601
|
Lan K, Yan R, Zhu K, Li W, Xu Z, Dang C, Li K. Itraconazole inhibits the proliferation of gastric cancer cells in vitro and improves patient survival. Oncol Lett 2018; 16:3651-3657. [PMID: 30127974 PMCID: PMC6096255 DOI: 10.3892/ol.2018.9072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 02/28/2018] [Indexed: 12/17/2022] Open
Abstract
Itraconazole is a Food and Drug Administration-approved antifungal drug belonging to the azole family. Recent studies reported that itraconazole has potential anticancer activity. Whether combining itraconazole with other anticancer compounds such as 5-fluorouracil (5-FU), a potent drug used in the treatment of gastric cancer, is unknown and warrants further study. In the present study, SGC-7901 gastric cancer cells were chosen to assess the anticancer effects of itraconazole in combination with 5-FU. Cell proliferation was assessed by a Cell Counting Kit-8 assay, and apoptosis was assessed by Annexin V/propidium iodide (PI) staining and flow cytometry. Cell cycle distribution was determined by PI staining and flow cytometer. Single-cell gel electrophoresis was used to estimate DNA damage. Medical records of patients with gastric cancer were retrospectively reviewed, and the patients treated with itraconazole were selected for the present study. Itraconazole treatment inhibited the proliferation and altered cell cycle in SGC-7901 cells while promoting early apoptosis and DNA damage. These effects were promoted in cells treated with both itraconazole and 5-FU. Combination itraconazole and 5-FU treatment showed a synergetic anticancer effect in SGC-7901 cells. in vivo, itraconazole was able to improve the outcome of 5-FU-based chemotherapy. Itraconazole alone and in combination with 5-FU was able to inhibit the growth of gastric cancer in vitro, and it was able to prolong the survival of patients with gastric cancer.
Collapse
Affiliation(s)
- Ke Lan
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Ron Yan
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Kun Zhu
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Wenhan Li
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Zisen Xu
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Chengxue Dang
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Kang Li
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
602
|
Peeters M, Cervantes A, Moreno Vera S, Taieb J. Trifluridine/tipiracil: an emerging strategy for the management of gastrointestinal cancers. Future Oncol 2018; 14:1629-1645. [DOI: 10.2217/fon-2018-0147] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Fluoropyrimidines are currently the backbone of treatment for gastrointestinal (GI) cancers but development of resistance to these agents remains a major problem. Trifluridine/tipiracil is an oral chemotherapeutic agent recently approved for third-line treatment of chemorefractory metastatic colorectal cancer. This article reviews the clinical value of trifluridine/tipiracil as a monotherapy, including recent trials in GI cancers, and the potential benefit of combining it with other agents in patients with GI cancers, including the preclinical rationale for combination therapy and recently completed and ongoing clinical trials. Data gathered so far suggest that trifluridine/tipiracil has the potential to form the chemotherapeutic backbone in the continuum of care for GI cancers in the future.
Collapse
Affiliation(s)
| | - Andrés Cervantes
- CIBERONC, Department of Medical Oncology, Biomedical Research institute INCLIVA, University of Valencia, Blasco Ibáñez 17, 46010 Valencia, Spain
| | | | - Julien Taieb
- Sorbonne Paris Cité, Hôpital Européen Georges Pompidou (HEGP), Paris – Descartes University, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France
| |
Collapse
|
603
|
Mizrak Kaya D, Nogueras-Gonzáles GM, Harada K, Amlashi FG, Thomas I, Rogers JE, Bhutani MS, Lee JH, Weston B, Minsky BD, Estrella JS, Blum Murphy MA, Matamoros A, Devine CE, Das P, Badgwell BD, Ajani JA. Potentially curable gastric adenocarcinoma treated without surgery. Eur J Cancer 2018; 98:23-29. [DOI: 10.1016/j.ejca.2018.04.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/03/2018] [Accepted: 04/22/2018] [Indexed: 11/30/2022]
|
604
|
Cost-effectiveness analysis of capecitabine monotherapy versus capecitabine plus oxaliplatin in elderly patients with advanced gastric cancer. PLoS One 2018; 13:e0199553. [PMID: 29953476 PMCID: PMC6023138 DOI: 10.1371/journal.pone.0199553] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 06/08/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND There is no single standard chemotherapy regimen for elderly patients with advanced gastric cancer (AGC). A phase III trial has confirmed that both capecitabine monotherapy and capecitabine plus oxaliplatin are well tolerated for elderly patients with AGC, but their economic influence in China is unknown. OBJECTIVE The purpose of this cost-effectiveness analysis was to estimate the effects of capecitabine monotherapy and capecitabine plus oxaliplatin in elderly patients with AGC on health and economic outcomes in China. METHODS We created a Markov model based on data from a Korean clinical phase III trial to analyze the cost-effectiveness of the treatment of elderly patients in the capecitabine monotherapy (X) group and capecitabine plus oxaliplatin (XELOX) group. The costs were obtained from published reports and the local health system. The utilities were assumed on the basis of the published literature. Costs, quality-adjusted life years (QALYs), and incremental cost-effectiveness ratios (ICER) were estimated. One-way and probabilistic sensitivity analyses (Monte Carlo simulations) were performed. RESULTS In the cost-effectiveness analysis, X had a lower total cost ($45,731.68) and cost-effectiveness ratio ($65,918.93/QALY). The one-way sensitivity analysis suggested that the most influential parameter was the risk of requiring second-line chemotherapy in XELOX group. The probabilistic sensitivity analysis predicted that the X regimen was cost-effective 100% of the time, given a willingness-to-pay threshold of $26,598. CONCLUSIONS Our findings show that the XELOX regimen is less cost-effective compared to the X regimen for elderly patients with AGC in China from a Chinese healthcare perspective.
Collapse
|
605
|
Uguen A, Troncone G. A review on the Idylla platform: towards the assessment of actionable genomic alterations in one day. J Clin Pathol 2018; 71:757-762. [DOI: 10.1136/jclinpath-2018-205189] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 12/27/2022]
|
606
|
Zhong Q, Chen QY, Li P, Xie JW, Wang JB, Lin JX, Lu J, Cao LL, Lin M, Tu RH, Zheng CH, Huang CM. Prediction of Conditional Probability of Survival After Surgery for Gastric Cancer: A Study Based on Eastern and Western Large Data Sets. Surgery 2018; 163:1307-1316. [PMID: 29685636 DOI: 10.1016/j.surg.2018.02.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 01/27/2018] [Accepted: 02/06/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND The dynamic prognosis of patients who have undergone curative surgery for gastric cancer has yet to be reported. Our objective was to devise an accurate tool for predicting the conditional probability of survival for these patients. METHODS We analyzed 11,551 gastric cancer patients from the Surveillance, Epidemiology, and End Results database. Two-thirds of the patients were selected randomly for the development set and one-third for the validation set. Two nomograms were constructed to predict the conditional probability of overall survival and the conditional probability of disease-specific survival, using conditional survival methods. We then applied these nomograms to the 4,001 patients in the database from Fujian Medical University Union Hospital, Fuzhou, China, one of the most active Chinese institutes. RESULTS The 5-year conditional probability of overall survival of the patients was 41.6% immediately after resection and increased to 52.8%, 68.2%, and 80.4% at 1, 2, and 3 years after gastrectomy. The 5-year conditional probability of disease-specific survival "increased" from 48.9% at the time of gastrectomy to 59.8%, 74.7%, and 85.5% for patients surviving 1, 2, and 3 years, respectively. Sex; race; age; depth of tumor invasion; lymph node metastasis; and tumor size, site, and grade were associated with overall survival and disease-specific survival (P <.05). Within the Surveillance, Epidemiology, and End Results validation set, the accuracy of the conditional probability of overall survival nomogram was 0.77, 0.81, 0.82, and 0.82 at 1, 3, 5, and 10 years after gastrectomy, respectively. Within the other validation set from the Fujian Medical University Union Hospital (n = 4,001), the accuracy of the conditional probability of overall survival nomogram was 0.76, 0.79, 0.77, and 0.77 at 1, 3, 5, and 10 years, respectively. The accuracy of the conditional probability of disease-specific survival model was also favorable. The calibration curve demonstrated good agreement between the predicted and observed survival rates. CONCLUSION Based on the large Eastern and Western data sets, we developed and validated the first conditional nomogram for prediction of conditional probability of survival for patients with gastric cancer to allow consideration of the duration of survivorship.
Collapse
Affiliation(s)
- Qing Zhong
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
607
|
Ikoma N, Hofstetter WL, Estrella JS, Das P, Minsky BD, Fournier KF, Mansfield PF, Ajani JA, Badgwell BD. The ypT category does not impact overall survival in node negative gastric cancer. J Surg Oncol 2018; 117:1721-1728. [PMID: 29949666 PMCID: PMC7703861 DOI: 10.1002/jso.25081] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 03/30/2018] [Indexed: 01/26/2023]
Abstract
BACKGROUND Although the eighth-edition American Joint Committee on Cancer (AJCC) gastric cancer staging system created ypTNM staging for patients who underwent preoperative therapy, the ideal ypTNM grouping is unknown. We sought to investigate risk factors for OS in ypT0-3N0M0 gastric cancer. METHODS From an institutional database of The University of Texas MD Anderson Cancer Center and the National Cancer Database (NCDB), we identified patients with ypT0-3N0M0 gastric adenocarcinoma who underwent R0 gastrectomy after chemotherapy or chemoradiation during 1995-2015 (MD Anderson) or 2006-2014 (NCDB). RESULTS The study included 175 MD Anderson and 3200 NCDB patients. By multivariable analysis, ypT category was not associated with OS (hazard ratio [HR] for ypT3 vs ypT1: MD Anderson, 0.83 [95% CI, 0.36-1.92], P = 0.669; NCDB, 0.96 [95% CI, 0.85-1.08], P = 0.472). cN-positive disease was not associated with OS in the MD Anderson cohort (HR, 0.96 [95% CI, 0.55-1.67]; P = 0.873) but was weakly associated with shorter OS in the NCDB cohort (HR, 1.11 [95% CI, 1.01-1.21]; P = 0.031). CONCLUSIONS The ypT category does not impact OS in ypT0-3N0M0 gastric cancer. The impact of cN status on OS appeared limited. These findings should be considered in future systems of post-neoadjuvant pathologic staging.
Collapse
Affiliation(s)
- Naruhiko Ikoma
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wayne L. Hofstetter
- Department Cardiac and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeannelyn S. Estrella
- Department Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Prajnan Das
- Department Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bruce D. Minsky
- Department Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Keith F. Fournier
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paul F. Mansfield
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jaffer A. Ajani
- Department Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Brian D. Badgwell
- Departments of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
608
|
Zhang CD, Yamashita H, Zhang S, Seto Y. Reevaluation of laparoscopic versus open distal gastrectomy for early gastric cancer in Asia: A meta-analysis of randomized controlled trials. Int J Surg 2018; 56:31-43. [PMID: 29860125 DOI: 10.1016/j.ijsu.2018.05.733] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 05/20/2018] [Accepted: 05/27/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND The benefits and risks of laparoscopic distal gastrectomy (LADG) are not yet sufficiently clear for acceptance as a standard treatment of early gastric cancer. Previous meta-analyses were not powered to reach definitive conclusions. MATERIALS AND METHODS Randomized controlled trials comparing LADG with open distal gastrectomy (ODG) for early gastric cancer in Asia and published between January 1994 and January 2018 were retrieved from PubMed, Embase, the Cochrane Library, and Google Scholar. Patient characteristics, oncological safety and efficacy, and surgical safety were evaluated following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) and Grading of Recommendations Assessment, Development and Evaluation guidelines (GRADE) guidelines. Trial Sequential Analysis (TSA) reduced random error and reinforced the reliability and strength of evidence. RESULTS Eight trials including 2666 participants were selected. LADG benefits were an 11.6 cm shorter incision (95% CI: -13.31 to -9.88 cm; P < 0.0001), 103.81 ml less blood loss (95% CI: -133.68 to -73.94; P < 0.0001), 1.73 times less analgesic use (95% CI: -2.21 to -1.24; P < 0.0001), 0.51 days shorter time to first flatus (95% CI: -0.88 to -0.15 days; P = 0.006), lower risk of wound dehiscence (RR = 0.24, 95% CI: 0.08-0.78; P = 0.02), lower risk of surgical adverse events (RR = 0.69, 95% CI: 0.53-0.91; P = 0.008), and lower risk of respiratory complications (RR = 0.40; 95% CI: 0.20-0.79; P = 0.009) than ODG. LADG had 2.22 fewer resected lymph nodes (95% CI: -4.33 to -0.12; P = 0.04) and 76.61 min longer procedures (76.61 min, 95% CI: 57.74-95.47 min; P < 0.0001). CONCLUSIONS In Asian patients, LADG had similar mortality and oncological safety, better surgical safety, less operative morbidity, less trauma, and faster recovery than ODG. It has a high role to play in node-negative cases due to better short-term outcomes but less nodal harvest. It is a recommended alternative treatment for experienced surgeons in high-volume centers.
Collapse
Affiliation(s)
- Chun-Dong Zhang
- Department of Gastrointestinal Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan; Department of Gastrointestinal Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hiroharu Yamashita
- Department of Gastrointestinal Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Shun Zhang
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.
| |
Collapse
|
609
|
Feng J, Qin S. The synergistic effects of Apatinib combined with cytotoxic chemotherapeutic agents on gastric cancer cells and in a fluorescence imaging gastric cancer xenograft model. Onco Targets Ther 2018; 11:3047-3057. [PMID: 29872316 PMCID: PMC5975614 DOI: 10.2147/ott.s159935] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Introduction Methylsulfonic apatinib (hereinafter referred to as Apatinib) is a small-molecule angiogenesis inhibitor highly and selectively targeted to vascular endothelial growth factor receptor-2. At present, a series of basic and clinical studies have confirmed that Apatinib mono-therapy can inhibit the growth of different carcinomas. Our experiment aimed to determine whether there is a synergistic effect between the combination of the traditional cytotoxic chemotherapy drugs paclitaxel (TAX), oxaliplatin (L-OHP), 5-fluorouracil (5-FU), and Apatinib. Materials and methods We evaluated the combined effect using cytological experiments and a fluorescence imaging xenograft model. In vitro, the inhibition of cell proliferation increased notably when Apatinib was combined with TAX, L-OHP, and 5-FU. Then, for the mechanistic research, we selected the optimal dose of drugs that also had a synergistic effect. Apatinib combined with the aforementioned drugs, especially the combination of Apatinib and 5-FU, decreased the invasion and migration ability of the cells and increased the apoptosis ratio; expression of the anti-apoptotic protein Bcl-2 significantly decreased, and expression of the pro-apoptotic protein Bax increased. In vivo, when Apatinib was combined with TAX, L-OHP, and 5-FU, the volume of the xenograft model was significantly inhibited, the strength of the green fluorescence was weakened and the microvessel density decreased. Results The combination of Apatinib with TAX and 5-FU was synergistic (coefficient of drug interaction <1); the combination effect of Apatinib and L-OHP was only additive, with a shorter associated survival time. Conclusion The combination of Apatinib and classical chemotherapy drugs may be an optimal choice for gastric cancer treatment.
Collapse
Affiliation(s)
- Jiuhuan Feng
- Institute of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shukui Qin
- Oncology Center of PLA, 81 Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
610
|
Chen B, Wang J, Chen Y, Gu X, Feng X. The MDM2 rs937283 A > G variant significantly increases the risk of lung and gastric cancer in Chinese population. Int J Clin Oncol 2018; 23:867-876. [PMID: 29777315 DOI: 10.1007/s10147-018-1295-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/14/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Currently, the MDM2 promoter rs937283 A > G variant that is able to alter MDM2 gene expression has been widely studied to explore the association of MDM2 with cancer risk. In this report, we investigate the association of MDM2 rs937283 A > G variant with risk of lung cancer (LC) and gastric cancer (GC) in a Chinese population of Hubei province, which was followed by a meta-analysis. METHODS The genotyping of rs937283 was performed by polymerase chain reaction-restriction fragment length polymorphism and confirmed by sequencing. RESULTS The results of the present study showed that rs937283 was significantly associated with the risk of LC, and the factors of age, gender, smoking status and drinking status would affect such association. However, rs937283 was only associated with the risk of GC in male, smoking and drinking subgroups. The following meta-analysis demonstrated that rs937283 was associated with the overall cancer risk particularly in Chinese population, which reinforced our present finding. Moreover, the meta-analysis according to cancer types revealed that rs937283 was associated with retinoblastoma risk, but not squamous cell carcinoma risk. CONCLUSION Collectively, the MDM2 rs937283 A > G variant may be a valuable risk factor or diagnostic biomarker for Chinese cancer patients.
Collapse
Affiliation(s)
- Bifeng Chen
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China.
| | - Jieling Wang
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Yucan Chen
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Xiuli Gu
- Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Wuhan Tongji Reproductive Medicine Hospital, Wuhan, China
| | - Xianhong Feng
- Clinical Laboratory, Wuhan Xinzhou District People's Hospital, Wuhan, China.
| |
Collapse
|
611
|
Ishigami H, Fujiwara Y, Fukushima R, Nashimoto A, Yabusaki H, Imano M, Imamoto H, Kodera Y, Uenosono Y, Amagai K, Kadowaki S, Miwa H, Yamaguchi H, Yamaguchi T, Miyaji T, Kitayama J. Phase III Trial Comparing Intraperitoneal and Intravenous Paclitaxel Plus S-1 Versus Cisplatin Plus S-1 in Patients With Gastric Cancer With Peritoneal Metastasis: PHOENIX-GC Trial. J Clin Oncol 2018; 36:1922-1929. [PMID: 29746229 DOI: 10.1200/jco.2018.77.8613] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Purpose Intraperitoneal paclitaxel plus systemic chemotherapy demonstrated promising clinical effects in patients with gastric cancer with peritoneal metastasis. We aimed to verify its superiority over standard systemic chemotherapy in overall survival. Patients and Methods This randomized phase III trial enrolled patients with gastric cancer with peritoneal metastasis who had received no or short-term (< 2 months) chemotherapy. Patients were randomly assigned at a two-to-one ratio to receive intraperitoneal and intravenous paclitaxel plus S-1 (IP; intraperitoneal paclitaxel 20 mg/m2 and intravenous paclitaxel 50 mg/m2 on days 1 and 8 plus S-1 80 mg/m2 per day on days 1 to 14 for a 3-week cycle) or S-1 plus cisplatin (SP; S-1 80 mg/m2 per day on days 1 to 21 plus cisplatin 60 mg/m2 on day 8 for a 5-week cycle), stratified by center, previous chemotherapy, and extent of peritoneal metastasis. The primary end point was overall survival. Secondary end points were response rate, 3-year overall survival rate, and safety. Results We enrolled 183 patients and performed efficacy analyses in 164 eligible patients. Baseline characteristics were balanced between the arms, except that patients in the IP arm had significantly more ascites. The median survival times for the IP and SP arms were 17.7 and 15.2 months, respectively (hazard ratio, 0.72; 95% CI, 0.49 to 1.04; stratified log-rank P = .080). In the sensitivity analysis adjusted for baseline ascites, the hazard ratio was 0.59 (95% CI, 0.39 to 0.87; P = .008). The 3-year overall survival rate was 21.9% (95% CI, 14.9% to 29.9%) in the IP arm and 6.0% (95% CI, 1.6% to 14.9%) in the SP arm. Both regimens were well tolerated. Conclusion This trial failed to show statistical superiority of intraperitoneal paclitaxel plus systemic chemotherapy. However, the exploratory analyses suggested possible clinical benefits of intraperitoneal paclitaxel for gastric cancer.
Collapse
Affiliation(s)
- Hironori Ishigami
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Yoshiyuki Fujiwara
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Ryoji Fukushima
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Atsushi Nashimoto
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Hiroshi Yabusaki
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Motohiro Imano
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Haruhiko Imamoto
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Yasuhiro Kodera
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Yoshikazu Uenosono
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Kenji Amagai
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Shigenori Kadowaki
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Hiroto Miwa
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Hironori Yamaguchi
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Takuhiro Yamaguchi
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Tempei Miyaji
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| | - Joji Kitayama
- Hironori Ishigami and Tempei Miyaji, The University of Tokyo; Ryoji Fukushima, Teikyo University, Tokyo; Yoshiyuki Fujiwara, Osaka International Cancer Institute; Motohiro Imano and Haruhiko Imamoto, Kindai University, Osaka; Atsushi Nashimoto, Nanbugo General Hospital; Hiroshi Yabusaki, Niigata Cancer Center Hospital, Niigata; Yasuhiro Kodera, Nagoya University; Shigenori Kadowaki, Aichi Cancer Center Hospital, Aichi; Yoshikazu Uenosono, Kagoshima University, Kagoshima; Kenji Amagai, Ibaraki Prefectural Central Hospital, Ibaraki; Hiroto Miwa, Hyogo College of Medicine, Hyogo; Hironori Yamaguchi and Joji Kitayama, Jichi Medical University, Tochigi; and Takuhiro Yamaguchi, Tohoku University, Miyagi, Japan
| |
Collapse
|
612
|
Abstract
A postoperative complications rate of nearly 50% has compelled oesophago-gastric practice to adopt minimally invasive techniques such as robotic surgery
Collapse
Affiliation(s)
- Y A Qureshi
- Department of Oesophago-Gastric Surgery, University College London Hospital , London
| | - B Mohammadi
- Department of Oesophago-Gastric Surgery, University College London Hospital , London
| |
Collapse
|
613
|
Gil-Delgado MA, Lucidarme O, Bachet JB, Mahi N, Khayat D. Long-Term Survival in Gastroesophageal Junction Adenocarcinoma: Ramucirumab. Case Rep Gastroenterol 2018; 12:532-539. [PMID: 30283289 PMCID: PMC6167727 DOI: 10.1159/000490906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 06/05/2018] [Indexed: 01/22/2023] Open
Abstract
We report a case of long-term survival with complete response of liver metastases within RAINBOW, a randomized, controlled trial of ramucirumab 8 mg/kg intravenously (days 1, 15) versus placebo, both plus paclitaxel 80 mg/m2 intravenously (days 1, 8, 15), every 4 weeks in patients with previously treated advanced gastroesophageal junction adenocarcinoma. A 64-year-old man with gastroesophageal junction adenocarcinoma and liver metastases received first-line folinic acid, 5-fluorouracil plus oxaliplatin (FOLFOX) following jejunostomy. On liver progression, he enrolled in RAINBOW (April 2012), receiving ramucirumab. In November 2013, positron emission tomography scan was consistent with complete metabolic response, confirmed by a follow-up scan in March 2016.
Collapse
Affiliation(s)
| | - Olivier Lucidarme
- Radiology Department, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Sorbonne University, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale (LIB), Paris, France
| | - Jean Baptiste Bachet
- Sorbonne University, UPMC University, Gastrointestinal Diseases Service, Groupe Hospitalier Pitié Salpêtrière, Paris, France
| | - Naima Mahi
- Eli Lilly France, Neuilly-sur-Seine, France
| | - David Khayat
- Medical Oncology Service, Groupe Hospitalier Pitié Salpêtrière, Paris, France
| |
Collapse
|
614
|
Distal versus total gastrectomy for middle and lower-third gastric cancer: A systematic review and meta-analysis. Int J Surg 2018; 53:163-170. [DOI: 10.1016/j.ijsu.2018.03.047] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 02/07/2018] [Accepted: 03/23/2018] [Indexed: 02/06/2023]
|
615
|
Feng X, Wang J, Gu X, Zhang J, Li X, Tao Z, Chen J, Chen B. Association of DNMT3B -283T>C polymorphism with risk of lung and gastric cancer: a case-control study and a meta-analysis. Int J Biol Markers 2018; 33:195-200. [PMID: 29027179 DOI: 10.5301/ijbm.5000306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE To investigate the association of DNMT3B -283T>C polymorphism with the risk of lung or gastric cancer, which was followed by a meta-analysis. METHODS The genotyping of -283T>C was performed by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) and was confirmed by sequencing. RESULTS The results of this case-control study showed that -283T>C was not associated with the risk of lung or gastric cancer, and further stratified analysis according to age, gender, smoking status, and alcohol status confirmed the present finding. However, data from a meta-analysis in the Asian population revealed a significant association between -283T>C and lung cancer risk in the allelic model (C vs. T: odds ratio [OR] = 1.28, 95% confidence interval [CI], 1.06-1.55, p = 0.01) and two genetic models (CC vs. TC: OR = 1.29, 95% CI, 1.04-1.59, p = 0.02; CC vs. TC + TT: OR = 1.30, 95% CI, 1.06-1.60, p = 0.01). CONCLUSIONS These results provided evidence that the DNMT3B -283T>C polymorphism might significantly contribute to the lung cancer risk in the Asian population, but not the gastric cancer risk in the Chinese population.
Collapse
Affiliation(s)
- Xianhong Feng
- 1 Clinical Laboratory, Wuhan Xinzhou District People's Hospital, Wuhan - PR China
| | - Jingdong Wang
- 2 Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan - PR China
| | - Xiuli Gu
- 3 Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan - PR China
| | - Jingli Zhang
- 2 Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan - PR China
| | - Xiaolin Li
- 1 Clinical Laboratory, Wuhan Xinzhou District People's Hospital, Wuhan - PR China
| | - Zhi Tao
- 1 Clinical Laboratory, Wuhan Xinzhou District People's Hospital, Wuhan - PR China
| | - Jiebing Chen
- 1 Clinical Laboratory, Wuhan Xinzhou District People's Hospital, Wuhan - PR China
| | - Bifeng Chen
- 2 Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan - PR China
| |
Collapse
|
616
|
Jaruvongvanich V, Sempokuya T, Wijarnpreecha K, Ungprasert P. Continued versus interrupted aspirin use and bleeding risk after endoscopic submucosal dissection of gastric neoplasms: a meta-analysis. Ann Gastroenterol 2018; 31:344-349. [PMID: 29720860 PMCID: PMC5924857 DOI: 10.20524/aog.2018.0251] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/22/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Balancing the risk of bleeding and thromboembolic events for patients who use aspirin and need to undergo endoscopic submucosal dissection (ESD) for gastric neoplasms is a delicate process. The current guidelines from different associations provide inconsistent recommendations. METHODS MEDLINE and EMBASE databases were searched through August 2017 for studies that compared the risk of post-ESD bleeding in patients who continued aspirin vs. those who discontinued aspirin preoperatively. Pooled odds ratios (OR) and 95% confidence intervals (CI) were calculated using a random-effect model, generic inverse variance method. The between-study heterogeneity was quantified using the Q statistic and I2 . RESULTS A total of five studies that included 700 patients were identified. Our meta-analysis could not demonstrate a significantly increased risk of post-ESD bleeding among the aspirin-continued group compared to the aspirin-interrupted group, the pooled OR being 1.81 (95%CI 0.85-3.83). The statistical heterogeneity was insignificant, with an I2 of 25%. Nine thrombotic events occurred in the aspirin-interrupted group whereas none occurred in the aspirin-continued group. CONCLUSIONS This meta-analysis could not demonstrate that continuation of aspirin significantly increases the risk of post-ESD bleeding. However, the analysis was restricted by the small sample size and the observational nature of the primary studies. Randomized controlled trials are still needed to clarify this risk.
Collapse
Affiliation(s)
- Veeravich Jaruvongvanich
- Department of Internal Medicine, University of Hawaii, Honolulu, HI, USA (Veeravich Jaruvongvanich, Tomoki Sempokuya)
- Department of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand (Veeravich Jaruvongvanich)
| | - Tomoki Sempokuya
- Department of Internal Medicine, University of Hawaii, Honolulu, HI, USA (Veeravich Jaruvongvanich, Tomoki Sempokuya)
| | - Karn Wijarnpreecha
- Department of Internal Medicine, Bassett Medical Center and Columbia University College of Physicians and Surgeons, Cooperstown, NY, USA (Karn Wijarnpreecha)
| | - Patompong Ungprasert
- Department of Division of Clinical Epidemiology, Siriraj Medical Research Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand (Patompong Ungprasert)
| |
Collapse
|
617
|
Abstract
Gastric adenocarcinoma (GAC) is estimated as the fifteenth most common cancer in the USA. Incidence rate has been gradually decreasing, but prognosis remains dismal. For patients with locally advanced GAC (stage > T1B and < T4B), multimodality therapies, such as surgery, chemotherapy, and radiation therapy, are needed. Perioperative chemotherapy or postoperative chemoradiation/chemotherapy is recommended. For metastatic GAC patients, combination of two cytotoxics (platinum compound and fluoropyrimidine) has become a common place in the USA, and when HER2 is positive, trastuzumab is added. When GAC progresses after the first line therapy, additional biomarkers (microsatellite instability and programmed death ligand 1) should be tested so that checkpoint inhibitors can be used. Overall, the options for advanced GAC patients are limited and more research is needed.
Collapse
Affiliation(s)
- Kazuto Harada
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA.,Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto 860-8556, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto 860-8556, Japan
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
618
|
Shah MA, Starodub A, Sharma S, Berlin J, Patel M, Wainberg ZA, Chaves J, Gordon M, Windsor K, Brachmann CB, Huang X, Vosganian G, Maltzman JD, Smith V, Silverman JA, Lenz HJ, Bendell JC. Andecaliximab/GS-5745 Alone and Combined with mFOLFOX6 in Advanced Gastric and Gastroesophageal Junction Adenocarcinoma: Results from a Phase I Study. Clin Cancer Res 2018; 24:3829-3837. [PMID: 29691300 DOI: 10.1158/1078-0432.ccr-17-2469] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 03/06/2018] [Accepted: 04/19/2018] [Indexed: 12/21/2022]
Abstract
Purpose: Matrix metalloproteinase-9 (MMP9) is implicated in protumorigenic processes. Andecaliximab (GS-5745, a monoclonal antibody targeting MMP9) was evaluated as monotherapy and in combination with mFOLFOX6.Patients and Methods: Three dosages of andecaliximab monotherapy [200, 600, and 1800 mg i.v. every 2 weeks (q2w)] were investigated in patients with advanced solid tumors (n = 13 in a 3+3 design). After determining a recommended dose, patients with advanced HER2-negative gastric/gastroesophageal junction (GEJ) adenocarcinoma (n = 40) received 800 mg andecaliximab + mFOLFOX6 q2w. Pharmacokinetics, pharmacodynamics, safety, and efficacy were assessed.Results: Andecaliximab monotherapy demonstrated no dose-limiting toxicity (DLT) in any cohort, displaying target-mediated drug disposition at the lowest dose (200 mg) and linear pharmacokinetics at higher doses. Based on target engagement, recommended doses for further study are 800 mg q2w or 1,200 mg q3w. Maximal andecaliximab target binding, defined as undetectable andecaliximab-free MMP9 in plasma, was observed in the gastric/GEJ adenocarcinoma cohort. We observed no unusual toxicity, although there were four deaths on study not attributed to andecaliximab treatment. In first-line patients (n = 36), median progression-free survival (PFS) was 9.9 months [95% confidence interval (CI), 5-13.9 months], and the overall response rate (ORR) was 50%. Among all patients (n = 40), median PFS was 7.8 (90% CI, 5.5-13.9) months, and ORR was 48%, with a median duration of response of 8.4 months.Conclusions: Andecaliximab monotherapy achieved target engagement without DLT. Andecaliximab + mFOLFOX6 showed encouraging clinical activity without additional toxicity in patients with HER2-negative gastric/GEJ adenocarcinoma. A phase III study evaluating mFOLFOX6 ± andecaliximab in this setting is ongoing. Clin Cancer Res; 24(16); 3829-37. ©2018 AACR.
Collapse
Affiliation(s)
- Manish A Shah
- Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York.
| | - Alexander Starodub
- Parkview Comprehensive Cancer Institute/Parkview Health, Fort Wayne, Indiana
| | - Sunil Sharma
- University of Utah Huntsman Cancer Institute, Salt Lake City, Utah
| | - Jordan Berlin
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Manish Patel
- Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, Florida
| | - Zev A Wainberg
- Division of Hematology Oncology, Department of Medicine, UCLA School of Medicine, Los Angeles, California
| | - Jorge Chaves
- Northwest Medical Specialties PLLC, Tacoma, Washington
| | | | | | | | - Xi Huang
- Gilead Sciences, Inc., Foster City, California
| | | | | | | | | | | | - Johanna C Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee
| |
Collapse
|
619
|
Capillary morphogenesis gene 2 maintains gastric cancer stem-like cell phenotype by activating a Wnt/β-catenin pathway. Oncogene 2018; 37:3953-3966. [PMID: 29662192 PMCID: PMC6053357 DOI: 10.1038/s41388-018-0226-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/29/2017] [Accepted: 02/23/2018] [Indexed: 02/07/2023]
Abstract
A growing body of evidence shows that the development and progression of gastric cancer (GC) is mainly associated to the presence of gastric cancer stem-like cells (GCSLCs). However, it is unclear how GCSLC population is maintained. This study aimed to explore the role of capillary morphogenesis gene 2 (CMG2) in GCSLC maintenance and the relevance to GC progression. We found that CMG2 was highly expressed in GC tissues and the expression levels were associated with the invasion depth and lymph node metastasis of GC, and inversely correlated with the survival of GC patients. Sorted CMG2High GC cells preferentially clustered in CD44High stem-like cell population, which expressed high levels of stemness-related genes with increased capabilities of self-renewal and tumorigenicity. Depletion of CMG2 gene resulted in reduction of GCSLC population with attenuated stemness and decrease of invasive and metastatic capabilities with subdued epithelial–mesenchymal transition phenotype in GC cells. Mechanistically, CMG2 interacted with LRP6 in GCSLCs to activate a Wnt/β-catenin pathway. Thus, our results demonstrate that CMG2 promotes GC progression by maintaining GCSLCs and can serve as a new prognostic indicator and a target for human GC therapy.
Collapse
|
620
|
Cisło M, Filip AA, Arnold Offerhaus GJ, Ciseł B, Rawicz-Pruszyński K, Skierucha M, Polkowski WP. Distinct molecular subtypes of gastric cancer: from Laurén to molecular pathology. Oncotarget 2018; 9:19427-19442. [PMID: 29721214 PMCID: PMC5922408 DOI: 10.18632/oncotarget.24827] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/27/2018] [Indexed: 12/12/2022] Open
Abstract
In Western countries the majority of gastric cancers (GC) are usually diagnosed in advanced stages reporting a 5-year survival rate of only 26%. The Laurén classification of GC was most widely used in clinical practice since it reflects GC morphology, epidemiology, tumor biology, clinical management and outcome. Despite the initial promise of individualizing antitumor treatment, the management of GC still remains relatively broad and general. Apart from clinical staging, molecular profiling enables targeting of the identified underlying alterations, rather than histology. In contrast to breast carcinoma, molecular classification of GC does not yet imply treatment modality. Molecular classifications of GC and their therapeutic implications are therefore extensively studied. The current proposed molecular divisions of GC come from three different parts of the world where different standard treatment modalities for advanced GC are recommended. Wider use of GC molecular subtyping may solve problems, such as susceptibility to novel systemic therapy regimens or selection of patients for aggressive surgery and targeted adjuvant/conversion therapy. In any case, the rapid entry of novel molecular targeted therapies into routine oncology practice clearly underscores the urgent need for clinicians to be aware of these new possibilities.
Collapse
Affiliation(s)
- Magdalena Cisło
- Department of Surgical Oncology, Medical University of Lublin, Lublin, Poland
| | - Agata Anna Filip
- Department of Cancer Genetics and Cytogenetics Laboratory, Medical University of Lublin, Lublin, Poland
| | | | - Bogumiła Ciseł
- Department of Surgical Oncology, Medical University of Lublin, Lublin, Poland
| | | | - Małgorzata Skierucha
- Department of Surgical Oncology, Medical University of Lublin, Lublin, Poland
- Department of Human Anatomy, Medical University of Lublin, Lublin, Poland
| | | |
Collapse
|
621
|
Liu S, Shi H, Ji C, Zheng H, Pan X, Guan W, Chen L, Sun Y, Tang L, Guan Y, Li W, Ge Y, He J, Liu S, Zhou Z. Preoperative CT texture analysis of gastric cancer: correlations with postoperative TNM staging. Clin Radiol 2018; 73:756.e1-756.e9. [PMID: 29625746 DOI: 10.1016/j.crad.2018.03.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/08/2018] [Indexed: 02/07/2023]
Abstract
AIM To explore the role of computed tomography (CT) texture analysis in predicting pathologic stage of gastric cancers. MATERIALS AND METHODS Preoperative enhanced CT images of 153 patients (112 men, 41 women) with gastric cancers were reviewed retrospectively. Regions of interest (ROIs) were manually drawn along the margin of the lesion on the section where it appeared largest on the arterial and venous CT images, which yielded texture parameters, including mean, maximum frequency, mode, skewness, kurtosis, and entropy. Correlations between texture parameters and pathological stage were analysed with Spearman's correlation test. The diagnostic performance of CT texture parameters in differentiating different stages was evaluated using receiver operating characteristic (ROC) analysis. RESULTS Maximum frequency in the arterial phase and mean, maximum frequency, mode in the venous phase correlated positively with T stage, N stage, and overall stage (all p<0.05) of gastric cancer. Entropy in the venous phase also correlated positively with N stage (p=0.009) and overall stage (p=0.032). Skewness in the arterial phase had the highest area under the ROC curve (AUC) of 0.822 in identifying early from advanced gastric cancers. Multivariate analysis identified four parameters, including maximum frequency, skewness, entropy in the venous phase, and differentiation degree from biopsy, for predicting lymph node metastasis of gastric cancer. The multivariate model could distinguish gastric cancers with and without lymph node metastasis with an AUC of 0.892. CONCLUSION Multiple CT texture parameters, especially those in the venous phase, correlated well with pathological stage and hold great potential in predicting lymph node metastasis of gastric cancers.
Collapse
Affiliation(s)
- S Liu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - H Shi
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - C Ji
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - H Zheng
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - X Pan
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - W Guan
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - L Chen
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Y Sun
- Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - L Tang
- Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Y Guan
- School of Electronic Science and Engineering, Nanjing University, Nanjing, 210046, China
| | - W Li
- School of Electronic Science and Engineering, Nanjing University, Nanjing, 210046, China
| | - Y Ge
- School of Electronic Science and Engineering, Nanjing University, Nanjing, 210046, China
| | - J He
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - S Liu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - Z Zhou
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|
622
|
Luo H, Peng L, Wang N, Zhang J, Zheng X, Sun Y, Fan C, Ge H. Early brain metastasis of advanced gastric cancer with a pathological complete response to neoadjuvant chemotherapy followed by surgery: A case report and literature review. Cancer Biol Ther 2018; 19:875-878. [PMID: 29580147 DOI: 10.1080/15384047.2018.1456600] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Advanced gastric cancer with a pathological complete response to neoadjuvant chemotherapy and surgery followed by early brain metastasis is rare. A 52-y-old male patient who was diagnosed with advanced gastric cancer (cT4N2M0, stage ШB). Radiological examinations after three cycles of preoperative chemotherapy with a modified FOLFOX6 (mFOLFOX6) regimen showed a partial response (PR) had been achieved. The patient underwent curative surgery consisting of proximal gastrectomy, and D2 lymph node dissection. The lack of abnormal gastric cancer cells in the primary lesion or lymph nodes confirmed a pathological complete response (pCR). Postoperative chemotherapy with oral S-1 was administrated. However, during the second cycles of postoperative chemotherapy, the patient experienced headaches, projectile vomiting and convulsion. Upon further examination, a tumor representing metastasis to the brain was recognized by cranial enhanced magnetic resonance imaging (MRI) examination and cytopathology of cerebrospinal fluid. In addition to documenting the case report, we reviewed the literature associated to features of metastatic brain malignancies that form from gastric cancer. In short, advanced gastric cancer patents achieved pCR after preoperative chemotherapy typically have good prognosis; however, great attention should be paid on detecting metastatic events.
Collapse
Affiliation(s)
- Hui Luo
- a Division of Graduate, The second clinical medical school and the second affiliated hospital of Zhengzhou University , Zhengzhou , China.,c Department of Radiation Oncology , The affiliated cancer hospital of Zhengzhou University , Zhengzhou , China
| | - Liangqun Peng
- b Department of General Surgery , The affiliated cancer hospital of Zhengzhou University , Zhengzhou , China
| | - Nan Wang
- c Department of Radiation Oncology , The affiliated cancer hospital of Zhengzhou University , Zhengzhou , China
| | - Jiangong Zhang
- c Department of Radiation Oncology , The affiliated cancer hospital of Zhengzhou University , Zhengzhou , China
| | - Xiaoli Zheng
- c Department of Radiation Oncology , The affiliated cancer hospital of Zhengzhou University , Zhengzhou , China
| | - Yanan Sun
- c Department of Radiation Oncology , The affiliated cancer hospital of Zhengzhou University , Zhengzhou , China
| | - Chengcheng Fan
- c Department of Radiation Oncology , The affiliated cancer hospital of Zhengzhou University , Zhengzhou , China
| | - Hong Ge
- c Department of Radiation Oncology , The affiliated cancer hospital of Zhengzhou University , Zhengzhou , China
| |
Collapse
|
623
|
Impact of peri-operative blood transfusion on post-operative infections after radical gastrectomy for gastric cancer: a propensity score matching analysis focusing on the timing, amount of transfusion and role of leukocyte depletion. J Cancer Res Clin Oncol 2018; 144:1143-1154. [PMID: 29572591 PMCID: PMC5948291 DOI: 10.1007/s00432-018-2630-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/20/2018] [Indexed: 01/26/2023]
Abstract
Purpose Allogeneic blood transfusions (BTF) are sometimes inevitable during radical gastrectomy with lymphadenectomy for advanced gastric cancer. The aim of this retrospective study was to investigate the association between BTF and post-operative infections, focusing on the impact of timing, amount of transfusion and the role of leukocyte depletion. Methods The study cohort was 2064 patients who underwent gastrectomy for gastric cancer from November 2010 to August 2017. The association between BTF and post-operative infections was estimated by univariate and multivariate analyses after propensity score matching. Subgroup analysis was performed according to the timing and amount of transfusion, and leukocyte depletion or not. Results Out of a total 2064 patients, 426 (20.6%) received peri-operative BTF. After one-to-one matching, 361 pairs of patients were included for further analysis, of who 68 (9.4%) developed infections. Multivariate analysis identified that an operation time ≥ 240 min, combined multi-organ resection, BTF and BMI ≥ 25 kg/m2 were independent risk factors for post-operative infection. Patients given a high-volume (> 7.5 U), intra-operatively of leukocyte-non-depleted BTF had the highest risk of developing infections clarified by subgroup analysis. Conclusion Infection was the most common complication following gastrectomy for gastric cancer and BTF was identified as an independent risk factor by propensity score matching and multivariate analyses. The timing, amount of transfusion and leukocyte depletion had an impact on the incidence of infection. To decrease infection, BTF should be avoided where possible, particularly during operation, with a large amount and leukocyte-not-depleted blood.
Collapse
|
624
|
Choi IJ, Kook MC, Kim YI, Cho SJ, Lee JY, Kim CG, Park B, Nam BH. Helicobacter pylori Therapy for the Prevention of Metachronous Gastric Cancer. N Engl J Med 2018; 378:1085-1095. [PMID: 29562147 DOI: 10.1056/nejmoa1708423] [Citation(s) in RCA: 479] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Patients with early gastric cancers that are limited to gastric mucosa or submucosa usually have an advanced loss of mucosal glandular tissue (glandular atrophy) and are at high risk for subsequent (metachronous) development of new gastric cancer. The long-term effects of treatment to eradicate Helicobacter pylori on histologic improvement and the prevention of metachronous gastric cancer remain unclear. METHODS In this prospective, double-blind, placebo-controlled, randomized trial, we assigned 470 patients who had undergone endoscopic resection of early gastric cancer or high-grade adenoma to receive either H. pylori eradication therapy with antibiotics or placebo. Two primary outcomes were the incidence of metachronous gastric cancer detected on endoscopy performed at the 1-year follow-up or later and improvement from baseline in the grade of glandular atrophy in the gastric corpus lesser curvature at the 3-year follow-up. RESULTS A total of 396 patients were included in the modified intention-to-treat analysis population (194 in the treatment group and 202 in placebo group). During a median follow-up of 5.9 years, metachronous gastric cancer developed in 14 patients (7.2%) in the treatment group and in 27 patients (13.4%) in the placebo group (hazard ratio in the treatment group, 0.50; 95% confidence interval, 0.26 to 0.94; P=0.03). Among the 327 patients in the subgroup that underwent histologic analysis, improvement from baseline in the atrophy grade at the gastric corpus lesser curvature was observed in 48.4% of the patients in the treatment group and in 15.0% of those in the placebo group (P<0.001). There were no serious adverse events; mild adverse events were more common in the treatment group (42.0% vs. 10.2%, P<0.001). CONCLUSIONS Patients with early gastric cancer who received H. pylori treatment had lower rates of metachronous gastric cancer and more improvement from baseline in the grade of gastric corpus atrophy than patients who received placebo. (Funded by the National Cancer Center, South Korea; ClinicalTrials.gov number, NCT02407119 .).
Collapse
Affiliation(s)
- Il Ju Choi
- From the Center for Gastric Cancer (I.J.C., M.-C.K., Y.-I.K., S.-J.C., J.Y.L., C.G.K.) and the Biometrics Research Branch, Research Institute (B.P., B.-H.N.), National Cancer Center, Goyang, South Korea
| | - Myeong-Cherl Kook
- From the Center for Gastric Cancer (I.J.C., M.-C.K., Y.-I.K., S.-J.C., J.Y.L., C.G.K.) and the Biometrics Research Branch, Research Institute (B.P., B.-H.N.), National Cancer Center, Goyang, South Korea
| | - Young-Il Kim
- From the Center for Gastric Cancer (I.J.C., M.-C.K., Y.-I.K., S.-J.C., J.Y.L., C.G.K.) and the Biometrics Research Branch, Research Institute (B.P., B.-H.N.), National Cancer Center, Goyang, South Korea
| | - Soo-Jeong Cho
- From the Center for Gastric Cancer (I.J.C., M.-C.K., Y.-I.K., S.-J.C., J.Y.L., C.G.K.) and the Biometrics Research Branch, Research Institute (B.P., B.-H.N.), National Cancer Center, Goyang, South Korea
| | - Jong Yeul Lee
- From the Center for Gastric Cancer (I.J.C., M.-C.K., Y.-I.K., S.-J.C., J.Y.L., C.G.K.) and the Biometrics Research Branch, Research Institute (B.P., B.-H.N.), National Cancer Center, Goyang, South Korea
| | - Chan Gyoo Kim
- From the Center for Gastric Cancer (I.J.C., M.-C.K., Y.-I.K., S.-J.C., J.Y.L., C.G.K.) and the Biometrics Research Branch, Research Institute (B.P., B.-H.N.), National Cancer Center, Goyang, South Korea
| | - Boram Park
- From the Center for Gastric Cancer (I.J.C., M.-C.K., Y.-I.K., S.-J.C., J.Y.L., C.G.K.) and the Biometrics Research Branch, Research Institute (B.P., B.-H.N.), National Cancer Center, Goyang, South Korea
| | - Byung-Ho Nam
- From the Center for Gastric Cancer (I.J.C., M.-C.K., Y.-I.K., S.-J.C., J.Y.L., C.G.K.) and the Biometrics Research Branch, Research Institute (B.P., B.-H.N.), National Cancer Center, Goyang, South Korea
| |
Collapse
|
625
|
Ter Veer E, Creemers A, de Waal L, van Oijen MGH, van Laarhoven HWM. Comparing cytotoxic backbones for first-line trastuzumab-containing regimens in human epidermal growth factor receptor 2-positive advanced oesophagogastric cancer: A meta-analysis. Int J Cancer 2018; 143:438-448. [PMID: 29451302 DOI: 10.1002/ijc.31325] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 12/13/2017] [Accepted: 02/02/2018] [Indexed: 12/14/2022]
Abstract
According to the Trastuzumab for Gastric Cancer (ToGA) study, trastuzumab plus cisplatin and capecitabine/5-fluorouracil (5-FU) is standard first-line treatment for human epidermal growth factor receptor 2 (HER2)-positive advanced oesophagogastric cancer. We examined the relative efficacy and safety of alternative trastuzumab-based cytotoxic backbone regimens compared to the standard ToGA regimen using meta-analysis. We searched Medline, EMBASE, CENTRAL and ASCO and ESMO up to March 2017 for studies investigating alternative first-line trastuzumab-based regimens for HER2-positive oesophagogastric cancer, defined as high protein expression IHC3+ or IHC2+ and gene amplification by in situ hybridisation. We compared primary outcome overall survival (OS) of alternative trastuzumab-based regimens to the ToGA regimen. Hazard ratios (HRs) and 95% confidence intervals (95%CI) were calculated by extraction of the published Kaplan-Meier curves. Incidence counts and toxicity sample-sizes were extracted for adverse events and compared using single-arm proportion meta-analysis in R. Fifteen studies (N = 557 patients) were included. OS was significantly longer with regimen trastuzumab plus doublet oxaliplatin and capecitabine/5-FU (median OS = 20.7 months) versus ToGA (16.0 months, HR = 0.75, 95% CI = 0.59-0.99) and was less toxic. Trastuzumab plus doublet cisplatin and S-1 showed no OS difference versus ToGA, but showed a different toxicity profile, including less hand-foot syndrome. Trastuzumab plus cisplatin or capecitabine as singlet backbone showed significantly worse survival and more toxicity versus ToGA regimen. Trastuzumab with triplet cytotoxic backbones or with bevacizumab and doublet cytotoxic backbone showed no survival benefit and more toxicity. In conclusion, trastuzumab plus doublet cytotoxic backbone containing oxaliplatin is preferable over the ToGA regimen with cisplatin. S-1 can substitute capecitabine or 5-FU when specific toxicities are encountered.
Collapse
Affiliation(s)
- Emil Ter Veer
- Department of Medical Oncology, Cancer Center Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Aafke Creemers
- Department of Medical Oncology, Cancer Center Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Laura de Waal
- Department of Medical Oncology, Cancer Center Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Martijn G H van Oijen
- Department of Medical Oncology, Cancer Center Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Cancer Center Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
626
|
Mizrak Kaya D, Harada K, Amlashi FG, Vasilakopoulou M, Ajani JA. Customization of therapy for gastroesophageal adenocarcinoma patients. Chronic Dis Transl Med 2018; 4:8-17. [PMID: 29756119 PMCID: PMC5938285 DOI: 10.1016/j.cdtm.2018.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Indexed: 12/18/2022] Open
Abstract
Gastroesophageal adenocarcinomas (GEACs) remain a global health problem. These are most often diagnosed at advanced stage and the estimated 5-year relative survival rate is about 5%. Although cure is not possible for patients with advanced GEAC, systemic therapy (chemotherapy or biochemotherapy) can palliate symptoms, improve survival and provide a better quality of life. One of the most promising options for some patients with advanced stage GEAC is immunotherapy, which can result in durable responses. Numerous phase III trials evaluating targeted therapies in different lines are ongoing and it is hoped that better biomarkers will emerge to identify patients who can benefit from targeted agents and immunotherapy in the future. Surgery remains as the corner stone for localized GEAC and adjunctive therapies can increase the survival rates by about 10%. The high toxicity and low completion rates of adjuvant therapy led to the strategies of preoperative treatment. With the results of ongoing pre-operative therapy trials we will be able to determine the optimal adjunctive approach for resectable GEAC.
Collapse
Affiliation(s)
| | | | | | | | - Jaffer A. Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
627
|
Zhang D, Wu J, Wang K, Duan X, Liu S, Zhang B. Which are the best Chinese herbal injections combined with XELOX regimen for gastric cancer?: A PRISMA-compliant network meta-analysis. Medicine (Baltimore) 2018; 97:e0127. [PMID: 29561411 PMCID: PMC5895335 DOI: 10.1097/md.0000000000010127] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/16/2018] [Accepted: 02/18/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The optimal Chinese herbal injections (CHIs) combined with XELOX regimen for patients with gastric cancer remains elusive. The aim of our network meta-analysis (NMA) is to explore the best options among different CHIs for gastric cancer. METHODS PubMed, Embase, the Cochrane Library, the China National Knowledge Infrastructure Database (CNKI), Wan-fang Database, Cqvip Database (VIP), China Biology Medicine disc (CBMdisc) were searched to identify RCTs which focused on CHIs against gastric cancer. The quality assessment of included randomized controlled trials (RCTs) was conducted by the Cochrane risk of bias tool. Standard pair-wise and Bayesian NMAs were performed to compare the efficacy and safety of different CHIs combined with the XELOX regimen via Stata 13.0 and WinBUGS1.4 software. RESULTS A total of 2316 records were searched, the network of evidence included 26 eligible RCTs involving 13 types of CHIs and 2154 patients. The results suggested that Shenqifuzheng+ XELOX, Huachansu+ XELOX, Kangai+ XELOX, Javanica oil emulsion+ XELOX, Aidi injection+ XELOX might be the optimal treatment for gastric cancer in improving the performance status than using XELOX regimen single, with odds ratios (OR) and 95% confidence intervals (CIs) of 2.74 (1.24, 6.17), 8.27 (1.74, 42.43), 4.28 (1.80, 10.48), 5.14 (1.87, 16.28), 0.20 (0.090, 0.44). At the aspects of ADRs (adverse reactions), Compound Kushen+ XELOX, Lentinan+ XELOX, Xiaoaiping injection+ XELOX could obviously relieve leukopenia than only receiving XELOX regimen, and their ORs and 95% CIs were 5.62 (1.41, 36.24), 8.16 (2.25, 29.43), 5.69 (1.85, 15.77). Furthermore, Disodium cantharidinate and vitamin B6+ XELOX, Shenqifuzheng+ XELOX, Kangai+ XELOX, Lentinan+ XELOX could obviously relieve the nausea and vomiting than receiving the XELOX regimen alone, with ORs and 95% CIs of 5.29 (1.30, 23.96), 2.50 (1.16, 5.26), 2.42 (1.06, 5.63), 9.04 (3.24, 26.73). Nevertheless, CHIs combined with XELOX regimen did not confer higher better clinical effectiveness rate over receiving XELOX regimen alone, with nonstatistically significant between-group differences. CONCLUSIONS As the available evidence suggested that CHIs combined with XELOX regimen could provide treatment benefits for patients with gastric cancer. Among 13 types of CHIs, Javanica oil emulsion and Compound Kushen injection is the optimal treatment in improving the clinical effectiveness rate and performance status, and Lentinan injection was superior in relieving ADRs.
Collapse
|
628
|
A Novel Nomogram for Predicting Postsurgical Intra-abdominal Infection in Gastric Cancer Patients: a Prospective Study. J Gastrointest Surg 2018; 22:421-429. [PMID: 29330724 DOI: 10.1007/s11605-017-3580-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/06/2017] [Indexed: 01/31/2023]
Abstract
BACKGROUND This study aimed to determine the relationship between intra-abdominal infection (IAI) and sarcopenia prospectively and to construct a nomogram to identify patients at a high risk of IAI. METHODS We conducted a prospective study of 682 consecutive patients with gastric cancer who underwent radical gastrectomy. The sarcopenia elements, including lumbar skeletal muscle index, handgrip strength, and gait speed, were measured before surgery. Factors contributing to IAI were determined through univariate and multivariate analysis. A nomogram consisting of the independent risk factors was constructed to quantify the individual risk of IAI. RESULTS Of the 682 patients enrolled in this study, 132 patients were diagnosed with sarcopenia and 61 were diagnosed with IAI. Logistic analysis revealed that sarcopenia, tumor size, pathological type, and multivisceral resection were independent prognostic factors for IAI. The nomogram model for IAI was able to reliably quantify the risk of IAI with a strong optimism-adjusted discrimination (concordance index, 0.736). CONCLUSIONS Sarcopenia is an independent predictor of IAI. Our nomogram was a simple and practical instrument to quantify the individual risk of IAI and could be used to identify patients at a high risk.
Collapse
|
629
|
Ikoma N, Cormier JN, Feig B, Du XL, Yamal JM, Hofstetter W, Das P, Ajani JA, Roland C, Fournier K, Royal R, Mansfield P, Badgwell BD. Racial disparities in preoperative chemotherapy use in gastric cancer patients in the United States: Analysis of the National Cancer Data Base, 2006-2014. Cancer 2018; 124:998-1007. [PMID: 29393964 PMCID: PMC5937252 DOI: 10.1002/cncr.31155] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/03/2017] [Accepted: 10/23/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND No studies have investigated whether race/ethnicity is associated with the recommended use of preoperative chemotherapy or subsequent outcomes in gastric cancer. To determine whether there is such an association, analyses of patients with gastric cancer in the National Cancer Data Base (NCDB) were performed. METHODS Patients with clinical T2-4bN0-1M0 gastric adenocarcinoma, as defined by the eighth edition of the American Joint Committee on Cancer staging manual, who underwent gastrectomy from 2006 to 2014 were identified from the NCDB. Multiple logistic regression was conducted to examine factors associated with preoperative chemotherapy use. RESULTS This study identified 16,945 patients who met the criteria, and 8286 of these patients (49%) underwent preoperative chemotherapy. The use of preoperative chemotherapy remarkably increased over the study period, from 34% in 2006 to 65% in 2014. Preoperative chemotherapy was more commonly used for cardia tumors than noncardia tumors (83% vs 44% in 2014). In a multivariable analysis, races and ethnicities other than non-Hispanic (NH) white race were associated with less frequent use of preoperative chemotherapy in comparison with NH whites after adjustments for social, tumor, and hospital factors. The insurance status and the education level mediated an enhanced effect of racial/ethnic disparities in preoperative chemotherapy use. The use of preoperative chemotherapy and radiation therapy was associated with reduced racial/ethnic disparities in overall survival. CONCLUSIONS Racial/ethnic disparities in the use of preoperative chemotherapy and in outcomes exist among patients with gastric cancer in the United States. Efforts to improve the access to high-quality cancer care in minority groups may reduce racial disparities in gastric cancer in the United States. Cancer 2018;124:998-1007. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- Naruhiko Ikoma
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Janice N Cormier
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Barry Feig
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xianglin L. Du
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Jose-Miguel Yamal
- Department of Biostatistics, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Wayne Hofstetter
- Department of Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jaffer A. Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Christina Roland
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Keith Fournier
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard Royal
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Paul Mansfield
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Brian D. Badgwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
630
|
Jabo B, Selleck MJ, Morgan JW, Lum SS, Bahjri KA, Aljehani M, Garberoglio CA, Reeves ME, Namm JP, Solomon NL, Luca F, Dyke C, Senthil M. Comparison of perioperative chemotherapy with adjuvant chemoradiotherapy for resectable gastric cancer: findings from a population-based study. J Gastrointest Oncol 2018; 9:35-45. [PMID: 29564169 DOI: 10.21037/jgo.2017.10.13] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background Both perioperative chemotherapy (PC) and adjuvant chemoradiotherapy (CRT) improve survival in resectable gastric cancer; however, these treatments have never been formally compared. Our objective was to evaluate treatment trends and compare survival outcomes for gastric cancer patients treated with surgery and either PC or CRT. Methods We performed a retrospective population-based cohort study between 2007 through 2013 using California Cancer Registry data. Patients diagnosed with stage IB-III gastric adenocarcinoma and treated with total or partial gastrectomy were eligible for this study. Based on the type of treatment received, patients were grouped into surgery-only, PC, or CRT. Primary and secondary outcomes were overall survival (OS) and gastric cancer-specific survival (GCCS) respectively. Mortality hazards ratios (HRs) for each of these outcomes were computed using propensity score weighted and covariate-adjusted Cox regression models, stratified by clinical node status. Results Of 2,146 patients who underwent surgical resection, 1,067 had surgery-only, while 771 and 308 received PC or CRT, respectively. Median OS was 25, 33, and 52 months for surgery-only, PC, and CRT, respectively; P<0.001. Overall, patients treated with PC had significantly poorer survival compared to CRT (HR =1.45; 95% CI: 1.22-1.73). PC was also associated with higher mortality in patients with signet ring histology (HR =1.66; 95% CI: 1.21-2.28) and clinical node negative cancer (HR =1.85; 95% CI: 1.32-2.60). Survival was not different between PC vs. CRT in clinical node positive patients (HR =1.29; 95% CI: 0.84-2.08). Of note, the percentage of patients receiving PC increased from 17.5% in 2007-2008, to 41.5% in 2013-2014; P<0.001. Conclusions Despite the rapid adoption of PC, overall, CRT is associated with better survival than PC. Specifically, clinical node negative and signet ring histology patients had better survival when treated with CRT compared to PC. Based on these findings, we recommend against indiscriminate adoption of PC and consideration for CRT over PC in clinical node negative patients.
Collapse
Affiliation(s)
- Brice Jabo
- School of Public Health, Loma Linda University, Loma Linda, CA, USA
| | - Matthew J Selleck
- Division of Surgical Oncology, Loma Linda University, Loma Linda, CA, USA
| | - John W Morgan
- Surveillance Epidemiology and End Results (SEER) Cancer Registry, Cancer Registry of Greater California and California Cancer Registry, Sacramento, Loma Linda, CA, USA
| | - Sharon S Lum
- Division of Surgical Oncology, Loma Linda University, Loma Linda, CA, USA
| | - Khaled A Bahjri
- School of Public Health, Loma Linda University, Loma Linda, CA, USA
| | - Mayada Aljehani
- School of Public Health, Loma Linda University, Loma Linda, CA, USA
| | | | - Mark E Reeves
- Division of Surgical Oncology, Loma Linda University, Loma Linda, CA, USA
| | - Jukes P Namm
- Division of Surgical Oncology, Loma Linda University, Loma Linda, CA, USA
| | | | - Fabrizio Luca
- Division of Surgical Oncology, Loma Linda University, Loma Linda, CA, USA
| | - Crickett Dyke
- Surveillance Epidemiology and End Results (SEER) Cancer Registry, Cancer Registry of Greater California and California Cancer Registry, Sacramento, Loma Linda, CA, USA
| | - Maheswari Senthil
- Division of Surgical Oncology, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
631
|
Nguyen PM, Putoczki TL. Could the inhibition of IL-17 or IL-18 be a potential therapeutic opportunity for gastric cancer? Cytokine 2018; 118:8-18. [PMID: 29396054 DOI: 10.1016/j.cyto.2018.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 02/07/2023]
Abstract
Chronic inflammation is recognized as a key tumor-promoting factor in a number of epithelial cancers, including gastric cancer (GC). The production of pro-inflammatory cytokines in the tumor microenvironment by both the innate and the adaptive immune response can activate signaling pathways that are associated with increased cell survival and proliferation of cancer cells. Among the cytokines that have most commonly been linked to inflammation-associated cancers, are the Th17 cell-associated cytokines IL-17A, IL-23, IL-22, and the IL-1 family members IL-1β and IL-18. However, whether their contribution to inflammation-associated cancers is universal, or specific to individual types of cancers, remains to be elucidated. This review will explore our current understanding of the known roles of these cytokines in gastritis and discuss how their therapeutic inhibition may be useful for GC.
Collapse
Affiliation(s)
- Paul M Nguyen
- Walter and Eliza Hall Institute of Medical Research, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Victoria 3052, Australia
| | - Tracy L Putoczki
- Walter and Eliza Hall Institute of Medical Research, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Victoria 3052, Australia.
| |
Collapse
|
632
|
Altman AM, Hui JYC, Tuttle TM. Quality-of-life implications of risk-reducing cancer surgery. Br J Surg 2018; 105:e121-e130. [DOI: 10.1002/bjs.10725] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 09/04/2017] [Indexed: 01/15/2023]
Abstract
Abstract
Background
Modern advances in genetic sequencing techniques have allowed for increased availability of genetic testing for hereditary cancer syndromes. Consequently, more people are being identified as mutation carriers and becoming aware of their increased risk of malignancy. Testing is commonplace for many inheritable cancer syndromes, and with that comes the knowledge of being a gene carrier for some patients. With increased risk of malignancy, many guidelines recommend that gene carriers partake in risk reduction strategies, including risk-reducing surgery for some syndromes. This review explores the quality-of-life consequences of genetic testing and risk-reducing surgery.
Methods
A narrative review of PubMed/MEDLINE was performed, focusing on the health-related quality-of-life implications of surgery for hereditary breast and ovarian cancer, familial adenomatous polyposis and hereditary diffuse gastric cancer.
Results
Risk-reducing surgery almost uniformly decreases cancer anxiety and affects patients' quality of life.
Conclusion
Although the overwhelming quality-of-life implications of surgery are neutral to positive, risk-reducing surgery is irreversible and can be associated with short- and long-term side-effects.
Collapse
Affiliation(s)
- A M Altman
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - J Y C Hui
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - T M Tuttle
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
633
|
Li Z, Li Z, Zhang L, Liu Q, Wang Z, Zhang Z, Xiao G, Fu W, Wang X, Ye Y, Yu J, Li F, Chen L, Wang S, Ji J. Staging laparoscopy for locally advanced gastric cancer in Chinese patients: a multicenter prospective registry study. BMC Cancer 2018; 18:63. [PMID: 29321007 PMCID: PMC5764004 DOI: 10.1186/s12885-017-3791-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 11/15/2017] [Indexed: 01/01/2023] Open
Abstract
Background Staging laparoscopy(SL) is a recommended technique for the staging of Gastric Cancer(GC) and provides the indication for a radical surgery. Considering the medical practice in China, the standardized and regular usage of SL is yet to be spread. However, existing guidelines vary and make an ambiguity of indication for SL. Besides, the specific indication for Chinese patients remains a niche. This study aims to the essential, missing information of Chinese patients and tries to normalize the indication of LS in medical practice in China. Methods The study is a prospective, multicenter cohort study being conducted in China with a total of 450 patients, all diagnosed with locally advanced gastric cancer (cT2-4 N0-3 M0, no evidence of intra-abdominal dissemination) through Computed Tomography(CT) and/or Endoscopic Ultrasonography(EUS). Peritoneal lavage is regularly performed during the SL. Multivariate Cox regression model and receiver-operator characteristic(ROC) analysis will be used to analyze the significant risk factors of intra-abdominal metastasis(including peritoneal dissemination and a positive cytological result). Discussion This confirmatory study will provide us with the specific positive rate of intraabdominal metastasis of GC in China, compared with empirical evidence of 20%. We expect this trial will contribute to our discovery of the specific risk factors of intra-abdominal metastasis of Chinese patients and to the stimulating and performing of minimally invasive surgical procedures. Trial registration ClinicalTrials.gov: registration number NCT02172690.
Collapse
Affiliation(s)
- Ziyu Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Zhemin Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Lianhai Zhang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Qian Liu
- Cancer Hospital and Institute, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Zhenjun Wang
- Department of General Surgery, Capital Medical University Affiliated Beijing Chao-Yang Hospital, Beijing, 100020, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Gang Xiao
- Department of General Surgery, Beijing Hospital of Ministry of Health, Beijing, 100730, China
| | - Wei Fu
- Department of General Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Xin Wang
- Department of General Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People's Hospital, Beijing, 100044, China
| | - Jianchun Yu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, 100730, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Lin Chen
- Department of General Surgery, The General Hospital of PLA, Beijing, 100853, China
| | - Shibin Wang
- Department of General Surgery, The First Affiliated Hospital, General Hospital of PLA, Beijing, 100048, China
| | - Jiafu Ji
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, 100142, China. .,Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
634
|
Jaruvongvanich V, Sempokuya T, Wijarnpreecha K, Ungprasert P. Heparin-Bridging Therapy and Risk of Bleeding After Endoscopic Submucosal Dissection for Gastric Neoplasms: a Meta-Analysis. J Gastrointest Cancer 2018; 49:16-20. [DOI: 10.1007/s12029-017-0049-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
635
|
Xiao H, Liu W, Quan H, Ouyang Y. Peri-Operative Blood Transfusion Does Not Influence Overall and Disease-Free Survival After Radical Gastrectomy for Stage II/III Gastric Cancer: a Propensity Score Matching Analysis. J Gastrointest Surg 2018; 22:1489-1500. [PMID: 29777453 PMCID: PMC6132396 DOI: 10.1007/s11605-018-3808-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/07/2018] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Whether peri-operative blood transfusions (BTF) negatively impact long-term survival after gastrectomy for gastric cancer (GC) remains controversial. The aim of this retrospective study was to investigate independent predictive factors of BTF and the potential impact of BTF on overall survival (OS) and disease-free survival (DFS) in patients who underwent radical gastrectomy for stage II/III GC. METHODS Of 1020 patients who underwent gastrectomy for stage II/III GC from November 2010 to December 2015, 231 (22.6%) patients received BTF. The independent predictive factors of BTF were identified using univariate and multivariate analyses. Cox regression and propensity score matching (PSM) analyses of OS and DFS in patients who received BTF or not were compared. RESULTS Multivariate analysis revealed that age, pre-operative hemoglobin levels, tumor size, operation time, combined multi-organ resection, and intra-operative blood loss were independent predictive factors for BTF. PSM analysis created 205 pairs of patients. BTF was significantly associated with decreased OS (P = 0.025) and DFS (P = 0.034) in the entire cohort before PSM. After PSM, there was no longer a significant association between BTF and OS (P = 0.850) or DFS (P = 0.880). BTF was not identified as an independent risk factor for OS or DFS by multivariate Cox regression analysis. CONCLUSIONS The present study revealed that BTF did not influence OS and DFS after radical gastrectomy for stage II/III GC. Worse oncological outcomes were caused by clinical circumstances requiring blood transfusions, including longer operation time and advanced tumor stage, not due to BTF itself.
Collapse
Affiliation(s)
- Hua Xiao
- 0000 0001 0379 7164grid.216417.7Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wu Liu
- 0000 0001 0379 7164grid.216417.7Department of Gastroenterology and Urology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Hu Quan
- 0000 0001 0379 7164grid.216417.7Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yongzhong Ouyang
- 0000 0001 0379 7164grid.216417.7Department of Gastroduodenal and Pancreatic Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
636
|
Ilson DH, van Hillegersberg R. Management of Patients With Adenocarcinoma or Squamous Cancer of the Esophagus. Gastroenterology 2018; 154:437-451. [PMID: 29037469 DOI: 10.1053/j.gastro.2017.09.048] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/09/2017] [Accepted: 09/12/2017] [Indexed: 02/07/2023]
Abstract
Esophageal cancer is characterized by early and frequent metastasis. Surgery is the primary treatment for early-stage disease, whereas patients with patients with locally advanced disease receive perioperative chemotherapy or chemoradiotherapy. Squamous cancers can be treated with primary chemoradiotherapy without surgery, depending on their response to therapy and patient tolerance for subsequent surgery. Chemotherapy with a fluorinated pyrimidine and a platinum agent, followed by later treatment with taxanes and irinotecan, provides some benefit. Agents that inhibit the erb-b2 receptor tyrosine kinase 2 (ERBB2 or HER2), or vascular endothelial growth factor, including trastuzumab, ramucirumab, and apatinib, increase response and survival times. Esophageal adenocarcinomas have mutations in tumor protein p53 and mutations that activate receptor-associated tyrosine kinase, vascular endothelial growth factor, and cell cycle pathways, whereas esophageal squamous tumors have a distinct set of mutations. Esophageal cancers develop systems to evade anti-tumor immune responses, but studies are needed to determine how immune checkpoint modification contributes to esophageal tumor development.
Collapse
Affiliation(s)
- David H Ilson
- Memorial Sloan Kettering Cancer Center, New York, New York.
| | | |
Collapse
|
637
|
Sawaki A, Yamada Y, Yamaguchi K, Nishina T, Doi T, Satoh T, Chin K, Boku N, Omuro Y, Komatsu Y, Hamamoto Y, Koizumi W, Saji S, Shah MA, Van Cutsem E, Kang YK, Iwasaki J, Kuriki H, Ohtsuka W, Ohtsu A. Regional differences in advanced gastric cancer: exploratory analyses of the AVAGAST placebo arm. Gastric Cancer 2018; 21:429-438. [PMID: 29058097 PMCID: PMC5906488 DOI: 10.1007/s10120-017-0773-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 10/06/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND AVAGAST was an international, randomized, placebo-controlled phase III study of chemotherapy with or without bevacizumab as first-line therapy for patients with advanced gastric cancer. We performed exploratory analyses to evaluate regional differences observed in the trial. METHODS Analyses were performed in the placebo plus chemotherapy arm (intention-to-treat population). Chemotherapy was cisplatin 80 mg/m2 for six cycles plus capecitabine (1000 mg/m2 orally bid days 1-14) or 5-fluorouracil (800 mg/m2/day continuous IV infusion days 1-5) every 3 weeks until disease progression or unacceptable toxicity. RESULTS Overall, 387 patients were assigned to placebo plus chemotherapy (eastern Europe/South America, n = 118; USA/western Europe, n = 81; Korea/other Asia, n = 94; Japan, n = 94). At baseline, poor performance status, liver metastases, and larger tumors were most frequent in eastern Europe/South America and least frequent in Japan. Patients received subsequent chemotherapy after disease progression as follows: eastern Europe/South America (14%); USA/western Europe (37%); Korea/other Asia (61%); and Japan (77%). Hazard ratios for overall survival versus USA/western Europe were 1.47 (95% CI, 1.09-1.99) for eastern Europe/South America, 0.91 (95% CI, 0.67-1.25) for Korea/other Asia, and 0.87 (95% CI, 0.64-1.19) for Japan. CONCLUSIONS Regional differences in the healthcare environment may have contributed to the differences in overall survival observed in the AVAGAST study.
Collapse
Affiliation(s)
- Akira Sawaki
- Aichi Cancer Center Hospital, Aichi, Japan.
- Department of Medical Oncology, School of Medicine, Fujita Health University, Toyoake, Aichi, 470-1192, Japan.
| | | | - Kensei Yamaguchi
- Saitama Cancer Center Hospital, Saitama, Japan
- Cancer Institute Hospital of JFCR, Tokyo, Japan
| | | | - Toshihiko Doi
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Taroh Satoh
- Faculty of Medicine, Kinki University Hospital, Osaka, Japan
- Osaka University Hospital, Osaka, Japan
| | - Keisho Chin
- Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - Narikazu Boku
- National Cancer Center Hospital, Tokyo, Japan
- Shizuoka Cancer Center, Shizuoka, Japan
| | - Yasushi Omuro
- Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, Tokyo, Japan
| | | | | | - Wasaburo Koizumi
- Kitasato University East Hospital, Kanagawa, Japan
- Kitasato University Hospital, Tokyo, Japan
| | - Shigehira Saji
- International Medical Center, Saitama Medical University, Saitama, Japan
- Fukushima Medical University, Fukushima, Japan
| | - Manish A Shah
- Weill Cornell Medical College/New York Hospital, New York, USA
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg, Leuven and KULeuven, Louvain, Belgium
| | - Yoon-Koo Kang
- Asan Medical Center, University of Ulsan, Seoul, South Korea
| | | | | | | | - Atsushi Ohtsu
- National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
638
|
Zhang L, Xing X, Meng F, Wang Y, Zhong D. Oral fluoropyrimidine versus intravenous 5-fluorouracil for the treatment of advanced gastric and colorectal cancer: Meta-analysis. J Gastroenterol Hepatol 2018; 33:209-225. [PMID: 28608993 DOI: 10.1111/jgh.13845] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/07/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIM 5-Fluorouracil (5-Fu) is one of the most commonly prescribed antineoplastic agents against gastric and colorectal cancers. Continuous infusion would be the optimal way of its administration, however, may usually cause thrombosis, infection, and prolonged hospital stay. Oral fluoropyrimidines would be an attractive alternative, but their efficiency and toxicities for the treatment of gastric and colorectal cancer are still obscure as compared with infusion 5-Fu. METHODS Literature retrieval, trials selection and assessment, data collection, and statistic analysis were performed according to the Cochrane Handbook. The outcome measures were tumor response rate, progression-free survival, overall survival, and adverse effects. RESULTS Twenty-nine randomized controlled trials, comprising totally 15 154 patients, were included. Meta-analysis showed similar overall outcome in terms of response rate (1.01; 95% confidence interval [CI], 0.92-1.12), progression-free survival (hazard ratio 1.00; 95%CI, 0.94-1.06), and overall survival (hazard ratio 0.96; 95%CI, 0.92-1.01) between oral fluoropyrimidine-based and intravenous 5-Fu-based regimens in gastric and colorectal cancer patients. The risk of grade 3/4 neutropenia, thrombocytopenia, and stomatitis was more prominent in intravenous 5-Fu-based regimens; while more frequent grade 3/4 hand-foot syndrome, diarrhea, and anorexia were detected in oral fluoropyrimidine-based regimens. CONCLUSIONS Oral-fluoropyrimidines showed equivalent response and similar survival outcomes, but different toxicity profiles, as compared with intravenous 5-Fu. Thus, it would be a more convenient and adjustable alternative in treatment of advanced gastric and colorectal cancer.
Collapse
Affiliation(s)
- Linlin Zhang
- Department of Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoli Xing
- Tianjin Fifth Central Hospital, Tianjin, China
| | - Fanlu Meng
- Department of Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Wang
- Department of Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Diansheng Zhong
- Department of Oncology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
639
|
Lee B, Hutchinson R, Wong HL, Tie J, Putoczki T, Tran B, Gibbs P, Christie M. Emerging biomarkers for immunomodulatory cancer treatment of upper gastrointestinal, pancreatic and hepatic cancers. Semin Cancer Biol 2017; 52:241-252. [PMID: 29258858 DOI: 10.1016/j.semcancer.2017.12.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 12/14/2022]
Abstract
Carcinomas of the oesophagus, stomach, pancreas and liver are common and account for a disproportionately high number of cancer deaths. There is a need for new treatment options for patients with advanced disease. Immunomodulatory treatments including immune checkpoint blockade offer a promising new approach, with efficacy shown in other solid tumour types. However, only a small proportion of patients with carcinomas of the oesophagus, stomach, pancreas and liver have responded to single agent checkpoint inhibitors, and there is a need for markers that are predictive of response to guide treatment of individual patients. Predictive markers may include epidemiological factors such as ethnicity, the genomic status of the tumour, circulating markers, expression of immune checkpoint molecules, and other features of the stromal/immune response at the site of the tumour. This review will focus on predictive biomarkers for immune checkpoint blockade in oesophageal, gastric, pancreatic and hepatocellular carcinomas, including the genomic context and immune landscape in which they occur. Pancreatic carcinomas are largely resistant to immune checkpoint inhibition in trials to date, therefore emerging immunomodulatory treatments in this tumour type are also reviewed.
Collapse
Affiliation(s)
- Belinda Lee
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Oncology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ryan Hutchinson
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Hui-Li Wong
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Jeanne Tie
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Tracy Putoczki
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ben Tran
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Peter Gibbs
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Oncology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Michael Christie
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Pathology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
| |
Collapse
|
640
|
Mizrak Kaya D, Nogueras-González GM, Harada K, Amlashi FG, Roy-Chowdhuri S, Estrella JS, Das P, Lee JH, Weston B, Bhutani MS, Matamoros A, Thomas I, Lin Q, Badgwell BD, Ajani JA. Risk of peritoneal metastases in patients who had negative peritoneal staging and received therapy for localized gastric adenocarcinoma. J Surg Oncol 2017; 117:678-684. [PMID: 29205363 DOI: 10.1002/jso.24912] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/15/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Positive peritoneal cytology (+PCyt) or gross carcinomatosis (GPC) carries a poor prognosis. Laparoscopic staging to detect +PCyt/GPC is recommended for all ≥T1b gastric adenocarcinoma (GAC). The natural history of patients with GAC who have baseline -PCyt and then undergo multimodality therapy is not well documented, particularly for the risk of subsequent GPC. METHODS We identified 238 GAC patients with baseline -PCyt who were followed for the development of peritoneal carcinomatosis (PC). Standard statistical methods were employed. RESULTS Of 238 patients, 192 had attempted surgery after preoperative therapy. Of these, 13 patients (6.8%) had GPC and one had liver metastases, thus surgery was aborted. We followed 164 patients who had an R0 resection. The median follow-up duration was 3.4 (range, 0.6-18) years. The rate of PC was 13.4%, (22/164 patients) and the median time to PC was 15.6 months. Female gender was associated with PC on multivariate analysis. The 5-year OS rate for patients without subsequent PC was 75%. Conclusion Even with baseline -Cyt, ∼25% of patients develop PC following multimodality therapy. Patients who do not develop PC have an excellent OS rate. Further research is warranted to detect PC at baseline by the use of biomarkers.
Collapse
Affiliation(s)
- Dilsa Mizrak Kaya
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | | | - Kazuto Harada
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Fatemeh G Amlashi
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Sinchita Roy-Chowdhuri
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Jeannelyn S Estrella
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Jeffrey H Lee
- Department of Gastroenterology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Brian Weston
- Department of Gastroenterology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Manoop S Bhutani
- Department of Gastroenterology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Aurelio Matamoros
- Department of Radiology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Irene Thomas
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Quan Lin
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Brian D Badgwell
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
641
|
Kang YK, Boku N, Satoh T, Ryu MH, Chao Y, Kato K, Chung HC, Chen JS, Muro K, Kang WK, Yeh KH, Yoshikawa T, Oh SC, Bai LY, Tamura T, Lee KW, Hamamoto Y, Kim JG, Chin K, Oh DY, Minashi K, Cho JY, Tsuda M, Chen LT. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017; 390:2461-2471. [PMID: 28993052 DOI: 10.1016/s0140-6736(17)31827-5] [Citation(s) in RCA: 1693] [Impact Index Per Article: 211.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/19/2017] [Accepted: 06/22/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, two or more previous regimens of chemotherapy have a poor prognosis, and current guidelines do not recommend any specific treatments for these patients. We assessed the efficacy and safety of nivolumab, a fully human IgG4 monoclonal antibody inhibitor of programmed death-1 (PD-1), in patients with advanced gastric or gastro-oesophageal junction cancer who had been previously been treated with two or more chemotherapy regimens. METHODS In this randomised, double-blind, placebo-controlled, phase 3 trial done at 49 clinical sites in Japan, South Korea, and Taiwan, eligible patients (aged ≥20 years with unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, standard therapy [including two or more previous chemotherapy regimens], with an Eastern Cooperative Oncology Group [ECOG] performance status of 0-1, and naive to anti-PD-1 therapy or other therapeutic antibodies and pharmacotherapies for the regulation of T cells) were recruited. Patients were randomly assigned (2:1) using an interactive web response system to receive 3 mg/kg nivolumab or placebo intravenously every 2 weeks, stratified by country, ECOG performance status, and number of organs with metastases. Study treatment was continued until progressive disease per investigator assessment or onset of toxicities requiring permanent discontinuation. Patients and investigators were masked to group assignment. The primary endpoint was overall survival in the intention-to-treat population. Safety was analysed in all patients who received at least one dose of study treatment. This study is ongoing but not recruiting new patients, and is registered with ClinicalTrials.gov, number NCT02267343. FINDINGS Between Nov 4, 2014, and Feb 26, 2016, we randomly assigned 493 patients to receive nivolumab (n=330) or placebo (n=163). At the data cutoff (Aug 13, 2016), median follow-up in surviving patients was 8·87 months (IQR 6·57-12·37) in the nivolumab group and 8·59 months (5·65-11·37) in the placebo group. Median overall survival was 5·26 months (95% CI 4·60-6·37) in the nivolumab group and 4·14 months (3·42-4·86) in the placebo group (hazard ratio 0·63, 95% CI 0·51-0·78; p<0·0001). 12-month overall survival rates were 26·2% (95% CI 20·7-32·0) with nivolumab and 10·9% (6·2-17·0) with placebo. Grade 3 or 4 treatment-related adverse events occurred in 34 (10%) of 330 patients who received nivolumab and seven (4%) of 161 patients who received placebo; treatment-related adverse events led to death in five (2%) of 330 patients in the nivolumab group and two (1%) of 161 patients in the placebo group. No new safety signals were observed. INTERPRETATION In this phase 3 study, the survival benefits indicate that nivolumab might be a new treatment option for heavily pretreated patients with advanced gastric or gastro-oesophageal junction cancer. Ongoing trials that include non-Asian patients are investigating nivolumab for advanced gastric or gastro-oesophageal junction cancer in various settings and earlier treatment lines. FUNDING Ono Pharmaceutical and Bristol-Myers Squibb.
Collapse
Affiliation(s)
- Yoon-Koo Kang
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Narikazu Boku
- Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Taroh Satoh
- Frontier Science for Cancer and Chemotherapy, Osaka University Graduate School of Medicine, Suita, Japan
| | - Min-Hee Ryu
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Yee Chao
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ken Kato
- Division of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hyun Cheol Chung
- Division of Medical Oncology, Medical Oncology Yonsei Cancer Center, Song-Dang Institute for Cancer Research, Yonsei University College of Medicine, Yonsei University Health System, Seoul, South Korea
| | - Jen-Shi Chen
- Division of Hematology and Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Won Ki Kang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kun-Huei Yeh
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Takaki Yoshikawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Sang Cheul Oh
- Division of Hematology and Oncology, Department of Internal Medicine, College of Medicine, Korea University, Seoul, South Korea
| | - Li-Yuan Bai
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Takao Tamura
- Department of Medical Oncology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Keun-Wook Lee
- Division of Hematology and Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
| | - Yasuo Hamamoto
- Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Jong Gwang Kim
- Kyungpook National University School of Medicine, Daegu, South Korea
| | - Keisho Chin
- Department of Gastroenterology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Keiko Minashi
- Department of Clinical Trial Promotion, Chiba Cancer Center, Chiba, Japan
| | - Jae Yong Cho
- Department of Medical Oncology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Masahiro Tsuda
- Department of Gastroenterological Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
642
|
Chen B, Wang J, Gu X, Zhang J, Zhang J, Feng X. The DNMT3B -579G>T Polymorphism Is Significantly Associated With the Risk of Gastric Cancer but not Lung Cancer in Chinese Population. Technol Cancer Res Treat 2017; 16:1259-1265. [PMID: 29332452 PMCID: PMC5762089 DOI: 10.1177/1533034617740475] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/12/2017] [Accepted: 10/09/2017] [Indexed: 01/09/2023] Open
Abstract
The -149C>T and -579G>T, 2 single nucleotide polymorphisms in de novo methyltransferase 3B gene promoter, have been previously reported to potentially alter the promoter activity and to influence cancer risk. However, the results from previous studies remain conflicting rather than conclusive. In view of this, we conducted a case-control study and then a meta-analysis to examine the association between these 2 single-nucleotide polymorphisms with risk of lung and gastric cancer in Chinese population. The genotyping was performed by polymerase chain reaction-restriction fragment length polymorphism and confirmed by sequencing. In this case-control study, no significant association with lung or gastric cancer risk was observed for -149C>T, while -579G>T was significantly correlated with the risk of gastric cancer but not lung cancer. Moreover, haplotype analysis showed that haplotype -149T/-579 T, which carried the risk -579 T allele, significantly increased the susceptibility to gastric cancer. However, none of the haplotypes was associated with the risk of lung cancer. The following meta-analysis involved only Chinese population and further confirmed the significant association of -579G>T with gastric cancer but not lung cancer and suggested no significant association between -149C>T and risk of lung or gastric cancer. Collectively, DNMT3B -579G>T polymorphism is associated with gastric cancer risk in Chinese population, and the -579G>T may be used as a genetic biomarker to predict the risk of gastric cancer in Chinese population.
Collapse
Affiliation(s)
- Bifeng Chen
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Jingdong Wang
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Xiuli Gu
- Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Wuhan Tongji Reproductive Medicine Hospital, Wuhan, Hubei, China
| | - Jingli Zhang
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Jiankun Zhang
- Department of Biological Science and Technology, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei, China
| | - Xianhong Feng
- Clinical Laboratory, Wuhan Xinzhou District People’s Hospital, Wuhan, China
| |
Collapse
|
643
|
Strong VE, Russo A, Yoon SS, Brennan MF, Coit DG, Zheng CH, Li P, Huang CM. Comparison of Young Patients with Gastric Cancer in the United States and China. Ann Surg Oncol 2017; 24:3964-3971. [PMID: 29052085 DOI: 10.1245/s10434-017-6073-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Indexed: 08/29/2023]
Abstract
BACKGROUND This study aimed to compare the clinicopathologic characteristics and stage-specific prognosis of young patients with gastric cancer (GC) after curative resection (R0) in the United States and China. METHODS Data were collected on young patients (age ≤40 years) undergoing R0 resection at one U.S. (n = 79) and one Chinese (n = 257) institution. Patient, surgical, and pathologic variables and stage-specific survival rates were compared. Factors associated with 5-year disease-specific survival (DSS) were determined via multivariate analysis. RESULTS Tumor location was most often proximal in U.S. patients and distal in Chinese patients. The Chinese patients had more advanced-stage tumors, with a greater number of positive lymph nodes identified. Preoperative chemotherapy was administered more often in the United States. The 5-year overall survival (p = 0.07) and DSS (p = 0.07) did not differ statistically between the U.S. and Chinese cohorts. Among the patients with early GC receiving surgery alone, DSS did not differ significantly between the two cohorts (p = 0.44). Among the patients with advanced GC, DSS was comparable between the U.S. patients receiving preoperative chemotherapy plus surgery and the Chinese patients receiving surgery plus postoperative chemotherapy (p = 0.85). Lauren classification, depth of invasion, number of metastatic lymph nodes, and type of gastrectomy, but not country, were independent predictors of DSS. CONCLUSIONS Tumor features and therapeutic strategies among young patients with GC differ between the United States and China. Survival is comparable between young patients with advanced GC receiving preoperative chemotherapy plus surgery in the United States and those receiving surgery plus postoperative chemotherapy in China, suggesting that the outcomes for young patients with GC are stage dependent but not country specific.
Collapse
Affiliation(s)
- Vivian E Strong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ashley Russo
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sam S Yoon
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Murray F Brennan
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel G Coit
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| |
Collapse
|
644
|
Chao J, Lin J, Frankel P, Clark AJ, Wiley DT, Garmey E, Fakih M, Lim D, Chung V, Luevanos E, Eliasof S, Davis ME, Yen Y. Pilot trial of CRLX101 in patients with advanced, chemotherapy-refractory gastroesophageal cancer. J Gastrointest Oncol 2017; 8:962-969. [PMID: 29299355 DOI: 10.21037/jgo.2017.08.10] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background CRLX101 is an investigational nanoparticle-drug conjugate with a camptothecin payload. Preclinical evidence indicated preferential uptake in tumors, and tumor xenograft models demonstrate superiority of CRLX101 over irinotecan. A pilot trial was conducted at recommended phase 2 dosing (RP2D) using the bimonthly schedule to assess preferential uptake of CRLX101 in tumor vs. adjacent normal tissue in endoscopically accessible tumors in chemotherapy-refractory gastroesophageal cancer. Results from the biopsies were previously reported and herein we present the clinical outcomes. Methods Patients initiated CRLX101 dosed at RP2D (15 mg/m2) on days 1 and 15 of a 28-day cycle. Detection of preferential CRLX101 tumor uptake was the primary endpoint and objective response rate (ORR) was a secondary endpoint. With a sample size of ten patients, the study had 90% power to detect ≥1 responder if the true response rate is ≥21%. Results Between Dec. 2012 and Dec. 2014, ten patients with chemotherapy-refractory (median 2 prior lines of therapy, range 1-4) gastric adenocarcinoma were enrolled. The median time-to-progression was 1.7 months. Best response was seen in one patient with stable disease (SD) for 8 cycles. Only ≥ grade 3 drug-related toxicity occurred in one patient with grade 3 cardiac chest pain who was able to resume therapy after CRLX101 was reduced to 12 mg/m2. Conclusions Bimonthly CRLX101 demonstrated minimal activity with SD as best response in this heavily pretreated population. Future efforts with CRLX101 in gastric cancer should focus on combination and more dose-intensive strategies given its favorable toxicity profile and evidence of preferential tumor uptake.
Collapse
Affiliation(s)
- Joseph Chao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - James Lin
- Division of Gastroenterology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Paul Frankel
- Department of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Andrew J Clark
- Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Devin T Wiley
- Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | | | - Marwan Fakih
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Dean Lim
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Vincent Chung
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Eloise Luevanos
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Mark E Davis
- Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Yun Yen
- The Graduate Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
645
|
Li Z, Ji G, Bai B, Yu D, Liu Y, Lian B, Zhao Q. Laparoscopy-assisted distal gastrectomy versus laparoscopy-assisted total gastrectomy with D2 lymph node dissection for middle-third advanced gastric cancer. Surg Endosc 2017; 32:2255-2262. [PMID: 29098430 DOI: 10.1007/s00464-017-5919-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/07/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND There still remains controversy for the choice of resection extent for gastric cancer involving the middle-third of the stomach. The aim of this study was to compare the technical feasibility and long-term outcomes of laparoscopy-assisted distal gastrectomy (LADG) versus laparoscopy-assisted total gastrectomy (LATG) for middle-third advanced gastric cancer (AGC) and to determine which is the optimal surgical procedure. METHODS For this study, clinical data for 379 patients who underwent LADG or LATG with D2 lymph node dissection between April 2005 and June 2014 were analyzed retrospectively. The short- and long-term outcomes were compared between the propensity score-matched groups. RESULTS The LADG group had a significantly shorter operating time (212.74 vs. 241.79 min, P < 0.001), less estimated blood loss (114.38 vs. 181.51 ml, P = 0.000), shorter first flatus and postoperative hospital stay. Additionally, the total cost of hospitalization was significantly higher in the LATG group than LADG group (71187.58 vs. 65783.25 RMB, P = 0.000). There were no significant differences in postoperative complications rate between the LADG group and the LATG group. The 5-year overall survival (OS) rates were 64.4% in the LADG group and 61.0% in the LATG group (P = 0.548). The resection extent was not an independent prognostic factor for the OS. CONCLUSIONS LADG with D2 nodal dissection is a feasible treatment strategy for middle-third AGC with better short-term outcomes and similar long-term survival rates compared with LATG. We recommended that DG should be the optimal surgical procedure for middle one-third AGC under the premise of negative proximal resection margin.
Collapse
Affiliation(s)
- Zhengyan Li
- Department of Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 127 Changle West Road, Xian, 710032, China
| | - Gang Ji
- Department of Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 127 Changle West Road, Xian, 710032, China
| | - Bin Bai
- Department of Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 127 Changle West Road, Xian, 710032, China
| | - Deliang Yu
- Department of Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 127 Changle West Road, Xian, 710032, China
| | - Yezhou Liu
- Department of Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 127 Changle West Road, Xian, 710032, China
| | - Bo Lian
- Department of Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 127 Changle West Road, Xian, 710032, China
| | - Qingchuan Zhao
- Department of Surgery, Xijing Hospital of Digestive Diseases, The Fourth Military Medical University, No. 127 Changle West Road, Xian, 710032, China.
| |
Collapse
|
646
|
Marchesi F, De Sario G, Cecchini S, Tartamella F, Riccò M, Romboli A. Laparoscopic subtotal gastrectomy for the treatment of advanced gastric cancer: a comparison with open procedure at the beginning of the learning curve. ACTA BIO-MEDICA : ATENEI PARMENSIS 2017; 88:302-309. [PMID: 29083335 PMCID: PMC6142852 DOI: 10.23750/abm.v88i3.6541] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 06/14/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND In the last decades, after some initial concern, laparoscopic subtotal gastrectomy (LSG) is gaining popularity also for the treatment of advanced gastric cancer (AGC). The aim of this study is to compare a single surgeon initial experience on LSG and open subtotal gastrectomy in terms of surgical safety and radicality, postoperative recovery and midterm oncological outcomes. METHODS a case control study was conducted matching the first 13 LSG for AGC with 13 open procedures performed by the same surgeon. Operative and pathological data, postoperative parameters and midterm oncological outcomes were analyzed. RESULTS There was no significant difference in mortality (0%) and morbidity, while the laparoscopic approach allowed lower analgesic consumption and faster bowel movement recovery. Operation time was significantly higher in LSG patients (301.5 vs 232 min, p: 0.023), with an evident learning curve effect. Both groups had a high rate of adequate lymph node harvest, but the number was significantly higher in LSG group (p: 0.033). No significant difference in survival was registered. Multivariate analysis identified age at diagnosis, diffuse-type tumor, pN and LODDS as independent predictors of worse prognosis. CONCLUSIONS LSG can be safely performed for the treatment of AGC, allowing faster postoperative recovery.
Collapse
Affiliation(s)
- Federico Marchesi
- Dipartimento di Medicina e Chirurgia, sezione di Clinica Chirurgica Generale, Università degli studi di Parma, Via Gramsci n.14, 43100 Parma Italia..
| | | | | | | | | | | |
Collapse
|
647
|
Laterza MM, Pompella L, Petrillo A, Tirino G, Pappalardo A, Orditura M, Troiani T, Ciardiello F, Di Martino N, De Vita F. Efficacy of a triplet and doublet-based chemotherapy as first-line therapy in patients with HER2-negative metastatic gastric cancer: a retrospective analysis from the clinical practice. Med Oncol 2017; 34:186. [DOI: 10.1007/s12032-017-1046-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 10/13/2017] [Indexed: 11/30/2022]
|
648
|
Li B, Wang L, Li Z, Wang W, Zhi X, Huang X, Zhang Q, Chen Z, Zhang X, He Z, Xu J, Zhang L, Xu H, Zhang D, Xu Z. miR-3174 Contributes to Apoptosis and Autophagic Cell Death Defects in Gastric Cancer Cells by Targeting ARHGAP10. MOLECULAR THERAPY-NUCLEIC ACIDS 2017; 9:294-311. [PMID: 29246308 PMCID: PMC5684471 DOI: 10.1016/j.omtn.2017.10.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 10/13/2017] [Accepted: 10/13/2017] [Indexed: 01/04/2023]
Abstract
Gastric cancer (GC) is a major health problem worldwide because of its high morbidity and mortality. Considering the well-established roles of miRNA in the regulation of GC carcinogenesis and progression, we screened differentially expressed microRNAs (miRNAs) by using The Cancer Genome Atlas (TCGA) and the GEO databases. We found that miR-3174 was the most significantly differentially expressed miRNA in GC. Ectopic miR-3174 expression was also detected in clinical GC patient samples and cell lines and associated with poor patient prognosis. Apoptosis and autophagic cell death are two types of programmed cell death, whereas both are deficient in gastric cancer. Our functional analyses demonstrated that miR-3174 inhibited mitochondria-dependent apoptosis and autophagic cell death in GC. Moreover, high expression of miR-3174 also resulted in Cisplatin resistance in GC cells. Using bioinformatics analyses combined with in vitro and in vivo experiments, we determined that miR-3174 directly targets ARHGAP10. Notably, ARHGAP10 promoted mitochondria-dependent apoptosis by enhancing p53 expression, which was followed by Bax trans-activation and caspase cleavage. ARHGAP10 also facilitated autophagic cell death by suppressing mammalian target of rapamycin complex 1 (mTOC1) activity. Our results reveal a potential miRNA-based clinical therapeutic target that may also serve as a predictive marker for GC.
Collapse
Affiliation(s)
- Bowen Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Lu Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Zheng Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Weizhi Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Xiaofei Zhi
- Department of General Surgery, The Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Xiaoxu Huang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Qiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Zheng Chen
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Xuan Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Zhongyuan He
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Jianghao Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Lu Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Hao Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Diancai Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
649
|
Franck C, Canbay A, Malfertheiner P, Venerito M. Neoadjuvant treatment with cisplatin and S-1 in elderly patients with oesophagogastric adenocarcinoma and locoregional disease: Two case reports and review of the literature. Mol Clin Oncol 2017; 7:1069-1072. [PMID: 29285376 DOI: 10.3892/mco.2017.1445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/21/2017] [Indexed: 12/21/2022] Open
Abstract
Perioperative platinum/fluoropyrimidine-based chemotherapy is the therapeutic standard for oesophagogastric cancer (OAC) patients with locoregional disease. The preoperative condition directly affects postoperative prognosis; thus, particularly for elderly patients, a perioperative regimen with a favourable side effect profile is highly desirable. In the palliative setting, the combination of cisplatin and S-1 (Cis/S-1) was found to be as effective as cisplatin/5-fluorouracil, but with a more favourable side effect profile. However, no data on this combination have been reported in the perioperative setting in Caucasian patients. To the best of our knowledge, this is the first report on the treatment outcome of two elderly Caucasian OAC patients with locoregional disease receiving two neoadjuvant 4-week cycles of intravenous Cis/S-1. Both patients tolerated the doublet therapy well. No treatment delay or dose reduction was required. In both cases, preoperative staging revealed a clear response and complete surgical resection could be performed without any complications.
Collapse
Affiliation(s)
- Caspar Franck
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, D-39120 Magdeburg, Germany
| | - Ali Canbay
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, D-39120 Magdeburg, Germany
| | - Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, D-39120 Magdeburg, Germany
| | - Marino Venerito
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, D-39120 Magdeburg, Germany
| |
Collapse
|
650
|
Elimova E, Slack RS, Chen HC, Planjery V, Shiozaki H, Shimodaira Y, Charalampakis N, Lin Q, Harada K, Wadhwa R, Estrella JS, Kaya DM, Sagebiel T, Lee JH, Weston B, Bhutani M, Murphy MB, Matamoros A, Minsky B, Das P, Mansfield PF, Badgwell BD, Ajani JA. Patterns of relapse in patients with localized gastric adenocarcinoma who had surgery with or without adjunctive therapy: costs and effectiveness of surveillance. Oncotarget 2017; 8:81430-81440. [PMID: 29113402 PMCID: PMC5655297 DOI: 10.18632/oncotarget.19226] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 06/02/2017] [Indexed: 02/06/2023] Open
Abstract
PURPOSE After therapy of localized gastric adenocarcinoma (GAC) patients, the costs of surveillance, relapse patterns, and possibility of salvage are unknown. MATERIALS AND METHODS We identified 246 patients, who after having a negative peritoneal staging, received therapy (any therapy which included surgery) and were surveyed (every 3-6 months in the first 3 years, then yearly; ∼10 CTs and ∼7 endoscopies per patient). We used the 2016 Medicare dollars reimbursed as the "costs" for surveillance. RESULTS Common features were: Caucasians (57%), men (60%), poorly differentiated histology (76%), preoperative chemotherapy (74%), preoperative chemoradiation (59%), and had surgery (100%). At a median follow-up of 3.7 years (range, 0.1 to 18.3), the median overall survival (OS) was 9.2 years (95% CI, 6.0 to 11.2). Tumor grade (p = 0.02), p/yp stage (p < 0.001), % residual GAC (p = 0.05), the R status (p = 0.01), total gastrectomy (p = 0.001), and relapse type (p = 0.02) were associated with OS. Relapse occurred in 79 (32%) patients (only 8% were local-regional) and 90% occurred within 36 months of surgery. P/yp stage (p < 0.001) and total gastrectomy (p = 0.01) were independent prognosticators for OS in the multivariate analysis. Only 1 relapsed patient had successful salvage therapy. The estimated reimbursement for imaging studies and endoscopies was $1,761,221.91 (marked underestimation of actual costs). CONCLUSIONS The median OS of localized GAC patients was excellent with infrequent local-regional relapses. Rigorous surveillance had a low yield and high "costs". Our data suggest that less frequent surveillance intervals and limiting expensive investigations to symptomatic patients may be warranted.
Collapse
Affiliation(s)
- Elena Elimova
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
- Department of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Rebecca S. Slack
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Hsiang-Chun Chen
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Venkatram Planjery
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Hironori Shiozaki
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Yusuke Shimodaira
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Nick Charalampakis
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Quan Lin
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Kazuto Harada
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Roopma Wadhwa
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jeannelyn S. Estrella
- Department of Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Dilsa Mizrak Kaya
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Tara Sagebiel
- Department of Diagnostic Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey H. Lee
- Department of Gastroenterology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Brian Weston
- Department of Gastroenterology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Manoop Bhutani
- Department of Gastroenterology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Mariela Blum Murphy
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Aurelio Matamoros
- Department of Diagnostic Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Bruce Minsky
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Paul F. Mansfield
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Brian D. Badgwell
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jaffer A. Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|