3001
|
Resection of at-risk mesenteric lymph nodes is associated with improved survival in patients with small bowel neuroendocrine tumors. World J Surg 2014; 37:1695-700. [PMID: 23657749 DOI: 10.1007/s00268-013-1918-8] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Neuroendocrine tumors of the small intestine commonly metastasize to regional lymph nodes (LNs). Single-institution reports suggest that removal of LNs improves outcome, but comprehensive data are lacking. We hypothesized that the extent of lymphadenectomy reported in a large administrative database would be associated with overall survival for jejunal and ileal neuroendocrine tumors. METHODS A search of the Surveillance Epidemiology and End Results database was performed for patients with jejunal and ileal neuroendocrine tumors from 1977 to 2004. Descriptive patient characteristics were collected to include age at diagnosis, sex, race, grade, primary tumor size, LN status, number of LNs resected, presence of distant metastasis, and the type of operation. Statistical analyses were limited to patients with only one primary tumor to exclude patients with other malignancies. Univariate and multivariate analyses were performed to analyze the number of LNs resected and the LN ratio (number of positive LNs/total number of LNs removed) to determine the effect on cancer-specific survival. RESULTS Altogether, 1,364 patients were included in this analysis. Removal of any LNs was associated with improved cancer-specific survival when compared to patients with no LN removal reported (p = 0.0027) on univariate analysis. Among those who had any LNs removed, a median of eight LNs were identified in resection specimens with a median LN ratio of 0.29 (range 0-1). On multivariate analysis (adjusting for age and tumor size), patients with >7 LNs removed experienced better cancer-specific survival than those with ≤ 7 LNs removed (median survival not reached vs. 140 months): hazard ratio and 95 % confidence interval were 0.573 (0.402, 0.817) (p = 0.002). CONCLUSIONS This review of a large number of surgical patients demonstrates that regional mesenteric lymphadenectomy in conjunction with resection of the primary tumor is associated with improved survival of patients with small bowel neuroendocrine tumors.
Collapse
|
3002
|
Bao A, Li Y, Tong Y, Zheng H, Wu W, Wei C. 1,25-Dihydroxyvitamin D₃ and cisplatin synergistically induce apoptosis and cell cycle arrest in gastric cancer cells. Int J Mol Med 2014; 33:1177-84. [PMID: 24573222 DOI: 10.3892/ijmm.2014.1664] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 02/19/2014] [Indexed: 12/20/2022] Open
Abstract
1,25-Dihydroxyvitamin D3 [1,25(OH)2D3] plays an anticancer role in multiple types of cancer and potentiates the cytotoxic effects of several common chemotherapeutic agents. The hypercalcemia caused by 1,25(OH)2D3 alone or resistance to cisplatin weaken the anticancer effects of vitamin D. Thus, in this study, we aimed to investigate the synergistic effects of 1,25(OH)2D3 and cisplatin on the apoptosis and cell cycle progression of gastric cancer cells. BGC-823 human gastric cancer cells were treated with 1,25(OH)2D3 or cisplatin alone, or a combination of both agents. Cell apoptosis was assessed by TUNEL assay and flow cytometry. The expression of the apoptosis-related proteins, poly(ADP-ribose) polymerase (PARP), Bax, Bcl-2, caspase-3 and caspase-8, was examined using immunoblot analysis. ERK and AKT phosphorylation were examined by immunoblot analysis. The cell cycle distribution was determined by propidium iodide staining and flow cytometric analysis. p21 and p27 protein expression was also examined using immunoblot analysis. Our results revealed that co-treatment with 1,25(OH)2D3 enhanced cisplatin-induced apoptosis and upregulated the expression of Bax, and promoted the cleavage of PARP and caspase-3. The phosphorylation levels of ERK and AKT were reduced following combined treatment with 1,25(OH)2D3 and cisplatin. The percentage of cells in the G0/G1 phase was greater in the cells treated with the combined treatment than in those treated with either 1,25(OH)2D3 or cisplatin alone. p21 and p27 expression was upregulated following co-treatment with both agents. The results of this study suggest that 1,25(OH)2D3 potentiates cisplatin-mediated cell growth inhibition and cell apoptosis, which involves the upregulation of Bax, a decrease in ERK and AKT phosphorylation levels, and increased p21 and p27 levels.
Collapse
Affiliation(s)
- Anyu Bao
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yan Li
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yongqing Tong
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hongyun Zheng
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Wu
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Chuandong Wei
- Department of Clinical Laboratory, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| |
Collapse
|
3003
|
Ross JS, Wang K, Gay L, Al-Rohil R, Rand JV, Jones DM, Lee HJ, Sheehan CE, Otto GA, Palmer G, Yelensky R, Lipson D, Morosini D, Hawryluk M, Catenacci DVT, Miller VA, Churi C, Ali S, Stephens PJ. New routes to targeted therapy of intrahepatic cholangiocarcinomas revealed by next-generation sequencing. Oncologist 2014; 19:235-42. [PMID: 24563076 DOI: 10.1634/theoncologist.2013-0352] [Citation(s) in RCA: 362] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) is a subtype of primary liver cancer that is rarely curable by surgery and is rapidly increasing in incidence. Relapsed ICC has a poor prognosis, and current systemic nontargeted therapies are commonly extrapolated from those used in other gastrointestinal malignancies. We hypothesized that genomic profiling of clinical ICC samples would identify genomic alterations that are linked to targeted therapies and that could facilitate a personalized approach to therapy. METHODS DNA sequencing of hybridization-captured libraries was performed for 3,320 exons of 182 cancer-related genes and 36 introns of 14 genes frequently rearranged in cancer. Sample DNA was isolated from 40 μm of 28 formalin-fixed paraffin-embedded ICC specimens and sequenced to high coverage. RESULTS The most commonly observed alterations were within ARID1A (36%), IDH1/2 (36%), and TP53 (36%) as well as amplification of MCL1 (21%). Twenty cases (71%) harbored at least one potentially actionable alteration, including FGFR2 (14%), KRAS (11%), PTEN (11%), CDKN2A (7%), CDK6 (7%), ERBB3 (7%), MET (7%), NRAS (7%), BRCA1 (4%), BRCA2 (4%), NF1 (4%), PIK3CA (4%), PTCH1 (4%), and TSC1 (4%). Four (14%) of the ICC cases featured novel gene fusions involving the tyrosine kinases FGFR2 and NTRK1 (FGFR2-KIAA1598, FGFR2-BICC1, FGFR2-TACC3, and RABGAP1L-NTRK1). CONCLUSION Two thirds of patients in this study harbored genomic alterations that are associated with targeted therapies and that have the potential to personalize therapy selection for to individual patients.
Collapse
Affiliation(s)
- Jeffrey S Ross
- Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York, USA; Foundation Medicine, Inc., Cambridge, Massachusetts, USA; University of Chicago, Chicago, Illinois, USA; The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3004
|
Angiopoietins and non-vascular endothelial growth factor antiangiogenic targets in advanced renal cell carcinoma. Cancer J 2014; 19:307-10. [PMID: 23867511 DOI: 10.1097/ppo.0b013e31829d5d15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The treatment of metastatic renal cell carcinoma has evolved from an era dominated by immune modulation to an era of antiangiogenesis agents. Blockade of vascular endothelial growth factor-mediated pathways and mammalian target of rapamycin pathways has accounted for most of these gains. Although these agents have offered dramatic improvements in survival for kidney cancer patients, resistance inevitably occurs, and new classes of agents are needed to continue to improve outcomes in this setting. We discuss several alternative pathways of angiogenesis, which are being investigated as targets to overcome treatment resistance, including angiopoietin family proteins, fibroblast growth factor, platelet-derived growth factor, and vascular disrupting agents.
Collapse
|
3005
|
Chhatrala R, Thanavala Y, Iyer R. Targeted therapy in gastrointestinal malignancies. J Carcinog 2014; 13:4. [PMID: 24737952 PMCID: PMC3986534 DOI: 10.4103/1477-3163.127639] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 12/15/2013] [Indexed: 12/13/2022] Open
Abstract
Increased understanding of cancer pathogenesis has identified several pathways that serve as potential targets for novel targeted agents in development. The selection of targeted cancer therapy based on biomarkers has instigated a new era of personalized medicine and changed the way we practice oncology. Many targeted agents are approved for treatment of gastrointestinal malignancies most targeting tumor angiogenesis, and many more are in different phases of development. Here we briefly summarize nine different targeted agents that are approved currently in the U.S. and several other agents currently being studied in various gastrointestinal cancers.
Collapse
Affiliation(s)
- Ravi Chhatrala
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University, Richmond, VA, USA
| | - Yasmin Thanavala
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Renuka Iyer
- Department of Medicine, Division of Medical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
3006
|
Karanicolas PJ, Metrakos P, Chan K, Asmis T, Chen E, Kingham TP, Kemeny N, Porter G, Fields RC, Pingpank J, Dixon E, Wei A, Cleary S, Zogopoulos G, Dey C, D'Angelica M, Fong Y, Dowden S, Ko YJ. Hepatic arterial infusion pump chemotherapy in the management of colorectal liver metastases: expert consensus statement. ACTA ACUST UNITED AC 2014; 21:e129-36. [PMID: 24523610 DOI: 10.3747/co.21.1577] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite significant improvements in systemic therapy for patients with colorectal liver metastases (crlms), response rates in the first-line setting are not optimal, and response rates in the second-line setting remain disappointing. Hepatic arterial infusion pump (haip) chemotherapy has been extensively studied in patients with crlms, but it remains infrequently used. We convened an expert panel to discuss the role of haip in the contemporary management of patients with crlm. Using a consensus process, we developed these statements: haip chemotherapy should be given in combination with systemic chemotherapy.haip chemotherapy should be offered in the context of a multidisciplinary program that includes expertise in hepatobiliary surgery, medical oncology, interventional radiology, nursing, and nuclear medicine.haip chemotherapy in combination with systemic therapy should be considered in patients with unresectable crlms who have progressed on first-line systemic treatment. In addition, haip chemotherapy is acceptable as first-line treatment in patients with unresectable colorectal liver metastases.haip chemotherapy is not recommended in the setting of extrahepatic disease outside the context of a clinical trial.haip chemotherapy in combination with systemic therapy is an option for select patients with resected colorectal liver metastases. These consensus statements provide a framework that clinicians who treat patients with crlm can use when considering treatment with haip.
Collapse
Affiliation(s)
- P J Karanicolas
- Ontario: Departments of Surgery (Karanicolas, Wei, Cleary), Medicine (Chan, Ko), and Medical Imaging (Dey), University of Toronto, Toronto; Departments of Surgery (Karanicolas), Medicine (Chan, Ko), and Medical Imaging (Dey), Sunnybrook Health Sciences Centre, Toronto; Departments of Medicine (Chen) and Surgery (Wei, Cleary), University Health Network, Toronto; Department of Medicine (Asmis), University of Ottawa, Ottawa
| | - P Metrakos
- Quebec: Department of Surgery (Metrakos, Zogopoulos), McGill University, Montreal
| | - K Chan
- Ontario: Departments of Surgery (Karanicolas, Wei, Cleary), Medicine (Chan, Ko), and Medical Imaging (Dey), University of Toronto, Toronto; Departments of Surgery (Karanicolas), Medicine (Chan, Ko), and Medical Imaging (Dey), Sunnybrook Health Sciences Centre, Toronto; Departments of Medicine (Chen) and Surgery (Wei, Cleary), University Health Network, Toronto; Department of Medicine (Asmis), University of Ottawa, Ottawa
| | - T Asmis
- Ontario: Departments of Surgery (Karanicolas, Wei, Cleary), Medicine (Chan, Ko), and Medical Imaging (Dey), University of Toronto, Toronto; Departments of Surgery (Karanicolas), Medicine (Chan, Ko), and Medical Imaging (Dey), Sunnybrook Health Sciences Centre, Toronto; Departments of Medicine (Chen) and Surgery (Wei, Cleary), University Health Network, Toronto; Department of Medicine (Asmis), University of Ottawa, Ottawa
| | - E Chen
- Ontario: Departments of Surgery (Karanicolas, Wei, Cleary), Medicine (Chan, Ko), and Medical Imaging (Dey), University of Toronto, Toronto; Departments of Surgery (Karanicolas), Medicine (Chan, Ko), and Medical Imaging (Dey), Sunnybrook Health Sciences Centre, Toronto; Departments of Medicine (Chen) and Surgery (Wei, Cleary), University Health Network, Toronto; Department of Medicine (Asmis), University of Ottawa, Ottawa
| | - T P Kingham
- New York State: Departments of Surgery (Kingham, Fong, D'Angelica) and Medicine (Kemeny), Memorial Sloan-Kettering Cancer Center, New York, NY, U.S.A
| | - N Kemeny
- New York State: Departments of Surgery (Kingham, Fong, D'Angelica) and Medicine (Kemeny), Memorial Sloan-Kettering Cancer Center, New York, NY, U.S.A
| | - G Porter
- Nova Scotia: Department of Surgery (Porter), Dalhousie University, Halifax
| | - R C Fields
- Missouri: Department of Surgery (Fields), Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis
| | - J Pingpank
- Pennsylvania: Department of Surgery (Pingpank), University of Pittsburgh, Pittsburgh
| | - E Dixon
- Alberta: Departments of Surgery (Dixon) and Medicine (Dowden), University of Calgary, Calgary
| | - A Wei
- Ontario: Departments of Surgery (Karanicolas, Wei, Cleary), Medicine (Chan, Ko), and Medical Imaging (Dey), University of Toronto, Toronto; Departments of Surgery (Karanicolas), Medicine (Chan, Ko), and Medical Imaging (Dey), Sunnybrook Health Sciences Centre, Toronto; Departments of Medicine (Chen) and Surgery (Wei, Cleary), University Health Network, Toronto; Department of Medicine (Asmis), University of Ottawa, Ottawa
| | - S Cleary
- Ontario: Departments of Surgery (Karanicolas, Wei, Cleary), Medicine (Chan, Ko), and Medical Imaging (Dey), University of Toronto, Toronto; Departments of Surgery (Karanicolas), Medicine (Chan, Ko), and Medical Imaging (Dey), Sunnybrook Health Sciences Centre, Toronto; Departments of Medicine (Chen) and Surgery (Wei, Cleary), University Health Network, Toronto; Department of Medicine (Asmis), University of Ottawa, Ottawa
| | - G Zogopoulos
- Quebec: Department of Surgery (Metrakos, Zogopoulos), McGill University, Montreal
| | - C Dey
- Ontario: Departments of Surgery (Karanicolas, Wei, Cleary), Medicine (Chan, Ko), and Medical Imaging (Dey), University of Toronto, Toronto; Departments of Surgery (Karanicolas), Medicine (Chan, Ko), and Medical Imaging (Dey), Sunnybrook Health Sciences Centre, Toronto; Departments of Medicine (Chen) and Surgery (Wei, Cleary), University Health Network, Toronto; Department of Medicine (Asmis), University of Ottawa, Ottawa
| | - M D'Angelica
- New York State: Departments of Surgery (Kingham, Fong, D'Angelica) and Medicine (Kemeny), Memorial Sloan-Kettering Cancer Center, New York, NY, U.S.A
| | - Y Fong
- New York State: Departments of Surgery (Kingham, Fong, D'Angelica) and Medicine (Kemeny), Memorial Sloan-Kettering Cancer Center, New York, NY, U.S.A
| | - S Dowden
- Alberta: Departments of Surgery (Dixon) and Medicine (Dowden), University of Calgary, Calgary
| | - Y J Ko
- Ontario: Departments of Surgery (Karanicolas, Wei, Cleary), Medicine (Chan, Ko), and Medical Imaging (Dey), University of Toronto, Toronto; Departments of Surgery (Karanicolas), Medicine (Chan, Ko), and Medical Imaging (Dey), Sunnybrook Health Sciences Centre, Toronto; Departments of Medicine (Chen) and Surgery (Wei, Cleary), University Health Network, Toronto; Department of Medicine (Asmis), University of Ottawa, Ottawa
| |
Collapse
|
3007
|
Weng W, Feng J, Qin H, Ma Y. Molecular therapy of colorectal cancer: progress and future directions. Int J Cancer 2014; 136:493-502. [PMID: 24420815 DOI: 10.1002/ijc.28722] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/08/2014] [Indexed: 12/23/2022]
Abstract
Colorectal cancer (CRC) remains one of the most common types of cancer and leading causes of cancer death worldwide. Although the introduction of cytotoxic drugs such as oxaliplatin, irinotecan and fluorouracil has improved the treatment of advanced CRC, the individual response to chemoradiotherapy varies tremendously from one patient to another. However, recent progress in CRC molecular therapies may provide new insight into the treatment of this disease. Currently, components of the EGFR, VEGF, Wnt and NF-kB pathways are the most important targets for CRC therapy. This review chronicles the development of molecular CRC therapies over the past few decades. We also provide an update on the current progress of research concerning the molecular pathways leading to CRC and discuss the possible implications for CRC therapy.
Collapse
Affiliation(s)
- Wenhao Weng
- Department of Clinical laboratory, Shanghai Tenth People's Hospital Affiliated with Tongji University, Shanghai, People's Republic of China
| | | | | | | |
Collapse
|
3008
|
Ezziddin S, Khalaf F, Vanezi M, Haslerud T, Mayer K, Al Zreiqat A, Willinek W, Biersack HJ, Sabet A. Outcome of peptide receptor radionuclide therapy with 177Lu-octreotate in advanced grade 1/2 pancreatic neuroendocrine tumours. Eur J Nucl Med Mol Imaging 2014; 41:925-33. [PMID: 24504504 DOI: 10.1007/s00259-013-2677-3] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 12/20/2013] [Indexed: 02/07/2023]
Abstract
PURPOSE The clinical benefit of peptide receptor radionuclide therapy (PRRT) in patients with pancreatic neuroendocrine tumours (pNET) has not yet been well described and defined in its full extent due to limited data in this tumour subgroup. This study was intended to obtain robust, comparative data on the outcome and toxicity of standardized PRRT with (177)Lu-octreotate in a well-characterized population of patients with advanced pNET of grade 1/2 (G1/2). METHODS We retrospectively analysed a cohort of 68 pNET patients with inoperable metastatic disease consecutively treated with (177)Lu-octreotate (four intended cycles at 3-monthly intervals; mean activity per cycle 8.0 GBq). Of these 68 patients, 46 (67.6 %) had documented morphological tumour progression during the 12 months before initiation of treatment, and PRRT was the first-line systemic therapy in 35 patients (51.5 %). Response was evaluated according to modified Southwest Oncology Group (SWOG) criteria and additionally with Response Criteria in Solid Tumors (RECIST) 1.1. Survival was analysed using Kaplan-Meier curves and Cox proportional hazards model for univariate and multivariate analyses. Toxicity was assessed by standard follow-up laboratory work-up including blood count, and liver and renal function, supplemented with serial (99m)Tc-DTPA clearance measurements. RESULTS The median follow-up period was 58 months (range 4 - 112). Reversible haematotoxicity (grade 3 or more) occurred in four patients (5.9 %). No significant nephrotoxicity (grade 3 or more) was observed. Treatment responses (SWOG criteria) consisted of a partial response in 41 patients (60.3 %), a minor response in 8 (11.8 %), stable disease in 9 (13.2 %), and progressive disease in 10 (14.7 %). Median progression-free survival (PFS) and overall survival (OS) were 34 (95 % CI 26 - 42) and 53 months (95 % CI 46 - 60), respectively. A G1 proliferation status was associated with longer PFS (p = 0.04) and OS (p = 0.044) in the multivariate analysis. Variables linked to impaired OS, on the other hand, were a reduced performance status (Karnofsky score ≤ 70 %, p = 0.007), a high hepatic tumour burden (≥ 25 % liver volume, p = 0.017), and an elevated plasma level of neuron-specific enolase (NSE >15 ng/ml, p = 0.035). CONCLUSION The outstanding response rates and survival outcomes suggest that PRRT is highly effective in advanced G1/2 pNET when compared to data of other treatment modalities. Independent predictors of survival are the tumour proliferation index, the patient's performance status, tumour burden and baseline plasma NSE level.
Collapse
Affiliation(s)
- Samer Ezziddin
- Department of Nuclear Medicine, University Hospital Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
3009
|
Prise en charge des troubles métaboliques observés avec évérolimus chez les patients atteints de tumeurs neuroendocrines bien différenciées non résécables : propositions d’experts. Bull Cancer 2014; 101:175-83. [DOI: 10.1684/bdc.2014.1887] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
3010
|
Foubert F, Matysiak-Budnik T, Touchefeu Y. Options for metastatic colorectal cancer beyond the second line of treatment. Dig Liver Dis 2014; 46:105-12. [PMID: 23954144 DOI: 10.1016/j.dld.2013.07.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 06/11/2013] [Accepted: 07/01/2013] [Indexed: 02/07/2023]
Abstract
Colorectal cancer is the third most common cancer, with recent advances in the management of unresectable metastatic lesions. The aim of this review is to discuss the remaining options for heavily pretreated patients with unresectable metastatic colorectal cancer. Beyond second-line treatment, two epidermal growth factor receptor (EGFR) inhibitors, cetuximab and panitumumab, have a demonstrated clinical interest in patients with KRAS wild-type tumours. However, few data exist in patients pretreated with an anti-EFGR and who are being rechallenged with anti-EGFR drugs. Reintroduction of chemotherapy should be considered. In September 2012, regorafenib, a multi-kinase inhibitor was approved by the US Federal Drug Administration for patients refractory to other standard treatments. In the case of metastases limited to the liver, transarterial chemoembolization, hepatic artery infusion and radioembolization could also be discussed in selected patients. With the multiplication of therapeutic options in first-line, second-line treatment, and beyond, the concept of subsequent lines of chemotherapy should be replaced by a multiline strategy, dependent on the patient and on tumour biology. A better understanding of the tumour biology and predictive factors for the response to these therapies is needed, and further strategic trials are urgently warranted.
Collapse
Affiliation(s)
- Fanny Foubert
- Gastroenterology Department, Gastrointestinal Oncology Unit, University Hospital, Nantes, France
| | - Tamara Matysiak-Budnik
- Gastroenterology Department, Gastrointestinal Oncology Unit, University Hospital, Nantes, France
| | - Yann Touchefeu
- Gastroenterology Department, Gastrointestinal Oncology Unit, University Hospital, Nantes, France.
| |
Collapse
|
3011
|
Vergès B, Walter T, Cariou B. Endocrine side effects of anti-cancer drugs: effects of anti-cancer targeted therapies on lipid and glucose metabolism. Eur J Endocrinol 2014; 170:R43-55. [PMID: 24154684 DOI: 10.1530/eje-13-0586] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
During the past years, targeted therapies for cancer have been developed using drugs that have significant metabolic consequences. Among them, the mammalian target of rapamycin (mTOR) inhibitors and, to a much lesser extent, the tyrosine kinase inhibitors (TKIs) are involved. mTOR plays a key role in the regulation of cell growth as well as lipid and glucose metabolism. Treatment with mTOR inhibitors is associated with a significant increase in plasma triglycerides and LDL cholesterol. mTOR inhibitors seem to increase plasma triglycerides by reducing the activity of the lipoprotein lipase which is in charge of the catabolism of triglyceride-rich lipoproteins. The increase in LDL cholesterol observed with mTOR inhibitors seems to be due to a decrease in LDL catabolism secondary to a reduction of LDL receptor expression. In addition, treatment with mTOR inhibitors is associated with a high incidence of hyperglycemia, ranging from 13 to 50% in the clinical trials. The mechanisms responsible for hyperglycemia with new onset diabetes are not clear, but are likely due to the combination of impaired insulin secretion and insulin resistance. TKIs do not induce hyperlipidemia but alter glucose homeostasis. Treatment with TKIs may be associated either with hyperglycemia or hypoglycemia. The molecular mechanism by which TKIs control glucose homeostasis remains unknown. Owing to the metabolic consequences of these agents used as targeted anti-cancer therapies, a specific and personalized follow-up of blood glucose and lipids is recommended when using mTOR inhibitors and of blood glucose when using TKIs.
Collapse
Affiliation(s)
- Bruno Vergès
- Service Endocrinologie, Diabétologie et Maladies Métaboliques, INSERM CRI 866, Hôpital du Bocage, CHU Dijon, Université de Bourgogne, 21000 Dijon, France
| | | | | |
Collapse
|
3012
|
Sunakawa Y, Furuse J, Okusaka T, Ikeda M, Nagashima F, Ueno H, Mitsunaga S, Hashizume K, Ito Y, Sasaki Y. Regorafenib in Japanese patients with solid tumors: phase I study of safety, efficacy, and pharmacokinetics. Invest New Drugs 2014; 32:104-112. [PMID: 23553067 PMCID: PMC3913857 DOI: 10.1007/s10637-013-9953-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 03/18/2013] [Indexed: 12/19/2022]
Abstract
The safety, pharmacokinetics, and antitumor activity of the multikinase inhibitor regorafenib in Japanese patients was assessed in this multicenter, single-arm, phase I trial. Fifteen patients with treatment-refractory advanced solid tumors received regorafenib 160 mg once daily for the first 3 weeks of each 4-week cycle until disease progression, unacceptable toxicity, or investigator or patient decision to stop. The median duration of treatment was 2.1 months (range, 0.9-20.1 months). At data cutoff, one patient was still receiving regorafenib in cycle 21. Reasons for treatment discontinuation were disease progression (n = 12) and adverse events (liver enzyme elevation n = 1; anemia n = 1). Adverse events necessitated dose reduction in six patients, interruption of daily treatment in seven patients, and cycle delay in four patients. All patients experienced at least one drug-related adverse event, particularly gastrointestinal (87 %), dermatologic (73 %), or hematologic (67 %) events. There was no significant change in time to maximum concentration or terminal half-life of regorafenib and its active metabolites M2 and M5 between single dosing and 21-day continuous dosing. The area under the concentration-time curve was 2.1-fold higher for regorafenib, 5.2-fold higher for M2, and 37.3-fold higher for M5, and the maximum concentration was 2.0-fold, 4.8-fold, and 36.0-fold higher, respectively, after continuous dosing than after single dosing. One patient had a partial response (duration 10.5 months) and seven patients had stable disease. This study indicates that regorafenib 160 mg orally once daily (21 days on/7 days off treatment) can be given to Japanese patients who have solid tumors, without undue toxicity.
Collapse
Affiliation(s)
- Yu Sunakawa
- International Medical Center-Comprehensive Cancer Center, Saitama Medical University, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
3013
|
Gil Delgado M, Delgado FM, Khayat D. Role of aflibercept in the treatment of advanced colorectal cancer. COLORECTAL CANCER 2014. [DOI: 10.2217/crc.13.78] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
SUMMARY Colorectal cancer is the third most common cancer in men, representing 10% of cases, and the second most common cancer in women, representing 9.4% of cases, worldwide. It is estimated that 50–60% of patients will develop metastases during the course of their disease, while 15–25% will present with synchronous metastatic lesions, reducing the probability of 5-year survival to 12%. Colon cancer represents the fourth most common cause of death from cancer worldwide and the second most common cause in developed countries. First-line therapy of advanced or metastatic colorectal carcinoma (mCRC) usually consists of the administration of oxaliplatin or irinotecan in combination with leucovorin and 5-fluorouracil. In the first- or second-line settings, monoclonal antibodies can be added to chemotherapy. Bevacizumab (anti-VEGF monoclonal antibody) is utilized with fluoropyrimidine-containing regimens in combination with oxaliplatin or irinotecan. Cetuximab can be administered in combination with irinotecan or as a single agent in patients who have wild-type KRAS. Panitumumab has also been approved for third-line single-agent therapy in KRAS wild-type patients and recently in first- and second-line therapy in combination with chemotherapy. There are no standard second-line treatments of mCRC specifically used in combination with FOLFIRI, and no drugs have been approved for patients with prior bevacizumab treatment. Aflibercept is a new angiogenesis inhibitor with a unique mechanism of action. Aflibercept is a recombinant fusion protein consisting of VEGF-binding portions from extracellular domains of the human VEGFR1 and 2 fused to the Fc portion of human IgG1. Aflibercept acts as a soluble decoy receptor that binds to VEGF-A with higher affinity than its native receptors, as well as the related ligands PIGF and VEGF-B. Aflibercept interferes with the biological actions of VEGF by ‘trapping’ VEGF and preventing it from interacting with its receptors on endothelial cells, blocking the activation of VEGFRs and the subsequent proliferation of endothelial cells; this results in inhibition of the growth of new vessels. The role of aflibercept in the management of mCRC was studied in the VELOUR trial, which showed that its addition to the FOLFIRI regimen resulted in a survival benefit, giving a statistically significant log-rank test with p = 0.0032 (hazard ratio: 0.817; 95.34% CI: 0.713–0.937). This benefit was supported by subgroup and sensitivity analyses, the increased median progression-free survival, and improved response rates observed in the aflibercept arm. Aflibercept toxicity is acceptable, manageable and within the established range of other VEGF inhibitors.
Collapse
Affiliation(s)
- Marian Gil Delgado
- Groupe Hospitalier Pitié Salpêtrière, Service d’Oncologie Médicale, 47 Boulevard de l’Hôpital, 75013 Paris, France
| | - François Michel Delgado
- Groupe Hospitalier Pitié Salpêtrière, Service d’Oncologie Médicale, 47 Boulevard de l’Hôpital, 75013 Paris, France
| | - David Khayat
- Groupe Hospitalier Pitié Salpêtrière, Service d’Oncologie Médicale, 47 Boulevard de l’Hôpital, 75013 Paris, France
| |
Collapse
|
3014
|
Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis 2014; 17:471-94. [PMID: 24482243 PMCID: PMC4061466 DOI: 10.1007/s10456-014-9420-y] [Citation(s) in RCA: 529] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 01/15/2014] [Indexed: 12/17/2022]
Abstract
Tumours require a vascular supply to grow and can achieve this via the expression of pro-angiogenic growth factors, including members of the vascular endothelial growth factor (VEGF) family of ligands. Since one or more of the VEGF ligand family is overexpressed in most solid cancers, there was great optimism that inhibition of the VEGF pathway would represent an effective anti-angiogenic therapy for most tumour types. Encouragingly, VEGF pathway targeted drugs such as bevacizumab, sunitinib and aflibercept have shown activity in certain settings. However, inhibition of VEGF signalling is not effective in all cancers, prompting the need to further understand how the vasculature can be effectively targeted in tumours. Here we present a succinct review of the progress with VEGF-targeted therapy and the unresolved questions that exist in the field: including its use in different disease stages (metastatic, adjuvant, neoadjuvant), interactions with chemotherapy, duration and scheduling of therapy, potential predictive biomarkers and proposed mechanisms of resistance, including paradoxical effects such as enhanced tumour aggressiveness. In terms of future directions, we discuss the need to delineate further the complexities of tumour vascularisation if we are to develop more effective and personalised anti-angiogenic therapies.
Collapse
|
3015
|
Sherman SK, Maxwell JE, Carr JC, Wang D, O'Dorisio MS, O'Dorisio TM, Howe JR. GIPR expression in gastric and duodenal neuroendocrine tumors. J Surg Res 2014; 190:587-93. [PMID: 24565507 DOI: 10.1016/j.jss.2014.01.044] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 01/16/2014] [Accepted: 01/24/2014] [Indexed: 01/14/2023]
Abstract
BACKGROUND Compounds targeting somatostatin-receptor-type-2 (SSTR2) are useful for small bowel neuroendocrine tumor (SBNET) and pancreatic neuroendocrine tumor (PNET) imaging and treatment. We recently characterized expression of 13 cell surface receptor genes in SBNETs and PNETs, identifying three drug targets (GIPR, OXTR, and OPRK1). This study set out to characterize expression of this gene panel in the less common neuroendocrine tumors of the stomach and duodenum (gastric and duodenal neuroendocrine tumors [GDNETs]). METHODS Primary tumors and adjacent normal tissue were collected at surgery, RNA was extracted, and expression of 13 target genes was determined by quantitative polymerase chain reaction. Expression was normalized to GAPDH and POLR2A internal control genes. Expression relative to normal tissue (ddCT) and absolute expression (dCT) were calculated. Wilcoxon tests compared median expression with false discovery rate correction for multiple comparisons. RESULTS Gene expression was similar in two gastric and seven duodenal tumors, and these were analyzed together. Like SBNETs (n = 63) and PNETs (n = 51), GDNETs showed significant overexpression compared with normal tissue of BRS3, GIPR, GRM1, GPR113, OPRK1, and SSTR2 (P < 0.05 for all). Of these, SSTR2 had the highest absolute expression in GDNETs (median dCT 4.0). Absolute expression of BRS3, GRM1, GPR113, and OPRK1 was significantly lower than SSTR2 in GDNETs (P < 0.05 for all), whereas expression of GIPR was similar to SSTR2 (median 4.3, P = 0.4). CONCLUSIONS As in SBNETs and PNETs, GIPR shows absolute expression close to SSTR2 but has greater overexpression relative to normal tissue (21.1 versus 3.5-fold overexpression). We conclude that GIPR could provide an improved signal-to-noise ratio for imaging versus SSTR2 and represents a promising novel therapeutic target in GDNETs.
Collapse
Affiliation(s)
- Scott K Sherman
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Jessica E Maxwell
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Jennifer C Carr
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Donghong Wang
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - M Sue O'Dorisio
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Thomas M O'Dorisio
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - James R Howe
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa.
| |
Collapse
|
3016
|
Lee J, Hong YS, Hong JY, Han SW, Kim TW, Kang HJ, Kim TY, Kim KP, Kim SH, Do IG, Kim KM, Sohn I, Park SH, Park JO, Lim HY, Cho YB, Lee WY, Yun SH, Kim HC, Park YS, Kang WK. Effect of simvastatin plus cetuximab/irinotecan for KRAS mutant colorectal cancer and predictive value of the RAS signature for treatment response to cetuximab. Invest New Drugs 2014; 32:535-41. [DOI: 10.1007/s10637-014-0065-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 01/10/2014] [Indexed: 12/13/2022]
|
3017
|
Colorectal cancer in 2013: Towards improved drugs, combinations and patient selection. Nat Rev Clin Oncol 2014; 11:79-80. [PMID: 24445520 DOI: 10.1038/nrclinonc.2013.254] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
3018
|
Le Tourneau C, Paoletti X, Coquan E, Sablin MP, Zoubir M, Tannock IF. Critical Evaluation of Disease Stabilization As a Measure of Activity of Systemic Therapy: Lessons From Trials With Arms in Which Patients Do Not Receive Active Treatment. J Clin Oncol 2014; 32:260-3. [DOI: 10.1200/jco.2013.53.5518] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Christophe Le Tourneau
- Institut Curie and Institut National de la Sante et de la Recherche Medicale U900, Paris, France
| | - Xavier Paoletti
- Institut Curie and Institut National de la Sante et de la Recherche Medicale U900, Paris, France
| | | | | | | | - Ian F. Tannock
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
3019
|
Pancione M, Giordano G, Remo A, Febbraro A, Sabatino L, Manfrin E, Ceccarelli M, Colantuoni V. Immune escape mechanisms in colorectal cancer pathogenesis and liver metastasis. J Immunol Res 2014; 2014:686879. [PMID: 24741617 PMCID: PMC3987978 DOI: 10.1155/2014/686879] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 11/18/2013] [Accepted: 11/18/2013] [Indexed: 02/08/2023] Open
Abstract
Over the past decade, growing evidence indicates that the tumor microenvironment (TME) contributes with genomic/epigenomic aberrations of malignant cells to enhance cancer cells survival, invasion, and dissemination. Many factors, produced or de novo synthesized by immune, stromal, or malignant cells, acting in a paracrine and autocrine fashion, remodel TME and the adaptive immune response culminating in metastasis. Taking into account the recent accomplishments in the field of immune oncology and using metastatic colorectal cancer (mCRC) as a model, we propose that the evasion of the immune surveillance and metastatic spread can be achieved through a number of mechanisms that include (a) intrinsic plasticity and adaptability of immune and malignant cells to paracrine and autocrine stimuli or genotoxic stresses; (b) alteration of positional schemes of myeloid-lineage cells, produced by factors controlling the balance between tumour-suppressing and tumour-promoting activities; (c) acquisition by cancer cells of aberrant immune-phenotypic traits (NT5E/CD73, CD68, and CD163) that enhance the interactions among TME components through the production of immune-suppressive mediators. These properties may represent the driving force of metastatic progression and thus clinically exploitable for cancer prevention and therapy. In this review we summarize results and suggest new hypotheses that favour the growing impact of tumor-infiltrating immune cells on tumour progression, metastasis, and therapy resistance.
Collapse
Affiliation(s)
- Massimo Pancione
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
| | - Guido Giordano
- Medical Oncology Unit, Fatebenefratelli Hospital, 82100 Benevento, Italy
| | - Andrea Remo
- Department of Pathology “Mater Salutis” Hospital, 37045 Legnago (VR), Italy
| | - Antonio Febbraro
- Medical Oncology Unit, Fatebenefratelli Hospital, 82100 Benevento, Italy
| | - Lina Sabatino
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
| | - Erminia Manfrin
- Department of Surgery and Oncology, University of Verona, 37129 Verona, Italy
| | - Michele Ceccarelli
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
- Bioinformatics Lab, BIOGEM scrl, 83031 Ariano Irpino (AV), Italy
| | - Vittorio Colantuoni
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
| |
Collapse
|
3020
|
Ezziddin S, Attassi M, Yong-Hing CJ, Ahmadzadehfar H, Willinek W, Grünwald F, Guhlke S, Biersack HJ, Sabet A. Predictors of Long-Term Outcome in Patients with Well-Differentiated Gastroenteropancreatic Neuroendocrine Tumors After Peptide Receptor Radionuclide Therapy with 177Lu-Octreotate. J Nucl Med 2014; 55:183-90. [DOI: 10.2967/jnumed.113.125336] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
3021
|
Ahmed S, Howel D, Debrah S. The influence of age on the outcome of treatment of elderly patients with colorectal cancer. J Geriatr Oncol 2014; 5:133-40. [PMID: 24495704 DOI: 10.1016/j.jgo.2013.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 08/22/2013] [Accepted: 12/31/2013] [Indexed: 12/13/2022]
Abstract
OBJECTIVES We investigated factors associated with post-operative mortality rates in those aged ≥60, and in particular, the relative survival of age bands within this group. METHODS Secondary analysis of a large comprehensive cohort of the elderly treated for colorectal cancer in the North of England during 1998-2003. We investigated seven risk factors associated with 30-day and 6-month post-operative mortality from colorectal surgery. RESULTS 6083 patients aged ≥60 underwent colorectal cancer surgery. Approximately 8% had died within 30 days of surgery and 17% had died within 6 months. Thirty-day mortality was greater in the elderly (80 years+) compared to the young-old (60-69 years) (adjusted OR: 3.2, 95% CI 2.4 to 4.4). There was neither a significant difference between the proportions offered curative resections across the age-groups, nor was there a significant association between intent of surgery and 30-day mortality. Six-month mortality rose with age, but the association was stronger in those having curative surgery (adjusted OR: 3.8, 95% CI 2.8 to 5.2) than palliative surgery (adjusted OR: 1.5, 95% CI 1.1 to 2.1). Mortality from emergency surgery at 6-months was particularly high in elderly females. CONCLUSIONS This large population study adds more weight to the findings that age itself is a major risk factor in the outcome of colorectal surgery in elderly and that 30-day mortality underestimates the longer-term outcome in this age group. There was no significant association between radical resections and 30-day mortality in elderly patients compared to the younger age groups; however, a disproportionately higher mortality at 6 months was seen in elderly female patients.
Collapse
Affiliation(s)
- Sohail Ahmed
- Darlington Memorial Hospital, Hollyhurst Road, Darlington DL3-6HX, UK.
| | - Denise Howel
- Institute of Health & Society, Newcastle University, Baddiley-Clark Building, Richardson Road, Newcastle-upon-Tyne NE2-4AX, UK.
| | - Samuel Debrah
- Darlington Memorial Hospital, Darlington DL3-6HX, UK.
| | | |
Collapse
|
3022
|
Bekaii-Saab T, Wu C. Seeing the forest through the trees: a systematic review of the safety and efficacy of combination chemotherapies used in the treatment of metastatic colorectal cancer. Crit Rev Oncol Hematol 2014; 91:9-34. [PMID: 24534706 DOI: 10.1016/j.critrevonc.2014.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 12/20/2013] [Accepted: 01/09/2014] [Indexed: 12/15/2022] Open
Abstract
Combinations of fluoropyrimidines with oxaliplatin or irinotecan plus a biologic agent are standard treatments for metastatic colorectal cancer (mCRC). Recent approvals of first-line cetuximab, second-line ziv-aflibercept, and regorafenib as salvage therapy have increased the complexity of the treatment armamentarium. To define optimal regimens, we systematically reviewed combination chemotherapy trials (N=83). Descriptive analyses focusing on fluoropyrimidine formulation, oxaliplatin vs irinotecan combinations, and compatibility with biologics indicated the following: infusional 5-fluorouracil (5-FU) yielded better efficacy and safety than bolus 5-FU. Capecitabine had similar outcomes and better safety than 5-FU with oxaliplatin but not irinotecan. First-line oxaliplatin and irinotecan appeared equivalent. Antiangiogenics, such as bevacizumab and ziv-aflibercept, and epidermal growth factor receptor-targeted monoclonal antibodies cetuximab and panitumumab further improved efficacy. The treatment landscape for mCRC has become complex, and we are approaching individualized therapy based on predictive factors, including KRAS mutational status. Appropriate administration of chemotherapy/biologic combinations is critical.
Collapse
Affiliation(s)
- Tanios Bekaii-Saab
- The Ohio State University Comprehensive Cancer Center, A454 Starling Loving Hall, 320 West 10th Avenue, Columbus, OH 43210, United States.
| | - Christina Wu
- The Ohio State University Comprehensive Cancer Center, A454 Starling Loving Hall, 320 West 10th Avenue, Columbus, OH 43210, United States.
| |
Collapse
|
3023
|
Hocking CM, Townsend AR, Price TJ. Panitumumab in metastatic colorectal cancer. Expert Rev Anticancer Ther 2014; 13:781-93. [DOI: 10.1586/14737140.2013.811064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
3024
|
|
3025
|
Sorafenib and irinotecan (NEXIRI) as second- or later-line treatment for patients with metastatic colorectal cancer and KRAS-mutated tumours: a multicentre Phase I/II trial. Br J Cancer 2014; 110:1148-54. [PMID: 24407191 PMCID: PMC3950852 DOI: 10.1038/bjc.2013.813] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 10/21/2013] [Accepted: 12/10/2013] [Indexed: 12/28/2022] Open
Abstract
Background: This trial evaluated the feasibility and efficacy of combined sorafenib and irinotecan (NEXIRI) as second- or later-line treatment of patients with KRAS-mutated metastatic colorectal cancer (mCRC), who had progressed after irinotecan-based chemotherapy. Methods: In Phase I, in a 3+3 dose escalation schedule, patients received irinotecan (125, 150 or 180 mg m−2 every 2 weeks), in combination with 400 mg sorafenib b.d. The primary end point was the maximum-tolerated dose of irinotecan. In Phase II, the primary end point was disease control rate (DCR). Secondary end points were progression-free survival (PFS), overall survival (OS) and toxicity. Results: Phase I included 10 patients (median age 63 (49–73)); no dose-limiting toxicity was seen. In Phase II, 54 patients (median age 60 (43–80) years) received irinotecan 180 mg m−2 every 2 weeks with sorafenib 400 mg b.d. Nine patients (17%) remained on full-dose sorafenib. The DCR was 64.9% (95% CI, 51–77). Median PFS and OS were 3.7 (95% CI, 3.2–4.7) and 8.0 (95% CI, 4.8–9.7) months, respectively. Toxicities included Grade 3 diarrhoea (37%), neutropenia (18%), hand-foot syndrome (13%) and Grade 4 neutropenia (17%). Conclusion: The NEXIRI regimen showed promising activity as second- or later-line treatment in this heavily pretreated mCRC population (ClinicalTrials.gov NCT00989469).
Collapse
|
3026
|
Qi WX, Tang LN, Shen Z, Yao Y. Treatment-related mortality with aflibercept in cancer patients: a meta-analysis. Eur J Clin Pharmacol 2014; 70:461-7. [PMID: 24390630 DOI: 10.1007/s00228-013-1633-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 12/17/2013] [Indexed: 12/21/2022]
Abstract
PURPOSE Aflibercept, a fully humanized vascular endothelial growth factor (VEGF)-targeted agent, has emerged as an effective therapy in the treatment of various solid tumors. We carried out an up-to-date meta-analysis to determine the risk of fatal adverse events (FAEs) in cancer patients treated with aflibercept. METHODS We searched databases such as PubMed and Web of Science, and abstracts presented at the American Society of Clinical Oncology (ASCO) and the European Society of Medical Oncology (ESMO) meetings for records up to August 2013 to identify relevant studies. Eligible studies included prospective phase II and III trials evaluating aflibercept in cancer patients with adequate data on FAEs. Statistical analyses were conducted to calculate the summary incidence, odds ratio (OR) and 95 % confidence intervals (CIs) by using either random effects or fixed-effect models according to the heterogeneity of included studies. RESULTS A total of 3,060 patients with a variety of solid tumors from ten clinical trials were included in our analysis. The overall incidence of FAEs associated with aflibercept was 5.1 % (95%CI: 3.8-6.8 %). The use of aflibercept significantly increased the risk of FAEs compared to patients treated with control medication (OR 1.81, 95 % CI: 1.20-2.72, p = 0.004). Additionally, the most common causes of FAEs were infection (38.8 %), hemorrhage (5.9 %) and GI perforation (5.9 %), respectively. CONCLUSIONS With available evidence, the use of aflibercept is associated with an increased risk of FAEs compared to controls. Further studies are still needed to investigate this association. In the appropriate clinical scenario, the use of aflibercept remains justified in its approved indications.
Collapse
Affiliation(s)
- Wei-Xiang Qi
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yishan road, Shanghai, 200233, China
| | | | | | | |
Collapse
|
3027
|
Fendrich V, Wichmann S, Wiese D, Waldmann J, Lauth M, Rexin P, L-Lopez C, Schlitt HJ, Bartsch DK, Lang SA. Inhibition of heat shock protein 90 with AUY922 represses tumor growth in a transgenic mouse model of islet cell neoplasms. Neuroendocrinology 2014; 100:300-9. [PMID: 25301256 DOI: 10.1159/000368610] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 09/18/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND This study was designed to evaluate the role of heat shock protein 90 (HSP90) in tumor progression of murine islet cell tumors. Blockade of HSP90 has recently been proposed as a therapeutic target, but effects in models of islet cell tumors with AUY922, a newly developed HSP90 inhibitor, have not been examined. MATERIAL AND METHODS The carcinoid cell line BON-1 and the HSP90 inhibitor AUY922 were used to determine effects on signaling and growth in vitro. In vivo transgenic RIP1-Tag2 mice, which develop islet cell neoplasms, were treated with vehicle or AUY922 (25 mg/kg/twice per week) from week 5 until death. The resected pancreata were evaluated macroscopically and microscopically by immunohistochemistry. Quantitative real-time PCR was performed for HSP90 targets with RNA from islets isolated from treated and untreated RIP1-Tag2 mice. RESULTS HSP90 blockade impaired constitutive and growth factor-induced signaling in vitro. Moreover, HSP90 inhibition attenuated in vitro cell growth in a dose-dependent manner. In vivo, AUY922 significantly reduced tumor volume by 92% compared to untreated controls (p = 0.000), and median survival in the used transgenic mouse model was prolonged (110 vs. 119 days; p = 0.75). Quantitative real-time PCR for downstream target genes of HSP90 demonstrated significant downregulation in the islet cell tumors of RIP1-Tag2 mice treated with AUY922, confirming our ability to achieve effective pharmacologic levels of AUY922 within the desired tissue site in vivo. CONCLUSION This is the first study to show that the HSP90 antagonist AUY922 may provide a new option for therapy of islet cell neoplasms.
Collapse
Affiliation(s)
- Volker Fendrich
- Department of Surgery, Philipps University Marburg, Marburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
3028
|
Kesavan M, Claringbold PG, Turner JH. Hematological toxicity of combined 177Lu-octreotate radiopeptide chemotherapy of gastroenteropancreatic neuroendocrine tumors in long-term follow-up. Neuroendocrinology 2014; 99:108-17. [PMID: 24714208 DOI: 10.1159/000362558] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 03/28/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND The combination of radiopeptide therapy [peptide receptor radionuclide therapy (PRRT)] with radiosensitizing chemotherapy of gastroenteropancreatic neuroendocrine tumors (GEP NETs) may improve efficacy, but has the potential to increase myelotoxicity. In a prospective clinical study of GEP NET patients treated with (177)Lu-octreotate PRRT in combination with capecitabine and temozolomide, as a prelude to a planned Australasian Gastro-Intestinal Trials Group (AGITG) international randomized controlled trial, we characterized the incidence and degree of hematological toxicity. MATERIALS AND METHODS Well-differentiated progressive metastatic GEP NETs in 65 patients were treated with 4 cycles of 7.8 GBq (177)Lu-octreotate, 1,650 mg/m(2) capecitabine (n = 28) and 1,500 mg/m(2) capecitabine with 200 mg/m(2) temozolomide (n = 37), and monitored for hematological toxicity over a 5-year period. RESULTS Short-term, self-limited hematological toxicity grade 3/4 comprised anemia in 1 patient (3.5%) in the 28 patient-cohort of patients treated with (177)Lu-octreotate and capecitabine. One of these patients (3.5%) later developed significant anemia and one developed thrombocytopenia (3.5%) over a median follow-up of 60 months (SD 20). The incidence of short-term grade 3/4 reversible myelosuppression in 37 patients after (177)Lu-octreotate/capecitabine/temozolomide was zero. Long- term follow-up for a median of 36 months (SD 11) showed significant thrombocytopenia in 2.7% and neutropenia in 2.7% of the patients and anemia in 10.8% of the patients (n = 4). The 3-year median hemoglobin and platelet and neutrophil counts trended downwards, but remained within normal ranges. Two patients in this cohort developed myelodysplastic syndrome. CONCLUSION The modest reversible hematological toxicity of PRRT of GEP NETs is not significantly increased by the addition of radiosensitizing chemotherapy with capecitabine and temozolomide in combination with (177)Lu-octreotate, which has the potential to enhance the efficacy of radiopeptide therapy.
Collapse
Affiliation(s)
- Murali Kesavan
- Department of Hematology, Fremantle Hospital, The University of Western Australia, Fremantle, W.A., Australia
| | | | | |
Collapse
|
3029
|
Yamamoto Y, Matsui J, Matsushima T, Obaishi H, Miyazaki K, Nakamura K, Tohyama O, Semba T, Yamaguchi A, Hoshi S, Mimura F, Haneda T, Fukuda Y, Kamata JI, Takahashi K, Matsukura M, Wakabayashi T, Asada M, Nomoto KI, Watanabe T, Dezso Z, Yoshimatsu K, Funahashi Y, Tsuruoka A. Lenvatinib, an angiogenesis inhibitor targeting VEGFR/FGFR, shows broad antitumor activity in human tumor xenograft models associated with microvessel density and pericyte coverage. Vasc Cell 2014; 6:18. [PMID: 25197551 PMCID: PMC4156793 DOI: 10.1186/2045-824x-6-18] [Citation(s) in RCA: 368] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 08/04/2014] [Indexed: 12/13/2022] Open
Abstract
Background Lenvatinib is an oral inhibitor of multiple receptor tyrosine kinases (RTKs) targeting vascular endothelial growth factor receptor (VEGFR1-3), fibroblast growth factor receptor (FGFR1-4), platelet growth factor receptor α (PDGFR α), RET and KIT. Antiangiogenesis activity of lenvatinib in VEGF- and FGF-driven angiogenesis models in both in vitro and in vivo was determined. Roles of tumor vasculature (microvessel density (MVD) and pericyte coverage) as biomarkers for lenvatinib were also examined in this study. Method We evaluated antiangiogenesis activity of lenvatinib against VEGF- and FGF-driven proliferation and tube formation of HUVECs in vitro. Effects of lenvatinib on in vivo angiogenesis, which was enhanced by overexpressed VEGF or FGF in human pancreatic cancer KP-1 cells, were examined in the mouse dorsal air sac assay. We determined antitumor activity of lenvatinib in a broad panel of human tumor xenograft models to test if vascular score, which consisted of high MVD and low pericyte coverage, was associated with sensitivity to lenvatinib treatment. Vascular score was also analyzed using human tumor specimens with 18 different types of human primary tumors. Result Lenvatinib inhibited VEGF- and FGF-driven proliferation and tube formation of HUVECs in vitro. In vivo angiogenesis induced by overexpressed VEGF (KP-1/VEGF transfectants) or FGF (KP-1/FGF transfectants) was significantly suppressed with oral treatments of lenvatinib. Lenvatinib showed significant antitumor activity in KP-1/VEGF and five 5 of 7 different types of human tumor xenograft models at between 1 to 100 mg/kg. We divided 19 human tumor xenograft models into lenvatinib-sensitive (tumor-shrinkage) and relatively resistant (slow-growth) subgroups based on sensitivity to lenvatinib treatments at 100 mg/kg. IHC analysis showed that vascular score was significantly higher in sensitive subgroup than relatively resistant subgroup (p < 0.0004). Among 18 types of human primary tumors, kidney cancer had the highest MVD, while liver cancer had the lowest pericyte coverage, and cancers in Kidney and Stomach had highest vascular score. Conclusion These results indicated that Lenvatinib inhibited VEGF- and FGF-driven angiogenesis and showed a broad spectrum of antitumor activity with a wide therapeutic window. MVD and pericyte-coverage of tumor vasculature might be biomarkers and suggest cases that would respond for lenvatinib therapy.
Collapse
|
3030
|
Wong A, Ma BBY. Personalizing therapy for colorectal cancer. Clin Gastroenterol Hepatol 2014; 12:139-44. [PMID: 24025538 DOI: 10.1016/j.cgh.2013.08.040] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 08/26/2013] [Accepted: 08/26/2013] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is the third most commonly diagnosed cancer worldwide. Several important scientific discoveries in the molecular biology of CRC have changed clinical practice in oncology. These included the comprehensive genome-wide profiling of CRC by the Cancer Genome Atlas Network, the discovery of mutations along the RAS-RAF signaling pathway as major determinants of response to antibodies against the epidermal growth factor receptor, the elucidation of new molecular subsets of CRC or gene signatures that may predict clinical outcome after adjuvant chemotherapy, and the innovative targeting of the family of vascular endothelial growth factor and receptors. These new data have allowed oncologists to individualize drug therapy on the basis of a patient's tumor's unique molecular profile, especially in the management of metastatic CRC. This review article will discuss the progress of personalized medicine in the contemporary management of CRC.
Collapse
Affiliation(s)
- Ashley Wong
- Sir Y. K. Pao Centre for Cancer, Department of Clinical Oncology, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Brigette B Y Ma
- Sir Y. K. Pao Centre for Cancer, Department of Clinical Oncology, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
3031
|
Cingarlini S, Bonomi M, Trentin C, Corbo V, Scarpa A, Tortora G. Profiling mTOR Pathway in Neuroendocrine Tumors. MANAGEMENT OF NEUROENDOCRINE TUMORS OF THE PANCREAS AND DIGESTIVE TRACT 2014:9-27. [DOI: 10.1007/978-2-8178-0430-9_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
3032
|
Abstract
BACKGROUND Breast cancer cells can develop resistance to standard hormonal treatment and chemotherapy with the activation of the mTOR pathway; this is supported by results of preclinical and clinical studies. In clinical trials, the addition of everolimus to hormonal treatment or anti-HER2 treatment improved the outcomes of breast cancer patients. The aim of this review is to discuss the efficacy and safety data of everolimus in all categories of breast cancer in recent published studies. SCOPE Everolimus showed positive results in clinical studies. A literature search was made from PubMed, ASCO and San Antonio Breast Cancer Symposium Meeting abstracts by using the following search key words: 'everolimus', 'RAD001', 'mTOR inhibitor', 'breast cancer' 'endocrine therapy resistance' and 'HER-2 targeted therapies'. The last search was on June 10, 2013. The most important limitation of our review is that most of the data on everolimus rely on phase I and II trials. FINDINGS Preclinical studies showed that mTOR activation can be the responsible mechanism in all subgroups of breast cancer. Results of both the TAMRAD and BOLERO-2 studies have showed that mTOR inhibition in combination with endocrine therapy can be a new treatment strategy for MBC patients who are resistant to aromatase inhibitors. In the BOLERO-2 study, time to deterioration in health-related quality of life was also significantly higher in the everolimus and exemestane arm compared to the exemestane plus placebo arm. The recently completed BOLERO-3 study showed that mTOR inhibition in combination with trastuzumab plus vinorelbine treatment significantly improved PFS compared to trastuzumab plus vinorelbine alone in trastuzumab-resistant MBC patients. CONCLUSION Recent trials have shown that everolimus has produced promising anti-tumor activity in combination with trastuzumab in HER2-positive metastatic breast cancer and in combination with exemestane in patients with hormone-receptor-positive metastatic breast cancer who had recurrence or progression while receiving a nonsteroidal aromatase inhibitor. Results of ongoing studies with everolimus show evidence that using everolimus in earlier stages of the disease, namely in the adjuvant and neoadjuvant settings, could be benefical.
Collapse
Affiliation(s)
- Mehmet A N Sendur
- Ankara Numune Education and Research Hospital, Department of Medical Oncology , Ankara , Turkey
| | | | | | | |
Collapse
|
3033
|
Rusthoven CG, Schefter TE. Rationale for ablation of oligometastatic disease and the role of stereotactic body radiation therapy for hepatic metastases. Hepat Oncol 2014; 1:81-94. [PMID: 30190943 PMCID: PMC6114003 DOI: 10.2217/hep.13.12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Management paradigms for metastatic solid tumors are evolving. Once regarded as uniformly incurable, today there is recognition of an intermediate oligometastatic state, where ablation of metastatic foci may improve disease control and prolong survival. In the setting of limited colorectal liver metastases, hepatic resection has resulted in favorable long-term outcomes, but is technically unsuitable for most patients. Stereotactic body radiation therapy represents an effective, noninvasive means of tumor ablation, supported by a large body of prospective evidence specific to hepatic metastases. This review examines the current rationale for ablation of oligometastatic disease, including various objectives beyond indefinite disease-free survival. The role of stereotactic body radiation therapy for ablation of hepatic metastases is then comprehensively reviewed.
Collapse
Affiliation(s)
- Chad G Rusthoven
- Department of Radiation Oncology, University of Colorado Denver, 1665 North Aurora Court, Suite 1032, Mail Stop F706, Aurora, CO 80045, USA
| | - Tracey E Schefter
- Department of Radiation Oncology, University of Colorado Denver, 1665 North Aurora Court, Suite 1032, Mail Stop F706, Aurora, CO 80045, USA
| |
Collapse
|
3034
|
Abstract
Everolimus (RAD001, Afinitor®) is an oral protein kinase inhibitor of the mammalian target of rapamycin (mTOR) serine/threonine kinase signal transduction pathway. The mTOR pathway regulates cell growth, proliferation, and survival and is frequently deregulated in cancer. Everolimus has been approved by the FDA and the EMA for the treatment of advanced renal cell carcinoma (RCC), subependymal giant cell astrocytoma (SEGA) associated with tuberous sclerosis (TSC), pancreatic neuroendocrine tumors (PNET), in combination with exemestane in advanced hormone-receptor (HR)-positive, HER2-negative breast cancer. Everolimus shows promising clinical activity in additional indications. Multiple phase 2 and phase 3 trials of everolimus alone or in combination are ongoing and will help to further elucidate the role of mTOR in oncology. For a review on everolimus as immunosuppressant, please consult other sources.
Collapse
Affiliation(s)
- Jens Hasskarl
- Department Innere Medizin, Klinik für Innere Medizin I, Schwerpunkt Hämatologie, Onkologie und Stammzelltransplantation, Hugstetter Str. 55, 79102, Freiburg, Germany,
| |
Collapse
|
3035
|
Hocking CM, Price TJ. Panitumumab in the management of patients with KRAS wild-type metastatic colorectal cancer. Therap Adv Gastroenterol 2014; 7:20-37. [PMID: 24381645 PMCID: PMC3871277 DOI: 10.1177/1756283x13498660] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The past 15 years has seen a marked increase in available therapeutic options for patients with metastatic colorectal cancer resulting in improvements in median survival from 12 to 24 months. One of these new options is panitumumab, which is a fully humanized monoclonal antibody that binds to the epidermal growth factor receptor of tumor cells and inhibits downstream cell signaling with antitumor effects of inhibition of tumor growth, induction of apoptosis and inhibition of angiogenesis. Large randomized clinical trials have demonstrated significant improvements in tumor response rates and progression-free survival when panitumumab is combined with chemotherapy and as monotherapy in chemorefractory metastatic colorectal cancer. Clinical benefit with panitumumab is limited to patients with nonmutated KRAS tumors. Rash is a common toxicity of panitumumab treatment but can potentially be ameliorated with the use of prophylactic strategies. The role of panitumumab in the overall treatment of metastatic colorectal cancer is evolving and future clinical trials will focus on improved patient selection through use of novel predictive biomarkers, and the optimal timing of treatment.
Collapse
Affiliation(s)
- Christopher M Hocking
- Department of Medical Oncology, The Queen Elizabeth Hospital, Woodville, SA, Australia
| | - Timothy J Price
- Department of Medical Oncology, TQEH, Woodville, Woodville Road, Woodville, SA 5011, Australia
| |
Collapse
|
3036
|
Abstract
Colorectal cancer is one of the most frequent solid tumors in the Western world. Treatment options are dependent on the stage of the disease, the performance status of the patient, and increasingly the molecular makeup of the tumor. In countries with surveillance programs, the incidence rate as well as the mortality rate has gone down because of the earlier stages at which the tumors are detected. For rectal cancer, standard of care differs from that of colon cancer with regard to perioperative treatment. In the metastatic setting, treatment options are uniform for colorectal cancer. Over the years, treatment options have emerged from single-agent 5-fluorouracil (5-FU) treatment to combination regimens using 5-FU and oxaliplatin or irinotecan or both. Treatment efficacy in the metastatic setting has been increased with the introduction of targeted substances. These include (a) the anti-vascular endothelial growth factor-A (anti-VEGF-A) antibody bevacizumab, (b) the anti-epidermal growth factor receptor (anti-EGFR) antibodies cetuximab and panitumumab, (c) the anti-angiogenic multi-kinase inhibitor regorafenib, and (d) the anti-angiogenic compound aflibercept. Anti-EGFR antibodies have shown efficacy only in the subpopulations of tumors that do not have any mutation in KRAS and NRAS exon 2, 3, 4. Physicians have the choice in the first line to use anti-EGFR or anti-VEGF inhibitors in combination with chemotherapy based on treatment goals and patient performance. In recent years, tumor location has been shown to be prognostic and predictive for clinical outcome. Right-sided sporadic colon cancers differ significantly in molecular characteristics and, with the exception of microsatellite instability (MSI-H) tumors, are associated with poor prognosis. Tumors based on hereditary non-polyposis colorectal cancer, on the other hand, have excellent prognosis in stage II and III disease. Recent efforts have focused on the molecular classification of colorectal cancer with the purpose of establishing molecularly defined subgroups.
Collapse
Affiliation(s)
- Sebastian Stintzing
- Department of Hematology and Oncology, University Hospital Grosshadern, University of Munich Marchioninistraße 15, 81377 Munich Germany
| |
Collapse
|
3037
|
Walenkamp A, Crespo G, Fierro Maya F, Fossmark R, Igaz P, Rinke A, Tamagno G, Vitale G, Öberg K, Meyer T. Hallmarks of gastrointestinal neuroendocrine tumours: implications for treatment. Endocr Relat Cancer 2014; 21:R445-60. [PMID: 25296914 DOI: 10.1530/erc-14-0106] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In the past few years, there have been advances in the treatment of neuroendocrine tumours (NETs) and improvements in our understanding of NET biology. However, the benefits to patients have been relatively modest and much remains yet to be done. The 'Hallmarks of Cancer', as defined by Hanahan and Weinberg, provide a conceptual framework for understanding the aberrations that underlie tumourigenesis and to help identify potential targets for therapy. In this study, our objective is to review the major molecular characteristics of NETs, based on the recently modified 'Hallmarks of Cancer', and highlight areas that require further research.
Collapse
Affiliation(s)
- Annemiek Walenkamp
- Department of Medical OncologyUniversity Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The NetherlandsDepartment of Medical OncologyHospital Universitario de Burgos, Avenida Islas Baleares 3, 09006 Burgos, SpainDepartment of Endocrine OncologyNational Cancer Institute, Bogotá, ColombiaDepartment of Cancer Research and Molecular MedicineNorwegian University of Science and Technology, 7491 Trondheim, Norway2nd Department of MedicineSemmelweis University, 46, Szentkiralyi Street, H-1088 Budapest, HungaryDepartment of GastroenterologyUniversity Hospital Marburg, Baldinger Strasse, Marburg D-35043, GermanyDepartment of General Internal MedicineSt Columcille's Hospital, Loughlinstown - Co., Dublin, IrelandDepartment of Clinical Sciences and Community Health (DISCCO)University of Milan, Milan, ItalyLaboratory of Endocrine and Metabolic ResearchIstituto Auxologico Italiano IRCCS, Via Zucchi 18, Cusano Milanino (MI) 20095, ItalyDepartment of Endocrine OncologyUniversity Hospital, Uppsala, SwedenUCL Cancer InstituteUCL, Huntley Street, London WC1E 6BT, UK
| | - Guillermo Crespo
- Department of Medical OncologyUniversity Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The NetherlandsDepartment of Medical OncologyHospital Universitario de Burgos, Avenida Islas Baleares 3, 09006 Burgos, SpainDepartment of Endocrine OncologyNational Cancer Institute, Bogotá, ColombiaDepartment of Cancer Research and Molecular MedicineNorwegian University of Science and Technology, 7491 Trondheim, Norway2nd Department of MedicineSemmelweis University, 46, Szentkiralyi Street, H-1088 Budapest, HungaryDepartment of GastroenterologyUniversity Hospital Marburg, Baldinger Strasse, Marburg D-35043, GermanyDepartment of General Internal MedicineSt Columcille's Hospital, Loughlinstown - Co., Dublin, IrelandDepartment of Clinical Sciences and Community Health (DISCCO)University of Milan, Milan, ItalyLaboratory of Endocrine and Metabolic ResearchIstituto Auxologico Italiano IRCCS, Via Zucchi 18, Cusano Milanino (MI) 20095, ItalyDepartment of Endocrine OncologyUniversity Hospital, Uppsala, SwedenUCL Cancer InstituteUCL, Huntley Street, London WC1E 6BT, UK
| | - Felipe Fierro Maya
- Department of Medical OncologyUniversity Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The NetherlandsDepartment of Medical OncologyHospital Universitario de Burgos, Avenida Islas Baleares 3, 09006 Burgos, SpainDepartment of Endocrine OncologyNational Cancer Institute, Bogotá, ColombiaDepartment of Cancer Research and Molecular MedicineNorwegian University of Science and Technology, 7491 Trondheim, Norway2nd Department of MedicineSemmelweis University, 46, Szentkiralyi Street, H-1088 Budapest, HungaryDepartment of GastroenterologyUniversity Hospital Marburg, Baldinger Strasse, Marburg D-35043, GermanyDepartment of General Internal MedicineSt Columcille's Hospital, Loughlinstown - Co., Dublin, IrelandDepartment of Clinical Sciences and Community Health (DISCCO)University of Milan, Milan, ItalyLaboratory of Endocrine and Metabolic ResearchIstituto Auxologico Italiano IRCCS, Via Zucchi 18, Cusano Milanino (MI) 20095, ItalyDepartment of Endocrine OncologyUniversity Hospital, Uppsala, SwedenUCL Cancer InstituteUCL, Huntley Street, London WC1E 6BT, UK
| | - Reidar Fossmark
- Department of Medical OncologyUniversity Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The NetherlandsDepartment of Medical OncologyHospital Universitario de Burgos, Avenida Islas Baleares 3, 09006 Burgos, SpainDepartment of Endocrine OncologyNational Cancer Institute, Bogotá, ColombiaDepartment of Cancer Research and Molecular MedicineNorwegian University of Science and Technology, 7491 Trondheim, Norway2nd Department of MedicineSemmelweis University, 46, Szentkiralyi Street, H-1088 Budapest, HungaryDepartment of GastroenterologyUniversity Hospital Marburg, Baldinger Strasse, Marburg D-35043, GermanyDepartment of General Internal MedicineSt Columcille's Hospital, Loughlinstown - Co., Dublin, IrelandDepartment of Clinical Sciences and Community Health (DISCCO)University of Milan, Milan, ItalyLaboratory of Endocrine and Metabolic ResearchIstituto Auxologico Italiano IRCCS, Via Zucchi 18, Cusano Milanino (MI) 20095, ItalyDepartment of Endocrine OncologyUniversity Hospital, Uppsala, SwedenUCL Cancer InstituteUCL, Huntley Street, London WC1E 6BT, UK
| | - Peter Igaz
- Department of Medical OncologyUniversity Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The NetherlandsDepartment of Medical OncologyHospital Universitario de Burgos, Avenida Islas Baleares 3, 09006 Burgos, SpainDepartment of Endocrine OncologyNational Cancer Institute, Bogotá, ColombiaDepartment of Cancer Research and Molecular MedicineNorwegian University of Science and Technology, 7491 Trondheim, Norway2nd Department of MedicineSemmelweis University, 46, Szentkiralyi Street, H-1088 Budapest, HungaryDepartment of GastroenterologyUniversity Hospital Marburg, Baldinger Strasse, Marburg D-35043, GermanyDepartment of General Internal MedicineSt Columcille's Hospital, Loughlinstown - Co., Dublin, IrelandDepartment of Clinical Sciences and Community Health (DISCCO)University of Milan, Milan, ItalyLaboratory of Endocrine and Metabolic ResearchIstituto Auxologico Italiano IRCCS, Via Zucchi 18, Cusano Milanino (MI) 20095, ItalyDepartment of Endocrine OncologyUniversity Hospital, Uppsala, SwedenUCL Cancer InstituteUCL, Huntley Street, London WC1E 6BT, UK
| | - Anja Rinke
- Department of Medical OncologyUniversity Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The NetherlandsDepartment of Medical OncologyHospital Universitario de Burgos, Avenida Islas Baleares 3, 09006 Burgos, SpainDepartment of Endocrine OncologyNational Cancer Institute, Bogotá, ColombiaDepartment of Cancer Research and Molecular MedicineNorwegian University of Science and Technology, 7491 Trondheim, Norway2nd Department of MedicineSemmelweis University, 46, Szentkiralyi Street, H-1088 Budapest, HungaryDepartment of GastroenterologyUniversity Hospital Marburg, Baldinger Strasse, Marburg D-35043, GermanyDepartment of General Internal MedicineSt Columcille's Hospital, Loughlinstown - Co., Dublin, IrelandDepartment of Clinical Sciences and Community Health (DISCCO)University of Milan, Milan, ItalyLaboratory of Endocrine and Metabolic ResearchIstituto Auxologico Italiano IRCCS, Via Zucchi 18, Cusano Milanino (MI) 20095, ItalyDepartment of Endocrine OncologyUniversity Hospital, Uppsala, SwedenUCL Cancer InstituteUCL, Huntley Street, London WC1E 6BT, UK
| | - Gianluca Tamagno
- Department of Medical OncologyUniversity Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The NetherlandsDepartment of Medical OncologyHospital Universitario de Burgos, Avenida Islas Baleares 3, 09006 Burgos, SpainDepartment of Endocrine OncologyNational Cancer Institute, Bogotá, ColombiaDepartment of Cancer Research and Molecular MedicineNorwegian University of Science and Technology, 7491 Trondheim, Norway2nd Department of MedicineSemmelweis University, 46, Szentkiralyi Street, H-1088 Budapest, HungaryDepartment of GastroenterologyUniversity Hospital Marburg, Baldinger Strasse, Marburg D-35043, GermanyDepartment of General Internal MedicineSt Columcille's Hospital, Loughlinstown - Co., Dublin, IrelandDepartment of Clinical Sciences and Community Health (DISCCO)University of Milan, Milan, ItalyLaboratory of Endocrine and Metabolic ResearchIstituto Auxologico Italiano IRCCS, Via Zucchi 18, Cusano Milanino (MI) 20095, ItalyDepartment of Endocrine OncologyUniversity Hospital, Uppsala, SwedenUCL Cancer InstituteUCL, Huntley Street, London WC1E 6BT, UK
| | - Giovanni Vitale
- Department of Medical OncologyUniversity Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The NetherlandsDepartment of Medical OncologyHospital Universitario de Burgos, Avenida Islas Baleares 3, 09006 Burgos, SpainDepartment of Endocrine OncologyNational Cancer Institute, Bogotá, ColombiaDepartment of Cancer Research and Molecular MedicineNorwegian University of Science and Technology, 7491 Trondheim, Norway2nd Department of MedicineSemmelweis University, 46, Szentkiralyi Street, H-1088 Budapest, HungaryDepartment of GastroenterologyUniversity Hospital Marburg, Baldinger Strasse, Marburg D-35043, GermanyDepartment of General Internal MedicineSt Columcille's Hospital, Loughlinstown - Co., Dublin, IrelandDepartment of Clinical Sciences and Community Health (DISCCO)University of Milan, Milan, ItalyLaboratory of Endocrine and Metabolic ResearchIstituto Auxologico Italiano IRCCS, Via Zucchi 18, Cusano Milanino (MI) 20095, ItalyDepartment of Endocrine OncologyUniversity Hospital, Uppsala, SwedenUCL Cancer InstituteUCL, Huntley Street, London WC1E 6BT, UK Department of Medical OncologyUniversity Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The NetherlandsDepartment of Medical OncologyHospital Universitario de Burgos, Avenida Islas Baleares 3, 09006 Burgos, SpainDepartment of Endocrine OncologyNational Cancer Institute, Bogotá, ColombiaDepartment of Cancer Research and Molecular MedicineNorwegian University of Science and Technology, 7491 Trondheim, Norway2nd Department of MedicineSemmelweis University, 46, Szentkiralyi Street, H-1088 Budapest, HungaryDepartment of GastroenterologyUniversity Hospital Marburg, Baldinger Strasse, Marburg D-35043, GermanyDepartment of General Internal MedicineSt Columcille's Hospital, Loughlinstown - Co., Dublin, IrelandDepartment of Clinical Sciences and Community Health (DISCCO)University of Milan, Milan, ItalyLaboratory of Endocrine and Metabolic ResearchIstituto Au
| | - Kjell Öberg
- Department of Medical OncologyUniversity Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The NetherlandsDepartment of Medical OncologyHospital Universitario de Burgos, Avenida Islas Baleares 3, 09006 Burgos, SpainDepartment of Endocrine OncologyNational Cancer Institute, Bogotá, ColombiaDepartment of Cancer Research and Molecular MedicineNorwegian University of Science and Technology, 7491 Trondheim, Norway2nd Department of MedicineSemmelweis University, 46, Szentkiralyi Street, H-1088 Budapest, HungaryDepartment of GastroenterologyUniversity Hospital Marburg, Baldinger Strasse, Marburg D-35043, GermanyDepartment of General Internal MedicineSt Columcille's Hospital, Loughlinstown - Co., Dublin, IrelandDepartment of Clinical Sciences and Community Health (DISCCO)University of Milan, Milan, ItalyLaboratory of Endocrine and Metabolic ResearchIstituto Auxologico Italiano IRCCS, Via Zucchi 18, Cusano Milanino (MI) 20095, ItalyDepartment of Endocrine OncologyUniversity Hospital, Uppsala, SwedenUCL Cancer InstituteUCL, Huntley Street, London WC1E 6BT, UK
| | - Tim Meyer
- Department of Medical OncologyUniversity Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The NetherlandsDepartment of Medical OncologyHospital Universitario de Burgos, Avenida Islas Baleares 3, 09006 Burgos, SpainDepartment of Endocrine OncologyNational Cancer Institute, Bogotá, ColombiaDepartment of Cancer Research and Molecular MedicineNorwegian University of Science and Technology, 7491 Trondheim, Norway2nd Department of MedicineSemmelweis University, 46, Szentkiralyi Street, H-1088 Budapest, HungaryDepartment of GastroenterologyUniversity Hospital Marburg, Baldinger Strasse, Marburg D-35043, GermanyDepartment of General Internal MedicineSt Columcille's Hospital, Loughlinstown - Co., Dublin, IrelandDepartment of Clinical Sciences and Community Health (DISCCO)University of Milan, Milan, ItalyLaboratory of Endocrine and Metabolic ResearchIstituto Auxologico Italiano IRCCS, Via Zucchi 18, Cusano Milanino (MI) 20095, ItalyDepartment of Endocrine OncologyUniversity Hospital, Uppsala, SwedenUCL Cancer InstituteUCL, Huntley Street, London WC1E 6BT, UK
| |
Collapse
|
3038
|
Akbar A, Bhatti ABH, Khattak S, Syed AA, Kazmi AS, Jamshed A. Outcome of rectal cancer in patients aged 30 years or less in the Pakistani population. Asian Pac J Cancer Prev 2014; 15:6339-6342. [PMID: 25124621 DOI: 10.7314/apjcp.2014.15.15.6339] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The incidence of rectal cancer is increasing in younger age groups. Limited data is available regarding survival outcome in younger patients with conflicting results from western world. The goal of this study was to determine survival in patients with rectal cancer<30 years of age and compare it with their older counterparts in the Pakistani population. MATERIALS AND METHODS A retrospective chart review of patients operated for rectal adenocarcinoma between January 2005 and December 2010 was performed. Patients were divided into two groups, Group 1 aged ≤30 years and Group 2 aged >30 years. Patient characteristics, surgical procedure, histopathological details and number of loco-regional and distant failures were compared. Expected 5 year survival was calculated using Kaplan Meier curves and significance was determined using the Log rank test. RESULTS There were 38 patients in group 1 and 144 in group 2. A significantly high number of younger patients presented with poorly differentiated histology (44.7% vs 9.7%) (p=0.0001) and advanced pathological stage (63.1% vs 38.1%) (p=0.04). Predicted overall 5 year survival was 38% versus 57% in groups I and II, respectively (p=0.05). Disease free survival was 37% versus 52% and was significantly different (p=0.007). CONCLUSIONS Early onset rectal cancer is associated with poor pathological features and a worse outcome in Pakistani population.
Collapse
Affiliation(s)
- Ali Akbar
- Department of Surgical Oncology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, Pakistan E-mail :
| | | | | | | | | | | |
Collapse
|
3039
|
Gurzu S, Szentirmay Z, Bara T, Bara T, Jung I. Myxoid variant of adrenocortical carcinoma: a report of two illustrative cases and a brief review of the literature. Pathology 2014; 46:83-85. [PMID: 24300732 DOI: 10.1097/pat.0000000000000035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Simona Gurzu
- 1Department of Pathology 2Department of Surgery, University of Medicine and Pharmacy of Tirgu-Mures, Romania 3Center of Tumors, National Institute of Oncology, Budapest, Hungary
| | | | | | | | | |
Collapse
|
3040
|
Abstract
Regorafenib (BAY 73-4506, Stivarga®) is an oral diphenylurea multikinase inhibitor that targets angiogenic (VEGFR1-3, TIE2), stromal (PDGFR-β, FGFR), and oncogenic receptor tyrosine kinases (KIT, RET, and RAF). Regorafenib is the first small-molecule multikinase inhibitor to achieve survival benefits in metastatic colorectal cancer that has progressed after all standard therapies. Consequently, regorafenib was FDA approved for this indication. In addition, regorafenib treatment resulted in a significant improvement in progression-free survival (PFS) compared with placebo in patients with metastatic gastrointestinal stromal tumors (GIST) after progression on standard treatments and is also an FDA approved indication. Currently, regorafenib is examined in several clinical trials (mostly phase II) in different tumor entities, including renal cell carcinoma (RCC), hepatocellular carcinoma (HCC), and soft tissue sarcoma (STS).
Collapse
Affiliation(s)
- Thomas J Ettrich
- Department of Internal Medicine I, University of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany,
| | | |
Collapse
|
3041
|
Actualités 2013 : le point de vue du comité de rédaction du Bulletin du Cancer. Bull Cancer 2014; 101:75-92. [DOI: 10.1684/bdc.2013.1874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
3042
|
Xu J, Tian D. Hematologic toxicities associated with mTOR inhibitors temsirolimus and everolimus in cancer patients: a systematic review and meta-analysis. Curr Med Res Opin 2014; 30:67-74. [PMID: 24028709 DOI: 10.1185/03007995.2013.844116] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Mammalian target of rapamycin (mTOR) inhibitors, temsirolimus and everolimus, are currently approved for the treatment of several malignancies. Hematological toxicities have been reported with these drugs, but overall incidence and relative risk remains undefined. We perform an up-to-date meta-analysis to determine the incidence and risk of hematologic toxicities associated with mTOR inhibitors. METHODS Several databases were searched, including PubMed, Embase and Cochrane databases. Eligible studies included prospective phase II and III trials of temsirolimus and everolimus with adequate safety data profile reporting anemia, leucopenia, neutropenia or thrombocytopenia. Overall incidence rates, relative risk (RR), and 95% confidence intervals (CI) were calculated by using either random effects or fixed effects models according to the heterogeneity of included studies. RESULTS A total of 5436 patients with a variety of solid tumors from 26 clinical trials were included for the meta-analysis. The overall incidences of mTOR inhibitor associated all-grade and high-grade hematologic toxicities were, respectively: anemia--38.8% and 7.5%; leucopenia--19.6% and 1.8%; neutropenia--14.9% and 5.6%; thrombocytopenia--33.1% and 3.6%. Compared to placebo/control arms, mTOR inhibitors were associated with a significantly increased risk of all-grade (RR 2.05, 95% CI: 1.52-2.77; p < 0.001) and high-grade anemia (RR 1.57, 95% CI: 1.20-2.05; p = 0.001), all-grade (RR 6.03, 95% CI: 2.76-13.14; p < 0.001) and high-grade thrombocytopenia (RR 2.73, 95% CI: 1.87-3.99; p < 0.001). Additionally, a non-significantly increased risk of all-grade leucopenia (RR 1.46, 95% CI: 0.66-3.23; p = 0.34) and neutropenia (RR 1.77, 95% CI: 0.80-3.93; p = 0.16) was observed in the mTOR inhibitor group, while the risk of high-grade leucopenia (RR 0.53, 95% CI: 0.31-0.90, p = 0.019) and neutropenia (RR 0.96, 95% CI: 0.62-1.51; p = 0.87) did not increase. Similar results were also observed in sub-group analysis according to mTOR inhibitor based regimens. CONCLUSIONS The use of mTOR inhibitors is associated with a significant increase in the risk of developing all-grade and high-grade anemia and thrombocytopenia compared with placebo/control arms.
Collapse
Affiliation(s)
- Jian Xu
- Department of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
| | | |
Collapse
|
3043
|
Ku GY, Ilson DH. Emerging tyrosine kinase inhibitors for esophageal cancer. Expert Opin Emerg Drugs 2013; 18:219-30. [PMID: 23725567 DOI: 10.1517/14728214.2013.805203] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Because of the poor prognosis for patients with esophagogastric cancers (EGCs), increasing attention has focused on targeted agents. AREAS COVERED Targets include epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), Her2, mammalian target of rapamycin (mTOR), and MET. We briefly discuss preclinical data and the rationale for targeting these pathways and summarize the results of clinical trials of tyrosine kinase inhibitors (TKIs) against these targets. EXPERT OPINION While anti-EGFR therapy has been extensively investigated, completed Phase III trials suggest that this is not a promising target. A Phase III trial of an anti-VEGF antibody failed to show improvement in the primary endpoint of overall survival but response rates and progression-free survival were improved; a Phase III trial of an anti-VEGF receptor 2 antibody in second-line therapy did show improved survival. As such, Phase II and III evaluations of anti-VEGF TKIs are ongoing. The only Food and Drug Administration-approved targeted therapy in EGC is trastuzumab, an anti-Her2 antibody, and the results of a Phase III evaluation of lapatinib, an anti-Her2 TKI, are awaited. Phase III evaluation of an mTOR inhibitor has been negative. Finally, MET inhibition appears to have significant clinical potential and early testing of MET TKIs is underway.
Collapse
Affiliation(s)
- Geoffrey Y Ku
- Memorial Sloan-Kettering Cancer Center, Department of Medicine, Gastrointestinal Oncology Service, 300 East 66th Street, New York, NY 10065, USA
| | | |
Collapse
|
3044
|
Pietrantonio F, Perrone F, de Braud F, Castano A, Maggi C, Bossi I, Gevorgyan A, Biondani P, Pacifici M, Busico A, Gariboldi M, Festinese F, Tamborini E, Di Bartolomeo M. Activity of temozolomide in patients with advanced chemorefractory colorectal cancer and MGMT promoter methylation. Ann Oncol 2013; 25:404-8. [PMID: 24379162 DOI: 10.1093/annonc/mdt547] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND No evidence-based treatment options are available for patients with advanced colorectal cancer (CRC) progressing after standard therapies. MGMT is involved in repair of DNA damage and MGMT promoter methylation may predict benefit from alkylating agents such as temozolomide. The aim of our study was to evaluate the activity of temozolomide in terms of response rate in patients with metastatic CRC and MGMT methylation, after failure of approved treatments. PATIENTS AND METHODS Patients were enrolled in a monocentre, open-label, phase II study and treated with temozolomide at a dose of 150 mg/m2/day for 5 consecutive days in 4-weekly cycles. The treatment was continued for at least six cycles or until progressive disease. RESULTS Thirty-two patients were enrolled from August 2012 to July 2013. Treatment was well tolerated with one grade 4 thrombocytopenia and no other grade≥3 toxicities. No complete response occurred. The objective response rate was 12%, reaching the pre-specified level for promising activity. Median progression-free survival and overall survival were 1.8 and 8.4 months, respectively. Patients with KRAS, BRAF and NRAS wild-type CRC showed significantly higher response when compared with those with any RAS or BRAF mutation (44% versus 0%; P=0.004). TP53 status had no influence on the primary end point. CONCLUSIONS Temozolomide is tolerable and active in heavily pre-treated patients with advanced CRC and MGMT promoter methylation. Further studies in biomolecularly enriched populations or in a randomized setting are necessary to demonstrate the efficacy of temozolomide after failure of standard treatments.
Collapse
|
3045
|
Carpizo DR, Gensure RH, Yu X, Gendel VM, Greene SJ, Moore DF, Jabbour SK, Nosher JL. Pilot study of angiogenic response to yttrium-90 radioembolization with resin microspheres. J Vasc Interv Radiol 2013; 25:297-306.e1. [PMID: 24360887 DOI: 10.1016/j.jvir.2013.10.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 10/12/2013] [Accepted: 10/18/2013] [Indexed: 12/18/2022] Open
Abstract
PURPOSE To investigate the impact of radioembolization with yttrium-90 resin microspheres on the regulation of angiogenesis through observation of serial changes in a spectrum of angiogenic markers and other cytokines after therapy. MATERIALS AND METHODS This prospective pilot study enrolled 22 patients with liver-dominant disease deriving from biopsy-proven hepatocellular carcinoma (HCC) (n = 7) or metastatic colorectal carcinoma (mCRC) (n = 15). Circulating angiogenic markers were measured from serum samples drawn at baseline and at time points after therapy ranging from 6 hours to 120 days. Using multiplex enzyme-linked immunosorbent assay, several classic angiogenesis factors (vascular endothelial growth factor [VEGF], angiopoietin-2 [Ang-2], basic fibroblast growth factor [bFGF], platelet-derived growth factor subunit BB [PDGF-BB], thrombospondin-1 [Tsp-1]) and nonclassic factors (follistatin, leptin, interleukin [IL]-8) were evaluated. RESULTS Increases in cytokine levels ≥ 50% over baseline were observed in more than half of all patients studied for many cytokines, including classic angiogenic factors such as VEGF, Ang-2, and Tsp-1 as well as nonclassic factors IL-8 and follistatin (range, 36%-82% for all cytokines). Baseline cytokine levels in patients with overall survival (OS) < 6 months differed significantly from patients with longer survival for Ang-2 (P = .033) and IL-8 (P = .041). Patients with OS ≤ 6 months exhibited transient increases in VEGF and PDGF-BB after therapy compared with patients with OS > 6 months. CONCLUSIONS Radioembolization is associated with early transient increases in many angiogenic cytokines. In this small sample size, some of these changes were associated with worse OS. This research has important implications for future studies of radioembolization with antiangiogenic therapy performed during and after the procedure.
Collapse
Affiliation(s)
- Darren R Carpizo
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Biomedical and Health Sciences, New Brunswick, NJ 08903-0019
| | - Rebekah H Gensure
- Center for Biomedical Imaging & Informatics, Rutgers Cancer Institute of New Jersey, Rutgers Biomedical and Health Sciences, New Brunswick, NJ 08903-0019
| | - Xin Yu
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Biomedical and Health Sciences, New Brunswick, NJ 08903-0019
| | - Vyacheslav M Gendel
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, One Robert Wood Johnson Place MEB 404, PO Box 19, New Brunswick, NJ 08903-0019
| | - Samuel J Greene
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, One Robert Wood Johnson Place MEB 404, PO Box 19, New Brunswick, NJ 08903-0019
| | - Dirk F Moore
- Department of Biostatistics, School of Public Health, Rutgers Biomedical and Health Sciences, New Brunswick, NJ 08903-0019
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Biomedical and Health Sciences, New Brunswick, NJ 08903-0019
| | - John L Nosher
- Department of Radiology, Rutgers Robert Wood Johnson Medical School, One Robert Wood Johnson Place MEB 404, PO Box 19, New Brunswick, NJ 08903-0019.
| |
Collapse
|
3046
|
Dysphonia induced by anti-angiogenic compounds. Invest New Drugs 2013; 32:774-82. [PMID: 24343672 DOI: 10.1007/s10637-013-0049-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 11/05/2013] [Indexed: 12/18/2022]
Abstract
The number of studies reporting the benefit of angiogenesis inhibition is steadily increasing. Anti-angiogenic drugs, used as monotherapy or in association with chemotherapy, have been shown to benefit patients with several different malignancies. Despite the benefits of these therapies, however, each drug has different side effects. This review is specifically focused on analyzing the frequency of one of the complications the most frequently overlooked by physicians, dysphonia. Perhaps this side effect is overlooked because it is not life-threatening, but dysphonia may nevertheless affect quality of life considerably. We reviewed 88 studies concerning treatment with anti-angiogenics (bevacizumab, aflibercept, sunitinib, sorafenib, pazopanib, axitinib and regorafenib) presently approved for clinical use, to review the incidence of dysphonia or voice changes in phase I, II and III closed clinical studies reported in ClinicalTrials.gov until March 2013. We found that almost all studies reported certain degree of dysphonia in the trial arms associated with anti-angiogenic treatment. We discuss these findings in light of the fact that it is not an uncommon side effect in patients exposed to these kinds of drugs. Particularly for treatments with axitinib, aflibercept and regorafenib, the angiogenesis inhibition possibly plays a role by altering the larynx in some way and modifying vocal fold vibrations, leading to dysphonia.
Collapse
|
3047
|
Abstract
The combination of cytotoxic chemotherapy and antiangiogenic agents has become a conventional treatment option for patients with metastatic colorectal cancer. Ziv-aflibercept is a fusion protein which acts as a decoy receptor for vascular endothelial growth factor (VEGF)-A, VEGF-B, and placental growth factor (PlGF); it was approved in combination with 5-fluorouracil, leucovorin, and irinotecan (FOLFIRI) for the treatment of patients with metastatic colorectal cancer that is resistant to or has progressed after an oxaliplatin-containing fluoropyrimidine-based regimen. Herein we review the role of tumor angiogenesis as the rationale for antiangiogenic therapy, the clinical data associated with ziv-aflibercept, and its current role as a treatment option compared to other antiangiogenic agents, such as bevacizumab and regorafenib.
Collapse
Affiliation(s)
- Anuj Patel
- Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Weijing Sun
- Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
3048
|
Stabilized epoxygenated fatty acids regulate inflammation, pain, angiogenesis and cancer. Prog Lipid Res 2013; 53:108-23. [PMID: 24345640 DOI: 10.1016/j.plipres.2013.11.003] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 11/26/2013] [Indexed: 12/21/2022]
Abstract
Epoxygenated fatty acids (EpFAs), which are lipid mediators produced by cytochrome P450 epoxygenases from polyunsaturated fatty acids, are important signaling molecules known to regulate various biological processes including inflammation, pain and angiogenesis. The EpFAs are further metabolized by soluble epoxide hydrolase (sEH) to form fatty acid diols which are usually less-active. Pharmacological inhibitors of sEH that stabilize endogenous EpFAs are being considered for human clinical uses. Here we review the biology of ω-3 and ω-6 EpFAs on inflammation, pain, angiogenesis and tumorigenesis.
Collapse
|
3049
|
De Wit M, Boers-Doets CB, Saettini A, Vermeersch K, de Juan CR, Ouwerkerk J, Raynard SS, Bazin A, Cremolini C. Prevention and management of adverse events related to regorafenib. Support Care Cancer 2013; 22:837-46. [PMID: 24337717 PMCID: PMC3913844 DOI: 10.1007/s00520-013-2085-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 11/26/2013] [Indexed: 12/21/2022]
Abstract
Regorafenib is an oral multikinase inhibitor that has shown antitumor activity in a range of solid tumors. Based on data from phase III clinical trials, regorafenib is indicated for the treatment of adult patients with metastatic colorectal cancer who have previously been treated with, or are not considered candidates for, other available therapies, and in patients with advanced gastrointestinal stromal tumors that cannot be surgically removed and no longer respond to other appropriate treatments. A panel of oncology nurses, research coordinators, and other medical oncology experts, experienced in the care of patients treated with regorafenib, met to discuss the best practice for the management of regorafenib-associated adverse events (AEs). The panel agreed that, in clinical trials and daily practice with regorafenib, AEs are common but mostly manageable. The most common and/or important AEs associated with regorafenib were considered to be hand-foot skin reaction, rash or desquamation, stomatitis, diarrhea, hypertension, liver abnormalities, and fatigue. This manuscript describes the experience and recommendations of the panel for managing these AEs in everyday clinical practice. Appropriate education, monitoring, and management are considered essential for reducing the incidence, duration, and severity of regorafenib-associated AEs.
Collapse
Affiliation(s)
- Mieke De Wit
- Digestive Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium,
| | | | | | | | | | | | | | | | | |
Collapse
|
3050
|
Aprile G, Lutrino SE, Ferrari L, Casagrande M, Bonotto M, Ongaro E, Puglisi F. Evidence-based appraisal of the upfront treatment for unresectable metastatic colorectal cancer patients. World J Gastroenterol 2013; 19:8474-88. [PMID: 24379565 PMCID: PMC3870493 DOI: 10.3748/wjg.v19.i46.8474] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 11/13/2013] [Accepted: 12/03/2013] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is a significant health problem, with around 1 million new cases and 500000 deaths every year worldwide. Over the last two decades, the use of novel therapies and more complex treatment strategies have contributed to progressively increase the median survival of patients with unresectable advanced CRC up to approximately 30 mo. The availability of additional therapeutic options, however, has created new challenges and generated more complicated treatment algorithms. Moreover, several clinically important points are still in debate in first-line, such as the optimal treatment intensity, the most appropriate maintenance strategy, the preferred biologic to be used upfront in patients with KRAS wild-type CRC, and the need for more detailed information on tumor biology. In this moving landscape, this review analyses why the first-line treatment decision is crucial and how the choice may impact on further treatment lines. In addition, it focuses on results of major phase III randomized trials.
Collapse
|