1
|
Chen P, Wang Q, Xie J, Kwok HF. Signaling networks and the feasibility of computational analysis in gastroenteropancreatic neuroendocrine tumors. Semin Cancer Biol 2019; 58:80-89. [DOI: 10.1016/j.semcancer.2019.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/25/2019] [Accepted: 04/29/2019] [Indexed: 12/22/2022]
|
2
|
Beyens M, Vandamme T, Peeters M, Van Camp G, Op de Beeck K. Resistance to targeted treatment of gastroenteropancreatic neuroendocrine tumors. Endocr Relat Cancer 2019; 26:R109-R130. [PMID: 32022503 DOI: 10.1530/erc-18-0420] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The mammalian target of rapamycin (mTOR) is part of the phosphoinositide-3-kinase (PI3K)/protein kinase B (Akt)/mTOR signaling. The PI3K/Akt/mTOR pathway has a pivotal role in the oncogenesis of neuroendocrine tumors (NETs). In addition, vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) drive angiogenesis in NETs and therefore contributes to neuroendocrine tumor development. Hence, mTOR and angiogenesis inhibitors have been developed. Everolimus, a first-generation mTOR inhibitor, has shown significant survival benefit in advanced gastroenteropancreatic NETs. Sunitinib, a pan-tyrosine kinase inhibitor that targets the VEGF receptor, has proven to increase progression-free survival in advanced pancreatic NETs. Nevertheless, primary and acquired resistance to rapalogs and sunitinib has limited the clinical benefit for NET patients. Despite the identification of multiple molecular mechanisms of resistance, no predictive biomarker has made it to the clinic. This review is focused on the mTOR signaling and angiogenesis in NET, the molecular mechanisms of primary and acquired resistance to everolimus and sunitinib and how to overcome this resistance by alternative drug compounds.
Collapse
Affiliation(s)
- Matthias Beyens
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium
| | - Timon Vandamme
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium
- Section of Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Marc Peeters
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Guy Van Camp
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium
| | - Ken Op de Beeck
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
3
|
Abstract
Pancreatic neuroendocrine tumors are rare tumors of the pancreas originating from the islets of the Langerhans. These tumors comprise 1% to 3% of all newly diagnosed pancreatic cancers every year and have a unique heterogeneity in clinical presentation. Whole-genome sequencing has led to an increased understanding of the molecular biology of these tumors. In this review, we will summarize the current knowledge of the signaling pathways involved in the tumorigenesis of pancreatic neuroendocrine tumors as well as the major studies targeting these pathways at preclinical and clinical levels.
Collapse
|
4
|
Koenig A, Krug S, Mueller D, Barth PJ, Koenig U, Scharf M, Ellenrieder V, Michl P, Moll R, Homayunfar K, Kann PH, Stroebel P, Gress TM, Rinke A. Clinicopathological hallmarks and biomarkers of colorectal neuroendocrine neoplasms. PLoS One 2017; 12:e0188876. [PMID: 29232390 PMCID: PMC5726657 DOI: 10.1371/journal.pone.0188876] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 11/14/2017] [Indexed: 12/29/2022] Open
Abstract
Chromogranin A (CgA) is a well-established marker for diagnosis and follow up of patients with gastroenteropancreatic neuroendocrine neoplasms (GEP-NEN). Recently, it has been shown that plasma levels of CgA correlate with tumor load and predict survival of patients with NEN of the small bowel. It is assumed that this is as well valid for NEN of the colon and rectum, however, this is not supported by data. To evaluate this assumption, we analyzed 62 patients with NEN of the colon and rectum listed in the Marburg GEP-NEN registry for clinicopathological characteristics, expression and plasma levels of CgA. The present study demonstrates that immunohistochemical CgA and synaptophysin are good markers for histological diagnosis in patients with NEN of the colon and rectum. However, plasma CgA is a poor marker to follow-up these patients because only a minority exhibited increased levels which did not increase significantly during tumor progression. In contrast to NEN of the small bowel, there is no correlation of CgA plasma levels with tumor burden or survival. Patients with NEN of the colon and rectum displayed a relatively good prognosis resulting in a median survival of 8.5 years. However, a subset of patients affected by G3 neoplasms, exhibited a poorer prognosis with a median survival of 2.5 years. Taken together, CgA is a valuable marker for immunohistochemical diagnosis, but CgA plasma concentration is not suitable to mirror tumor burden or prognosis in patients with NEN of the colon and rectum.
Collapse
Affiliation(s)
- Alexander Koenig
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Goettingen, Goettingen, Germany
- * E-mail: (AK); (AR)
| | - Sebastian Krug
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
- Department of Internal Medicine I, University Halle, Halle, Germany
| | - Daniela Mueller
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
| | - Peter J. Barth
- Gerhard-Domagk-Institute of Pathology, University of Muenster, Muenster, Germany
- Department of Pathology, Philipps-University of Marburg, Marburg, Germany
| | - Ute Koenig
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Michael Scharf
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Patrick Michl
- Department of Internal Medicine I, University Halle, Halle, Germany
| | - Roland Moll
- Department of Pathology, Philipps-University of Marburg, Marburg, Germany
| | - Kia Homayunfar
- Department of General-, Visceral- and Pediatric Surgery University Medical Center Goettingen, Goettingen, Germany
| | - Peter Herbert Kann
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
| | - Philipp Stroebel
- Department of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Thomas M. Gress
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
| | - Anja Rinke
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Marburg, Germany
- * E-mail: (AK); (AR)
| |
Collapse
|
5
|
Takikawa M, Ohki R. A vicious partnership between AKT and PHLDA3 to facilitate neuroendocrine tumors. Cancer Sci 2017; 108:1101-1108. [PMID: 28295876 PMCID: PMC5480075 DOI: 10.1111/cas.13235] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/01/2017] [Accepted: 03/06/2017] [Indexed: 12/19/2022] Open
Abstract
Pancreatic neuroendocrine tumors (PanNET) are rare cancers that generally have a poor prognosis. Accurate diagnosis and proper treatment of these tumors requires a better understanding of the molecular mechanisms underlying the development of PanNET. It has been shown that the mTOR inhibitor everolimus can improve the progression‐free survival of PanNET patients, suggesting that inhibition of the PI3K‐Akt‐mTOR pathway may suppress the progression of PanNET. PHLDA3 is a novel tumor suppressor protein that inhibits Akt activation by competition for binding to PIP3. Our analysis of PanNET revealed frequent loss‐of‐heterozygosity and DNA methylation at the PHLDA3 locus, resulting in strong suppression of PHLDA3 transcription. Such alterations in the PHLDA3 gene were also frequently found in lung neuroendocrine tumors (NET), suggesting the possibility that various types of NET have in common the functional loss of the PHLDA3 gene.
Collapse
Affiliation(s)
- Masahiro Takikawa
- Division of Rare Cancer Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Rieko Ohki
- Division of Rare Cancer Research, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
6
|
Translational research in neuroendocrine tumors: pitfalls and opportunities. Oncogene 2017; 36:1899-1907. [PMID: 27641330 DOI: 10.1038/onc.2016.316] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/19/2016] [Accepted: 07/22/2016] [Indexed: 12/16/2022]
Abstract
Interest in research on neuroendocrine tumors (NETs) has grown in the past 10 years, coinciding with improvements in our understanding of the molecular pathogenesis of NETs. In addition, NETs have become one of the most exciting settings for drug development. Two targeted agents for the management of advanced pancreatic NETs have been approved, but the development of targeted agents for NETs is limited by problems with both patient selection and demonstration of activity. In this review, we analyze these limitations and discuss ways to increase the predictive value of preclinical models for target discovery and drug development. The role of translational research and 'omics' methodologies is emphasized, with the final aim of developing personalized medicine. Because NETs usually grow slowly and metastatic tumors are found at easily accessible locations, and owing to improvements in techniques for liquid biopsies, NETs provide a unique opportunity to obtain tumor samples at all stages of the evolution of the disease and to adapt treatment to changes in tumor biology. Combining clinical and translational research is essential to achieve progress in the NET field. Slow growth and genetic stability limit and challenge both the availability and further development of preclinical models of NETs, one of the most crucial unmet research needs in the field. Finally, we suggest some useful approaches for improving clinical drug development for NETs: moving from classical RECIST-based response end points to survival parameters; searching for different criteria to define response rates (for example, antiangiogenic effects and metabolic responses); implementing randomized phase II studies to avoid single-arm phase II studies that produce limited data on drug efficacy; and using predictive biomarkers for patient selection.
Collapse
|
7
|
Roviello G, Zanotti L, Venturini S, Bottini A, Generali D. Role of targeted agents in neuroendocrine tumors: Results from a meta-analysis. Cancer Biol Ther 2016; 17:883-8. [PMID: 27414404 DOI: 10.1080/15384047.2016.1210735] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Several randomized phase III trials in neuroendocrine tumors (NETs) showed the clinical role of new targeted agents and their impact on tumor response and outcome of whose patients affected by advanced NET. In this study, we summarize the available clinical data related to clinical efficacy of targeted therapies in the treatment of advanced NETs. METHODS A meta-analysis of randomized studies in accordance with the PRISMA guidelines was performed after searching the databases of PubMed, the Cochrane Library, and the ASCO University Meeting for relevant publications. RESULTS One thousand 9 hundred and 8 cases were included in the meta-analysis; among these, 1012 were in the experimental arm and 896 were in the control arm. The pooled analysis of the use of target agents in NETs revealed significantly increased of progression free survival compared to control group (hazard ratio = 0.59, 95% CI:0.42-0.84; P = 0.003). Subgroup analysis of patients according to tumor site showed a difference in favor of pancreatic neuroendocrine tumors. Moreover, targeted therapies improved the overall survival (hazard ratio = 0.79, 95%CI: 0.63-0.98; P = 0.03), and response rate (hazard ratio = 3.33, 95% CI 2.02-5.49; P < 0.00001) in all types of NETs. CONCLUSION Our analysis supports the routine use of targeted agents for treatment of neuroendocrine tumors with particular regards to the pancreatic neuroendocrine tumors.
Collapse
Affiliation(s)
- Giandomenico Roviello
- a Department of Molecular and Translational Medicine, Section of Pharmacology and University Center DIFF-Drug Innovation Forward Future , University of Brescia , Brescia , Italy.,b Unit of Molecular Therapy and Pharmacogenomic , AO Azienda Istituti Ospitalieri di Cremona , Cremona , Italy
| | - Laura Zanotti
- b Unit of Molecular Therapy and Pharmacogenomic , AO Azienda Istituti Ospitalieri di Cremona , Cremona , Italy
| | - Sergio Venturini
- c Centre for Research on Health and Social Care Management (CeRGAS) , Bocconi University , Milan , Italy
| | - Alberto Bottini
- b Unit of Molecular Therapy and Pharmacogenomic , AO Azienda Istituti Ospitalieri di Cremona , Cremona , Italy
| | - Daniele Generali
- b Unit of Molecular Therapy and Pharmacogenomic , AO Azienda Istituti Ospitalieri di Cremona , Cremona , Italy.,d Department of Medical , Surgery and Health Sciences, University of Trieste , Trieste , Italy
| |
Collapse
|
8
|
Abstract
Neuroendocrine tumors (NETs) represent a heterogeneous group of diseases with varied natural history and prognosis depending upon the organ of origin and grade of aggressiveness. The most widely used biomarker to determine disease burden and monitor response to treatment is chromogranin A (CgA), but it is far from being the optimal predictive and prognostic biomarker in NETs. Biological understanding and derived treatment options for NETs have changed markedly in recent years. Over the last decade, the genomic landscape of these tumors has been extensively investigated. This has resulted in the discovery of mutations and expression anomalies in genes and pathways such as the PI3K/Akt/mTOR, DAXX/ATRX, and MEN1, which are promising predictive and prognostic biomarkers and future candidates for targeted therapies. Additionally, the study of tumor stroma and environment are one of the most promising fields for discovery of potential new targets and biomarkers.
Collapse
|
9
|
Manfredi GI, Dicitore A, Gaudenzi G, Caraglia M, Persani L, Vitale G. PI3K/Akt/mTOR signaling in medullary thyroid cancer: a promising molecular target for cancer therapy. Endocrine 2015; 48:363-70. [PMID: 25115638 DOI: 10.1007/s12020-014-0380-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 08/01/2014] [Indexed: 02/06/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway is a central hub for the regulation of cell proliferation, apoptosis, cell cycle, metabolism, and angiogenesis. Several studies have recently suggested that the PI3K/Akt/mTOR signaling pathway is implicated in the pathogenesis and progression of neuroendocrine tumors. Medullary thyroid cancer (MTC) is a neuroendocrine tumor developing from the C cells of the thyroid. Mutations in the RET proto-oncogene are involved in the pathogenesis of several forms of MTC. The deregulation of the PI3K/Akt/mTOR pathway seems to contribute to the tumorigenic activity of RET proto-oncogene mutations. Targeting this pathway through specific inhibitors at simple or multiple sites may represent an attractive potential therapeutic approach for patients with advanced MTCs. The aim of this review is to examine the role of the PI3K/Akt/mTOR pathway in the development and progression of MTC and the new therapeutic options that target this signaling pathway.
Collapse
Affiliation(s)
- Gloria Irene Manfredi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | | | | | | | | |
Collapse
|
10
|
Abstract
Pancreatic neuroendocrine neoplasms include mainly well-differentiated neuroendocrine tumors but also rare poorly differentiated neuroendocrine carcinomas. Molecular mechanisms underlying pancreatic neuroendocrine tumorigenesis have recently been elucidated. While alterations in the chromatin remodeling and PI3K/Akt/mTOR pathways are present in most well-differentiated pancreatic neuroendocrine tumors, mutations in TP53 and RB may contribute to the development of pancreatic poorly differentiated neuroendocrine carcinomas. With these discoveries, new molecular targeted therapies have become available and show promise in some patients with pancreatic well-differentiated neuroendocrine tumor.
Collapse
Affiliation(s)
- Chanjuan Shi
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David S Klimstra
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, New York 10065.
| |
Collapse
|
11
|
Krug S, Kühnemuth B, Griesmann H, Neesse A, Mühlberg L, Boch M, Kortenhaus J, Fendrich V, Wiese D, Sipos B, Friemel J, Gress TM, Michl P. CUX1: a modulator of tumour aggressiveness in pancreatic neuroendocrine neoplasms. Endocr Relat Cancer 2014; 21:879-90. [PMID: 25248790 DOI: 10.1530/erc-14-0152] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Pancreatic neuroendocrine neoplasms (PNENs) constitute a rare tumour entity, and prognosis and treatment options depend on tumour-mediating hallmarks such as angiogenesis, proliferation rate and resistance to apoptosis. The molecular pathways that determine the malignant phenotype are still insufficiently understood and this has limited the use of effective combination therapies in the past. In this study, we aimed to characterise the effect of the oncogenic transcription factor Cut homeobox 1 (CUX1) on proliferation, resistance to apoptosis and angiogenesis in murine and human PNENs. The expression and function of CUX1 were analysed using knockdown and overexpression strategies in Ins-1 and Bon-1 cells, xenograft models and a genetically engineered mouse model of insulinoma (RIP1Tag2). Regulation of angiogenesis was assessed using RNA profiling and functional tube-formation assays in HMEC-1 cells. Finally, CUX1 expression was assessed in a tissue microarray of 59 human insulinomas and correlated with clinicopathological data. CUX1 expression was upregulated during tumour progression in a time- and stage-dependent manner in the RIP1Tag2 model, and associated with pro-invasive and metastatic features of human insulinomas. Endogenous and recombinant CUX1 expression increased tumour cell proliferation, tumour growth, resistance to apoptosis, and angiogenesis in vitro and in vivo. Mechanistically, the pro-angiogenic effect of CUX1 was mediated via upregulation of effectors such as HIF1α and MMP9. CUX1 mediates an invasive pro-angiogenic phenotype and is associated with malignant behaviour in human insulinomas.
Collapse
Affiliation(s)
- Sebastian Krug
- Departments of GastroenterologyEndocrinology and MetabolismSurgeryPhilipps-University Marburg, Baldingerstraße, 35043 Marburg, GermanyDepartment of PathologyEberhard-Karls-University Tübingen, Tübingen, GermanyDepartment of PathologyUniversity Hospital Zurich, Zurich, Switzerland
| | - Benjamin Kühnemuth
- Departments of GastroenterologyEndocrinology and MetabolismSurgeryPhilipps-University Marburg, Baldingerstraße, 35043 Marburg, GermanyDepartment of PathologyEberhard-Karls-University Tübingen, Tübingen, GermanyDepartment of PathologyUniversity Hospital Zurich, Zurich, Switzerland
| | - Heidi Griesmann
- Departments of GastroenterologyEndocrinology and MetabolismSurgeryPhilipps-University Marburg, Baldingerstraße, 35043 Marburg, GermanyDepartment of PathologyEberhard-Karls-University Tübingen, Tübingen, GermanyDepartment of PathologyUniversity Hospital Zurich, Zurich, Switzerland
| | - Albrecht Neesse
- Departments of GastroenterologyEndocrinology and MetabolismSurgeryPhilipps-University Marburg, Baldingerstraße, 35043 Marburg, GermanyDepartment of PathologyEberhard-Karls-University Tübingen, Tübingen, GermanyDepartment of PathologyUniversity Hospital Zurich, Zurich, Switzerland
| | - Leonie Mühlberg
- Departments of GastroenterologyEndocrinology and MetabolismSurgeryPhilipps-University Marburg, Baldingerstraße, 35043 Marburg, GermanyDepartment of PathologyEberhard-Karls-University Tübingen, Tübingen, GermanyDepartment of PathologyUniversity Hospital Zurich, Zurich, Switzerland
| | - Michael Boch
- Departments of GastroenterologyEndocrinology and MetabolismSurgeryPhilipps-University Marburg, Baldingerstraße, 35043 Marburg, GermanyDepartment of PathologyEberhard-Karls-University Tübingen, Tübingen, GermanyDepartment of PathologyUniversity Hospital Zurich, Zurich, Switzerland
| | - Juliane Kortenhaus
- Departments of GastroenterologyEndocrinology and MetabolismSurgeryPhilipps-University Marburg, Baldingerstraße, 35043 Marburg, GermanyDepartment of PathologyEberhard-Karls-University Tübingen, Tübingen, GermanyDepartment of PathologyUniversity Hospital Zurich, Zurich, Switzerland
| | - Volker Fendrich
- Departments of GastroenterologyEndocrinology and MetabolismSurgeryPhilipps-University Marburg, Baldingerstraße, 35043 Marburg, GermanyDepartment of PathologyEberhard-Karls-University Tübingen, Tübingen, GermanyDepartment of PathologyUniversity Hospital Zurich, Zurich, Switzerland
| | - Dominik Wiese
- Departments of GastroenterologyEndocrinology and MetabolismSurgeryPhilipps-University Marburg, Baldingerstraße, 35043 Marburg, GermanyDepartment of PathologyEberhard-Karls-University Tübingen, Tübingen, GermanyDepartment of PathologyUniversity Hospital Zurich, Zurich, Switzerland
| | - Bence Sipos
- Departments of GastroenterologyEndocrinology and MetabolismSurgeryPhilipps-University Marburg, Baldingerstraße, 35043 Marburg, GermanyDepartment of PathologyEberhard-Karls-University Tübingen, Tübingen, GermanyDepartment of PathologyUniversity Hospital Zurich, Zurich, Switzerland
| | - Juliane Friemel
- Departments of GastroenterologyEndocrinology and MetabolismSurgeryPhilipps-University Marburg, Baldingerstraße, 35043 Marburg, GermanyDepartment of PathologyEberhard-Karls-University Tübingen, Tübingen, GermanyDepartment of PathologyUniversity Hospital Zurich, Zurich, Switzerland
| | - Thomas M Gress
- Departments of GastroenterologyEndocrinology and MetabolismSurgeryPhilipps-University Marburg, Baldingerstraße, 35043 Marburg, GermanyDepartment of PathologyEberhard-Karls-University Tübingen, Tübingen, GermanyDepartment of PathologyUniversity Hospital Zurich, Zurich, Switzerland
| | - Patrick Michl
- Departments of GastroenterologyEndocrinology and MetabolismSurgeryPhilipps-University Marburg, Baldingerstraße, 35043 Marburg, GermanyDepartment of PathologyEberhard-Karls-University Tübingen, Tübingen, GermanyDepartment of PathologyUniversity Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Cummins M, Pavlakis N. The use of targeted therapies in pancreatic neuroendocrine tumours: patient assessment, treatment administration, and management of adverse events. Ther Adv Med Oncol 2013; 5:286-300. [PMID: 23997829 DOI: 10.1177/1758834013501016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Together with the use of novel oral targeted therapies, a multidisciplinary approach can be used to effectively treat patients with advanced pancreatic neuroendocrine tumours (pNETs). Here we review the integration of the oncology nurse to the newly developed oral treatment setting for patients with pNETs. From the outset, the nurse must be involved in various processes, including performance of baseline assessments (e.g. blood pathology, cardiac and lung function testing, patient history) and general medical observations, treatment administration, dietary guidance, evaluation of comorbidities, and review of concomitant medications. Patient education and establishment of a strong partnership in care before the start of pNET therapy ultimately increase treatment adherence and reduce potential toxicities. Regular review of general patient status and disease progression and continuous monitoring of adverse events also help enhance treatment outcomes and subsequently improve quality of life. Nurses' knowledge of agent-specific toxicities and prompt, proactive management is a critical aspect of care. In essence, as the pNET treatment landscape evolves, the role of the healthcare professional in overall patient care must shift accordingly.
Collapse
Affiliation(s)
- Meredith Cummins
- Director of Nursing, Northern Cancer Institute, 49 Frenchs Forest Road, Frenchs Forest, NSW, 2086, Australia
| | | |
Collapse
|
13
|
Yao JC, Lagunes DR, Kulke MH. Targeted therapies in neuroendocrine tumors (NET): clinical trial challenges and lessons learned. Oncologist 2013; 18:525-32. [PMID: 23615698 DOI: 10.1634/theoncologist.2012-0434] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In the past 3 years, we have witnessed the completion of four randomized phase III studies in neuroendocrine tumors and the approval of two new drugs, everolimus and sunitinib, for the treatment of patients with well-differentiated pancreatic neuroendocrine tumors. These studies demonstrate a shift from case series and single-arm studies toward prospective, randomized controlled clinical trials and evidence-based therapy in the neuroendocrine tumor field. However, the clinical development of these agents also highlights the potential challenges awaiting other new drugs in this area. Herein, we discuss the strengths and weaknesses of the most recent phase II and phase III neuroendocrine tumor studies and discuss how limitations inherent in current trial design can lead to potential pitfalls. We also discuss how trial design can be improved, with the hope of increasing the number of drugs successfully developed to treat patients with neuroendocrine tumors.
Collapse
Affiliation(s)
- James C Yao
- The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
14
|
Wolin EM. PI3K/Akt/mTOR pathway inhibitors in the therapy of pancreatic neuroendocrine tumors. Cancer Lett 2013; 335:1-8. [PMID: 23419523 DOI: 10.1016/j.canlet.2013.02.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 02/06/2013] [Accepted: 02/08/2013] [Indexed: 02/07/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway is implicated in the pathogenesis of pancreatic neuroendocrine tumors (pNETs). Activation of this pathway is driven by aberrant tyrosine kinase receptor activities. Mutations in the PI3K/Akt/mTOR pathway occur in 15% of pNETs, and expression of genes of the PI3K/Akt/mTOR pathway is altered in the majority of pNETs. The mTOR inhibitor everolimus has been approved by the FDA for the treatment of pNET, but its efficacy may be limited by its inability to prevent mTORC2-mediated activation of Akt. Specific inhibitors of PI3K, Akt, or other pathway nodes, and their concomitant use with mTOR inhibitors, or agents with dual activity, may be more effective. Preclinical studies demonstrate that inhibitors of the PI3K pathway have antitumor activity in pNET cells, either through direct inhibition of individual pathway nodes or indirect inhibition of molecular chaperones such as heat-shock protein 90. Clinical studies are underway evaluating individual node and dual node inhibitors.
Collapse
Affiliation(s)
- Edward M Wolin
- Division of Hematology/Oncology, Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA.
| |
Collapse
|