1
|
Xia H, Zhu J, Men C, Wang A, Mao Q, Feng Y, Li J, Xu J, Cheng X, Shi H. Light-initiated aggregation of gold nanoparticles for synergistic chemo-photothermal tumor therapy. NANOSCALE ADVANCES 2023; 5:3053-3062. [PMID: 37260491 PMCID: PMC10228337 DOI: 10.1039/d3na00114h] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/24/2023] [Indexed: 06/02/2023]
Abstract
The combination of chemotherapy with photothermal therapy (PTT) has attracted extensive attention due to its excellent synergetic effect attributing to the fact that hyperthermia can effectively promote the tumor uptake of chemotherapeutic drugs. Herein, we propose a light-initiated gold nanoparticle (AuNP) aggregation boosting the uptake of chemotherapeutic drugs for enhanced chemo-photothermal tumor therapy. Novel light-responsive AuNPs (tm-AuNPs) were rationally designed and fabricated by conjugating both 2,5-diphenyltetrazole (Tz) and methacrylic acid (Ma) onto the surface of AuNPs with small size (∼20 nm). Upon the irradiation of 405 nm laser, AuNPs could be initiated to form aggregates specifically within tumors through the covalent cycloaddition reaction between Tz and Ma. Taking advantage of the controllable photothermal effect of Au aggregates under NIR excitation, improved enrichment of doxorubicin (DOX) in tumor tissues was realized, combined with PTT, resulting in outstanding synergetic anti-tumor efficacy in living mice. We thus believe that this light-initiated AuNP aggregation approach would offer a valuable and powerful tool for precisely synergistic chemo-photothermal tumor therapy.
Collapse
Affiliation(s)
- Huawei Xia
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University 199 Renai Road Suzhou 215123 China
| | - Jinfeng Zhu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University 199 Renai Road Suzhou 215123 China
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata Roma 00133 Italy
| | - Changhe Men
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University 199 Renai Road Suzhou 215123 China
| | - Anna Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University 199 Renai Road Suzhou 215123 China
| | - Qiulian Mao
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University 199 Renai Road Suzhou 215123 China
| | - Yali Feng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University 199 Renai Road Suzhou 215123 China
| | - Jiachen Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University 199 Renai Road Suzhou 215123 China
| | - Jingwei Xu
- Department of Cardiothoracic Surgery, Suzhou Municipal Hospital Institution Suzhou 215002 P. R. China
| | - Xiaju Cheng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University 199 Renai Road Suzhou 215123 China
| | - Haibin Shi
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University 199 Renai Road Suzhou 215123 China
| |
Collapse
|
2
|
Li H, Cheng Z, Jiang B, Shao X, Xu M. Prognosis value and positive association of Rab1A/IL4Rα aberrant expression in gastric cancer. Sci Rep 2023; 13:6964. [PMID: 37117331 PMCID: PMC10147632 DOI: 10.1038/s41598-023-33955-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/21/2023] [Indexed: 04/30/2023] Open
Abstract
Gastric cancer (GC) is the most common gastrointestinal cancer and the leading cause of worldwide cancer-associated mortality. Several GC patients are diagnosed at the advanced stage with an unsatisfactory 5-year survival rate. Rab1A was significantly associated with IL4Rα expression in non-small cell lung cancer. However, their potential correlation in expression and prognosis remains largely unknown in GC. In this study, Rab1A/IL-4Rα was significantly increased in GC than in para-cancerous tissues, and Rab1A/IL-4Rα overexpression caused poor prognosis among GC patients. Rab1A expression was significantly correlated with IL-4Rα expression in GC tissues, as determined by IHC analysis. In addition, the mRNA expression of Rab1A was closely linked with the IL-4Rα mRNA expression in GC tissue expressed by qPCR. Furthermore, the Kaplan-Meier analysis demonstrated that the group with negative Rab1A and IL-4Rα expression had longer 5-year survival rates than the other group. Besides, the group with positive Rab1A and IL-4Rα expression had a worse prognosis than the other group. Finally, nomograms revealed the overall 3 and 5-year survival determined crucial roles of Rab1A/IL-4Rα expression in predicting the prognosis of GC patients. Therefore, Rab1A/IL-4Rα is vital in GC, providing a novel perspective on targeted GC therapy.
Collapse
Affiliation(s)
- Haoran Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Zhengwu Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Bin Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Xinyu Shao
- Department of Gastroenterology, Suzhou Municipal Hospital, Affiliated Suzhou Hospital of Nanjing Medical University, No. 242 Guangji Road, Suzhou, 215006, Jiangsu, China.
| | - Menglin Xu
- Department of Oncology, The First Affiliated Hospital of Wannan Medical College, No. 2 Zheshan West Road, Jinghu District, Wuhu, 241000, Anhui, China.
| |
Collapse
|
3
|
Sun P, Antwi SO, Sartorius K, Zheng X, Li X. Tumor Microenvironment, Clinical Features, and Advances in Therapy for Bone Metastasis in Gastric Cancer. Cancers (Basel) 2022; 14:4888. [PMID: 36230816 PMCID: PMC9563035 DOI: 10.3390/cancers14194888] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/30/2022] [Accepted: 10/01/2022] [Indexed: 11/16/2022] Open
Abstract
Gastric cancer (GC) is one of the most malignant neoplasms worldwide, accounting for about 770,000 deaths in 2020. The incidence of gastric cancer bone metastasis (GC-BM) is low, about 0.9-13.4%, and GC patients develop GC-BM because of a suitable bone microenvironment. Osteoblasts, osteoclasts, and tumor cells interact with each other, secreting cytokines such as PTHrP, RANK-L, IL-6, and other growth factors that disrupt the normal bone balance and promote tumor growth. The functions and numbers of immune cells in the bone microenvironment are continuously inhibited, resulting in bone balance disorder due to the cytokines released from destroyed bone and growing tumor cells. Patients with GC-BM are generally younger than 65 years old and they often present with a later stage of the disease, as well as more aggressive tumors. They usually have shorter overall survival (OS) because of the occurrence of skeletal-related events (SREs) and undetected bone destruction due to the untimely bone inspection. Current treatments of GC-BM focus mainly on gastric cancer and SRE-related treatment. This article reviews the clinical features, possible molecular pathogeneses, and the most commonly used diagnostic methods and treatments of bone metastasis in gastric cancer.
Collapse
Affiliation(s)
- Pengcheng Sun
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou 213004, China
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou 213004, China
| | - Samuel O. Antwi
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
- The Africa Hepatopancreatobiliary Cancer Consortium (AHPBCC), Mayo Clinic, Jacksonville, FL 32224, USA
| | - Kurt Sartorius
- The Africa Hepatopancreatobiliary Cancer Consortium (AHPBCC), Mayo Clinic, Jacksonville, FL 32224, USA
- School of Laboratory Medicine and Molecular Sciences, College of Health Sciences, University of Kwazulu-Natal, Durban 4041, South Africa
- UKZN Gastrointestinal Cancer Research Unit, University of Kwazulu-Natal, Durban 4041, South Africa
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou 213004, China
| | - Xiaodong Li
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou 213004, China
- The Africa Hepatopancreatobiliary Cancer Consortium (AHPBCC), Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
4
|
Beeharry MK, Zhu ZL, Liu WT, Yao XX, Yan M, Zhu ZG. Prophylactic HIPEC with radical D2 gastrectomy improves survival and peritoneal recurrence rates for locally advanced gastric cancer: personal experience from a randomized case control study. BMC Cancer 2019; 19:932. [PMID: 31533660 PMCID: PMC6751672 DOI: 10.1186/s12885-019-6125-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023] Open
Abstract
Background To investigate the implications of prophylactic intraoperative Hyperthermic Intraperitoneal Chemotherapy (HIPEC) with D2 radical gastrectomy for locally advanced Gastric Cancer (AGC) in a randomized case control study. Method Eighty consecutive patients with locally AGC were randomly separated into 2 groups: HIPEC group (Curative Resection + intraoperative HIPEC with cisplatin 50 mg/m2 at 42.0 ± 1.0 °C for 60 min) and Control group (Curative Resection only). Intraoperative and post-operative events, clinical recovery, morbidity and the disease-free survival (DFS) rates were closely monitored. Results Faster recovery of bowel function (43 ± 5 h vs 68 ± 7, P < 0.05) and shorter postoperative stay (8d vs 14d, P < 0.05) were noted in the HIPEC group. Among the 40 HIPEC group patients, the highest intracranial temperature recorded during the procedure was 38.2 °C but the patient made an eventless recovery. Mild renal dysfunction, hyperbilirubinemia and mild liver dysfunction were recorded in the HIPEC group but their incidences were found to be statistically insignificant when compared with the control group (P > 0.05). The 3 year DFS rate analysis showed that the prophylactic HIPEC group had a higher DFS rate (93% vs 65%, P = 0.0054). The peritoneal recurrence rate was lower in the HIPEC group (3% vs 23%, P < 0.05). Conclusion Prophylactic HIPEC with radical D2 Gastrectomy improves survival and peritoneal recurrence rates for AGC with favorable post-operative recovery at low and acceptable morbidity.
Collapse
Affiliation(s)
- Maneesh Kumarsing Beeharry
- Department of Surgery, Ruijin Hospital affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zheng-Lun Zhu
- Department of Surgery, Ruijin Hospital affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wen-Tao Liu
- Department of Surgery, Ruijin Hospital affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xue-Xin Yao
- Department of Surgery, Ruijin Hospital affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Min Yan
- Department of Surgery, Ruijin Hospital affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zheng-Gang Zhu
- Department of Surgery, Ruijin Hospital affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
5
|
Abstract
AbstractRapid advances in pharmacotherapy and bioinformatics has led to the discovery and growing popularity of drug repositioning which includes re-investigating or recycling of existing drugs for new indications. There are innumerable advantages as well as challenges of drug repositioning. Since de-novo drug discovery takes plenty of time, effort and money, it has proved to a preferred alternative strategy for accelerated drug discovery. Moreover it is relatively inexpensive and carries minimal risk due to availability of previous pharmacological, safety and toxicology data. The strategies used are Known drug – new target/Drug focus/Drug-centric, Known target- new indication/Target focus/Target-centric and Disease focus/Disease-centric. Drug repositioning is a new breakthrough strategy to benefit patients by offering safer and effective treatment using shelved drugs.
Collapse
Affiliation(s)
- Man Mohan Mehndiratta
- Department of Neurology, Janakpuri Super Specialty Hospital, Janakpuri, New Delhi 110058, India
| | - Swati Wadhai
- Department of Pathology, Janakpuri Super Specialty Hospital, Janakpuri, New Delhi 110058, India
| | - Brij Tyagi
- Janakpuri Super Specialty Hospital, Janakpuri, New Delhi 110058, India
| | - Natasha Gulati
- Department of Pathology, Janakpuri Super Specialty Hospital, Janakpuri, New Delhi 110058, India
| | - Madhu Sinha
- Department of Pathology, Janakpuri Super Specialty Hospital, Janakpuri, New Delhi 110058, India
| |
Collapse
|
6
|
Personeni N, Baretti M, Bozzarelli S, Spaggiari P, Rubino L, Tronconi MC, Fumagalli Romario U, Rosati R, Giordano L, Roncalli M, Santoro A, Rimassa L. Assessment of HER2 status in patients with gastroesophageal adenocarcinoma treated with epirubicin-based chemotherapy: heterogeneity-related issues and prognostic implications. Gastric Cancer 2017; 20:428-437. [PMID: 27530622 DOI: 10.1007/s10120-016-0625-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 08/02/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND HER2 and topoisomerase 2 alpha (TOP2A) genomic status was previously reported to predict benefit from anthracyclines in breast cancer. We sought to define the prognostic impact and possible pitfalls related to these biomarkers in resectable gastroesophageal adenocarcinoma. METHODS HER2 and TOP2A gene amplification by fluorescent in situ hybridization and HER2 protein expression by immunohistochemistry (IHC) were assessed on whole tissue sections from 101 patients receiving peri- or postoperative epirubicin-based chemotherapy. In a subgroup of patients, at least two matched tumor blocks, originating either from surgical procedures (n = 88) or diagnostic biopsies (n = 32), were available for HER2 analyses by IHC. RESULTS Eighteen of 101 patients (17.8 %) were HER2 positive, whereas TOP2A was amplified in 4 of 84 patients (4.7 %). HER2 positivity was significantly associated with improved disease-free survival [HR = 0.47 (95 % CI 0.22-0.99), P = 0.046] and overall survival [HR = 0.33 (95 % CI 0.13-0.83), P < 0.018], independent of clinical-pathologic features. HER2 expression in matched tumor blocks from the same resection specimen was discordant in up to 11.8 % of pairs, while this rate increased up to 27.2 % when diagnostic biopsies and paired surgical samples were compared. CONCLUSIONS HER2 status is an independent prognostic biomarker in gastroesophageal adenocarcinomas receiving epirubicin-based chemotherapy. Compared to diagnostic biopsies, HER2 assessment in multiple resection specimens might lower the risk of sampling errors. These findings have several implications with respect to the optimal choice of the sample to be submitted to IHC testing of HER2.
Collapse
Affiliation(s)
- Nicola Personeni
- Humanitas Cancer Center, Humanitas Clinical and Research Center, Via Manzoni 56 Rozzano, 20089, Milan, Italy. .,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.
| | - Marina Baretti
- Humanitas Cancer Center, Humanitas Clinical and Research Center, Via Manzoni 56 Rozzano, 20089, Milan, Italy
| | - Silvia Bozzarelli
- Humanitas Cancer Center, Humanitas Clinical and Research Center, Via Manzoni 56 Rozzano, 20089, Milan, Italy
| | - Paola Spaggiari
- Department of Pathology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Luca Rubino
- Humanitas Cancer Center, Humanitas Clinical and Research Center, Via Manzoni 56 Rozzano, 20089, Milan, Italy
| | - Maria Chiara Tronconi
- Humanitas Cancer Center, Humanitas Clinical and Research Center, Via Manzoni 56 Rozzano, 20089, Milan, Italy
| | | | - Riccardo Rosati
- Department of General Surgery, Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Ospedale San Raffaele, Milan, Italy
| | - Laura Giordano
- Humanitas Cancer Center, Humanitas Clinical and Research Center, Via Manzoni 56 Rozzano, 20089, Milan, Italy
| | - Massimo Roncalli
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.,Department of Pathology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Armando Santoro
- Humanitas Cancer Center, Humanitas Clinical and Research Center, Via Manzoni 56 Rozzano, 20089, Milan, Italy.,Humanitas University, Rozzano, Milan, Italy
| | - Lorenza Rimassa
- Humanitas Cancer Center, Humanitas Clinical and Research Center, Via Manzoni 56 Rozzano, 20089, Milan, Italy
| |
Collapse
|
7
|
Wang H, Zhang H, Deng P, Liu C, Li D, Jie H, Zhang H, Zhou Z, Zhao YL. Tissue metabolic profiling of human gastric cancer assessed by (1)H NMR. BMC Cancer 2016; 16:371. [PMID: 27356757 PMCID: PMC4928316 DOI: 10.1186/s12885-016-2356-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 05/11/2016] [Indexed: 02/05/2023] Open
Abstract
Background Gastric cancer is the fourth most common cancer and the second most deadly cancer worldwide. Study on molecular mechanisms of carcinogenesis will play a significant role in diagnosing and treating gastric cancer. Metabolic profiling may offer the opportunity to understand the molecular mechanism of carcinogenesis and help to identify the potential biomarkers for the early diagnosis of gastric cancer. Methods In this study, we reported the metabolic profiling of tissue samples on a large cohort of human gastric cancer subjects (n = 125) and normal controls (n = 54) based on 1H nuclear magnetic resonance (1H NMR) together with multivariate statistical analyses (PCA, PLS-DA, OPLS-DA and ROC curve). Results The OPLS-DA model showed adequate discrimination between cancer tissues and normal controls, and meanwhile, the model excellently discriminated the stage-related of tissue samples (stage I, 30; stage II, 46; stage III, 37; stage IV, 12) and normal controls. A total of 48 endogenous distinguishing metabolites (VIP > 1 and p < 0.05) were identified, 13 of which were changed with the progression of gastric cancer. These modified metabolites revealed disturbance of glycolysis, glutaminolysis, TCA, amino acids and choline metabolism, which were correlated with the occurrence and development of human gastric cancer. The receiver operating characteristic diagnostic AUC of OPLS-DA model between cancer tissues and normal controls was 0.945. And the ROC curves among different stages cancer subjects and normal controls were gradually improved, the corresponding AUC values were 0.952, 0.994, 0.998 and 0.999, demonstrating the robust diagnostic power of this metabolic profiling approach. Conclusion As far as we know, the present study firstly identified the differential metabolites in various stages of gastric cancer tissues. And the AUC values were relatively high. So these results suggest that the metabolic profiling of gastric cancer tissues has great potential in detecting this disease and helping to understand its underlying metabolic mechanisms. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2356-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Huijuan Wang
- College of Medicine, Henan University, Kaifeng, 475004, Henan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Hailong Zhang
- College of Medicine, Henan University, Kaifeng, 475004, Henan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Pengchi Deng
- Analytical & Testing Center, Sichuan University, Chengdu, 610041, China
| | - Chunqi Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Dandan Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Hui Jie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Hu Zhang
- Department of Gastroenterology, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Zongguang Zhou
- Department of Gastrointestinal surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China.
| | - Ying-Lan Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
8
|
Qian J, Qian Y, Wang J, Gu B, Pei D, He S, Zhu F, Røe OD, Xu J, Liu L, Gu Y, Guo R, Yin Y, Shu Y, Chen X. A clinical prognostic scoring system for resectable gastric cancer to predict survival and benefit from paclitaxel- or oxaliplatin-based adjuvant chemotherapy. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:241-58. [PMID: 26966350 PMCID: PMC4771399 DOI: 10.2147/dddt.s88743] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Gastrectomy with D2 lymphadenectomy is a standard procedure of curative resection for gastric cancer (GC). The aim of this study was to develop a simple and reliable prognostic scoring system for GC treated with D2 gastrectomy combined with adjuvant chemotherapy. Methods A prognostic scoring system was established based on clinical and laboratory data from 579 patients with localized GC without distant metastasis treated with D2 gastrectomy and adjuvant chemotherapy. Results From the multivariate model for overall survival (OS), five factors were selected for the scoring system: ≥50% metastatic lymph node rate, positive lymphovascular invasion, pathologic TNM Stage II or III, ≥5 ng/mL preoperative carcinoembryonic antigen level, and <110 g/L preoperative hemoglobin. Two models were derived using different methods. Model A identified low- and high-risk patients for OS (P<0.001), while Model B differentiated low-, intermediate-, and high-risk patients for OS (P<0.001). Stage III patients in the low-risk group had higher survival probabilities than Stage II patients. Both Model A (area under the curve [AUC]: 0.74, 95% confidence interval [CI]: 0.69–0.78) and Model B (AUC: 0.79, 95% CI: 0.72–0.83) were better predictors compared with the pathologic TNM classification (AUC: 0.62, 95% CI: 0.59–0.71, P<0.001). Adjuvant paclitaxel- or oxaliplatin-based or triple chemotherapy showed significantly better outcomes in patients classified as high risk, but not in those with low and intermediate risk. Conclusion A clinical three-tier prognostic risk scoring system was established to predict OS of GC treated with D2 gastrectomy and adjuvant chemotherapy. The potential advantage of this scoring system is that it can identify high-risk patients in Stage II or III who may benefit from paclitaxel- or oxaliplatin-based regimens. Prospective studies are needed to confirm these results before they are applied clinically.
Collapse
Affiliation(s)
- Jing Qian
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Yingying Qian
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Jian Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Bing Gu
- Department of Laboratory Medicine, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, People's Republic of China; Medical Technology Institute, Xuzhou Medical College, Xuzhou, People's Republic of China
| | - Dong Pei
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Shaohua He
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Fang Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Oluf Dimitri Røe
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Department of Oncology, Clinical Cancer Research Center, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University Hospital, Aalborg, Denmark; Department of Surgery, Cancer Clinic, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Jin Xu
- Department of Molecular Cell Biology and Toxicology, Jiangsu Key Lab of Cancer Biomarkers, Prevention & Treatment, Cancer Center, Nanjing Medical University, Nanjing, People's Republic of China
| | - Lianke Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Renhua Guo
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| | - Xiaofeng Chen
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Xuzhou, People's Republic of China
| |
Collapse
|
9
|
Macrì A, Fleres F, Ieni A, Rossitto M, Mandolfino T, Micalizzi S, Iaropoli F, Mazzeo C, Trovato M, Cucinotta E, Saladino E. Wernicke encephalopathy as rare complication of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Int J Surg Case Rep 2015; 16:29-32. [PMID: 26410803 PMCID: PMC4643342 DOI: 10.1016/j.ijscr.2015.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 08/28/2015] [Accepted: 09/01/2015] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Peritoneal carcinomatosis of gastric origin is a frequent event with poor survival. A new promising approach is the association of the Cytoreductive Surgery (CRS) with the Hyperthermic Intraperitoneal Chemotherapy (HIPEC), which yet is characterized by high morbidity and mortality. We report, to our knowledge, the first case of Wernicke Encephalopathy (WE) complicating CRS plus HIPEC. WE, caused by a deficiency of thiamine, is characterized by ataxia, ocular motor cranial neuropathies and changes in consciousness. METHODS A patient affected by gastric cancer with peritoneal seeding, submitted to CRS plus HIPEC, in 4th post-operative day had manifested the appearance of flapping tremors, with positive manoeuvre of Mingazzini, impaired vision and mental confusion. The brain Magnetic Resonance Imaging (MRI) confirmed the clinical suspicion of WE. Even though the appropriate therapy was promptly applied, the patient died in 10th post-operative day. CONCLUSION WE is an uncommon neurological disorder. Only 16% of these patients inadequately treated recover fully, with a mortality rate of 10-20%. We consider useful to report this case, because it is the first time that WE is correlated to CRS plus HIPEC.
Collapse
Affiliation(s)
- Antonio Macrì
- Department of Human Pathology, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Francesco Fleres
- Department of Human Pathology, University of Messina, Via Consolare Valeria, 98125 Messina, Italy.
| | - Antonio Ieni
- Department of Human Pathology, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Maurizio Rossitto
- Department of Human Pathology, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Tommaso Mandolfino
- Anesthesiology and Neuroreanimation Unit, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Salvatore Micalizzi
- Anesthesiology and Neuroreanimation Unit, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Francesco Iaropoli
- Department of Human Pathology, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Carmelo Mazzeo
- Department of Human Pathology, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Massimo Trovato
- Department of Human Pathology, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Eugenio Cucinotta
- Department of Human Pathology, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Edoardo Saladino
- Department of Human Pathology, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| |
Collapse
|
10
|
Satoh T, Bang YJ, Gotovkin EA, Hamamoto Y, Kang YK, Moiseyenko VM, Ohtsu A, Van Cutsem E, Al-Sakaff N, Urspruch A, Hill J, Weber HA, Chung HC. Quality of life in the trastuzumab for gastric cancer trial. Oncologist 2014; 19:712-719. [PMID: 24951609 PMCID: PMC4077451 DOI: 10.1634/theoncologist.2014-0058] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 05/02/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The Trastuzumab for Gastric Cancer phase III trial demonstrated that combining trastuzumab with chemotherapy significantly improved overall survival compared with chemotherapy alone in HER2-positive advanced gastric or gastroesophageal junction cancer. We report health-related quality of life (HRQoL) and quality-adjusted time without symptoms of disease or toxicity (Q-TWiST) results from this trial. PATIENTS AND METHODS Patients were randomized to receive six cycles of chemotherapy given every 3 weeks (capecitabine or fluorouracil, plus cisplatin) either alone or combined with administration of trastuzumab every 3 weeks until disease progression. At each clinical visit, HRQoL was assessed using two European Organization for Research and Treatment of Cancer quality of life questionnaires, QLQ-C30 and QLQ-STO22. Q-TWiST methodology was applied retrospectively using the clinical data and utility coefficients. RESULTS Trastuzumab plus chemotherapy prolonged time to 10% definitive deterioration in all QLQ-C30 and QLQ-STO22 scores, including QLQ-C30 global health status versus chemotherapy alone, from 6.4 months to 10.2 months. In addition, trastuzumab plus chemotherapy extended Q-TWiST by 2.42 months compared with chemotherapy alone. CONCLUSION Compared with chemotherapy alone, trastuzumab plus chemotherapy prolongs time to deterioration of HRQoL and increases quality-adjusted survival in patients with HER2-positive gastric or gastroesophageal junction cancer.
Collapse
Affiliation(s)
- Taroh Satoh
- Osaka University Graduate School of Medicine, Osaka, Japan; Seoul National University College of Medicine, Seoul, Republic of Korea; Ivanovo Regional Oncology Dispensary, Ivanovo, Russian Federation; Tochigi Cancer Center, Utsunomiya, Japan; Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute of Oncology, St. Petersburg, Russian Federation; National Cancer Center Hospital East, Kashiwa, Japan; University Hospital Gasthuisberg, Leuven, Belgium; F. Hoffmann-La Roche Ltd., Basel, Switzerland; Roche Products, Sydney, New South Wales, Australia; Yonsei Cancer Center, Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yung-Jue Bang
- Osaka University Graduate School of Medicine, Osaka, Japan; Seoul National University College of Medicine, Seoul, Republic of Korea; Ivanovo Regional Oncology Dispensary, Ivanovo, Russian Federation; Tochigi Cancer Center, Utsunomiya, Japan; Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute of Oncology, St. Petersburg, Russian Federation; National Cancer Center Hospital East, Kashiwa, Japan; University Hospital Gasthuisberg, Leuven, Belgium; F. Hoffmann-La Roche Ltd., Basel, Switzerland; Roche Products, Sydney, New South Wales, Australia; Yonsei Cancer Center, Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Evgeny A Gotovkin
- Osaka University Graduate School of Medicine, Osaka, Japan; Seoul National University College of Medicine, Seoul, Republic of Korea; Ivanovo Regional Oncology Dispensary, Ivanovo, Russian Federation; Tochigi Cancer Center, Utsunomiya, Japan; Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute of Oncology, St. Petersburg, Russian Federation; National Cancer Center Hospital East, Kashiwa, Japan; University Hospital Gasthuisberg, Leuven, Belgium; F. Hoffmann-La Roche Ltd., Basel, Switzerland; Roche Products, Sydney, New South Wales, Australia; Yonsei Cancer Center, Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yasuo Hamamoto
- Osaka University Graduate School of Medicine, Osaka, Japan; Seoul National University College of Medicine, Seoul, Republic of Korea; Ivanovo Regional Oncology Dispensary, Ivanovo, Russian Federation; Tochigi Cancer Center, Utsunomiya, Japan; Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute of Oncology, St. Petersburg, Russian Federation; National Cancer Center Hospital East, Kashiwa, Japan; University Hospital Gasthuisberg, Leuven, Belgium; F. Hoffmann-La Roche Ltd., Basel, Switzerland; Roche Products, Sydney, New South Wales, Australia; Yonsei Cancer Center, Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoon-Koo Kang
- Osaka University Graduate School of Medicine, Osaka, Japan; Seoul National University College of Medicine, Seoul, Republic of Korea; Ivanovo Regional Oncology Dispensary, Ivanovo, Russian Federation; Tochigi Cancer Center, Utsunomiya, Japan; Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute of Oncology, St. Petersburg, Russian Federation; National Cancer Center Hospital East, Kashiwa, Japan; University Hospital Gasthuisberg, Leuven, Belgium; F. Hoffmann-La Roche Ltd., Basel, Switzerland; Roche Products, Sydney, New South Wales, Australia; Yonsei Cancer Center, Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Vladimir M Moiseyenko
- Osaka University Graduate School of Medicine, Osaka, Japan; Seoul National University College of Medicine, Seoul, Republic of Korea; Ivanovo Regional Oncology Dispensary, Ivanovo, Russian Federation; Tochigi Cancer Center, Utsunomiya, Japan; Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute of Oncology, St. Petersburg, Russian Federation; National Cancer Center Hospital East, Kashiwa, Japan; University Hospital Gasthuisberg, Leuven, Belgium; F. Hoffmann-La Roche Ltd., Basel, Switzerland; Roche Products, Sydney, New South Wales, Australia; Yonsei Cancer Center, Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Atsushi Ohtsu
- Osaka University Graduate School of Medicine, Osaka, Japan; Seoul National University College of Medicine, Seoul, Republic of Korea; Ivanovo Regional Oncology Dispensary, Ivanovo, Russian Federation; Tochigi Cancer Center, Utsunomiya, Japan; Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute of Oncology, St. Petersburg, Russian Federation; National Cancer Center Hospital East, Kashiwa, Japan; University Hospital Gasthuisberg, Leuven, Belgium; F. Hoffmann-La Roche Ltd., Basel, Switzerland; Roche Products, Sydney, New South Wales, Australia; Yonsei Cancer Center, Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eric Van Cutsem
- Osaka University Graduate School of Medicine, Osaka, Japan; Seoul National University College of Medicine, Seoul, Republic of Korea; Ivanovo Regional Oncology Dispensary, Ivanovo, Russian Federation; Tochigi Cancer Center, Utsunomiya, Japan; Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute of Oncology, St. Petersburg, Russian Federation; National Cancer Center Hospital East, Kashiwa, Japan; University Hospital Gasthuisberg, Leuven, Belgium; F. Hoffmann-La Roche Ltd., Basel, Switzerland; Roche Products, Sydney, New South Wales, Australia; Yonsei Cancer Center, Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Nedal Al-Sakaff
- Osaka University Graduate School of Medicine, Osaka, Japan; Seoul National University College of Medicine, Seoul, Republic of Korea; Ivanovo Regional Oncology Dispensary, Ivanovo, Russian Federation; Tochigi Cancer Center, Utsunomiya, Japan; Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute of Oncology, St. Petersburg, Russian Federation; National Cancer Center Hospital East, Kashiwa, Japan; University Hospital Gasthuisberg, Leuven, Belgium; F. Hoffmann-La Roche Ltd., Basel, Switzerland; Roche Products, Sydney, New South Wales, Australia; Yonsei Cancer Center, Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Alexa Urspruch
- Osaka University Graduate School of Medicine, Osaka, Japan; Seoul National University College of Medicine, Seoul, Republic of Korea; Ivanovo Regional Oncology Dispensary, Ivanovo, Russian Federation; Tochigi Cancer Center, Utsunomiya, Japan; Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute of Oncology, St. Petersburg, Russian Federation; National Cancer Center Hospital East, Kashiwa, Japan; University Hospital Gasthuisberg, Leuven, Belgium; F. Hoffmann-La Roche Ltd., Basel, Switzerland; Roche Products, Sydney, New South Wales, Australia; Yonsei Cancer Center, Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Julie Hill
- Osaka University Graduate School of Medicine, Osaka, Japan; Seoul National University College of Medicine, Seoul, Republic of Korea; Ivanovo Regional Oncology Dispensary, Ivanovo, Russian Federation; Tochigi Cancer Center, Utsunomiya, Japan; Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute of Oncology, St. Petersburg, Russian Federation; National Cancer Center Hospital East, Kashiwa, Japan; University Hospital Gasthuisberg, Leuven, Belgium; F. Hoffmann-La Roche Ltd., Basel, Switzerland; Roche Products, Sydney, New South Wales, Australia; Yonsei Cancer Center, Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Harald A Weber
- Osaka University Graduate School of Medicine, Osaka, Japan; Seoul National University College of Medicine, Seoul, Republic of Korea; Ivanovo Regional Oncology Dispensary, Ivanovo, Russian Federation; Tochigi Cancer Center, Utsunomiya, Japan; Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute of Oncology, St. Petersburg, Russian Federation; National Cancer Center Hospital East, Kashiwa, Japan; University Hospital Gasthuisberg, Leuven, Belgium; F. Hoffmann-La Roche Ltd., Basel, Switzerland; Roche Products, Sydney, New South Wales, Australia; Yonsei Cancer Center, Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun-Cheol Chung
- Osaka University Graduate School of Medicine, Osaka, Japan; Seoul National University College of Medicine, Seoul, Republic of Korea; Ivanovo Regional Oncology Dispensary, Ivanovo, Russian Federation; Tochigi Cancer Center, Utsunomiya, Japan; Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Institute of Oncology, St. Petersburg, Russian Federation; National Cancer Center Hospital East, Kashiwa, Japan; University Hospital Gasthuisberg, Leuven, Belgium; F. Hoffmann-La Roche Ltd., Basel, Switzerland; Roche Products, Sydney, New South Wales, Australia; Yonsei Cancer Center, Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
11
|
Iveson T, Donehower RC, Davidenko I, Tjulandin S, Deptala A, Harrison M, Nirni S, Lakshmaiah K, Thomas A, Jiang Y, Zhu M, Tang R, Anderson A, Dubey S, Oliner KS, Loh E. Rilotumumab in combination with epirubicin, cisplatin, and capecitabine as first-line treatment for gastric or oesophagogastric junction adenocarcinoma: an open-label, dose de-escalation phase 1b study and a double-blind, randomised phase 2 study. Lancet Oncol 2014; 15:1007-18. [PMID: 24965569 DOI: 10.1016/s1470-2045(14)70023-3] [Citation(s) in RCA: 233] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Dysregulation of the hepatocyte growth factor (HGF)/MET pathway promotes tumour growth and metastasis. Rilotumumab is a fully human, monoclonal antibody that neutralises HGF. We aimed to assess the safety, efficacy, biomarkers, and pharmacokinetics of rilotumumab combined with epirubicin, cisplatin, and capecitabine (ECX) in patients with advanced gastric or oesophagogastric junction cancer. METHODS We recruited patients (≥18 years old) with unresectable locally advanced or metastatic gastric or oesophagogastric junction adenocarcinoma, an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, who had not received previous systemic therapy, from 43 sites worldwide. Phase 1b was an open-label, dose de-escalation study to identify a safe dose of rilotumumab (initial dose 15 mg/kg intravenously on day 1) plus ECX (epirubicin 50 mg/m(2) intravenously on day 1, cisplatin 60 mg/m(2) intravenously on day 1, capecitabine 625 mg/m(2) twice a day orally on days 1-21, respectively), administered every 3 weeks. The phase 1b primary endpoint was the incidence of dose-limiting toxicities in all phase 1b patients who received at least one dose of rilotumumab and completed the dose-limiting toxicity assessment window (first cycle of therapy). Phase 2 was a double-blind study that randomly assigned patients (1:1:1) using an interactive voice response system to receive rilotumumab 15 mg/kg, rilotumumab 7·5 mg/kg, or placebo, plus ECX (doses as above), stratified by ECOG performance status and disease extent. The phase 2 primary endpoint was progression-free survival (PFS), analysed by intention to treat. The study is registered with ClinicalTrials.gov, number NCT00719550. FINDINGS Seven of the nine patients enrolled in the phase 1b study received at least one dose of rilotumumab 15 mg/kg, only two of whom had three dose-limiting toxicities: palmar-plantar erythrodysesthesia, cerebral ischaemia, and deep-vein thrombosis. In phase 2, 121 patients were randomly assigned (40 to rilotumumab 15 mg/kg; 42 to rilotumumab 7·5 mg/kg; 39 to placebo). Median PFS was 5·1 months (95% CI 2·9-7·0) in the rilotumumab 15 mg/kg group, 6·8 months (4·5-7·5) in the rilotumumab 7·5 mg/kg group, 5·7 months (4·5-7·0) in both rilotumumab groups combined, and 4·2 months (2·9-4·9) in the placebo group. The hazard ratio for PFS events compared with placebo was 0·69 (80% CI 0·49-0·97; p=0·164) for rilotumumab 15 mg/kg, 0·53 (80% CI 0·38-0·73; p=0·009) for rilotumumab 7·5 mg/kg, and 0·60 (80% CI 0·45-0·79; p=0·016) for combined rilotumumab. Any grade adverse events more common in the combined rilotumumab group than in the placebo group included haematological adverse events (neutropenia in 44 [54%] of 81 patients vs 13 [33%] of 39 patients; anaemia in 32 [40%] vs 11 [28%]; and thrombocytopenia in nine [11%] vs none), peripheral oedema (22 [27%] vs three [8%]), and venous thromboembolism (16 [20%] vs five [13%]). Grade 3-4 adverse events more common with rilotumumab included neutropenia (36 [44%] vs 11 [28%]) and venous thromboembolism (16 [20%] vs four [10%]). Serious adverse events were balanced between groups except for anaemia, which occurred more frequently in the combined rilotumumab group (ten [12%] vs none). INTERPRETATION Rilotumumab plus ECX had no unexpected safety signals and showed greater activity than placebo plus ECX. A phase 3 study of the combination in MET-positive gastric and oesophagogastric junction cancer is in progress. FUNDING Amgen Inc.
Collapse
Affiliation(s)
| | | | - Irina Davidenko
- State Institution of Public Health "Regional Clinical Oncology Dispensary", Krasnodar, Russia
| | | | - Andrzej Deptala
- Central Clinical Hospital MSW, Warsaw Medical University, Warsaw, Poland
| | | | - Somanath Nirni
- Indo-American Cancer Institute and Research Centre, Hyderabad, India
| | | | | | | | - Min Zhu
- Amgen Inc, Thousand Oaks, CA, USA
| | - Rui Tang
- Amgen Inc, Thousand Oaks, CA, USA
| | | | | | | | - Elwyn Loh
- Amgen Inc, South San Francisco, CA, USA
| |
Collapse
|
12
|
Hur H, Paik MJ, Xuan Y, Nguyen DT, Ham IH, Yun J, Cho YK, Lee G, Han SU. Quantitative measurement of organic acids in tissues from gastric cancer patients indicates increased glucose metabolism in gastric cancer. PLoS One 2014; 9:e98581. [PMID: 24911788 PMCID: PMC4049586 DOI: 10.1371/journal.pone.0098581] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/05/2014] [Indexed: 12/17/2022] Open
Abstract
The levels of organic acids representing metabolic pathway end products are important indicators of physiological status, and may be associated with metabolic changes in cancer. The aim of this study is to investigate the levels of organic acids in cancerous and normal tissues from gastric cancer patients and to confirm the role of metabolic alterations in gastric carcinogenesis. Organic acids in normal and cancerous tissues from forty-five patients with gastric adenocarcinoma were investigated by gas chromatography-mass spectrometry in selected ion monitoring mode as methoxime/tert-butyldimethylsilyl derivatives. We analysed the significant differences in the levels of organic acids in normal and cancer tissues and investigated the correlation of these levels in cancer tissues with clinicopathological features. The levels of Krebs cycle components, including α-ketoglutaric acid, succinic acid, fumaric acid, malic acid and oxaloacetic acid, were significantly increased in cancer tissues compared to normal tissues. In addition, the levels of glycolytic products, including pyruvic acid and lactic acid, as well as the levels of ketone bodies, including 3-hydroxybutyric acid, were also significantly increased in cancer tissues compared to normal tissues. The levels of ketone bodies in cancer tissues with differentiated histology and in intestinal-type cancer tissues were significantly increased. The organic acid profiling analysis described here may be a generally useful clinical tool for understanding the complexity of metabolic events in gastric adenocarcinoma, and organic acids may have potential as metabolic markers for the future discovery of diagnostic and therapeutic modalities.
Collapse
Affiliation(s)
- Hoon Hur
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
- Institute for Gastric Cancer Mechanism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Man Jeong Paik
- College of Pharmacy, Suncheon National University, Suncheon, Republic of Korea
| | - Yi Xuan
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Gastric Cancer and Soft Tissue Sarcoma, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
| | - Duc-Toan Nguyen
- National Institute of Drug Quality Control, Hoan Kiem, Ha Noi, Vietnam
| | - In-Hye Ham
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
- Institute for Gastric Cancer Mechanism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jisoo Yun
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
- Institute for Gastric Cancer Mechanism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Yong Kwan Cho
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Gwang Lee
- Institute for Gastric Cancer Mechanism, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Sang-Uk Han
- Department of Surgery, Ajou University School of Medicine, Suwon, Republic of Korea
- Institute for Gastric Cancer Mechanism, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
13
|
Schlesinger-Raab A, Mihaljevic AL, Egert S, Emeny RT, Jauch KW, Kleeff J, Novotny A, Nüssler NC, Rottmann M, Schepp W, Schmitt W, Schubert-Fritschle G, Weber B, Schuhmacher C, Engel J. Level of hospital care and outcome of gastric cancer: a population-based evaluation of the Munich Cancer Registry. J Cancer Res Clin Oncol 2014; 140:789-800. [PMID: 24633864 DOI: 10.1007/s00432-014-1632-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 02/28/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND Gastric cancer accounts for 5 % of cancer deaths. Successful implementation of guideline-recommended treatment procedures should result in population-based outcome improvements despite the still poor prognosis. In this context, the objective of this study was to compare the outcome of gastric cancer by different levels of hospital care. MATERIALS AND METHODS Total of 8,601 patients with invasive gastric cancer documented between 1998 and 2012 by the Munich Cancer Registry were evaluated. Tumour and therapy characteristics and outcome were analysed in regard to five levels of hospital care: three levels were defined for general hospitals (level I-III), while university hospitals and speciality hospitals were grouped as separate classes. Survival was investigated using the Kaplan-Meier-method, computing relative survival, and by multivariate Cox proportional hazard regression. RESULTS The average age differed between 66 years in university hospitals and 75 years in hospitals providing a basic level of care (level I). No survival differences were found for patients treated in different levels of hospital care in 75 % of the patient cohort, namely the M0 patients. A better survival could only be shown for patients with M1 at diagnosis when treated in a university or level III hospital compared to those treated in other hospitals. CONCLUSION The outcome difference of M1 patients is most likely caused by selection effects concerning health status differences and not by processes of health care attributable to level of hospital care. Thus, this study demonstrates and confirms appropriate treatment and care of gastric cancer over all levels of hospital care.
Collapse
Affiliation(s)
- Anne Schlesinger-Raab
- Department of Medical Informatics, Biometry and Epidemiology (IBE), Munich Cancer Registry (MCR) of the Munich Cancer Centre (MCC), Klinikum Großhadern, Ludwig-Maximilians-University (LMU), Marchioninistr. 15, 81377, Munich, Germany,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Su X, Zhan P, Gavine PR, Morgan S, Womack C, Ni X, Shen D, Bang YJ, Im SA, Ho Kim W, Jung EJ, Grabsch HI, Kilgour E. FGFR2 amplification has prognostic significance in gastric cancer: results from a large international multicentre study. Br J Cancer 2014; 110:967-975. [PMID: 24457912 PMCID: PMC3929881 DOI: 10.1038/bjc.2013.802] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 11/05/2013] [Accepted: 12/02/2013] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND In preclinical gastric cancer (GC) models, FGFR2 amplification was associated with increased tumour cell proliferation and survival, and drugs targeting this pathway are now in clinical trials. METHODS FGFR2 FISH was performed on 961 GCs from the United Kingdom, China and Korea, and the relationship with clinicopathological data and overlap with HER2 amplification were analysed. RESULTS The prevalence of FGFR2 amplification was similar between the three cohorts (UK 7.4%, China 4.6% and Korea 4.2%), and intratumoral heterogeneity was observed in 24% of FGFR2 amplified cases. FGFR2 amplification was associated with lymph node metastases (P<0.0001). FGFR2 amplification and polysomy were associated with poor overall survival (OS) in the Korean (OS: 1.83 vs 6.17 years, P=0.0073) and UK (OS: 0.45 vs 1.9 years, P<0.0001) cohorts, and FGFR2 amplification was an independent marker of poor survival in the UK cohort (P=0.0002). Co-amplification of FGFR2 and HER2 was rare, and when high-level amplifications did co-occur these were detected in distinct areas of the tumour. CONCLUSION A similar incidence of FGFR2 amplification was found in Asian and UK GCs and was associated with lymphatic invasion and poor prognosis. This study also shows that HER2 and FGFR2 amplifications are mostly exclusive.
Collapse
Affiliation(s)
- X Su
- AstraZeneca Asia & Emerging Markets, Innovative Medicines, Shanghai, China
| | - P Zhan
- AstraZeneca Asia & Emerging Markets, Innovative Medicines, Shanghai, China
| | - P R Gavine
- AstraZeneca Asia & Emerging Markets, Innovative Medicines, Shanghai, China
| | - S Morgan
- AstraZeneca, Oncology Innovative Medicines, Alderley Park, Macclesfield, UK
| | - C Womack
- AstraZeneca, Oncology Innovative Medicines, Alderley Park, Macclesfield, UK
| | - X Ni
- Department of General Surgery, Renji Hospital, School of Medicine, Jiaotong University, Shanghai, China
| | - D Shen
- Department of General Surgery, Renji Hospital, School of Medicine, Jiaotong University, Shanghai, China
| | - Y-J Bang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - S-A Im
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - W Ho Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - E-J Jung
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - H I Grabsch
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - E Kilgour
- AstraZeneca, Oncology Innovative Medicines, Alderley Park, Macclesfield, UK
| |
Collapse
|
15
|
Yao Z, Guo H, Yuan Y, Zhao Y, Yao S, Xu Y, Liu L, Liu T, Liu Y, Yang S. Retrospective analysis of docetaxel, oxaliplatin plus fluorouracil compared with epirubicin, cisplatin and fluorouracil as first-line therapy for advanced gastric cancer. J Chemother 2013; 26:117-21. [DOI: 10.1179/1973947813y.0000000120] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
16
|
Stenholm L, Stoehlmacher-Williams J, Al-Batran SE, Heussen N, Akin S, Pauligk C, Lehmann S, Senff T, Hofheinz RD, Ehninger G, Kramer M, Goekkurt E. Prognostic role of microRNA polymorphisms in advanced gastric cancer: a translational study of the Arbeitsgemeinschaft Internistische Onkologie (AIO). Ann Oncol 2013; 24:2581-2588. [PMID: 23975664 DOI: 10.1093/annonc/mdt330] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND To determine the prognostic role of selected microRNA (miRNA) polymorphisms in advanced gastric cancer (AGC). PATIENTS AND METHODS Six hundred and seventy-four AGC patients received 5-fluorouracil (F), leucovorin (L), oxaliplatin (O) or FL + cisplatin (P) or additional docetaxel (T) to FLO (FLOT) within four clinical trials. Polymorphisms of mir-26a1 (rs7372209), mir-27a (rs895819), mir-100 (rs1834306), mir-146a (rs2910164), mir-196-a2 (rs11614913), mir-219-1 (rs107822) and mir-423 (rs6505162) were genotyped. Variable selection for the final multivariate model (n = 487) was based on univariate and multivariate Cox-regression analyses with a cut-off P-value of ≤ 20%. RESULTS Genetic factors significantly associated with overall survival (OS) were rs7372209 (mir-26a1) variant genotypes (hazard ratio, HR 1.307 [95% confidence interval (CI) 1.031-1.656], P = 0.0272), rs895819 (mir-27a) variant genotypes (HR 1.304 [95% CI 1.031-1.650], P = 0.0270) and rs11614913 (mir-196a2) variant genotypes (HR 0.791 [95% CI 0.625-1.000], P = 0.0497). Clinical factors with significant impact on OS were Eastern Cooperative Oncology Group (ECOG) 2 performance status (HR 1.880 [95% CI 1.254-2.820], P = 0.0023), curative surgery of advanced disease (HR 0.235 [95% CI 0.123-0.449], P < 0.0001) and addition of docetaxel in locally AGC patients (HR 0.348 [95% CI 0.145-0.838], P = 0.0301). Combined analyses revealed an improved OS in patients without any unfavourable genotype of 18 months compared with 14, 12 and 10 months in patients with 1, 2 and 3 unfavourable genotypes, respectively (P = 0.0257). CONCLUSIONS These data suggest a significant impact of selected miRNA polymorphisms on prognosis in AGC.
Collapse
Affiliation(s)
- L Stenholm
- Department of Oncology, Haematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Aachen
| | - J Stoehlmacher-Williams
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, University of Dresden, Dresden
| | - S E Al-Batran
- Krankenhaus Nordwest, UCT-University Cancer Center, Frankfurt
| | - N Heussen
- Department of Medical Statistics, RWTH Aachen University, Aachen
| | - S Akin
- Department of Oncology, Haematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Aachen
| | - C Pauligk
- Krankenhaus Nordwest, UCT-University Cancer Center, Frankfurt
| | - S Lehmann
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, University of Dresden, Dresden
| | - T Senff
- Department of Oncology, Haematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Aachen
| | - R D Hofheinz
- III Medical Clinic, University Hospital Mannheim, University of Heidelberg, Mannheim
| | - G Ehninger
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, University of Dresden, Dresden
| | - M Kramer
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, University of Dresden, Dresden
| | - E Goekkurt
- Department of Oncology, Haematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Aachen; Haematologisch-Onkologische Praxis Eppendorf, Hamburg, Germany.
| |
Collapse
|
17
|
|
18
|
Bayrak M, Olmez OF, Kurt E, Cubukcu E, Evrensel T, Kanat O, Manavoglu O. Prognostic significance of c-erbB2 overexpression in patients with metastatic gastric cancer. Clin Transl Oncol 2013; 15:307-312. [PMID: 22911549 DOI: 10.1007/s12094-012-0921-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 07/19/2012] [Indexed: 12/18/2022]
Abstract
BACKGROUND Overexpression of the gene c-erbB2, which encodes a receptor tyrosine kinase, has been associated with prognosis and response to therapy in several solid tumors. This study was designed to test whether c-erb-B2 overexpression can be related to prognosis of patients with metastatic gastric cancer. METHODS Between 2005 and 2010, 46 cases of metastatic gastric cancer were evaluated immunohistochemically for c-erb-B2 overexpression. Overall survival (OS) and time-to-progression (TTP) served as the main outcome measures. RESULTS c-erbB2 was overexpressed in 19 (41.3 %) cases and 8 patients (17.4 %) had a c-erbB2 score of 3+ (a strong complete membrane staining observed in >10 % of the tumor cells). c-erbB2 expression was not associated with the clinicohistological characteristics of the study participants. The mean OS was 11.48 ± 1.03 months, whereas the mean TTP was 8.28 ± 0.8 months. Compared with patients with a score of 2+ or less (n = 38), those with a c-erbB2 score of 3+ (n = 8) had both a significantly lower OS (15.55 ± 1.63 vs. 8.22 ± 0.88 months, respectively, p < 0.05) and TTP (10.72 ± 1.81 vs. 6.11 ± 0.61 months, respectively, p < 0.05). After allowance for potential confounders, Cox regression analysis identified a c-erbB2 score of 3+ as an independent predictor of both OS (hazard ratio = 1.9; 95 % confidence interval = 1.1-3.7, p < 0.05) and TTP (hazard ratio = 1.8; 95 % confidence interval = 1.1-4.1, p < 0.05). CONCLUSION Our results suggest that c-erbB-2 overexpression may have a prognostic significance in patients with metastatic gastric cancer.
Collapse
Affiliation(s)
- Muharrem Bayrak
- Department of Medical Oncology, Uludag University Medical School, Bursa 16059, Turkey
| | | | | | | | | | | | | |
Collapse
|
19
|
Kanat O, O'Neil BH. Metastatic gastric cancer treatment: a little slow but worthy progress. Med Oncol 2013; 30:464. [PMID: 23335104 DOI: 10.1007/s12032-013-0464-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Accepted: 01/09/2013] [Indexed: 02/07/2023]
Abstract
Metastatic gastric cancer is incurable and remains one of the leading causes of cancer-related deaths around the world. Despite the significant progress in its systemic treatment, metastatic gastric cancer is still a major therapeutic challenge for oncologists. Newer chemotherapy regimens and the addition of molecularly targeted agents to chemotherapy seem to provide better clinical outcomes for patients with metastatic gastric cancer. The objective of this article is to review the current treatment approach for this formidable disease.
Collapse
Affiliation(s)
- Ozkan Kanat
- Faculty of Medicine, Department of Medical Oncology, Uludag University, Bursa, Turkey.
| | | |
Collapse
|
20
|
Cimino GD, Pan CX, Henderson PT. Personalized medicine for targeted and platinum-based chemotherapy of lung and bladder cancer. Bioanalysis 2013; 5:369-91. [PMID: 23394702 PMCID: PMC3644565 DOI: 10.4155/bio.12.325] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The personalized medicine revolution is occurring for cancer chemotherapy. Biomarkers are increasingly capable of distinguishing genotypic or phenotypic traits of individual tumors, and are being linked to the selection of treatment protocols. This review covers the molecular basis for biomarkers of response to targeted and cytotoxic lung and bladder cancer treatment with an emphasis on platinum-based chemotherapy. Platinum derivatives are a class of drugs commonly employed against solid tumors that kill cells by covalent attachment to DNA. Platinum-DNA adduct levels in patient tissues have been correlated to response and survival. The sensitivity and precision of adduct detection has increased to the point of enabling subtherapeutic dosing for diagnostics applications, termed diagnostic microdosing, prior to the initiation of full-dose therapy. The clinical status of this unique phenotypic marker for lung and bladder cancer applications is detailed along with discussion of future applications.
Collapse
Affiliation(s)
- George D Cimino
- Accelerated Medical Diagnostics, Inc., 2121 Second Street, B101, Davis, CA 95618, USA
| | - Chong-xian Pan
- University of California Davis, Department of Internal Medicine, Division of Hematology & Oncology & the UC Davis Comprehensive Cancer Center, 4501 X Street, Suite 3016, Sacramento, CA 94568, USA
- Hematology/Oncology, VA Northern California Health Care System, 10535 Hospital Way, Mather, CA 95655, USA
| | - Paul T Henderson
- Accelerated Medical Diagnostics, Inc., 2121 Second Street, B101, Davis, CA 95618, USA
- University of California Davis, Department of Internal Medicine, Division of Hematology & Oncology & the UC Davis Comprehensive Cancer Center, 4501 X Street, Suite 3016, Sacramento, CA 94568, USA
| |
Collapse
|
21
|
Shridhar R, Almhanna K, Hoffe SE, Fulp W, Weber J, Chuong MD, Meredith KL. Increased survival associated with surgery and radiation therapy in metastatic gastric cancer: a Surveillance, Epidemiology, and End Results database analysis. Cancer 2013; 119:1636-42. [PMID: 23361968 DOI: 10.1002/cncr.27927] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 11/13/2012] [Accepted: 11/13/2012] [Indexed: 12/27/2022]
Abstract
BACKGROUND Patients with metastatic gastric cancer have poor survival. The purpose of this study was to compare outcomes of metastatic gastric cancer patients stratified by surgery and radiation therapy. METHODS The Surveillance, Epidemiology, and End Results (SEER) database was accessed to identify patients with AJCC M1 stage IV gastric cancer (based on the American Joint Committee on Cancer Cancer Staging Manual, 6th edition) between 2004 thru 2008. Patients were divided into 4 groups: group 1, no surgery or radiation; group 2, radiation alone; group 3, surgery alone; group 4, surgery and radiation. Survival analysis was determined by Kaplan-Meier and log-rank analysis. Multivariate analysis (MVA) was analyzed by the Cox proportional hazard ratio model. RESULTS A total of 5072 patients were identified. Surgery and/or radiation were associated with a survival benefit. Median and 2-year survival for groups 1, 2, 3, and 4 was 7 months and 8.2%, 8 months and 8.9%, 10 months and 18.2%, and 16 months and 31.7%, respectively (P < .00001). MVA for all patients revealed that surgery and radiation were associated with decreased mortality whereas T-stage, N-stage, age, signet ring histology, and peritoneal metastases were associated with increased mortality. In patients treated with surgery, MVA showed that radiation was associated with decreased mortality, whereas T-stage, N-stage, age, removal of < 15 lymph nodes, signet ring histology, and peritoneal metastases was associated with increased mortality. Age was the only prognostic factor in patients who did not undergo surgery. CONCLUSIONS Surgery and radiation are associated with increased survival in a subset of patients with metastatic gastric cancer. Prospective trials will be needed to address the role and sequence of surgery and radiation in metastatic gastric cancer.
Collapse
Affiliation(s)
- Ravi Shridhar
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida 33612, USA.
| | | | | | | | | | | | | |
Collapse
|
22
|
Lee MJ, Hwang IG, Jang JS, Choi JH, Park BB, Chang MH, Kim ST, Park SH, Kang MH, Kang JH. Outcomes of third-line docetaxel-based chemotherapy in advanced gastric cancer who failed previous oxaliplatin-based and irinotecan-based chemotherapies. Cancer Res Treat 2012; 44:235-41. [PMID: 23341787 PMCID: PMC3546270 DOI: 10.4143/crt.2012.44.4.235] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 08/23/2012] [Indexed: 12/15/2022] Open
Abstract
Purpose Little is known about outcomes in the use of third-line chemotherapy in cases of advanced gastric cancer (AGC). The primary aim of this retrospective study was to evaluate outcomes of docetaxel-based chemotherapy in patients with AGC that progressed after both oxaliplatin-based and irinotecan-based regimens. Materials and Methods Eligible patients were those with AGC who had previous chemotherapy including fluoropyrimidine and oxaliplatin as well as fluoropyrimidine and irinotecan and who received subsequent docetaxel-based chemotherapy. Thirty-five patients were retrospectively recruited from 5 medical centers in Korea. Patients received either weekly or 3 weekly with docetaxel +/- cisplatin. Results Thirty-one out of 35 patients were evaluated for treatment response. A total of 94 cycles of chemotherapy (median, 2; range, 1 to 7) were administered. The overall response rate was 14.3%, and the disease control rate was 45.7%. The median progression-free survival (PFS) was 1.9 months (95% confidence interval [CI], 1.1 to 2.7 months). The median overall survival (OS) was 3.6 months (95% CI, 2.8 to 4.4 months). PFS and OS were significantly prolonged in patients of the Eastern Cooperative Oncology Group, with performance status of 0 or 1 in multivariate analysis (PFS: hazard ratio[HR], 0.411; 95% CI, 0.195 to 0.868; p=0.020 and OS: HR, 0.390; 95% CI, 0.184 to 0.826; p=0.014, respectively). Four of the 35 patients enrolled in the study died due to infection associated with neutropenia. Conclusion Our findings suggest that salvage docetaxel-based chemotherapy is a feasible treatment option for AGC patients with good performance status (PS), whereas chemotherapy for patients with poor PS (PS≤2) should be undertaken with caution for those who previously failed oxaliplatin- and irinotecan-based regimens.
Collapse
Affiliation(s)
- Min Jeong Lee
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Human diseases can be caused by complex mechanisms involving aberrations in numerous proteins and pathways. With recent advances in genomics, elucidating the molecular basis of disease on a personalized level has become an attainable goal. In many cases, relevant molecular targets will be identified for which approved drugs already exist, and the potential repositioning of these drugs to a new indication can be investigated. Repositioning is an accelerated route for drug discovery because existing drugs have established clinical and pharmacokinetic data. Personalized medicine and repositioning both aim to improve the productivity of current drug discovery pipelines, which expend enormous time and cost to develop new drugs, only to have them fail in clinical trials because of lack of efficacy or toxicity. Here, we discuss the current state of research in these two fields, focusing on recent large-scale efforts to systematically find repositioning candidates and elucidate individual disease mechanisms in cancer. We also discuss scenarios in which personalized drug repositioning could be particularly rewarding, such as for diseases that are rare or have specific mutations, as well as current challenges in this field. With an increasing number of drugs being approved for rare cancer subtypes, personalized medicine and repositioning approaches are poised to significantly alter the way we diagnose diseases, infer treatments and develop new drugs.
Collapse
Affiliation(s)
- Yvonne Y Li
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| | - Steven Jm Jones
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia V5Z 4S6, Canada
| |
Collapse
|