1
|
Muacevic A, Adler JR, Patel A, Catalano C, Abdelsayed G, Lalos A, Rustgi V. The Spontaneous Regression of Primary Gastrointestinal Malignancies: An Observational Review. Cureus 2022; 14:e32970. [PMID: 36712716 PMCID: PMC9879583 DOI: 10.7759/cureus.32970] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/25/2022] [Indexed: 12/27/2022] Open
Abstract
The spontaneous regression or remission (SR) of cancer, often described as the partial or complete disappearance of a malignant tumor in the absence of all medical treatment and therapy, is a well-documented phenomenon. With efforts ongoing to establish cancer treatments that limit undesirable outcomes and adverse effects, these uncommon occurrences of SR carry significant implications for novel therapies and warrant further investigation. While several case studies have reported instances of SR in gastrointestinal (GI) malignancies, a comprehensive review of previous manifestations of SR in the GI tract remains lacking. The inclusion criteria for the rare phenomenon are also in need of an appropriate update that takes recent scientific advancements and emerging new medical technologies into account. Our analysis of 390 cases of SR in the GI tract focuses primarily on neoplasms of the hepatobiliary system and proposes an updated version of the older inclusion criteria for spontaneous regression.
Collapse
|
2
|
Kohli P, Penumadu P, Srinivas BH, M S, Dubashi B, Kate V, Kumar H, R K, Balasubramanian A. Clinicopathological profile and its association with peritoneal disease among gastric cancer patients. Surg Oncol 2021; 38:101595. [PMID: 33991942 DOI: 10.1016/j.suronc.2021.101595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 03/30/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND There are no clinicopathological criteria or test to predict peritoneal metastasis either in primary or recurrent gastric cancer. The early prediction will help in altering or adding other adjuvant potential therapy modalities like HIPEC and maintenance chemotherapy. METHODS Paraffin based blocks of 110 gastric tumor specimens were subjected to IHC staining to assess VEGF, Her 2 neu, E cadherin, bcl 2 and p 53 expression and its association with peritoneal disease evaluated. RESULTS Her 2 neu uptake was present in 17.3%, bcl-2 expression in 19.1%, P53 expression in 40.9%, VEGF in 41.8% and E cadherin expression in 49.1% patients. On univariate analysis, a younger age(p = .029), female sex(p = .026), positive VEGF expression (p = .001) and p53 expression(p = .015) were significantly associated with peritoneal disease. A binomial logistic regression was performed to ascertain the effects of independent variables evaluated on univariate analysis. Of the 10 predictors variables, only three were statistically significant: tumor type, P53, and VEGF. Positive VEGF expression had 48.7, E cadherin 2.6 and Her2neu 1.5 times higher odds of exhibiting peritoneal disease. CONCLUSION A younger age, female sex, distal 2/3rd, diffuse variant, VEGF staining in >10% cells and decrease p53 expression were associated with peritoneal disease.
Collapse
Affiliation(s)
- Pavneet Kohli
- Department of Surgical Oncology, JIPMER, Puducherry, 6050006, India
| | - Prasanth Penumadu
- Department of Surgical Oncology, JIPMER, Puducherry, 6050006, India.
| | - B H Srinivas
- Department of Pathology, JIPMER, Puducherry, 605006, India
| | - Sivasanker M
- HPB Unit, Department of Surgery, Royal Liverpool University Hospitals NHS Trust, Merseyside, UK
| | - Biswajit Dubashi
- Department of Medical Oncology, JIPMER, Puducherry, 605006, India
| | - Vikram Kate
- Department of General Surgery, JIPMER, Puducherry, 605006, India
| | | | - Kalayarasan R
- Department of Surgical Gastroenterology, JIPMER, Puducherry, 605006, India
| | | |
Collapse
|
3
|
A global and physical mechanism of gastric cancer formation and progression. J Theor Biol 2021; 520:110643. [PMID: 33636204 DOI: 10.1016/j.jtbi.2021.110643] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/26/2020] [Accepted: 02/11/2021] [Indexed: 12/17/2022]
Abstract
Gastric cancer is regarded as a major health issue for human being nowadays. The Helicobacter pylori (H. pylori) infection has been found to accelerate the development of gastritis and gastric cancer. Significant efforts have been made towards the understanding of the biology of gastric cancer on both genetic and epigenetic levels. However the physical mechanism behind the gastric cancer formation is still elusive. In this study, we constructed a model for investigating gastric cancer formation by explored the gastric cancer landscape and the flow flux. We uncovered three stable state attractors on the landscape: normal, gastritis and gastric cancer. The definition of each attractor is based on the biological function and gene expression levels. The global stabilities and the switching processes were quantified through the barrier heights and dominant kinetic paths. To investigate the underlying mechanism of the process from normal through the gastritis to the gastric cancer caused by genetic or epigenetic factors, we simulate the oncogenesis of gastric cancer through changes of several gene regulation strengths and H. pylori infection. The simulated results can illustrate the developmental and metastasis process of gastric cancer. Different H. pylori infection degrees accelerating the process from gastritis to gastric cancer can be quantified. Then we applied global sensitivity analysis, one key gene and four key regulations were found. These results are consist with the experimental results and can be used to design the polygenic anti-cancer agents through multiple key genes or regulations. The landscape approach provides a physical and simple strategy for analyzing gastric cancer in a systematic and quantitative way. It also offers new insight into treatment strategy for gastric cancer by adjusting relevant polygenic genes and regulations.
Collapse
|
4
|
Oh HS, Eom DW, Kang GH, Ahn YC, Lee SJ, Kim JH, Jang HJ, Kim EJ, Oh KH, Ahn HJ. Prognostic implications of EGFR and HER-2 alteration assessed by immunohistochemistry and silver in situ hybridization in gastric cancer patients following curative resection. Gastric Cancer 2015; 17:402-11. [PMID: 23955257 DOI: 10.1007/s10120-013-0288-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 07/28/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND The aim of this study was to use immunohistochemistry (IHC) and silver in situ hybridization (SISH) to evaluate alterations in EGFR and HER2 in gastric cancer in order to determine the relationship with prognosis in gastric cancer patients following curative resection. PATIENTS AND METHODS In this study, we analyzed EGFR and HER-2 status by IHC and SISH in 254 stage I-III gastric cancer patients who underwent curative surgery. RESULTS Thirteen cases (2.48 %) showed EGFR alteration by IHC or SISH. EGFR alteration was associated with older age (P = 0.021), intestinal type (P = 0.040) and higher stage disease (P < 0.001). The patients with operable state gastric cancer who had EGFR alteration had an unfavorable prognosis, and multivariate analysis confirmed that EGFR alteration was an independent unfavorable prognostic factor. Twenty-seven cases (10.6 %) showed HER-2 alteration by IHC or SISH. HER-2 alteration was associated with older age (P = 0.006), well or moderately differentiated histology (P < 0.001) and intestinal type (P = 0.002). CONCLUSION HER-2 alteration is not an independent prognostic factor for curatively resectable gastric cancer. We observed EGFR alteration in a subset of cases with operable state gastric cancer and determined that it was associated with an unfavorable prognosis.
Collapse
Affiliation(s)
- Ho Suk Oh
- Department of Internal Medicine, GangNeung Asan Medical Center, University of Ulsan College of Medicine, Gangneung, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Ramos-Suzarte M, Lorenzo-Luaces P, Lazo NG, Perez ML, Soriano JL, Gonzalez CEV, Hernadez IM, Albuerne YÁ, Moreno BP, Alvarez ES, Callejo IP, Alert J, Martell JA, Gonzalez YS, Gonzalez YS, Astudillo de la Vega H, Ruiz-Garcia EB, Ramos TC. Treatment of malignant, non-resectable, epithelial origin esophageal tumours with the humanized anti-epidermal growth factor antibody nimotuzumab combined with radiation therapy and chemotherapy. Cancer Biol Ther 2012; 13:600-5. [PMID: 22555809 DOI: 10.4161/cbt.19849] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Over-expression of epidermal growth factor receptor in esophageal cancer is associated with poor prognosis. The present study was conducted to evaluate safety and preliminary efficacy of nimotuzumab, a humanized anti-EGFR antibody in combination with radiation and chemotherapy in advanced esophageal tumours. PATIENTS AND METHODS A Phase II clinical trial was conducted, where patients received cisplatin, 5-fluorouracil, and radiotherapy, either alone or combined with six weekly infusions of nimotuzumab at the dose of 200 mg. Safety was the primary endpoint. The antitumoral objective response rate was the secondary endpoint. Epidermal growth factor receptor expression, KRAS mutation status and anti-idiotypic response were also evaluated. RESULTS Sixty-three patients were included in the study. Thirty patients were entered into the control group, and thirty-three patients received the treatment with nimotuzumab. The antibody was very well tolerated. Objective response rate was 47.8 % (nimotuzumab group) and 15.4 % (control group). Disease control rate was 60.9 % (nimotuzumab group) and 26.9 % (control group). Response and disease control rate were higher in patients with EGFR overexpressing tumors. CONCLUSION Nimotuzumab plus chemoradiotherapy was safe and provided statistically significant objective response. A Phase III in patients with similar characteristics will be launched.
Collapse
|
6
|
De Vita F, Giuliani F, Silvestris N, Rossetti S, Pizzolorusso A, Santabarbara G, Galizia G, Colucci G, Ciardiello F, Orditura M. Current status of targeted therapies in advanced gastric cancer. Expert Opin Ther Targets 2012; 16 Suppl 2:S29-34. [PMID: 22443228 DOI: 10.1517/14728222.2011.652616] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION In metastatic gastric cancer, chemotherapy is the standard treatment because it prolongs survival when compared to best supportive care alone. However, even after the use of more effective regimens, the overall survival remains disappointing, justifying the need for new treatment options. AREAS COVERED Areas covered in this review include the most common molecular pathways, which have provided novel targets in gastric cancer therapy. These therapeutic strategies include EGFR inhibitors, anti-angiogenic agents, cell cycle inhibitors and apoptosis promoters. EXPERT OPINION Several mAbs and kinase inhibitors, especially those targeting EGFR and VEGF/VEGFR, have already demonstrated promising activity in gastric cancer. The Phase III ToGA trial reported an increase in overall survival for patients with human EGF receptor (HER)2-positive gastric cancer treated with chemotherapy and trastuzumab compared to chemotherapy alone. This means that accurate HER2 testing in gastric cancer is necessary. Final data of ongoing trials with novel agents will be critical to further progress with this cancer.
Collapse
Affiliation(s)
- Ferdinando De Vita
- Oncologia Medica, Seconda Università di Napoli, Via S.Pansini 5, 80131, Napoli, Italia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Albulescu R, Neagu M, Albulescu L, Tanase C. Tissular and soluble miRNAs for diagnostic and therapy improvement in digestive tract cancers. Expert Rev Mol Diagn 2011; 11:101-120. [PMID: 21171925 DOI: 10.1586/erm.10.106] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Digestive cancers (e.g., gastric, colorectal, pancreatic or hepatocarcinoma) are among the most frequently reported cancers in the world, and are characterized by invasivity, metastatic potential and poor outcomes. This group includes some of the most critical cancers (among them, are those ranked second to forth in cancer-related mortality) and, despite all sustained efforts, they maintain a profile of low survival rates and lack successful therapies. Discovery of biomarkers that improve disease characterization may make optimized or personalized therapy possible. Novel biomarkers are expected to provide, hopefully, less-invasive or noninvasive diagnostic tools that make possible earlier detection of disease. Also, they may provide a more reliable selection instrument in the drug discovery process. miRNAs, short noncoding RNAs, have emerged in the last few years as significant regulators of cellular activities, controlling protein expression at the post-transcriptional level, with a significant implication in pathology in general and, of most relevance, in cancers. Deregulation of miRNA expression levels and some genetic alterations were demonstrated in various cancers, including digestive cancers. Investigations in tissue samples have provided a considerable amount of knowledge, identifying altered expressions of miRNAs associated with tumorigenesis and tumor progression. Overexpression of some tumor-inducing or tumor-promoting miRNAs was demonstrated, as well as the downregulation of tumor-suppressor miRNAs. Both individual miRNAs, as well as sets of multiple miRNAs, were set up as candidate biomarkers for diagnostics or monitoring, offering relevant insights into tumorigenic mechanisms. Circulating miRNAs were demonstrated as valuable instruments in tumor diagnosis and the prognosis of digestive cancers (affecting the esophagus, stomach, intestine, colorectum, liver and pancreas), and are being investigated thoroughly in order to generate and validate less-invasive diagnostic tools with enhanced sensitivity.
Collapse
Affiliation(s)
- Radu Albulescu
- National Institute for Chemical Pharmaceutical R&D, 112 Vitan Ave, 031299, Bucharest, Romania.
| | | | | | | |
Collapse
|
8
|
A phase II study of weekly docetaxel and cisplatin plus oral tegafur/uracil and leucovorin as first-line chemotherapy in patients with locally advanced or metastatic gastric cancer. Br J Cancer 2010; 103:1343-8. [PMID: 20924378 PMCID: PMC2990611 DOI: 10.1038/sj.bjc.6605928] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background: Docetaxel plus cisplatin and 5-fluorouracil has become a new standard for treating advanced gastric cancer. However, high rates of severe neutropenia limit its application. Modification of the regimen could be the solution to get similar activity but less myelosuppression. Methods: Patients with histologically confirmed, locally advanced, or recurrent/metastatic gastric adenocarcinoma without previous chemotherapy were enrolled. This regimen consisted of docetaxel (Tyxan, TTY, Taipei, Taiwan) 30-min infusion at a dose of 36 mg m−2, followed by cisplatin 30 mg m−2 infusion over 1 h on days 1 and 8, and oral tegafur/uracil 300 mg m−2 per day plus leucovorin 90 mg per day on days 1–14, every 3 weeks. Tumour response was evaluated after every 2 cycles of treatment. Results: From August 2007 to March 2009, 45 patients were enrolled. The median age was 56 years (range: 22–75). Among the 40 patients evaluable for tumour response, one achieved a complete response, 22 had partial responses and 11 had stable disease. The overall response rates of the evaluable and intent-to-treat (ITT) populations were 58% (95% CI: 41–74%) and 53% (95% CI: 38–68%), respectively. The disease control rates in these populations were 85% (95% CI: 70–94%) and 82% (95% CI: 68–92%), respectively. In the ITT analysis, the median time to progression and overall survival were 6.8 and 13.9 months, respectively. Major grade 3–4 toxicities were neutropenia (51%), anaemia (22%), diarrhoea (16%), and infections (20%). No patient died of treatment-related toxicities. Conclusion: Concurrent weekly docetaxel and cisplatin plus oral tegafur/uracil and leucovorin are effective and well tolerated in the treatment of advanced gastric cancer.
Collapse
|
9
|
Vecchione L, Orditura M, Ciardiello F, De Vita F. Novel investigational drugs for gastric cancer. Expert Opin Investig Drugs 2010; 18:945-55. [PMID: 19466878 DOI: 10.1517/13543780902969455] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Gastric cancer still represents a leading cause of death worldwide. Several cytotoxic agents have demonstrated activity and combination regimens improve progression-free survival, overall survival and quality of life. Nevertheless, now there is no standard therapy for advanced gastric cancer patients. OBJECTIVE To evaluate the role of new investigational agents. METHODS We analysed Phase I, II and III studies that evaluated tailored drugs directed against the epidermal growth factor receptor (EGFR), the c-erbB2, the vascular endothelial growth factor (VEGF), the vascular endothelial growth factor receptor (VEGFR), the matrix metalloproteinases (MMP) and the mammalian target of rapamycin (mTOR). CONCLUSION Data from Phase II trials indicate the potential of improved efficacy of chemotherapy when administered in combination with bevacizumab and cetuximab. Trastuzumab results are ongoing, while marimastat has not obtained clinical developments even if it has demonstrated to be an active drug in this setting of patients.
Collapse
Affiliation(s)
- Loredana Vecchione
- Second University of Naples, School of Medicine, Division of Medical Oncology 'F. Magrassi & A. Lanzara', Department of Clinical and Experimental Medicine and Surgery, c/o II Policlinico Via S. Pansini 5, 80131 Naples, Italy
| | | | | | | |
Collapse
|
10
|
Brandt D, Volkmann X, Anstätt M, Länger F, Manns MP, Schulze-Osthoff K, Bantel H. Serum biomarkers of cell death for monitoring therapy response of gastrointestinal carcinomas. Eur J Cancer 2010; 46:1464-73. [PMID: 20202824 DOI: 10.1016/j.ejca.2010.01.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2009] [Revised: 01/24/2010] [Accepted: 01/29/2010] [Indexed: 12/22/2022]
Abstract
PURPOSE Antitumour treatments are thought to exert their therapeutic efficacy mainly by induction of apoptosis in tumour cells. In epithelial cells, caspases, the key enzymes of apoptosis, cleave the intermediate filament protein cytokeratin (CK)-18 into specific fragments that are released into circulating blood and can be detected by a specific ELISA. EXPERIMENTAL DESIGN To investigate the use of CK-18 fragments as a potential biomarker for the treatment response, we examined the association of serum CK-18 levels and clinical response in 35 patients with gastrointestinal cancers. RESULTS While both cleaved and total CK-18 levels were intrinsically elevated in tumour patients, they were further increased during 5-fluorouracil (5-FU)-based therapy. Importantly, the increased levels of CK-18 could discriminate between patients with different clinical response. Cancer patients with a partial response or stable disease revealed a significantly higher increase of cleaved CK-18 during chemotherapy as compared to patients with progressive disease. CONCLUSIONS Our results suggest that detection of circulating caspase-cleaved CK-18 might be a useful serum biomarker for monitoring treatment response and should merit further evaluation in larger patient groups.
Collapse
Affiliation(s)
- Doreen Brandt
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
11
|
de Geus-Oei LF, Vriens D, van Laarhoven HWM, van der Graaf WTA, Oyen WJG. Monitoring and predicting response to therapy with 18F-FDG PET in colorectal cancer: a systematic review. J Nucl Med 2009; 50 Suppl 1:43S-54S. [PMID: 19403879 DOI: 10.2967/jnumed.108.057224] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Molecular imaging with (18)F-FDG PET has been proven useful in the management of colorectal cancer. (18)F-FDG PET plays a pivotal role in staging before surgical resection of recurrent colorectal cancer and metastases, in the localization of recurrence in patients with an unexplained rise in serum carcinoembryonic antigen levels, and in the assessment of residual masses after treatment. Currently, there is increasing interest in the role of (18)F-FDG PET beyond staging. The technique appears to have significant potential for the characterization of tumors and for the prediction of prognosis in the context of treatment stratification and early assessment of tumor response to therapy. This systematic review provides an overview of the literature on the value of (18)F-FDG PET for monitoring and predicting the response to therapy in colorectal cancer. The review covers chemotherapy response monitoring in advanced colorectal cancer, monitoring of the effects of local ablative therapies, and preoperative radiotherapy and multimodality treatment response evaluation in primary rectal cancer. Given the added value of (18)F-FDG PET for these indications, implementation in clinical practice and systematic inclusion in therapeutic trials to exploit the potential of (18)F-FDG PET are warranted.
Collapse
Affiliation(s)
- Lioe-Fee de Geus-Oei
- Department of Nuclear Medicine, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | |
Collapse
|
12
|
Galizia G, Lieto E, Orditura M, Castellano P, Mura AL, Imperatore V, Pinto M, Zamboli A, De Vita F, Ferraraccio F. Prognostic Biomarkers and Targeted Therapy in Gastric Cancer: Reply. World J Surg 2008; 32:1227-1229. [DOI: 10.1007/s00268-007-9335-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Gennaro Galizia
- Division of Surgical Oncology, “F. Magrassi ‐ A. Lanzara” Department of Clinical and Experimental Medicine and Surgery Second University of Naples School of Medicine c/o II Policlinico, Edificio 17, Via Pansini, 5 80131 Naples Italy
| | - Eva Lieto
- Division of Surgical Oncology, “F. Magrassi ‐ A. Lanzara” Department of Clinical and Experimental Medicine and Surgery Second University of Naples School of Medicine c/o II Policlinico, Edificio 17, Via Pansini, 5 80131 Naples Italy
| | - Michele Orditura
- Division of Medical Oncology, “F. Magrassi ‐ A. Lanzara” Department of Clinical and Experimental Medicine and Surgery Second University of Naples School of Medicine c/o II Policlinico, Edificio 17, Via Pansini, 5 80131 Naples Italy
| | - Paolo Castellano
- Division of Surgical Oncology, “F. Magrassi ‐ A. Lanzara” Department of Clinical and Experimental Medicine and Surgery Second University of Naples School of Medicine c/o II Policlinico, Edificio 17, Via Pansini, 5 80131 Naples Italy
| | - Anna La Mura
- Division of Pathology, “F. Magrassi ‐ A. Lanzara” Department of Clinical and Experimental Medicine and Surgery Second University of Naples School of Medicine c/o II Policlinico, Edificio 17, Via Pansini, 5 80131 Naples Italy
| | - Vincenzo Imperatore
- Division of Surgical Oncology, “F. Magrassi ‐ A. Lanzara” Department of Clinical and Experimental Medicine and Surgery Second University of Naples School of Medicine c/o II Policlinico, Edificio 17, Via Pansini, 5 80131 Naples Italy
| | - Margherita Pinto
- Division of Surgical Oncology, “F. Magrassi ‐ A. Lanzara” Department of Clinical and Experimental Medicine and Surgery Second University of Naples School of Medicine c/o II Policlinico, Edificio 17, Via Pansini, 5 80131 Naples Italy
| | - Anna Zamboli
- Division of Surgical Oncology, “F. Magrassi ‐ A. Lanzara” Department of Clinical and Experimental Medicine and Surgery Second University of Naples School of Medicine c/o II Policlinico, Edificio 17, Via Pansini, 5 80131 Naples Italy
| | - Ferdinando De Vita
- Division of Medical Oncology, “F. Magrassi ‐ A. Lanzara” Department of Clinical and Experimental Medicine and Surgery Second University of Naples School of Medicine c/o II Policlinico, Edificio 17, Via Pansini, 5 80131 Naples Italy
| | - Francesca Ferraraccio
- Division of Pathology, “F. Magrassi ‐ A. Lanzara” Department of Clinical and Experimental Medicine and Surgery Second University of Naples School of Medicine c/o II Policlinico, Edificio 17, Via Pansini, 5 80131 Naples Italy
| |
Collapse
|
13
|
Lieto E, Ferraraccio F, Orditura M, Castellano P, Mura AL, Pinto M, Zamboli A, De Vita F, Galizia G. Expression of vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) is an independent prognostic indicator of worse outcome in gastric cancer patients. Ann Surg Oncol 2008; 15:69-79. [PMID: 17896140 DOI: 10.1245/s10434-007-9596-0] [Citation(s) in RCA: 189] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2007] [Revised: 08/03/2007] [Accepted: 08/06/2007] [Indexed: 12/12/2022]
Abstract
BACKGROUND Unlike other human tumors, gastric cancer remains a great therapeutic challenge since no standardized postoperative treatment exists. Knowledge of molecular pathways determining the behavior of individual gastric tumors seems to be crucial for therapeutic decisions, and evaluation of vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) expression might be critical for prognosis, assessment, and identification of patients that could be treated with tailored therapies. METHODS VEGF and EGFR determination was performed in 88 gastric cancer samples as well as 25 normal gastric mucosa specimens from non-cancer patients using a commercially available immunohistochemistry kit. In all samples, the correlation of VEGF and EGFR expression was investigated with each other, and with other prognostic indicators in all samples, and, finally, with survival rates in 69 patients undergoing potentially curative surgery. RESULTS Forty-eight per cent (42 cases) of gastric cancers expressed VEGF, and 44% (39 cases) stained for EGFR. In curatively treated patients, VEGF and EGFR expression was demonstrated to correlate with worse survival in both univariate and multivariate analyses. Molecular profiling was shown to more accurately estimate the risk of cancer-related death than TNM stage, and, of most interest, to allow sorting out high-risk patients within the same stage. CONCLUSIONS These findings provide evidence that contemporary evaluation of VEGF and EGFR expression may be crucial to select gastric cancer patients with poor prognosis who may benefit of tailored treatments.
Collapse
Affiliation(s)
- Eva Lieto
- Divisions of Surgical Oncology, F. Magrassi - A. Lanzara Department of Clinical and Experimental Medicine and Surgery, Second University of Naples School of Medicine, c/o II Policlinico, Edificio 17, Via Pansini, 5, 80131, Naples, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Galizia G, Lieto E, Orditura M, Castellano P, Mura AL, Imperatore V, Pinto M, Zamboli A, De Vita F, Ferraraccio F. Epidermal growth factor receptor (EGFR) expression is associated with a worse prognosis in gastric cancer patients undergoing curative surgery. World J Surg 2007; 31:1458-1468. [PMID: 17516110 DOI: 10.1007/s00268-007-9016-4] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND In gastric cancer, the recurrence rate is high even after curative surgery. A relevant issue is the identification of independent prognostic factors to select high-risk patients; such features can be used as predictive factors for tailored therapies. In this study we have investigated the role of epidermal growth factor receptor (EGFR) expression as a prognostic marker for predicting cancer behavior and clinical outcome in gastric cancer patients undergoing potentially curative surgery. METHODS Epidermal growth factor receptor determination using a commercially available immunohistochemistry (IHC) kit was performed in tissues from 82 gastric cancer patients receiving primary surgical treatment and in 25 normal gastric mucosa specimens from noncancer patients. The EGFR positivity was correlated with disease recurrence and survival in univariate and multivariate analyses. RESULTS Forty-four percent (36 cases) of gastric cancers were EGFR positive. In 66 curatively treated patients, EGFR expression correlated with disease recurrence and poorer survival in both univariate and multivariate analyses. In a multivariate model for predicting recurrence and survival, advanced tumor extension (T(3) or T(4)), nodal metastases, and EGFR expression were the only independent covariates. In particular, EGFR expression was shown to be a significant predictor of poor prognosis among gastric cancer patients having the same stage according to the current TNM staging system. CONCLUSIONS These findings suggest that EGFR expression may be useful in identifying high-risk gastric cancer patients undergoing potentially curative surgery. Multimodal treatments should be considered in the adjuvant treatment of these patients.
Collapse
Affiliation(s)
- Gennaro Galizia
- Division of Surgical Oncology, F. Magrassi - A. Lanzara Department of Clinical and Experimental Medicine and Surgery, Second University of Naples School of Medicine, c/o II Policlinico, Edificio 17, Via Pansini, Naples, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Löhr JM. Pancreatic cancer — Outlook: gene therapy. THE CHINESE-GERMAN JOURNAL OF CLINICAL ONCOLOGY 2007; 6:181-186. [DOI: 10.1007/s10330-007-0049-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
|
16
|
Yeo W, Boyer M, Chung HC, Ong SYK, Lim R, Zee B, Ma B, Lam KC, Mo FKF, Ng EKW, Ho R, Clarke S, Roh JK, Beale P, Rha SY, Jeung HC, Soo R, Goh BC, Chan ATC. Irofulven as first line therapy in recurrent or metastatic gastric cancer: a phase II multicenter study by the Cancer Therapeutics Research Group (CTRG). Cancer Chemother Pharmacol 2007; 59:295-300. [PMID: 16783579 DOI: 10.1007/s00280-006-0270-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Accepted: 05/11/2006] [Indexed: 11/27/2022]
Abstract
BACKGROUND The purpose of this study was to evaluate the tolerability and efficacy of irofulven, a DNA interacting acylfulvene analog, as first line therapy for patients with recurrent or metastatic gastric cancer. PATIENTS AND METHODS Twenty-three patients with recurrent or metastatic gastric cancer received irofulven at a dose of 0.45 mg/kg administered intravenously over 30-min infusion (up to a maximum of 50 mg), on days 1 and 8, every 3 weeks. RESULTS The median number of cycles delivered per patient was 2 (range 1-6). Two patients (9%) had >or= 1-week delay in administration of subsequent cycle of chemotherapy. For the day 8 chemotherapy, dose reductions were required in seven patients (30%); dose omitting occurred in five patients (22%). Grade 3/4 anemia and neutropenia occurred in 22 and 17% of patients, respectively. There was no grade 4 thrombocytopenia and no neutropenic fever was observed. Of the 20 evaluable patients, there were no responses observed, 3 patients had stable disease after 2 cycles of treatment which was not confirmed by a further assessment. Median overall survival was 6.05 months (95% CI 4.55-9.39). CONCLUSIONS Irofulven was tolerated at the dose of 0.45 mg/kg on days 1 and 8, every 3 weeks but showed no evidence of antitumor activity in patients with advanced gastric cancer.
Collapse
Affiliation(s)
- W Yeo
- Comprehensive Cancer Trials Unit, Department of Clinical Oncology, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
[18F]Fluorodeoxyglucose (FDG) positron emission tomography (PET) is a useful imaging tool in the evolving management of patients with colorectal carcinoma. This technique is able to measure and visualize metabolic changes in cancer cells. This feature results in the ability to distinguish viable tumor from scar tissue, in the detection of tumor foci at an earlier stage than possible by conventional anatomic imaging and in the measurement of alterations in tumor metabolism, indicative of tumor response to therapy. Nowadays, FDG-PET plays a pivotal role in staging patients before surgical resection of recurrence and metastases, in the localization of recurrence in patients with an unexplained rise in serum carcinoembryonic antigen and in assessment of residual masses after treatment. In the presurgical evaluation, FDG-PET may be best used in conjunction with anatomic imaging in order to combine the benefits of both anatomical (CT) and functional (PET) information, which leads to significant improvements in preoperative liver staging and preoperative judgment on the feasibility of resection. Integration of FDG-PET into the management algorithm of these categories of patients alters and improves therapeutic management, reduces morbidity due to futile surgery, leads to substantial cost savings and probably also to a better patient outcome. FDG-PET also appears to have great potential in monitoring the success of local ablative therapies soon after intervention and in the prediction and evaluation of response to radiotherapy, systemic therapy, and combinations thereof. This review aims to outline the current and future role of FDG-PET in the field of colorectal cancer.
Collapse
Affiliation(s)
- Lioe-Fee de Geus-Oei
- Department of Nuclear Medicine, Radboud University, Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
18
|
de Geus-Oei LF, Wiering B, Krabbe PFM, Ruers TJM, Punt CJA, Oyen WJG. FDG-PET for prediction of survival of patients with metastatic colorectal carcinoma. Ann Oncol 2006; 17:1650-5. [PMID: 16936185 DOI: 10.1093/annonc/mdl180] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The current study focuses on the prognostic value of pretreatment metabolic activity in metastases as measured with [(18)F]fluorodeoxyglucose positron emission tomography (FDG-PET), as an indicator of survival in colorectal cancer. PATIENTS AND METHODS In a prospective series of 152 patients with metastatic colorectal cancer, of whom 67 were treated with resection of metastases and 85 with chemotherapy, standardized uptake values (SUV) as measured with FDG-PET, were calculated prior to treatment. Survival probabilities were estimated by Cox proportional regression analysis. For Kaplan-Meier analysis SUV was stratified by the median value. Survival differences were assessed using the log-rank test. RESULTS SUV in metastases was a significant predictor for overall survival (hazard ratio 1.17, 95% confidence interval 1.06-1.30, P = 0.002), independent of the subsequent treatment. According to the median value of the patient population a low (SUV <4.26) and high uptake group (SUV >4.26) was defined. The median survival and the 2- and 3-year survival rates were 32 months, 59% and 45%, respectively, in the low-uptake group and 19 months, 37% and 28%, respectively, in the high-uptake group (P = 0.017). CONCLUSION A significant survival benefit was observed in patients with low FDG uptake in metastases of colorectal cancer.
Collapse
Affiliation(s)
- L F de Geus-Oei
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
19
|
Xu HY, Xu L, Gao JH, Yang JJ, Li KZ, Dou KF. Prokaryotic expression of anti-carcinoembryonic single-chain variable fragment and its value in detection of gastric carcinoma cells. Shijie Huaren Xiaohua Zazhi 2006; 14:1780-1784. [DOI: 10.11569/wcjd.v14.i18.1780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the prokaryotic expression of the anti-carcinoembryonic antigen (CEA) single-chain fragment variable (scFv) antibody T84.66 and its specific affinity to gastric cancer cell lines and tissues.
METHODS: The cDNA of anti-CEA scFv antibody was inserted into pCANTAB5E to obtain phage vector T84.66-scFv-pCANTAB5E, and then the vector was transferred into E. coli HB2151. Isopropyl-β-D-thiogalactoside (IPTG) was used to induce the expression of anti-CEA scFv antibody. SDS-PAGE and Western blot were used identify the anti-CEA scFv antibody. Human gastric cancer cells were cultured, and CEA was determined with the obtained scFv by immunohistochemistry in the cells and paraffin-embedded gastric carcinoma tissues.
RESULTS: SDS-PAGE and Western blot showed that the anti-CEA scFv antibody T84.66 was successfully expressed. T84.66 could bind to gastric carcinoma cell lines KATOⅢ, MKN45 and HGC-27, but not to SGC7901, GC803 and BGC823, suggesting that KATOⅢ, MKN45 and HGC-27 cells expressed CEA. For the 42 cases gastric carcinoma tissues, the positive rate of CEA in the early and progressive stage was 55% (6/11) and 61% (19/31), respectively, but no CEA expression was found in the 10 normal cases. CEA expression was significantly different between gastric cancer and normal tissues (P < 0.05).
CONCLUSION: The prokaryotic expression of anti-CEA scFv antibody T84.66 is successfully achieved, and can be used to identify CEA. CEA is highly expressed in gastric cancer, but not in normal mucosa.
Collapse
|
20
|
Chao Y, Li CP, Chao TY, Su WC, Hsieh RK, Wu MF, Yeh KH, Kao WY, Chen LT, Cheng AL. An open, multi-centre, phase II clinical trial to evaluate the efficacy and safety of paclitaxel, UFT, and leucovorin in patients with advanced gastric cancer. Br J Cancer 2006; 95:159-63. [PMID: 16804524 PMCID: PMC2360611 DOI: 10.1038/sj.bjc.6603225] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The aim of the study was to evaluate the response rate and safety of weekly paclitaxel (Taxol((R))) combination chemotherapy with UFT (tegafur, an oral 5-fluorouracil prodrug, and uracil at a 1 : 4 molar ratio) and leucovorin (LV) in patients with advanced gastric cancer. Patients with histologically confirmed, locally advanced or recurrent/metastatic gastric cancer were studied. Paclitaxel 1-h infusion at a dose of 100 mg m(-2) on days 1 and 8 and oral UFT 300 mg m(-2) day(-1) plus LV 90 mg day(-1) were given starting from day 1 for 14 days, followed by a 7-day period without treatment. Treatment was repeated every 21 days. From February 2003 to October 2004, 55 patients were enrolled. The median age was 62 years (range: 32-82). Among the 48 patients evaluated for tumour response, two achieved a complete response and 22 a partial response, with an overall response rate of 50% (95% confidence interval: 35-65%). All 55 patients were evaluated for survival and toxicities. Median time to progression and overall survival were 4.4 and 9.8 months, respectively. Major grade 3-4 toxicities were neutropenia in 25 patients (45%) and diarrhoea in eight patients (15%). Although treatment was discontinued owing to treatment-related toxicities in nine patients (16%), there was no treatment-related mortality. Weekly paclitaxel plus oral UFT/LV is effective, convenient, and well tolerated in treating patients with advanced gastric cancer.
Collapse
Affiliation(s)
- Y Chao
- Cancer Center, Taipei Veterans General Hospital, Taipei, Taiwan
- Central Clinic Hospital, Taipei, Taiwan
- National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - C P Li
- National Yang-Ming University School of Medicine, Taipei, Taiwan
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - T Y Chao
- Division of Hematology and Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - W C Su
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - R K Hsieh
- Division of Hematology and Oncology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - M F Wu
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - K H Yeh
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Oncology and Hematology, Department of Internal Medicine, Far Eastern Memorial Hospital, Taipei, Taiwan
| | - W Y Kao
- Division of Hematology and Oncology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - L T Chen
- Institute of Cancer Research, National Health Research Institutes, Taipei, Taiwan
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Institute of Cancer Research, National Health Research Institutes, Taipei Veterans General Hospital, Ward 191, No. 201, Sec. 2, Shih-Pai Road, Taipei 112, Taiwan. E-mail:
| | - A L Cheng
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Internal Medicine and Department of Oncology, National Taiwan University Hospital, No. 2, Chung-Shan S Rd, Taipei 100, Taiwan. E-mail:
| |
Collapse
|