101
|
Shah MA, Wainberg ZA, Catenacci DVT, Hochster HS, Ford J, Kunz P, Lee FC, Kallender H, Cecchi F, Rabe DC, Keer H, Martin AM, Liu Y, Gagnon R, Bonate P, Liu L, Gilmer T, Bottaro DP. Phase II study evaluating 2 dosing schedules of oral foretinib (GSK1363089), cMET/VEGFR2 inhibitor, in patients with metastatic gastric cancer. PLoS One 2013; 8:e54014. [PMID: 23516391 PMCID: PMC3597709 DOI: 10.1371/journal.pone.0054014] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 12/05/2012] [Indexed: 01/01/2023] Open
Abstract
PURPOSE The receptors for hepatocyte and vascular endothelial cell growth factors (MET and VEGFR2, respectively) are critical oncogenic mediators in gastric adenocarcinoma. The purpose is to examine the safety and efficacy of foretinib, an oral multikinase inhibitor targeting MET, RON, AXL, TIE-2, and VEGFR2 receptors, for the treatment of metastatic gastric adenocarcinoma. PATIENTS AND METHODS Foretinib safety and tolerability, and objective response rate (ORR) were evaluated in patients using intermittent (240 mg/day, for 5 days every 2 weeks) or daily (80 mg/day) dosing schedules. Thirty evaluable patients were required to achieve alpha = 0.10 and beta = 0.2 to test the alternative hypothesis that single-agent foretinib would result in an ORR of ≥ 25%. Up to 10 additional patients could be enrolled to ensure at least eight with MET amplification. Correlative studies included tumor MET amplification, MET signaling, pharmacokinetics and plasma biomarkers of foretinib activity. RESULTS From March 2007 until October 2009, 74 patients were enrolled; 74% male; median age, 61 years (range, 25-88); 93% had received prior therapy. Best response was stable disease (SD) in 10 (23%) patients receiving intermittent dosing and five (20%) receiving daily dosing; SD duration was 1.9-7.2 months (median 3.2 months). Of 67 patients with tumor samples, 3 had MET amplification, one of whom had SD. Treatment-related adverse events occurred in 91% of patients. Rates of hypertension (35% vs. 15%) and elevated aspartate aminotransferase (23% vs. 8%) were higher with intermittent dosing. In both patients with high baseline tumor phospho-MET (pMET), the pMET:total MET protein ratio decreased with foretinib treatment. CONCLUSION These results indicate that few gastric carcinomas are driven solely by MET and VEGFR2, and underscore the diverse molecular oncogenesis of this disease. Despite evidence of MET inhibition by foretinib, single-agent foretinib lacked efficacy in unselected patients with metastatic gastric cancer.
Collapse
Affiliation(s)
- Manish A Shah
- The Weill Cornell Medical College/New York-Presbyterian Hospital, New York, New York, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
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
|
103
|
Pan L, He M, Ma J, Tang W, Gao G, He R, Su H, Cui D. Phase and size controllable synthesis of NaYbF4 nanocrystals in oleic acid/ionic liquid two-phase system for targeted fluorescent imaging of gastric cancer. Theranostics 2013; 3:210-22. [PMID: 23471455 PMCID: PMC3590590 DOI: 10.7150/thno.5298] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 11/21/2012] [Indexed: 12/28/2022] Open
Abstract
Upconversion nanocrystals with small size and strong fluorescent signals own great potential in applications such as biomolecule-labeling, in vivo tracking and molecular imaging. Herein we reported that NaYbF4: 25%Gd, 2%Tm upconversion nanocrystals with small size and strong fluorescent signals were controllably synthesized by oleic acid (OA)/ ionic liquid (IL) two-phase system for targeted fluorescent imaging of gastric cancer in vivo. The optimal synthesis condition of NaYbF4: 25%Gd, 2%Tm upconversion nanocrystals by OA/IL two-phase system was established, adding more metal ion such as Na(+) ion could facilitate the size control and crystal-phase transition, more importantly, markedly enhancing fluorescent intensity of beta-phase nanocrystals compared with traditional methods. Alpha-phase NaYbF4, 2%Tm upconversion nanocrystals with less than 10nm in diameter and beta-phase NaYbF4: 25%Gd, 2%Tm upconversion nanocrystals with 30 nm or so in diameter and strong fluorescent signals were obtained, these synthesized nanocrystals exhibited very low cytotoxicity. Folic acid-conjugated silica-modified beta-phase NaYbF4: 25%Gd, 2%Tm upconversion nanocrystals were prepared, could actively target gastric cancer tissues implanted into nude mice in vivo, and realized targeted fluorescent imaging. Folic acid-conjugated silica-modified NaYbF4: 25%Gd, 2%Tm upconversion nanocrystals show great potential in applications such as targeted near infared radiation fluorescent imaging, magnetic resonance imaging and targeted therapy of gastric cancer in the near future.
Collapse
|
104
|
Wang YY, Zhang W, Qian S, Liu R, Kan ZX, Wang JH. The effect of locoregional transarterial infusion chemotherapy on liver metastasis after gastric cancer resection. J Int Med Res 2013; 40:1141-8. [PMID: 22906288 DOI: 10.1177/147323001204000334] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE A retrospective clinical study to evaluate the effect of transarterial infusion (TAI) chemotherapy and transarterial chemoembolization (TACE) on liver metastasis after gastric cancer resection. METHODS The study recruited patients who underwent surgical resection for stage T(2)N(2)M(0) gastric adenocarcinoma without liver metastasis. Patients chose to receive either TAI or systemic chemotherapy, based on the advantages and disadvantages of each regimen as explained by physicians. Both regimens comprised 100 mg/m2 oxaliplatin and 500 mg/m2 fluorodeoxy uridine, administered via TAI or a peripheral vein, commencing 30 days postsurgery. Patients who developed liver metastasis during the 3-year follow-up period were offered TACE. RESULTS Mean time from gastric cancer diagnosis to liver metastasis was significantly longer in the TAI group (n = 13) than in the systemic chemotherapy group (n = 29); 944 ± 231 days versus 506 ± 77 days. Patients who received TACE (n = 32) had a partial remission rate of 46.9% and a median survival of 14.7 months after diagnosis of liver metastasis. CONCLUSIONS Locoregional TAI chemotherapy was more effective in reducing liver metastasis after gastric cancer resection than conventional systemic chemotherapy. TACE is an effective treatment for liver metastasis.
Collapse
Affiliation(s)
- Y Y Wang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
105
|
Ando T, Hosokawa A, Kajiura S, Itaya Y, Ueda A, Fujinami H, Nishikawa J, Kobayashi T, Horikawa N, Tsukioka Y, Yabushita K, Note M, Ogawa K, Sugiyama T. Efficacy of weekly paclitaxel in patients with advanced gastric cancer refractory to docetaxel-based chemotherapy. Gastric Cancer 2012; 15:427-32. [PMID: 22252156 DOI: 10.1007/s10120-011-0135-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Accepted: 12/14/2011] [Indexed: 02/07/2023]
Abstract
BACKGROUND Only partial cross-resistance between docetaxel and paclitaxel has been demonstrated in breast and ovarian cancers. Whether weekly paclitaxel is effective in patients with advanced gastric cancer refractory to docetaxel-based chemotherapy remains unclear, and we aimed to clarify the efficacy and safety of weekly paclitaxel in such patients. METHODS Patients who had received docetaxel-based regimens were assigned to the prior-docetaxel group, and those who had never received docetaxel were designated as the non-docetaxel group. Paclitaxel at 80 mg/m(2) was administered by intravenous infusion in all patients, and this was repeated weekly for 3 weeks out of 4. RESULTS Between April 2006 and June 2011, 65 patients were studied: 26 in the prior-docetaxel group and 39 patients were non-docetaxel group. The median age, gender, performance status, histological type, history of gastrectomy, and the locations and numbers of metastatic sites did not differ significantly between the two groups. In the prior-docetaxel group, the response rate (RR) was 14.2% (3/21) among patients with measurable lesions, median progression-free survival (PFS) was 79 days [95% confidence interval (CI), 47-135 days], and overall survival (OS) was 123 days (95% CI, 90-215 days) from the initiation of paclitaxel treatment. In the non-docetaxel group, the RR was 11.5% (3/26) among patients with measurable lesions, PFS was 82 days (95% CI, 52-106 days), and OS was 143 days (95% CI, 121-178 days). The efficacy of weekly paclitaxel thus appeared to be similar in the two groups. CONCLUSIONS Weekly paclitaxel was modestly active in patients with gastric cancer refractory to docetaxel-based chemotherapy.
Collapse
Affiliation(s)
- Takayuki Ando
- Department of Gastroenterology and Hematology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
The kallikrein-related peptidase 13 (KLK13) gene is substantially up-regulated after exposure of gastric cancer cells to antineoplastic agents. Tumour Biol 2012; 33:2069-78. [DOI: 10.1007/s13277-012-0466-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 07/11/2012] [Indexed: 01/07/2023] Open
|
107
|
Abstract
Gastric cancer (GC) is currently the second leading cause of cancer death worldwide; unfortunately, most patients will present with locally advanced or metastatic disease. Despite recent progress in diagnosis, surgery, chemotherapy, and radiotherapy, prognosis remains poor. A better understanding of GC biology and signaling pathways is expected to improve GC therapy, and the integration of targeted therapies has recently become possible and appears to be promising. This article focuses on anti-Her-2 therapy, specifically trastuzumab, as well as other epidermal growth factor receptor antagonists such as cetuximab, panitumub, matuzumab, nimotzumab, gefitinib, and erlotinib. Additionally, drugs that target angiogenesis pathways are also under investigation, particulary bevacizumab, ramucirumab, sorafenib, sunitinib, and cediranib. Other targeted agents in preclinical or early clinical development include mTOR inhibitors, anti c-MET, polo-like kinase 1 inhibitors, anti-insulin-like growth factor, anti-heat shock proteins, and small molecules targeting Hedgehog signaling.
Collapse
Affiliation(s)
- Judith Meza-Junco
- Department of Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada
| | | |
Collapse
|
108
|
Ruan J, Ji J, Song H, Qian Q, Wang K, Wang C, Cui D. Fluorescent magnetic nanoparticle-labeled mesenchymal stem cells for targeted imaging and hyperthermia therapy of in vivo gastric cancer. NANOSCALE RESEARCH LETTERS 2012; 7:309. [PMID: 22709686 PMCID: PMC3441509 DOI: 10.1186/1556-276x-7-309] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 05/31/2012] [Indexed: 05/18/2023]
Abstract
How to find early gastric cancer cells in vivo is a great challenge for the diagnosis and therapy of gastric cancer. This study is aimed at investigating the feasibility of using fluorescent magnetic nanoparticle (FMNP)-labeled mesenchymal stem cells (MSCs) to realize targeted imaging and hyperthermia therapy of in vivo gastric cancer. The primary cultured mouse marrow MSCs were labeled with amino-modified FMNPs then intravenously injected into mouse model with subcutaneous gastric tumor, and then, the in vivo distribution of FMNP-labeled MSCs was observed by using fluorescence imaging system and magnetic resonance imaging system. After FMNP-labeled MSCs arrived in local tumor tissues, subcutaneous tumor tissues in nude mice were treated under external alternating magnetic field. The possible mechanism of MSCs targeting gastric cancer was investigated by using a micro-multiwell chemotaxis chamber assay. Results show that MSCs were labeled with FMNPs efficiently and kept stable fluorescent signal and magnetic properties within 14 days, FMNP-labeled MSCs could target and image in vivo gastric cancer cells after being intravenously injected for 14 days, FMNP-labeled MSCs could significantly inhibit the growth of in vivo gastric cancer because of hyperthermia effects, and CCL19/CCR7 and CXCL12/CXCR4 axis loops may play key roles in the targeting of MSCs to in vivo gastric cancer. In conclusion, FMNP-labeled MSCs could target in vivo gastric cancer cells and have great potential in applications such as imaging, diagnosis, and hyperthermia therapy of early gastric cancer in the near future.
Collapse
Affiliation(s)
- Jing Ruan
- Department of Bio-Nano Science and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Institute of Micro-Nano Science and Technology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Jiajia Ji
- Department of Bio-Nano Science and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Institute of Micro-Nano Science and Technology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Hua Song
- Department of Bio-Nano Science and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Institute of Micro-Nano Science and Technology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Qirong Qian
- Department of Orthopedics, Changzheng Hospital affiliated to Second Military Medical University, 451Fengyang Road, Shanghai, 200003, People's Republic of China
| | - Kan Wang
- Department of Bio-Nano Science and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Institute of Micro-Nano Science and Technology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Can Wang
- Department of Bio-Nano Science and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Institute of Micro-Nano Science and Technology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Daxiang Cui
- Department of Bio-Nano Science and Engineering, Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Institute of Micro-Nano Science and Technology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| |
Collapse
|
109
|
Fox SB, Kumarasinghe MP, Armes JE, Bilous M, Cummings MC, Farshid G, Fitzpatrick N, Francis GD, McCloud PI, Raymond W, Morey A. Gastric HER2 Testing Study (GaTHER): an evaluation of gastric/gastroesophageal junction cancer testing accuracy in Australia. Am J Surg Pathol 2012; 36:577-82. [PMID: 22314190 DOI: 10.1097/pas.0b013e318244adbb] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Trastuzumab provides a survival benefit in patients with human epidermal growth factor receptor 2 (HER2)-amplified/overexpressed advanced gastric and gastroesophageal junction cancers (GC/GJCs). However, the optimal method for testing is unclear. The aim of this study was to assess interlaboratory agreement on HER2 scoring in GC/GJC to aid the development of a robust testing algorithm for diagnostic practice in Australia. Nine laboratories assessed the HER2 status of 100 GC/GJC tissue samples by immunohistochemistry (IHC) and in situ hybridization (ISH) [chromogenic (CISH) or silver (SISH)] using both HER2 copy number and HER2:chr17 (chromosome 17) ratio. Results were compared with reference fluorescence ISH (FISH). Interlaboratory agreement on IHC3+ scoring was good (κ=0.76), and there was good/very good agreement between IHC (positivity defined as IHC3+) and ISH when HER2 copy number was used (κ=0.72 to 0.87). Agreement on CISH/SISH scoring was good/very good when HER2 copy number was used (κ=0.68 to 0.86), and agreement between CISH/SISH and FISH using HER2 copy number was very good (κ=0.88 to 0.91). Agreement was reduced when HER2:chr17 ratio was used. The good agreement for HER2 copy number determined by bright-field ISH suggests that this is the optimal method for testing in GC/GJC cases. An IHC3+ score was strongly predictive of a positive ISH result, although agreement for all IHC scores was only moderate, suggesting that IHC triage before ISH testing would be the most cost-effective strategy. However, because of the unique features of GC/GJC samples and the difficulty of ensuring consistent HER2 staining in the community setting, it is recommended that HER2 status in advanced GC/GJC be determined by both IHC and ISH in the same laboratory.
Collapse
Affiliation(s)
- Stephen B Fox
- Peter MacCallum Cancer Centre, Melbourne, Vic., Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Price TJ, Shapiro JD, Segelov E, Karapetis CS, Pavlakis N, Van Cutsem E, Shah MA, Kang YK, Tebbutt NC. Management of advanced gastric cancer. Expert Rev Gastroenterol Hepatol 2012; 6:199-208; quiz 209. [PMID: 22375525 DOI: 10.1586/egh.11.103] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The management of advanced gastric cancer has only evolved a little over the last 15 years: platinum and fluoropyrimidine chemotherapy remains the backbone of therapy with ongoing debate as to the benefit of triplet therapy with either an anthracycline or taxane. Recently published trials of biological agents, in particular those targeting the Her2 receptor, have provided some signs of improvement. This article summarizes the relevant literature, discusses the role of these agents, as well as geographical variations in use, and provides recommendations regarding both 'standard chemotherapy' and the role of biological agents in advanced gastric cancer. Given the relative lack of progress for gastric cancer over the last 15 years, the focus for the next 5 years should be on an improved understanding of the molecular basis of gastric cancer, thus allowing rational integration of new molecular agents.
Collapse
Affiliation(s)
- Timothy J Price
- Haematology Oncology Unit, The Queen Elizabeth Hospital, Woodville Rd, Woodville, SA 5011, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
111
|
HER2 testing in gastric and gastroesophageal junction cancers: a new therapeutic target and diagnostic challenge. Appl Immunohistochem Mol Morphol 2012; 19:506-8. [PMID: 22089490 DOI: 10.1097/pai.0b013e31822c3a0f] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adenocarcinomas of the stomach and gastroesophageal junction represent a major cause of cancer morbidity and mortality world wide. Complete surgical resection is the mainstay of treatment for nonmetastatic disease; however, many patients are not diagnosed until their disease is either locally advanced or metastatic and therefore unresectable. Clearly, there is an unmet clinical need for new therapeutic strategies, treatment options, and novel therapeutic targets. In a recent trial (Trastuzumab for GAstric cancer), patients assigned to the trastuzumab treatment protocol showed an improved overall survival over those not receiving treatment. Trastuzumab has recently been approved for treatment of advanced gastric and gastroesophageal junction cancers. Pathologists and diagnostic laboratories must be prepared for this new category of specimens requiring human epidermal growth factor receptor 2 testing, and have an awareness of the interpretive differences between breast and gastric cancers.
Collapse
|
112
|
Tafe LJ, Janjigian YY, Zaidinski M, Hedvat CV, Hameed MR, Tang LH, Hicks JB, Shah MA, Barbashina V. Human epidermal growth factor receptor 2 testing in gastroesophageal cancer: correlation between immunohistochemistry and fluorescence in situ hybridization. Arch Pathol Lab Med 2011; 135:1460-5. [PMID: 22032573 DOI: 10.5858/arpa.2010-0541-oa] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
CONTEXT Patients with advanced gastroesophageal cancer have poor survival with current therapy. Human epidermal growth factor receptor 2 (HER2) represents a promising therapeutic target, but the optimal HER2 testing strategy is not yet defined. OBJECTIVES To evaluate the concordance between immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) and to determine if the American Society of Clinical Oncology/College of American Pathologists HER2 scoring system is applicable to gastroesophageal carcinomas. DESIGN Formalin-fixed paraffin-embedded tumor samples from patients with advanced stage gastroesophageal cancer were tested by IHC and FISH and scored according to the American Society of Clinical Oncology/College of American Pathologists criteria for breast cancer. Concordance between IHC and FISH was evaluated. A subset of cases was subjected to array comparative genomic hybridization to verify the positive and negative HER2 results. RESULTS A total of 135 cases with paired IHC and FISH results were evaluated. The majority of samples (84%) were biopsies. HER2 amplification was detected in 20 tumors (15%). Using the American Society of Clinical Oncology/College of American Pathologists scoring system, IHC-FISH concordance was 97% for IHC 0, 93% for IHC 1+, and 100% for IHC 3+. Human epidermal growth factor receptor 2 positivity was strongly associated with tumor grade (moderately differentiated > poorly differentiated, P < .001) and histologic subtype (intestinal > diffuse, P = .007). Array comparative genomic hybridization analysis was successful in 31 tumors (14 FISH+ and 17 FISH-). Fluorescence in situ hybridization and array comparative genomic hybridization results were highly concordant in both HER2-positive and HER2-negative groups (93% and 100% concordance, respectively). CONCLUSIONS Human epidermal growth factor receptor 2 testing in gastroesophageal cancer can be performed using standard breast cancer procedures and the American Society of Clinical Oncology/College of American Pathologists scoring criteria. Although IHC 0 and IHC 3+ provide clear stratification, reliable separation of IHC 1+ and IHC 2+ may be difficult, especially in biopsy samples. The latter 2 groups are best referred to FISH for definitive classification.
Collapse
Affiliation(s)
- Laura J Tafe
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Costa NR, Paulo P, Caffrey T, Hollingsworth MA, Santos-Silva F. Impact of MUC1 mucin downregulation in the phenotypic characteristics of MKN45 gastric carcinoma cell line. PLoS One 2011; 6:e26970. [PMID: 22073229 PMCID: PMC3206881 DOI: 10.1371/journal.pone.0026970] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 10/07/2011] [Indexed: 12/19/2022] Open
Abstract
Background Gastric carcinoma is the second leading cause of cancer-associated death worldwide. The high mortality associated with this disease is in part due to limited knowledge about gastric carcinogenesis and a lack of available therapeutic and prevention strategies. MUC1 is a high molecular weight transmembrane mucin protein expressed at the apical surface of most glandular epithelial cells and a major component of the mucus layer above gastric mucosa. Overexpression of MUC1 is found in approximately 95% of human adenocarcinomas, where it is associated with oncogenic activity. The role of MUC1 in gastric cancer progression remains to be clarified. Methodology We downregulated MUC1 expression in a gastric carcinoma cell line by RNA interference and studied the effects on cellular proliferation (MTT assay), apoptosis (TUNEL assay), migration (migration assay), invasion (invasion assay) and aggregation (aggregation assay). Global gene expression was evaluated by microarray analysis to identify alterations that are regulated by MUC1 expression. In vivo assays were also performed in mice, in order to study the tumorigenicity of cells with and without MUC1 downregulation in MKN45 gastric carcinoma cell line. Results Downregulation of MUC1 expression increased proliferation and apoptosis as compared to controls, whereas cell-cell aggregation was decreased. No significant differences were found in terms of migration and invasion between the downregulated clones and the controls. Expression of TCN1, KLK6, ADAM29, LGAL4, TSPAN8 and SHPS-1 was found to be significantly different between MUC1 downregulated clones and the control cells. In vivo assays have shown that mice injected with MUC1 downregulated cells develop smaller tumours when compared to mice injected with the control cells. Conclusions These results indicate that MUC1 downregulation alters the phenotype and tumorigenicity of MKN45 gastric carcinoma cells and also the expression of several molecules that can be involved in tumorigenic events. Therefore, MUC1 should be further studied to better clarify its potential as a novel therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Natália R. Costa
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Paula Paulo
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Thomas Caffrey
- Eppley Institute for Research in Cancer and Allied Disease, Omaha, Nebraska, United States of America
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer and Allied Disease, Omaha, Nebraska, United States of America
| | - Filipe Santos-Silva
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- Medical Faculty, University of Porto, Porto, Portugal
- * E-mail:
| |
Collapse
|
114
|
Ohtsu A, Shah MA, Van Cutsem E, Rha SY, Sawaki A, Park SR, Lim HY, Yamada Y, Wu J, Langer B, Starnawski M, Kang YK. Bevacizumab in Combination With Chemotherapy As First-Line Therapy in Advanced Gastric Cancer: A Randomized, Double-Blind, Placebo-Controlled Phase III Study. J Clin Oncol 2011; 29:3968-76. [DOI: 10.1200/jco.2011.36.2236] [Citation(s) in RCA: 887] [Impact Index Per Article: 63.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose The Avastin in Gastric Cancer (AVAGAST) trial was a multinational, randomized, placebo-controlled trial designed to evaluate the efficacy of adding bevacizumab to capecitabine-cisplatin in the first-line treatment of advanced gastric cancer. Patients and Methods Patients received bevacizumab 7.5 mg/kg or placebo followed by cisplatin 80 mg/m2 on day 1 plus capecitabine 1,000 mg/m2 twice daily for 14 days every 3 weeks. Fluorouracil was permitted in patients unable to take oral medications. Cisplatin was given for six cycles; capecitabine and bevacizumab were administered until disease progression or unacceptable toxicity. The primary end point was overall survival (OS). Log-rank test was used to test the OS difference. Results In all, 774 patients were enrolled; 387 were assigned to each treatment group (intention-to-treat population), and 517 deaths were observed. Median OS was 12.1 months with bevacizumab plus fluoropyrimidine-cisplatin and 10.1 months with placebo plus fluoropyrimidine-cisplatin (hazard ratio 0.87; 95% CI, 0.73 to 1.03; P = .1002). Both median progression-free survival (6.7 v 5.3 months; hazard ratio, 0.80; 95% CI, 0.68 to 0.93; P = .0037) and overall response rate (46.0% v 37.4%; P = .0315) were significantly improved with bevacizumab versus placebo. Preplanned subgroup analyses revealed regional differences in efficacy outcomes. The most common grade 3 to 5 adverse events were neutropenia (35%, bevacizumab plus fluoropyrimidine-cisplatin; 37%, placebo plus fluoropyrimidine-cisplatin), anemia (10% v 14%), and decreased appetite (8% v 11%). No new bevacizumab-related safety signals were identified. Conclusion Although AVAGAST did not reach its primary objective, adding bevacizumab to chemotherapy was associated with significant increases in progression-free survival and overall response rate in the first-line treatment of advanced gastric cancer.
Collapse
Affiliation(s)
- Atsushi Ohtsu
- Atsushi Ohtsu, National Cancer Center Hospital East, Kashiwa, Chiba; Akira Sawaki, Aichi Cancer Center Hospital, Nagoya; Yasuhide Yamada, National Cancer Center Hospital, Tokyo, Japan; Manish A. Shah, Memorial Sloan-Kettering Cancer Center, New York, NY; Eric Van Cutsem, University Hospital Gasthuisberg, Leuven, Belgium; Sun Young Rha, Yonsei Cancer Center, Yonsei University College of Medicine; Ho Yeong Lim, Samsung Medical Center; Yoon-Koo Kang, Asan Medical Center, University of Ulsan College of
| | - Manish A. Shah
- Atsushi Ohtsu, National Cancer Center Hospital East, Kashiwa, Chiba; Akira Sawaki, Aichi Cancer Center Hospital, Nagoya; Yasuhide Yamada, National Cancer Center Hospital, Tokyo, Japan; Manish A. Shah, Memorial Sloan-Kettering Cancer Center, New York, NY; Eric Van Cutsem, University Hospital Gasthuisberg, Leuven, Belgium; Sun Young Rha, Yonsei Cancer Center, Yonsei University College of Medicine; Ho Yeong Lim, Samsung Medical Center; Yoon-Koo Kang, Asan Medical Center, University of Ulsan College of
| | - Eric Van Cutsem
- Atsushi Ohtsu, National Cancer Center Hospital East, Kashiwa, Chiba; Akira Sawaki, Aichi Cancer Center Hospital, Nagoya; Yasuhide Yamada, National Cancer Center Hospital, Tokyo, Japan; Manish A. Shah, Memorial Sloan-Kettering Cancer Center, New York, NY; Eric Van Cutsem, University Hospital Gasthuisberg, Leuven, Belgium; Sun Young Rha, Yonsei Cancer Center, Yonsei University College of Medicine; Ho Yeong Lim, Samsung Medical Center; Yoon-Koo Kang, Asan Medical Center, University of Ulsan College of
| | - Sun Young Rha
- Atsushi Ohtsu, National Cancer Center Hospital East, Kashiwa, Chiba; Akira Sawaki, Aichi Cancer Center Hospital, Nagoya; Yasuhide Yamada, National Cancer Center Hospital, Tokyo, Japan; Manish A. Shah, Memorial Sloan-Kettering Cancer Center, New York, NY; Eric Van Cutsem, University Hospital Gasthuisberg, Leuven, Belgium; Sun Young Rha, Yonsei Cancer Center, Yonsei University College of Medicine; Ho Yeong Lim, Samsung Medical Center; Yoon-Koo Kang, Asan Medical Center, University of Ulsan College of
| | - Akira Sawaki
- Atsushi Ohtsu, National Cancer Center Hospital East, Kashiwa, Chiba; Akira Sawaki, Aichi Cancer Center Hospital, Nagoya; Yasuhide Yamada, National Cancer Center Hospital, Tokyo, Japan; Manish A. Shah, Memorial Sloan-Kettering Cancer Center, New York, NY; Eric Van Cutsem, University Hospital Gasthuisberg, Leuven, Belgium; Sun Young Rha, Yonsei Cancer Center, Yonsei University College of Medicine; Ho Yeong Lim, Samsung Medical Center; Yoon-Koo Kang, Asan Medical Center, University of Ulsan College of
| | - Sook Ryun Park
- Atsushi Ohtsu, National Cancer Center Hospital East, Kashiwa, Chiba; Akira Sawaki, Aichi Cancer Center Hospital, Nagoya; Yasuhide Yamada, National Cancer Center Hospital, Tokyo, Japan; Manish A. Shah, Memorial Sloan-Kettering Cancer Center, New York, NY; Eric Van Cutsem, University Hospital Gasthuisberg, Leuven, Belgium; Sun Young Rha, Yonsei Cancer Center, Yonsei University College of Medicine; Ho Yeong Lim, Samsung Medical Center; Yoon-Koo Kang, Asan Medical Center, University of Ulsan College of
| | - Ho Yeong Lim
- Atsushi Ohtsu, National Cancer Center Hospital East, Kashiwa, Chiba; Akira Sawaki, Aichi Cancer Center Hospital, Nagoya; Yasuhide Yamada, National Cancer Center Hospital, Tokyo, Japan; Manish A. Shah, Memorial Sloan-Kettering Cancer Center, New York, NY; Eric Van Cutsem, University Hospital Gasthuisberg, Leuven, Belgium; Sun Young Rha, Yonsei Cancer Center, Yonsei University College of Medicine; Ho Yeong Lim, Samsung Medical Center; Yoon-Koo Kang, Asan Medical Center, University of Ulsan College of
| | - Yasuhide Yamada
- Atsushi Ohtsu, National Cancer Center Hospital East, Kashiwa, Chiba; Akira Sawaki, Aichi Cancer Center Hospital, Nagoya; Yasuhide Yamada, National Cancer Center Hospital, Tokyo, Japan; Manish A. Shah, Memorial Sloan-Kettering Cancer Center, New York, NY; Eric Van Cutsem, University Hospital Gasthuisberg, Leuven, Belgium; Sun Young Rha, Yonsei Cancer Center, Yonsei University College of Medicine; Ho Yeong Lim, Samsung Medical Center; Yoon-Koo Kang, Asan Medical Center, University of Ulsan College of
| | - Jian Wu
- Atsushi Ohtsu, National Cancer Center Hospital East, Kashiwa, Chiba; Akira Sawaki, Aichi Cancer Center Hospital, Nagoya; Yasuhide Yamada, National Cancer Center Hospital, Tokyo, Japan; Manish A. Shah, Memorial Sloan-Kettering Cancer Center, New York, NY; Eric Van Cutsem, University Hospital Gasthuisberg, Leuven, Belgium; Sun Young Rha, Yonsei Cancer Center, Yonsei University College of Medicine; Ho Yeong Lim, Samsung Medical Center; Yoon-Koo Kang, Asan Medical Center, University of Ulsan College of
| | - Bernd Langer
- Atsushi Ohtsu, National Cancer Center Hospital East, Kashiwa, Chiba; Akira Sawaki, Aichi Cancer Center Hospital, Nagoya; Yasuhide Yamada, National Cancer Center Hospital, Tokyo, Japan; Manish A. Shah, Memorial Sloan-Kettering Cancer Center, New York, NY; Eric Van Cutsem, University Hospital Gasthuisberg, Leuven, Belgium; Sun Young Rha, Yonsei Cancer Center, Yonsei University College of Medicine; Ho Yeong Lim, Samsung Medical Center; Yoon-Koo Kang, Asan Medical Center, University of Ulsan College of
| | - Michal Starnawski
- Atsushi Ohtsu, National Cancer Center Hospital East, Kashiwa, Chiba; Akira Sawaki, Aichi Cancer Center Hospital, Nagoya; Yasuhide Yamada, National Cancer Center Hospital, Tokyo, Japan; Manish A. Shah, Memorial Sloan-Kettering Cancer Center, New York, NY; Eric Van Cutsem, University Hospital Gasthuisberg, Leuven, Belgium; Sun Young Rha, Yonsei Cancer Center, Yonsei University College of Medicine; Ho Yeong Lim, Samsung Medical Center; Yoon-Koo Kang, Asan Medical Center, University of Ulsan College of
| | - Yoon-Koo Kang
- Atsushi Ohtsu, National Cancer Center Hospital East, Kashiwa, Chiba; Akira Sawaki, Aichi Cancer Center Hospital, Nagoya; Yasuhide Yamada, National Cancer Center Hospital, Tokyo, Japan; Manish A. Shah, Memorial Sloan-Kettering Cancer Center, New York, NY; Eric Van Cutsem, University Hospital Gasthuisberg, Leuven, Belgium; Sun Young Rha, Yonsei Cancer Center, Yonsei University College of Medicine; Ho Yeong Lim, Samsung Medical Center; Yoon-Koo Kang, Asan Medical Center, University of Ulsan College of
| |
Collapse
|
115
|
Ross JS. Update on HER2 testing for breast and upper gastrointestinal tract cancers. Biomark Med 2011; 5:307-18. [PMID: 21657840 DOI: 10.2217/bmm.11.31] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
With the regulatory approvals in Europe and the USA of trastuzumab-based anti-HER2 targeted therapy for upper gastrointestinal cancers in 2010, HER2 testing has now become universal for newly diagnosed cases of both breast cancer and adenocarcinomas of esophagus, stomach and gastroesophageal origin. In the 12 years or more since the approval of trastuzumab for breast cancer, general refinements in approaches to HER2 testing, including a greater understanding of the implications of preanalytic factors impacting the test results and the application of standardization of reporting of HER2 test results, have taken place. There has also been continuing development in breast cancer with the introduction of new HER2 tests, including non-FISH tests, dimerization assays, phosphorylated HER2 receptor tests, mRNA-based tests, HER2 gene sequencing tests and the application of HER2 testing to circulating tumor cells. Most recently, the introduction of HER2 testing for upper gastrointentinal malignancies has emphasized the need for performing and interpreting slide-based assays in a manner unique to these specimens and not to apply the breast cancer testing protocols to esophageal and gastric adenocarcinomas.
Collapse
|
116
|
Koltz BR, Hicks DG, Whitney-Miller CL. HER2 testing in gastric and esophageal adenocarcinoma: new diagnostic challenges arising from new therapeutic options. Biotech Histochem 2011; 87:40-5. [DOI: 10.3109/10520295.2011.591830] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
117
|
Abstract
Despite some advances in the past few years, the search for effective treatment modalities for advanced gastric and gastro-esophageal junction cancer is far from over. Available data clearly demonstrate that the development of new drugs will have little, if any, chance of success if it is not guided by in-depth knowledge of disease biology. However, using biologic agents to target key molecular pathways, such as those regulated by human epidermal growth factor receptor (HER) family members, may be effective. Indeed, the positive results achieved by the anti-HER2 agent trastuzumab in a phase III trial in HER2-positive patients support this approach. Many new anti-HER molecules are now under evaluation for the treatment of gastric and gastro-esophageal junction cancer, but so far attempts to identify reliable predictive factors from phase I and II trials have produced inconclusive results. In addition, large phase III trials are still being conducted in molecularly unselected populations. Refining patient selection is essential to maximize the benefit of targeted agents, to avoid significant toxicities and for the development of alternative therapeutic approaches in patients who have nonresponsive disease.
Collapse
|
118
|
Shah MA, Khanin R, Tang L, Janjigian YY, Klimstra DS, Gerdes H, Kelsen DP. Molecular classification of gastric cancer: a new paradigm. Clin Cancer Res 2011; 17:2693-701. [PMID: 21430069 PMCID: PMC3100216 DOI: 10.1158/1078-0432.ccr-10-2203] [Citation(s) in RCA: 241] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE Gastric cancer may be subdivided into 3 distinct subtypes--proximal, diffuse, and distal gastric cancer--based on histopathologic and anatomic criteria. Each subtype is associated with unique epidemiology. Our aim is to test the hypothesis that these distinct gastric cancer subtypes may also be distinguished by gene expression analysis. EXPERIMENTAL DESIGN Patients with localized gastric adenocarcinoma being screened for a phase II preoperative clinical trial (National Cancer Institute, NCI #5917) underwent endoscopic biopsy for fresh tumor procurement. Four to 6 targeted biopsies of the primary tumor were obtained. Macrodissection was carried out to ensure more than 80% carcinoma in the sample. HG-U133A GeneChip (Affymetrix) was used for cDNA expression analysis, and all arrays were processed and analyzed using the Bioconductor R-package. RESULTS Between November 2003 and January 2006, 57 patients were screened to identify 36 patients with localized gastric cancer who had adequate RNA for expression analysis. Using supervised analysis, we built a classifier to distinguish the 3 gastric cancer subtypes, successfully classifying each into tightly grouped clusters. Leave-one-out cross-validation error was 0.14, suggesting that more than 85% of samples were classified correctly. Gene set analysis with the false discovery rate set at 0.25 identified several pathways that were differentially regulated when comparing each gastric cancer subtype to adjacent normal stomach. CONCLUSIONS Subtypes of gastric cancer that have epidemiologic and histologic distinctions are also distinguished by gene expression data. These preliminary data suggest a new classification of gastric cancer with implications for improving our understanding of disease biology and identification of unique molecular drivers for each gastric cancer subtype.
Collapse
Affiliation(s)
- Manish A Shah
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | | | | | | | | | |
Collapse
|
119
|
Marimuthu A, Jacob HK, Jakharia A, Subbannayya Y, Keerthikumar S, Kashyap MK, Goel R, Balakrishnan L, Dwivedi S, Pathare S, Dikshit JB, Maharudraiah J, Singh S, Sameer Kumar GS, Vijayakumar M, Veerendra Kumar KV, Premalatha CS, Tata P, Hariharan R, Roa JC, Prasad T, Chaerkady R, Kumar RV, Pandey A. Gene Expression Profiling of Gastric Cancer. JOURNAL OF PROTEOMICS & BIOINFORMATICS 2011; 4:74-82. [PMID: 27030788 PMCID: PMC4809432 DOI: pmid/27030788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastric cancer is the second leading cause of cancer death worldwide, both in men and women. A genomewide gene expression analysis was carried out to identify differentially expressed genes in gastric adenocarcinoma tissues as compared to adjacent normal tissues. We used Agilent's whole human genome oligonucleotide microarray platform representing ~41,000 genes to carry out gene expression analysis. Two-color microarray analysis was employed to directly compare the expression of genes between tumor and normal tissues. Through this approach, we identified several previously known candidate genes along with a number of novel candidate genes in gastric cancer. Testican-1 (SPOCK1) was one of the novel molecules that was 10-fold upregulated in tumors. Using tissue microarrays, we validated the expression of testican-1 by immunohistochemical staining. It was overexpressed in 56% (160/282) of the cases tested. Pathway analysis led to the identification of several networks in which SPOCK1 was among the topmost networks of interacting genes. By gene enrichment analysis, we identified several genes involved in cell adhesion and cell proliferation to be significantly upregulated while those corresponding to metabolic pathways were significantly downregulated. The differentially expressed genes identified in this study are candidate biomarkers for gastric adenoacarcinoma.
Collapse
Affiliation(s)
- Arivusudar Marimuthu
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Manipal University, Madhav Nagar, Manipal, Karnataka 576104; India
| | - Harrys K.C. Jacob
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Manipal University, Madhav Nagar, Manipal, Karnataka 576104; India
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore 21205, Maryland, USA
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore 21205, Maryland, USA
| | - Aniruddha Jakharia
- Department of Zoology, Gauhati University, Guwahati 781014, Assam, India
- Imgenex India, Bhubaneswar 751024, Orissa, India
| | - Yashwanth Subbannayya
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Rajiv Gandhi University of Health Sciences, Bangalore, 560041, Karnataka, India
| | | | - Manoj Kumar Kashyap
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Department of Biotechnology, Kuvempu University, Shimoga 577451, Karnataka, India
| | - Renu Goel
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Department of Biotechnology, Kuvempu University, Shimoga 577451, Karnataka, India
| | - Lavanya Balakrishnan
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Department of Biotechnology, Kuvempu University, Shimoga 577451, Karnataka, India
| | - Sutopa Dwivedi
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- School of Biotechnology, Amrita Vishwa Vidyapeetham University, Kollam 690525, Kerala, India
| | | | | | - Jagadeesha Maharudraiah
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- RajaRajeswari Medical college, Bangalore, 560074, India
| | - Sujay Singh
- Imgenex India, Bhubaneswar 751024, Orissa, India
- Imgenex Corporation, San Diego 92121, California, USA
| | - Ghantasala S Sameer Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Department of Biotechnology, Kuvempu University, Shimoga 577451, Karnataka, India
| | - M. Vijayakumar
- Departments of Surgical Oncology, Kidwai Memorial Institute of Oncology, Bangalore 560029, Karnataka; India
| | | | | | - Pramila Tata
- Strand Life Sciences, Bangalore 560024, Karnataka, India
| | | | - Juan Carlos Roa
- Department of Pathology, Universidad de La Frontera, Temuco, Chile
| | - T.S.K Prasad
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Manipal University, Madhav Nagar, Manipal, Karnataka 576104; India
| | - Raghothama Chaerkady
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Manipal University, Madhav Nagar, Manipal, Karnataka 576104; India
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore 21205, Maryland, USA
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore 21205, Maryland, USA
| | - Rekha Vijay Kumar
- Department of Pathology, Kidwai Memorial Institute of Oncology, Bangalore 560029, Karnataka; India
- Corresponding authors: Akhilesh Pandey MD, PhD, McKusick-Nathans Institute of Genetic Medicine, 733 N. Broadway, BRB 527, Johns Hopkins University, Baltimore, MD 21205, Tel: 410-502-6662; Fax: 410-502-7544; , Rekha V. Kumar, MD, Department of Pathology, Kidwai Memorial Institute of Oncology, Bangalore, Karnataka 560029; India. Tel: 091-080-6560708; Fax: 091-080-6560723;
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore 21205, Maryland, USA
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore 21205, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore 21205, Maryland, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore 21205, Maryland, USA
- Corresponding authors: Akhilesh Pandey MD, PhD, McKusick-Nathans Institute of Genetic Medicine, 733 N. Broadway, BRB 527, Johns Hopkins University, Baltimore, MD 21205, Tel: 410-502-6662; Fax: 410-502-7544; , Rekha V. Kumar, MD, Department of Pathology, Kidwai Memorial Institute of Oncology, Bangalore, Karnataka 560029; India. Tel: 091-080-6560708; Fax: 091-080-6560723;
| |
Collapse
|
120
|
Shah MA, Jhawer M, Ilson DH, Lefkowitz RA, Robinson E, Capanu M, Kelsen DP. Phase II study of modified docetaxel, cisplatin, and fluorouracil with bevacizumab in patients with metastatic gastroesophageal adenocarcinoma. J Clin Oncol 2010; 29:868-74. [PMID: 21189380 DOI: 10.1200/jco.2010.32.0770] [Citation(s) in RCA: 157] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE To evaluate the safety and efficacy of a modified administration schedule of docetaxel, cisplatin, and fluorouracil (mDCF) with bevacizumab in patients with advanced gastroesophageal malignancies. PATIENTS AND METHODS Previously untreated patients with metastatic gastroesophageal adenocarcinoma received bevacizumab 10 mg/kg, docetaxel 40 mg/m², fluorouracil 400 mg/m², leucovorin 400 mg/m² on day 1, fluorouracil 1,000 mg/m²/d × 2 days intravenous continuous infusion beginning on day 1, and cisplatin 40 mg/m² on day 3. The primary objective was to improve 6-month progression-free survival (PFS) from 43% (historical DCF control) to 63% with the addition of bevacizumab. The target accrual was 44 patients to have 10% type I and II error rates. RESULTS In total, 44 eligible patients with cancer were enrolled from October 2006 to October 2008: 22 gastric, 20 gastroesophageal junction (GEJ), and two esophagus. In 39 patients with measurable disease, the confirmed response rate was 67% (95% CI, 50% to 81%). Six-month PFS was 79% (95% CI, 63% to 88%), and median PFS was 12 months (95% CI, 8.8 to 18.2 months). With 26-month follow-up, median overall survival (OS) was 16.8 months (95% CI, 12.1 to 26.1 months), and 2-year survival was 37%. Treatment-related grade 3 to 4 toxicity was as follows: neutropenia without fever (50%), fatigue (25%), venous thromboembolism (39%), and nausea, vomiting, mucositis, neuropathy, and febrile neutropenia less than 10% each. In subset analysis, diffuse gastric cancer had significantly worse PFS and OS, and the response rate in proximal/GEJ tumors was 85% (95% CI, 62% to 97%). CONCLUSION mDCF with bevacizumab appears tolerable and has notable patient outcomes in patients with advanced gastroesophageal adenocarcinoma. Six-month PFS was 79%, surpassing our predefined efficacy end point, and median and 2-year OS were 16.8 months and 37%, respectively.
Collapse
Affiliation(s)
- Manish A Shah
- Gastrointestinal Oncology Service, Memorial Sloan-Kettering Cancer Center, and the Weill School of Medicine, Cornell University, New York, NY, USA.
| | | | | | | | | | | | | |
Collapse
|