancer Registry (n = 2,204). Systemic treatment strategies were divided into monotherapy, doublet and triplet chemotherapy, and trastuzumab-containing regimens. Data on OS were available for all patients, on TTF for patients diagnosed from 2010 to 2015 (n = 1,700), and on toxicity for patients diagnosed from 2010 to 2014 (n = 1,221). OS and TTF were analyzed using multivariable Cox regression, with adjustment for relevant tumor and patient characteristics. Up to 45 different systemic treatment regimens were found to be administered, with a median TTF of 4.6 and OS of 7.5 months. Most patients (45%) were treated with doublet chemotherapy; 34% received triplets, 10% monotherapy and 10% a trastuzumab-containing regimen. The highest median OS was found in patients receiving a trastuzumab-containing regimen (11.9 months). Triplet chemotherapy showed equal survival rates compared to doublets (OS: HR 0.92, 95%CI 0.83-1.02; TTF: HR 0.92, 95%CI 0.82-1.04) but significantly more grade 3-5 toxicity than doublets (33% vs. 21%, respectively). In conclusion, heterogeneity of first-line palliative systemic treatment in metastatic esophagogastric cancer patients is striking. Based on our data, doublet chemotherapy is the preferred treatment strategy because of similar survival and less toxicity compared to triplets.
Collapse
Affiliation(s)
- Willemieke P M Dijksterhuis
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Research & Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - Rob H A Verhoeven
- Department of Research & Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands.,Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marije Slingerland
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nadia Haj Mohammad
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Judith de Vos-Geelen
- Division of Medical Oncology, Department of Internal Medicine, GROW - School for Oncology and Developmental Biology, Maastricht UMC+, Maastricht, The Netherlands
| | - Laurens V Beerepoot
- Department of Medical Oncology, Elisabeth-TweeSteden Hospital, Tilburg, The Netherlands
| | | | - Geert-Jan Creemers
- Department of Medical Oncology, Catharina Hospital, Eindhoven, The Netherlands
| | - Martijn G H van Oijen
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Research & Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
331
|
Maron SB, Chase LM, Lomnicki S, Kochanny S, Moore KL, Joshi SS, Landron S, Johnson J, Kiedrowski LA, Nagy RJ, Lanman RB, Kim ST, Lee J, Catenacci DVT. Circulating Tumor DNA Sequencing Analysis of Gastroesophageal Adenocarcinoma. Clin Cancer Res 2019; 25:7098-7112. [PMID: 31427281 DOI: 10.1158/1078-0432.ccr-19-1704] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/01/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE Gastroesophageal adenocarcinoma (GEA) has a poor prognosis and few therapeutic options. Utilizing a 73-gene plasma-based next-generation sequencing (NGS) cell-free circulating tumor DNA (ctDNA-NGS) test, we sought to evaluate the role of ctDNA-NGS in guiding clinical decision-making in GEA. EXPERIMENTAL DESIGN We evaluated a large cohort (n = 2,140 tests; 1,630 patients) of ctDNA-NGS results (including 369 clinically annotated patients). Patients were assessed for genomic alteration (GA) distribution and correlation with clinicopathologic characteristics and outcomes. RESULTS Treatment history, tumor site, and disease burden dictated tumor-DNA shedding and consequent ctDNA-NGS maximum somatic variant allele frequency. Patients with locally advanced disease having detectable ctDNA postoperatively experienced inferior median disease-free survival (P = 0.03). The genomic landscape was similar but not identical to tissue-NGS, reflecting temporospatial molecular heterogeneity, with some targetable GAs identified at higher frequency via ctDNA-NGS compared with previous primary tumor-NGS cohorts. Patients with known microsatellite instability-high (MSI-High) tumors were robustly detected with ctDNA-NGS. Predictive biomarker assessment was optimized by incorporating tissue-NGS and ctDNA-NGS assessment in a complementary manner. HER2 inhibition demonstrated a profound survival benefit in HER2-amplified patients by ctDNA-NGS and/or tissue-NGS (median overall survival, 26.3 vs. 7.4 months; P = 0.002), as did EGFR inhibition in EGFR-amplified patients (median overall survival, 21.1 vs. 14.4 months; P = 0.01). CONCLUSIONS ctDNA-NGS characterized GEA molecular heterogeneity and rendered important prognostic and predictive information, complementary to tissue-NGS.See related commentary by Frankell and Smyth, p. 6893.
Collapse
Affiliation(s)
- Steven B Maron
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Leah M Chase
- The University of Chicago Medical Center, Chicago, Illinois
| | | | - Sara Kochanny
- The University of Chicago Medical Center, Chicago, Illinois
| | - Kelly L Moore
- The University of Chicago Medical Center, Chicago, Illinois
| | - Smita S Joshi
- The University of Chicago Medical Center, Chicago, Illinois
| | - Stacie Landron
- The University of Chicago Medical Center, Chicago, Illinois
| | - Julie Johnson
- The University of Chicago Medical Center, Chicago, Illinois
| | - Lesli A Kiedrowski
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Rebecca J Nagy
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Richard B Lanman
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | | |
Collapse
|
332
|
Viúdez A, Carmona-Bayonas A, Gallego J, Lacalle A, Hernández R, Cano JM, Macías I, Custodio A, Martínez de Castro E, Sánchez A, Iglesia L, Reguera P, Visa L, Azkarate A, Sánchez-Cánovas M, Mangas M, Limón ML, Martínez-Torrón A, Asensio E, Ramchandani A, Martín-Carnicero A, Hurtado A, Cerdà P, Garrido M, Sánchez-Bayonas R, Serrano R, Jiménez-Fonseca P. Optimal duration of first-line chemotherapy for advanced gastric cancer: data from the AGAMENON registry. Clin Transl Oncol 2019; 22:734-750. [PMID: 31385226 DOI: 10.1007/s12094-019-02183-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND The optimal duration of first-line chemotherapy for patients with advanced gastric cancer is unknown. Diverse clinical trials have proposed different strategies including limited treatment, maintenance of some drugs, or treatment until progression. METHOD The sample comprises patients from the AGAMENON multicenter registry without progression after second evaluation of response. The objective was to explore the optimal duration of first-line chemotherapy. A frailty multi-state model was conducted. RESULTS 415 patients were divided into three strata: discontinuation of platinum and maintenance with fluoropyrimidine until progression (30%, n = 123), complete treatment withdrawal prior to progression (52%, n = 216), and full treatment until progression (18%, n = 76). The hazard of tumor progression decreased by 19% per month with the full treatment regimen. However, we found no evidence that fluoropyrimidine maintenance (hazard ratio [HR] 1.07, confidence interval [CI] 95%, 0.69-1.65) worsened progression-free survival (PFS) with respect to treatment until progression. Predictive factors for PFS were ECOG performance status, ≥ 3 metastatic sites, prior tumor response, and bone metastases. Toxicity grade 3/4 was more common in those who continued the full treatment until progression vs fluoropyrimidine maintenance (16% vs 6%). CONCLUSION The longer duration of the full initial regimen exerted a protective effect on the patients of this registry. Platinum discontinuation followed by fluoropyrimidine maintenance yields comparable efficacy to treatment up to PD, with a lower rate of serious adverse events.
Collapse
Affiliation(s)
- A Viúdez
- Medical Oncology Department, Complejo Hospitalario de Navarra (CHN), OncobionaTras Unit, Navarrabiomed, Navarrabiomed Biomedical Center, IdiSNA, Irunlarrea 3, 31008, Pamplona, Navarra, Spain.
| | - A Carmona-Bayonas
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, University of Murcia, IMIB, Murcia, Spain
| | - J Gallego
- Medical Oncology Department, Hospital Universitario de Elche, Elche, Spain
| | - A Lacalle
- Medical Oncology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - R Hernández
- Medical Oncology Department, Hospital Universitario de Canarias, Tenerife, Spain
| | - J M Cano
- Medical Oncology Department, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - I Macías
- Medical Oncology Department, Hospital Universitario Parc Tauli, Sabadell, Spain
| | - A Custodio
- Medical Oncology Department, Hospital Universitario La Paz, Madrid, Spain
| | - E Martínez de Castro
- Medical Oncology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - A Sánchez
- Medical Oncology Department, Hospital Universitario Doce de Octubre, Madrid, Spain
| | - L Iglesia
- Medical Oncology Department, Complejo Hospitalario de Orense, Orense, Spain
| | - P Reguera
- Medical Oncology Department, Hospital Universitario Ramón Y Cajal, Madrid, Spain
| | - L Visa
- Medical Oncology Department, Hospital Universitario El Mar, Barcelona, Spain
| | - A Azkarate
- Medical Oncology Department, Hospital Universitario Son Espases, Mallorca, Spain
| | - M Sánchez-Cánovas
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - M Mangas
- Medical Oncology Department, Hospital Galdakao-Usansolo, Galdakao-Usansolo, Spain
| | - M L Limón
- Medical Oncology Department, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - A Martínez-Torrón
- Pharmacy Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - E Asensio
- Medical Oncology Department, Hospital Universitario de Elche, Elche, Spain
| | - A Ramchandani
- Medical Oncology Department, Hospital Universitario Insular de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - A Martín-Carnicero
- Medical Oncology Department, Complejo Hospitalario San Millán, Logroño, Spain
| | - A Hurtado
- Medical Oncology Department, Hospital Universitario Fundación Alcorcón, Madrid, Spain
| | - P Cerdà
- Medical Oncology Department, Centro Médico Tecknon, Barcelona, Spain
| | - M Garrido
- Medical Oncology Department, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - R Sánchez-Bayonas
- Medical Oncology Department, Clínica Universidad de Navarra, Pamplona, Spain
| | - R Serrano
- Medical Oncology Department, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - P Jiménez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | |
Collapse
|
333
|
Ngai LL, ter Veer E, van den Boorn HG, van Herk EH, van Kleef JJ, van Oijen MGH, van Laarhoven HWM. TOXview: a novel graphical presentation of cancer treatment toxicity profiles. Acta Oncol 2019; 58:1138-1148. [PMID: 31017020 DOI: 10.1080/0284186x.2019.1601256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background: Toxicity profiles play a crucial role in the choice between specific palliative chemotherapy regimens. To optimize the quality of life for cancer patients, patients should be adequately informed about potential toxicities before undergoing chemotherapy. Therefore, we constructed TOXviews, a novel graphical presentation and overview of toxicity profiles to improve information provision about adverse events. As an example, we analyzed first-line chemotherapy regimens for advanced esophagogastric cancer (AEGC). Methods: We searched PubMed, EMBASE, CENTRAL, ASCO and ESMO for prospective phase II or III randomized controlled trials (RCTs) on palliative first-line systemic treatment for AEGC until February 2017. We extracted proportions of Common Terminology Criteria for Adverse Events grade 1-2 (mild) and 3-4 (severe) adverse events from each chemotherapy arm and pooled these by using single-arm meta-analysis. Toxicity profiles per chemotherapy regimen were visualized in bidirectional bar charts with pooled proportions plus 95% confidence intervals. For comparative analysis, chemotherapy regimens were grouped in singlets, doublets and triplets. Results: We included 92 RCTs with a total of 16,963 patients. TOXviews for 3 fluoropyrimidine singlets, 5 cisplatin-containing doublets (C-doublets), 10 fluoropyrimidine non-cisplatin containing doublets (F-doublets), 4 anthracycline-containing triplets (A-triplets) and 5 taxane-containing triplets (T-triplets) were constructed. C-doublets, A-triplets and T-triplets all showed an increased incidence of grade 3-4 adverse events and clinically relevant grade 1-2 adverse events compared to F-doublets. Conclusion: TOXview provides a new graphical presentation and overview of chemotherapy toxicities. TOXviews can be used to educate physicians about the incidences of AEs of systemic therapy and improve informed decision-making.
Collapse
Affiliation(s)
- Lok Lam Ngai
- Department of Medical Oncology, Cancer Centre Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Emil ter Veer
- Department of Medical Oncology, Cancer Centre Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Héctor G. van den Boorn
- Department of Medical Oncology, Cancer Centre Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - E. Hugo van Herk
- Department of Medical Oncology, Cancer Centre Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jessy Joy van Kleef
- Department of Medical Oncology, Cancer Centre Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Martijn G. H. van Oijen
- Department of Medical Oncology, Cancer Centre Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Hanneke W. M. van Laarhoven
- Department of Medical Oncology, Cancer Centre Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
334
|
Lee JG, Kim SA, Eun CS, Han DS, Kim YS, Choi BY, Song KS, Kim HJ, Park CH. Impact of age on stage-specific mortality in patients with gastric cancer: A long-term prospective cohort study. PLoS One 2019; 14:e0220660. [PMID: 31369631 PMCID: PMC6675285 DOI: 10.1371/journal.pone.0220660] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/20/2019] [Indexed: 12/31/2022] Open
Abstract
Controversies exist regarding the impact of age on gastric cancer-related mortality according to cancer stage. In our prospective cohort study, we evaluated the impact of age on stage-specific mortality in patients with gastric cancer. Between 2002 and 2006, patients with newly diagnosed gastric cancer were recruited from two university-affiliated hospitals in Korea. Follow-up data were updated regularly based on medical records and telephone surveys. Patients were classified into four subgroups according to age: <50, 50-59, 60-69, and 70-79 years. A total of 448 patients were followed up for 81.6 months (interquartile range, 25.0-139.3 months). The number of patients with stage I, II, III, and IV disease was 247, 74, 88, and 39, respectively. Overall, age was an independent risk factor for gastric cancer-specific mortality (hazard ratio [HR], [95% confidence interval (CI)]: 1.53 [0.91-2.57], 1.88 [1.21-2.91], and 2.64 [1.69-4.14] in the 50-59, 60-69, and 70-79 years groups, respectively, with the <50 years group as reference). In patients with stage I and II gastric cancer, the 70-79 years group was associated with a significantly higher rate of cancer-specific mortality than the <50 years group (stage I: HR [95% CI], 9.55 [2.11-43.12]; stage II: HR [95% CI], 7.17 [2.32-22.18]). However, age was not an independently associated factor for cancer-specific mortality in patients with stage III and IV gastric cancer. Although age was an independent risk factor for gastric cancer-related mortality in patients with gastric cancer, its impact may differ depending on the stage of cancer.
Collapse
Affiliation(s)
- Jae Gon Lee
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Shin Ah Kim
- Department of Food and Nutrition, Gangneung-Wonju National University, Gangneung, Korea
| | - Chang Soo Eun
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Dong Soo Han
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Yong Sung Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
- Korea & Department of Functional Genomics, University of Science and Technology (UST), Daejeon, Korea
| | - Bo Youl Choi
- Department of Preventive Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Kyu Sang Song
- Department of Pathology, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Hyun Ja Kim
- Department of Food and Nutrition, Gangneung-Wonju National University, Gangneung, Korea
- * E-mail: (HJK); (CHP)
| | - Chan Hyuk Park
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
- * E-mail: (HJK); (CHP)
| |
Collapse
|
335
|
Bjørklund G, Chirumbolo S, Dadar M, Pen JJ, Doşa MD, Pivina L, Semenova Y, Aaseth J. Insights on Nutrients as Analgesics in Chronic Pain. Curr Med Chem 2019; 27:6407-6423. [PMID: 31309880 DOI: 10.2174/0929867326666190712172015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/26/2019] [Accepted: 07/01/2019] [Indexed: 02/07/2023]
Abstract
Many serious inflammatory disorders and nutrient deficiencies induce chronic pain, and anti-inflammatory diets have been applied successfully to modify the inflammatory symptoms causing chronic pain. Numerous scientific data and clinical investigations have demonstrated that long-term inflammation could lead to an inappropriate or exaggerated sensibility to pain. In addition, some Non-steroidal Anti-inflammatory Drugs (NSAID), which directly act on the many enzymes involved in pain and inflammation, including cyclooxygenases, are used to dampen the algesic signal to the central nervous system, reducing the responses of soft C-fibers to pain stimuli. On the other hand, there are a few reports from both health authorities and physicians, reporting that decreased transmission of pain signals can be achieved and improved, depending on the patient's dietary habit. Many nutrients, as well as a suitable level of exercise (resistance training), are the best methods for improving the total mitochondrial capacity in muscle cells, which can lead to a reduction in sensitivity to pain, particularly by lowering the inflammatory signaling to C-fibers. According to the current literature, it could be proposed that chronic pain results from the changed ratio of neuropeptides, hormones, and poor nutritional status, often related to an underlying inflammatory disorder. The current review also evaluates the effective role of nutrition-related interventions on the severity of chronic pain. This review pointed out that nutritional interventions can have a positive effect on pain experience through the indirect inhibitory effect on prostaglandin E2 and attenuation of mitochondrial dysfunction caused by ischemia/reperfusion in skeletal muscle, improving the intracellular antioxidant defense system. These data highlight the need for more nutrition studies where chronic pain is the primary outcome, using accurate interventions. To date, no nutritional recommendation for chronic pain has been officially proposed. Therefore, the goal of this article is to explore pain management and pain modulation, searching for a mode of nutrition efficient in reducing pain.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy,CONEM Scientific Secretary, Verona, Italy
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Joeri J Pen
- Diabetes Clinic, Department of Internal Medicine, UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium,Department of Nutrition, UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Monica Daniela Doşa
- Department of Pharmacology, Faculty of Medicine, Ovidius University, Constanta, Romania
| | - Lyudmila Pivina
- Semey Medical University, Semey, Kazakhstan,CONEM Kazakhstan Environmental Health and Safety Research Group, Semey Medical University, Semey, Kazakhstan
| | - Yulia Semenova
- Semey Medical University, Semey, Kazakhstan,CONEM Kazakhstan Environmental Health and Safety Research Group, Semey Medical University, Semey, Kazakhstan
| | - Jan Aaseth
- Research Department, Innlandet Hospital Trust, Brumunddal, Norway,Faculty of Health and Social Science, Inland Norway University of Applied Sciences, Elverum, Norway
| |
Collapse
|
336
|
Gong X, Xu B, Zi L, Chen X. miR-625 reverses multidrug resistance in gastric cancer cells by directly targeting ALDH1A1. Cancer Manag Res 2019; 11:6615-6624. [PMID: 31410057 PMCID: PMC6643062 DOI: 10.2147/cmar.s208708] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/28/2019] [Indexed: 12/19/2022] Open
Abstract
Background: microRNAs (miRNAs) are emerging as critical regulators of multidrug resistance (MDR) in gastric cancer, a major cause of chemotherapy failure. miR-625 is downregulated in gastric cancer and negatively associated with metastasis. In the current study, we aimed to investigate whether miR-625 regulates MDR in gastric cancer. Methods: The level of miR-625 in gastric cancer cells with or without MDR was quantified by quantitative reverse transcription PCR (qRT-PCR) analysis. The sensitivity of gastric cancer cells to chemotherapeutic agents was assessed by MTT assay. The protein expression was determined by Western blot analysis, and the luciferase reporter assay was applied to confirm miR-625 regulation of the potential target. Results: miR-625 is downregulated in MDR gastric cancer cells compared with chemosensitive counterparts. In addition, miR-625 increases the sensitivity and promotes apoptosis of gastric cancer cells when treated with different chemotherapeutic agents. Moreover, miR-625 directly targets the aldehyde dehydrogenase 1A1 (ALDH1A1), and importantly, the restoration of ALDH1A1 expression rescues miR-625 effects on MDR in gastric cancer cells. Conclusion: miR-625 reverses MDR in gastric cancer cells by targeting ALDH1A1. Hence, our study identifies miR-625 as a novel regulator of MDR in gastric cancer cells, and implicates its potential application for overcoming MDR in gastric cancer chemotherapy.
Collapse
Affiliation(s)
- Xufei Gong
- Department of General Surgery, Linyi People's Hospital, Linyi, Shandong 276002, People's Republic of China
| | - Baoli Xu
- Department of General Surgery, Linyi People's Hospital, Linyi, Shandong 276002, People's Republic of China
| | - Li Zi
- Department of General Surgery, Linyi People's Hospital, Linyi, Shandong 276002, People's Republic of China
| | - Xinrui Chen
- Department of General Surgery, Linyi People's Hospital, Linyi, Shandong 276002, People's Republic of China
| |
Collapse
|
337
|
Novel Agents in Heavily Pretreated Metastatic Gastric Cancer: More Shadows Than Lights. JOURNAL OF ONCOLOGY 2019; 2019:5692317. [PMID: 31354820 PMCID: PMC6637676 DOI: 10.1155/2019/5692317] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/14/2019] [Accepted: 06/12/2019] [Indexed: 12/20/2022]
Abstract
Metastatic gastric cancer is still a disease with a poor prognosis. Recently, different novel agents (e.g., apatinib, nivolumab, TAS-102) have demonstrated a survival advantage compared with placebo for patients with heavily pretreated metastatic gastric cancer. Although the possible availability of active agents may be a desirable option in a very poor therapeutic scenario, clinical data from the recent studies with these drugs raise yet controversial issues. The purpose of this review is to briefly summarize the results of these novel drugs focusing on the limitations that bring some shadows on their positive therapeutic results.
Collapse
|
338
|
Özdemir BC, Moehler M, Syn NLX, Tai BC, Wagner AD. Immune checkpoint inhibitors plus chemotherapy versus chemotherapy or immune checkpoint inhibitors for first- or second-line treatment of advanced gastric and gastro-esophageal junction cancer. Hippokratia 2019. [DOI: 10.1002/14651858.cd013369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Berna C Özdemir
- Lausanne University Hospitals and Clinics; Department of Oncology; Rue de Bugnon 46 Lausanne Vaud Switzerland 1011
| | - Markus Moehler
- University Medical Center of the Johannes Gutenberg University; Department of Internal Medicine; Langenbeckstrasse 1 Mainz Germany 55131
| | - Nicholas LX Syn
- National University Cancer Institute; Department of Haematology-Oncology; 1E Kent Ridge Road NUHS Tower Block, Level 7 Singapore Singapore 119228
| | - Bee-Choo Tai
- National University of Singapore; Saw Swee Hock School of Public Health; 12 Science Drive 2 #10-03F Singapore Singapore 117549
| | - Anna Dorothea Wagner
- Lausanne University Hospitals and Clinics; Department of Oncology; Rue de Bugnon 46 Lausanne Vaud Switzerland 1011
| |
Collapse
|
339
|
Chan WL, Lam KO, So TH, Lee VHF, Kwong LWD. Third-line systemic treatment in advanced/metastatic gastric cancer: a comprehensive review. Ther Adv Med Oncol 2019; 11:1758835919859990. [PMID: 31285759 PMCID: PMC6600493 DOI: 10.1177/1758835919859990] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 06/06/2019] [Indexed: 12/11/2022] Open
Abstract
The management of advanced gastric cancer has improved over the past decade.
There is more evidence to support the efficacy of systemic treatment in
refractory gastric cancer beyond second-line treatment. Important randomized
controlled trials of chemotherapies, targeted agents and immunotherapies have
been reported. With the development of these novel therapies, clinicians can
better individualize treatment for patients beyond progression on second-line
therapy. However, there is no guideline on third-line therapy available for
clinicians. This review discussed the efficacy and safety data from the pivotal
trials of the agents proven to be effective in third-line settings, including
the quality of study design, level of evidence and subgroup analysis, and how
the data can help to guide clinicians on selecting the most appropriate
third-line therapy for their patients.
Collapse
Affiliation(s)
- Wing-Lok Chan
- Department of Clinical Oncology, Queen Mary Hospital, The University of Hong Kong, 1/F Professorial Block, 102 Pokfulam Road, Hong Kong
| | - Ka-On Lam
- Department of Clinical Oncology, Queen Mary Hospital, The University of Hong Kong, 1/F Professorial Block, 102 Pokfulam Road, Hong Kong
| | - Tsz-Him So
- Department of Clinical Oncology, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Victor Ho-Fun Lee
- Department of Clinical Oncology, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Lai-Wan Dora Kwong
- Department of Clinical Oncology, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| |
Collapse
|
340
|
Abstract
PURPOSE OF REVIEW Gastric adenocarcinoma is the fifth most common and the third most lethal cancer worldwide. Surgery is the only chance of cure, but recurrence is common, even with complete resection. RECENT FINDINGS Advances in diagnosis and staging, genomic classification, surgical resection and treatment of peritoneal disease, systemic chemotherapy and chemoradiation, and targeted and immune therapies have led to the current multidisciplinary approach to gastric adenocarcinoma. Treatment of gastric cancer is rapidly evolving in an effort to combat this challenging disease.
Collapse
Affiliation(s)
- Fabian M Johnston
- Section of Gastrointestinal Surgical Oncology, Peritoneal Surface Malignancy Program, Division of Surgical Oncology, Johns Hopkins University, 600 N. Wolfe Street/Blalock 606, Baltimore, MD, 21287, USA.
| | - Michael Beckman
- Department of Surgery, Johns Hopkins Hospital, 600 N. Wolfe Street/Blalock 665, Baltimore, MD, 21287, USA
| |
Collapse
|
341
|
Takeuchi M, Ajani JA, Fang X, Pfeiffer P, Takeuchi M, van Laarhoven HWM. Meta-Enrichment Analyses to Identify Advanced Gastric Cancer Patients Who Achieve a Higher Response to S-1/Cisplatin. Cancers (Basel) 2019; 11:cancers11060871. [PMID: 31234436 PMCID: PMC6627221 DOI: 10.3390/cancers11060871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/15/2019] [Accepted: 06/17/2019] [Indexed: 11/16/2022] Open
Abstract
The Multicenter phase III comparison of cisplatin/S-1 with cisplatin/infusional fluorouracil in advanced gastric or gastroesophageal adenocarcinoma study (FLAGS) and the Diffuse Gastric and Esophagogastric Junction Cancer S-1 Trial (DIGEST) have shown that patients with advanced gastric cancer treated with S-1/Cisplatin (CS) have similar overall survival (OS) compared to 5-fluorouracil/cisplatin (CF). The purpose of this analysis was to identify patients who may specifically benefit from CS using meta-enrichment analysis of the combined two datasets. Eleven clinico-pathological factors were selected and a high response enrichable population was determined. The efficacy of CS in the combined data set of 1365 patients (n = 1019 from FLAGS and n = 346 from DIGEST) was analyzed. We identified 683 patients (n = 374 from CS, n = 309 from CF) as the high response enrichable population who were classified as those with Eastern Cooperative Oncology Group Performance Status (ECOG PS) 1, more than two metastatic sites and low neutrophil-lymphocyte ratio (log(NL ratio)). In the high response enrichable population, the median OS in the CS group was 241 days compared to 210 days in the CF group (hazard ratio 0.776; 95% confidence interval 0.658 to 0.915; p-value 0.004). Through meta-enrichment analysis, the high response enrichable population to CS was identified. Our findings show the clinical importance of selecting the appropriate treatment based on specific patient characteristics.
Collapse
Affiliation(s)
- Madoka Takeuchi
- Faculty of Environment and Information Studies, Keio University, 5322 Endo, Fujisawa-shi, Kanagawa 252-0882, Japan.
| | - Jaffer A Ajani
- Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Xuemin Fang
- Clinical Medicine (Biostatistics), Kitasato University, Tokyo 108-8641, Japan.
| | - Per Pfeiffer
- Experimental research in medical cancer therapy, Odense University Hospital, 5000 Odense C, Denmark.
| | - Masahiro Takeuchi
- Clinical Medicine (Biostatistics), Kitasato University, Tokyo 108-8641, Japan.
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
342
|
Legué LM, Bernards N, Lemmens VE, de Hingh IH, Creemers GJ, van Erning FN. Palliative chemotherapy for patients with synchronous metastases of small-bowel adenocarcinoma: A reflection of daily practice. United European Gastroenterol J 2019; 7:1380-1388. [PMID: 31839964 DOI: 10.1177/2050640619858211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/28/2019] [Indexed: 12/27/2022] Open
Abstract
Background As small-bowel adenocarcinoma (SBA) is scarce, no standard systemic regimen in metastatic disease has been defined. Objective To obtain insights into the use and effects of palliative chemotherapy in patients with metastatic SBA in a population-based setting. Methods Data from the Netherlands Cancer Registry of patients with metastatic SBA between 2007 and 2016 were used (n = 522). For patients treated with palliative chemotherapy, differences in treatment regimens and survival were evaluated. Results Palliative chemotherapy was received by 38% of patients (n = 199). First-line combination chemotherapy was administered to 80% of patients, mainly CAPOX/FOLFOX. Single-agent chemotherapy mostly consisted of capecitabine. Second-line treatment, mostly irinotecan-based (58%), was prescribed to 27% of patients. Age 70 years or older was an adverse predictive factor for receiving first-line combination chemotherapy (odds ratio (OR) 0.2, 95% confidence interval (CI) 0.08-0.62) and second-line therapy (OR 0.3, 95% CI 0.10-0.72). Median overall survival with palliative chemotherapy was 9.3 months, compared with 3.0 months without. In subanalyses, patients who received only first-line treatment had a median overall survival of 5.6 and 7.0 months after single-agent and combination chemotherapy, respectively. Conclusion A minority of patients were treated with palliative chemotherapy. First-line treatment consisted predominantly of oxaliplatin-based combination chemotherapy, whereas second-line treatment was mainly irinotecan-based. Population-based median overall survival for selected patients treated with chemotherapy amounted to nine months.
Collapse
Affiliation(s)
- Laura M Legué
- Department of Medical Oncology, Catharina Hospital, Eindhoven, the Netherlands.,Department of Research, Netherlands Comprehensive Cancer Organisation, Eindhoven, the Netherlands
| | - Nienke Bernards
- Department of Medical Oncology, Catharina Hospital, Eindhoven, the Netherlands
| | - Valery Epp Lemmens
- Department of Research, Netherlands Comprehensive Cancer Organisation, Eindhoven, the Netherlands.,Department of Public Health, Erasmus MC University Medical Centre, Rotterdam, the Netherlands
| | - Ignace Hjt de Hingh
- Department of Research, Netherlands Comprehensive Cancer Organisation, Eindhoven, the Netherlands.,Department of Surgical Oncology, Catharina Hospital, Eindhoven, the Netherlands.,GROW, School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Geert-Jan Creemers
- Department of Medical Oncology, Catharina Hospital, Eindhoven, the Netherlands
| | - Felice N van Erning
- Department of Research, Netherlands Comprehensive Cancer Organisation, Eindhoven, the Netherlands
| |
Collapse
|
343
|
Schokker S, van der Woude SO, van Kleef JJ, van Zoen DJ, van Oijen MGH, Mearadji B, Beenen LFM, Stroes CI, Waasdorp C, Jibodh RA, Creemers A, Meijer SL, Hooijer GKJ, Punt CJA, Bijlsma MF, van Laarhoven HWM. Phase I Dose Escalation Study with Expansion Cohort of the Addition of Nab-Paclitaxel to Capecitabine and Oxaliplatin (CapOx) as First-Line Treatment of Metastatic Esophagogastric Adenocarcinoma (ACTION Study). Cancers (Basel) 2019; 11:cancers11060827. [PMID: 31207904 PMCID: PMC6627561 DOI: 10.3390/cancers11060827] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/31/2019] [Accepted: 06/11/2019] [Indexed: 02/07/2023] Open
Abstract
First-line triplet chemotherapy including a taxane may prolong survival in patients with metastatic esophagogastric cancer. The added toxicity of the taxane might be minimized by using nab-paclitaxel. The aim of this phase I study was to determine the feasibility of combining nab-paclitaxel with the standard of care in the Netherlands, capecitabine and oxaliplatin (CapOx). Patients with metastatic esophagogastric adenocarcinoma received oxaliplatin 65 mg/m2 on days 1 and 8, and capecitabine 1000 mg/m2 bid on days 1-14 in a 21-day cycle, with nab-paclitaxel on days 1 and 8 at four dose levels (60, 80, 100, and 120 mg/m2, respectively), using a standard 3 + 3 dose escalation phase, followed by a safety expansion cohort. Baseline tissue and serum markers for activated tumor stroma were assessed as biomarkers for response and survival. Twenty-six patients were included. The first two dose-limiting toxicities (i.e., diarrhea and dehydration) occurred at dose level 3. The resulting maximum tolerable dose (MTD) of 80 mg/m2 was used in the expansion cohort, but was reduced to 60 mg/m2 after three out of eight patients experienced diarrhea grade 3. The objective response rate was 54%. The median progression-free (PFS) and overall survival were 8.0 and 12.8 months, respectively. High baseline serum ADAM12 was associated with a significantly shorter PFS (p = 0.011). In conclusion, albeit that the addition of nab-paclitaxel 60 mg/m2 to CapOx may be better tolerated than other taxane triplets, relevant toxicity was observed. There is a rationale for preserving taxanes for later-line treatment. ADAM12 is a potential biomarker to predict survival, and warrants further investigation.
Collapse
Affiliation(s)
- Sandor Schokker
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Stephanie O van der Woude
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Jessy Joy van Kleef
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Daan J van Zoen
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Martijn G H van Oijen
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Banafsche Mearadji
- Department of Radiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Ludo F M Beenen
- Department of Radiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Charlotte I Stroes
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Cynthia Waasdorp
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - R Aarti Jibodh
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Aafke Creemers
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Sybren L Meijer
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Gerrit K J Hooijer
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Cornelis J A Punt
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Maarten F Bijlsma
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | - Hanneke W M van Laarhoven
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| |
Collapse
|
344
|
Rosenberg AJ, Rademaker A, Hochster HS, Ryan T, Hensing T, Shankaran V, Baddi L, Mahalingam D, Mulcahy MF, Benson AB. Docetaxel, Oxaliplatin, and 5-Fluorouracil (DOF) in Metastatic and Unresectable Gastric/Gastroesophageal Junction Adenocarcinoma: A Phase II Study with Long-Term Follow-Up. Oncologist 2019; 24:1039-e642. [PMID: 31138725 PMCID: PMC6693711 DOI: 10.1634/theoncologist.2019-0330] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 04/23/2019] [Indexed: 12/13/2022] Open
Abstract
Lessons Learned. Adding docetaxel to the modified FOLFOX7 backbone (DOF) is a feasible three‐drug combination therapy for advanced gastric cancer with high activity, providing evidence that leucovorin is not necessary in this setting. The DOF regimen represents an alternative to the FLOT (5‐FU 2,600 mg/m2 as 24‐hour infusion with leucovorin 200 mg/m2, oxaliplatin 85 mg/m2, and docetaxel 50 mg/m2) regimen that can be considered in select patients with advanced gastric cancer and is a potential choice in the curative setting.
Background. The combination of docetaxel, cisplatin, and 5‐fluorouracil (5‐FU) demonstrates high response rates in advanced gastric cancer, albeit with increased toxicity. Given the efficacy of platinum‐taxane‐fluoropyrimidine regimens, this phase II study evaluated the efficacy and toxicity of docetaxel, oxaliplatin, and 5‐FU (DOF) for the treatment of metastatic or unresectable gastric or gastroesophageal junction (GEJ) adenocarcinoma. Methods. Patients with metastatic or unresectable gastric or GEJ adenocarcinoma with no prior therapy for metastatic disease received docetaxel 50 mg/m2 on day 1, oxaliplatin 85 mg/m2 on day 1, and 5‐FU 2,400 mg/m2 continuous intravenous infusion over 46 hours; cycles were repeated every 2 weeks. The primary endpoint was overall response rate (ORR). Results. Forty‐four patients were enrolled. Assessment of treatment response and toxicity was feasible in 41 and 43 patients, respectively. ORR was 73.2% (68.3% partial response; 4.9% complete response). Therapy was discontinued for progressive disease in 53%, toxicity in 26%, and death on treatment in 16%. Two patients underwent surgical resection. Thirty‐three patients (76.7%) received at least seven cycles (7–34). Grade 3–4 toxicities occurred in 31 patients (72.1%), including neutropenia (23.3%), neurologic (20.9%), and diarrhea (14.0%). Median overall survival was 10.3 months. Conclusion. DOF demonstrates a high response rate, expected safety profile, and prolonged survival and remains an option for select patients with unresectable or metastatic gastric or GEJ adenocarcinoma.
Collapse
Affiliation(s)
- Ari Joseph Rosenberg
- Department of Medicine, Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Alfred Rademaker
- Department of Preventative Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Howard S Hochster
- Division of Medical Oncology, Section of Solid Tumor Oncology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Theresa Ryan
- Department of Medicine, Division of Hematology and Medical Oncology, New York University, New York New York, USA
| | - Thomas Hensing
- North Shore University Health System, Evanston, Illinois, USA
| | - Veena Shankaran
- Department of Medicine, Division of Oncology, Seattle Cancer Care Alliance, Seattle, Washington, USA
| | - Lisa Baddi
- US Oncology, Illinois Cancer Specialists, Chicago, Illinois, USA
| | - Devalingam Mahalingam
- Department of Medicine, Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Mary F Mulcahy
- Department of Medicine, Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Al B Benson
- Department of Medicine, Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
345
|
Zhang Z, Zhao W, Lin X, Gao J, Zhang Z, Shen L. Voltage-dependent calcium channel α2δ1 subunit is a specific candidate marker for identifying gastric cancer stem cells. Cancer Manag Res 2019; 11:4707-4718. [PMID: 31213895 PMCID: PMC6536713 DOI: 10.2147/cmar.s199329] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 04/03/2019] [Indexed: 12/22/2022] Open
Abstract
Purpose: Cancer stem cells (CSCs) are a subpopulation of cancer cells with self-renewal property and responsible for tumor malignancy, progression and drug resistance. Researches on CSC-specific markers in gastric cancer remain limited. Our current study explored the expression of voltage-dependent calcium channel α2δ1 subunit and the potential of using α2δ1 as a CSC marker in gastric cancer. We also compared the specificity of α2δ1 and CD44 in identifying gastric cancer stem cells (GCSCs). Materials and methods: Expression of α2δ1 was analyzed in gastric cancer cell lines, patient-derived xenograft (PDX) models and clinical samples of malignant ascites of gastric cancer patients. α2δ1+ gastric cancer cells were isolated from gastric cancer cell lines. CSC properties of α2δ1+ gastric cancer cells were then verified by subsequent tests both in vitro and in vivo. Results: The expression level of α2δ1 was found to differ drastically among gastric cancer cell lines, PDX models and clinical samples of malignant ascites. α2δ1+ gastric cancer cells sorted from HGC-27 and SGC-7901 cell lines demonstrated significant self-renewal properties, including tumorigenic capacity, sphere-formation capacity and asymmetric differentiation potential. Knockdown of α2δ1 in α2δ1+ HGC-27 significantly inhibited CSC properties and rendered HGC-27 more sensitive to chemotherapy. Flow cytometry showed that α2δ1+ gastric cancer cells accounted for a small fraction of CD44+ gastric cancer cells. Isolated CD44+α2δ1+ HGC-27 cells displayed more significant tumorigenic capacity and sphere-forming capacity compared with their CD44+α2δ1− counterparts. Conclusion: α2δ1+ gastric cancer cells possessed CSC properties. α2δ1 could be a proper marker in identifying GCSCs with superior specificity than CD44. The combination of α2δ1 and CD44 could be used to identify GCSCs with improved accuracy. Knockdown of α2δ1 combined with chemotherapy displayed higher therapeutic efficacy on gastric cancer cells, suggesting that α2δ1 could be a potential target for anticancer treatment.
Collapse
Affiliation(s)
- Ziran Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Wei Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Xiaoting Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Jing Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Zhiqian Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| |
Collapse
|
346
|
Efficacy and Safety of Xiao Ai Ping Injection Combined with Chemotherapy in Advanced Gastric Cancer: A Systematic Review and Meta-Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:3821053. [PMID: 31236124 PMCID: PMC6545757 DOI: 10.1155/2019/3821053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 04/16/2019] [Indexed: 12/24/2022]
Abstract
Xiao Ai Ping injection (XAPI), extracted from the Chinese herbal medicine Marsdenia tenacissima, is widely used in the adjuvant treatment of tumors in China. The present study aimed to evaluate the efficacy and safety of XAPI combined with chemotherapy for treating patients with advanced gastric cancer. Seven databases were searched for relevant studies published up to October 1, 2018, and Review Manager 5.3 software and Stata 12.0 software were used for meta-analysis. Fourteen studies, representing 1097 enrolled patients, were included in our analysis. Compared with chemotherapy alone, combination treatment with XAPI and the XELOX regimen (capecitabine plus oxaliplatin) was found to improve the objective response rate (ORR) [RR=1.36; 95%CI (1.10, 1.70); P=0.006], disease control rate (DCR) [RR=1.15; 95% CI (1.04, 1.28); P=0.010], and Karnofsky Performance Status (KPS) improvement rate [RR=1.51; 95%CI (1.14, 2.00); P=0.004] and to reduce the incidence of leukopenia [RR=0.68; 95%CI (0.55,0.84); P=0.0005], liver damage [RR=0.59; 95% CI (0.37, 0.92); P=0.02], renal impairment [RR=0.39; 95% CI (0.18, 0.85); P=0.02], and hand-foot syndrome [RR=0.56; 95%CI (0.35,0.90); P=0.02]. However, median progression-free survival (PFS), 1-year survival rate, and median overall survival (OS) were not extended by XAPI plus XELOX. Combination treatment with XAPI and the SOX regimen (tegafur plus oxaliplatin) did not improve ORR or DCR, but it did enhance the KPS improvement rate [RR=1.73; 95%CI (1.23,2.43); P=0.002] and reduce the incidence of nausea and vomiting [RR=0.66; 95% CI (0.50, 0.88); P=0.004]. XAPI in combination with the FOLFOX regimen (fluorouracil/calcium folinate/oxaliplatin) enhanced only the KPS improvement rate [RR=1.68; 95%CI (1.18,2.39); P=0.004] and had no significant effect on ORR or DCR or the incidence of adverse events. A single study reported that XAPI combined with the CPT-11 regimen (irinotecan) was superior to chemotherapy alone with respect to DCR and also reduced the incidence of leukopenia, liver damage, and hand-foot syndrome during chemotherapy, while prolonging PFS. Finally, one study reported that XAPI combined with the TP regimen (palitaxel plus cisplatin) improved ORR and KPS improvement rate to a greater extent than TP alone. Although the present review has some limitations, the findings suggest that XAPI combined with chemotherapy may represent a beneficial treatment strategy, particularly the combination of XAPI and XELOX.
Collapse
|
347
|
Zhao D, Klempner SJ, Chao J. Progress and challenges in HER2-positive gastroesophageal adenocarcinoma. J Hematol Oncol 2019; 12:50. [PMID: 31101074 PMCID: PMC6525398 DOI: 10.1186/s13045-019-0737-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 04/25/2019] [Indexed: 02/08/2023] Open
Abstract
HER2 expression remains an important biomarker to guide the addition of the monoclonal antibody trastuzumab to first-line systemic chemotherapy in unresectable, metastatic gastroesophageal adenocarcinomas (GEA). However, in contrast to breast cancer, other HER2-targeted strategies to date have not improved outcomes in this molecular subtype of GEA. Since the initial development of HER2 biomarker testing guidelines, significant spatial intratumoral heterogeneity of HER2 overexpression has been recognized as a major characteristic of this disease. In this review, we aim to survey the seminal positive and negative trials investigating HER2-targeted agents for GEA. We also highlight emerging data on the genomic and temporal heterogeneity of molecular resistance alterations that have yielded further insight into the heterogeneity of therapeutic responses. We conclude with an overview of promising novel agents and strategies which may refine the therapeutic landscape.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Bldg. 51, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| | - Samuel J Klempner
- The Angeles Clinic and Research Institute, Los Angeles, CA, 90025, USA.,Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Joseph Chao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Bldg. 51, 1500 E. Duarte Rd, Duarte, CA, 91010, USA.
| |
Collapse
|
348
|
Seesing MFJ, van der Veen A, Brenkman HJF, Stockmann HBAC, Nieuwenhuijzen GAP, Rosman C, van den Wildenberg FJH, van Berge Henegouwen MI, van Duijvendijk P, Wijnhoven BPL, Stoot JHMB, Lacle M, Ruurda JP, van Hillegersberg R. Resection of hepatic and pulmonary metastasis from metastatic esophageal and gastric cancer: a nationwide study. Dis Esophagus 2019; 32:5480096. [PMID: 31220859 PMCID: PMC7705435 DOI: 10.1093/dote/doz034] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/08/2019] [Accepted: 03/28/2019] [Indexed: 12/11/2022]
Abstract
The standard of care for gastroesophageal cancer patients with hepatic or pulmonary metastases is best supportive care or palliative chemotherapy. Occasionally, patients can be selected for curative treatment instead. This study aimed to evaluate patients who underwent a resection of hepatic or pulmonary metastasis with curative intent. The Dutch national registry for histo- and cytopathology was used to identify these patients. Data were retrieved from the individual patient files. Kaplan-Meier survival analysis was performed. Between 1991 and 2016, 32,057 patients received a gastrectomy or esophagectomy for gastroesophageal cancer in the Netherlands. Of these patients, 34 selected patients received a resection of hepatic metastasis (n = 19) or pulmonary metastasis (n = 15) in 21 different hospitals. Only 4 patients received neoadjuvant therapy before metastasectomy. The majority of patients had solitary, metachronous metastases. After metastasectomy, grade 3 (Clavien-Dindo) complications occurred in 7 patients and mortality in 1 patient. After resection of hepatic metastases, the median potential follow-up time was 54 months. Median overall survival (OS) was 28 months and the 1-, 3-, and 5- year OS was 84%, 41%, and 31%, respectively. After pulmonary metastases resection, the median potential follow-up time was 80 months. The median OS was not reached and the 1-, 3-, and 5- year OS was 67%, 53%, and 53%, respectively. In selected patients with gastroesophageal cancer with hepatic or pulmonary metastases, metastasectomy was performed with limited morbidity and mortality and offered a 5-year OS of 31-53%. Further prospective studies are required.
Collapse
Affiliation(s)
- M F J Seesing
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht
| | - A van der Veen
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht
| | - H J F Brenkman
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht
| | | | | | - C Rosman
- Department of Surgical Oncology, Radboud University Medical Center, Nijmegen
| | | | | | | | - B P L Wijnhoven
- Department of Surgical Oncology, Erasmus University Medical Center, Rotterdam
| | - J H M B Stoot
- Department of Surgery, Zuyderland Medical Center, Heerlen and Sittard-Geleen
| | - M Lacle
- Department of Pathology, University Medical Center Utrecht, Utrecht
| | - J P Ruurda
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht
| | - R van Hillegersberg
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht,Address correspondence to: Richard van Hillegersberg, MD, PhD, Department of Surgical Oncology, University Medical Center Utrecht, Heidelberglaan 100 G04.228, 3508 GA, Utrecht, the Netherlands.
| | | |
Collapse
|
349
|
Zhang F, Zhang Y, Jia Z, Wu H, Gu K. Oxaliplatin-Based Regimen is Superior to Cisplatin-Based Regimen in Tumour Remission as First-line Chemotherapy for Advanced Gastric Cancer: A Meta-Analysis. J Cancer 2019; 10:1923-1929. [PMID: 31205551 PMCID: PMC6547983 DOI: 10.7150/jca.28896] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/19/2019] [Indexed: 01/20/2023] Open
Abstract
Background: This study was initially designed to examine whether oxaliplatin-based regimen was superior to cisplatin-based regimen in tumour remission as first-line chemotherapy for advanced gastric cancer (GC). Methods: Literature in EMBASE, PUBMED, Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, American Society of Clinical Oncology (ASCO) and European Society of Medical Oncology (ESMO) was searched. Only phase II or III randomized controlled trials (RCTs) comparing the effectiveness and safety between oxaliplatin-based and cisplatin-based regimens as first-line treatment for advanced GC were selected. Odds ratios (ORs) with 95% confidence intervals (CIs) were reported. The primary endpoints were complete remission rate (CRR), partial remission rate (PRR), objective response rate (ORR), and disease control rate (DCR). The second endpoint was the toxicity response. Results: 2,140 patients from six phase II or III RCTs were included. Compared to cisplatin-based therapy, subjects who received oxaliplatin-based treatment had significantly higher PRR (OR: 1.25, 95%CI: 1.05-1.48, P=0.01, I2=0%), ORR (OR: 1.21, 95%CI: 1.02-1.44, P=0.03, I2=0%) and DCR (OR: 1.76, 95%CI: 1.31-2.38, P=0.0002, I2=25%), but not CRR (OR: 0.70, 95%CI: 0.37-1.31, P=0.27, I2=0%). In addition, oxaliplatin-based therapy significantly decreased all grades of leukopenia, neutropenia, anemia, febrile neutropenia, nausea, stomatitis, creatinine elevation and thromboembolism, as well as grades 3-4 of leukopenia, neutropenia, anemia and febrile neutropenia than cisplatin-based regimen. However, oxaliplatin-based treatment strikingly increased the risk of thrombocytopenia, sensory neuropathy, diarrhea, fatigue and liver dysfunction. Conclusions: Oxaliplatin-based regimen is superior to cisplatin-based regimen in tumour remission as first-line chemotherapy for advanced GC, and is associated with less toxicity and better tolerability.
Collapse
Affiliation(s)
| | | | | | | | - Kangsheng Gu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China
| |
Collapse
|
350
|
Duan S, Wang P, Liu F, Huang H, An W, Pan S, Wang X. Novel immune-risk score of gastric cancer: A molecular prediction model combining the value of immune-risk status and chemosensitivity. Cancer Med 2019; 8:2675-2685. [PMID: 30945466 PMCID: PMC6537086 DOI: 10.1002/cam4.2077] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/12/2019] [Accepted: 02/17/2019] [Indexed: 12/21/2022] Open
Abstract
Gastric cancer is still one of the most common and deadly malignancies in the world. Not all patients could benefit from chemotherapy or chemoradiotherapy due to tumor heterogeneity. Therefore, identifying different subgroups of patients is an important trend for obtaining more effective responses. However, few molecular classifications associated with chemosensitivity are based on immune–risk status. In this study, we obtained six key immune–related genes. Using these genes, we constructed a molecular model related to immune–risk status and calculated an individual immune–risk score. The score showed great efficiency and stability in predicting prognosis and identifying different subgroups where persons could benefit from postoperative adjuvant therapy. The patients could be divided into different risk groups based on the immune–related score. For patients in the low–risk group, both postoperative chemoradiotherapy and chemotherapy could significantly improve prognosis on overall survival (OS) and disease–free survival (DFS) (DFS, P < 0.001 and P = 0.041, respectively; OS, P < 0.001, P = 0.006, respectively) and chemoradiotherapy was significantly superior than simple chemotherapy (DFS, P = 0.031; OS, P = 0.027). For patients with an intermediate–risk score, postoperative chemoradiotherapy showed a statistically significant survival advantage over no anticancer treatment (P = 0.004 and P = 0.002, respectively), while chemotherapy did not. Compared with no adjuvant treatment, neither postoperative chemoradiotherapy nor chemotherapy made significant difference for patients in the high–risk group. Combining the value of immune–risk status and chemosensitivity, the immune–risk score could not only offer us prognostic evaluation and adjuvant treatment guidance, but also improve our understanding about the binding point between chemotherapy or chemoradiotherapy and the immune system, which may be helpful for further expanding the application of immunotherapy.
Collapse
Affiliation(s)
- Shijie Duan
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Pengliang Wang
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Funan Liu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hanwei Huang
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wen An
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Siwei Pan
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xin Wang
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|