1
|
Duszkiewicz R, Strzelczyk J, Chełmecka E, Strzelczyk JK. Evaluation of LRP6, SFRP3, and DVL1 Protein Concentrations in Serum of Patients with Gastroenteropancreatic or Bronchopulmonary Neuroendocrine Tumors. Cancers (Basel) 2024; 17:47. [PMID: 39796676 PMCID: PMC11718808 DOI: 10.3390/cancers17010047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/09/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Introduction: Neuroendocrine tumors are a diverse group of tumors predominantly found in the gastrointestinal tract or respiratory system. Methods: This retrospective study aimed to measure the serum concentrations of LRP6 (low-density lipoprotein receptor-related protein 6), SFRP3 (secreted frizzled-related protein 3), and DVL1 (segment polarity protein dishevelled homolog) using the ELISA method in patients with NETs (N = 80) and a control group (N = 62). We evaluated the results against various demographic, clinicopathological, and biochemical characteristics. Results: Our analyses revealed that the concentration of SFRP3 in patients with neuroendocrine tumors was significantly elevated (p < 0.001) compared to the control group. Additionally, DVL1 concentrations were significantly higher (p < 0.01) in patients with BP-NETs compared to GEP-NETs. Furthermore, DVL1 analysis showed a moderate negative correlation with chromogranin A (p < 0.001) and weak negative correlations with serotonin (p < 0.05) and 5-HIAA (p < 0.05). Significant negative correlations were also observed between DVL1 and age in the control group (p < 0.01), and between LRP6 and Ki-67 in the study group. Conclusions: These results suggest that changes in the SFRP3 and DVL1 pathways play a key role in NET development. Elevated levels of these proteins highlight their importance in tumor biology, with SFRP3 and DVL1 potentially being crucial in NET molecular mechanisms. Further research is needed to explore their roles and potential in diagnosis and treatment.
Collapse
Affiliation(s)
- Roksana Duszkiewicz
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana St., 41-808 Zabrze, Poland
| | - Janusz Strzelczyk
- Department of Endocrinology and Neuroendocrine Tumors, Department of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia 35 Ceglana St., 40-514 Katowice, Poland
| | - Elżbieta Chełmecka
- Department of Medical Statistics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 30 Ostrogórska St., 41-200 Sosnowiec, Poland
| | - Joanna Katarzyna Strzelczyk
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana St., 41-808 Zabrze, Poland
| |
Collapse
|
2
|
Ruz-Caracuel I, Pedraza-Arevalo S, Alonso-Gordoa T, Molina-Cerrillo J, Earl J, Sainz B. Everything you ever wanted to know about cancer stem cells in neuroendocrine neoplasms but were afraid to ask. ENDOCRINE ONCOLOGY (BRISTOL, ENGLAND) 2024; 4:e240006. [PMID: 39822777 PMCID: PMC11737516 DOI: 10.1530/eo-24-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/28/2024] [Accepted: 10/30/2024] [Indexed: 01/19/2025]
Abstract
While the role of cancer stem cells (CSCs) in tumorigenesis, chemoresistance, metastasis, and relapse has been extensively studied in solid tumors, such as adenocarcinomas or sarcomas, the same cannot be said for neuroendocrine neoplasms (NENs). While lagging, CSCs have been described in numerous NENs, including gastrointestinal and pancreatic NENs (PanNENs), and they have been found to play critical roles in tumor initiation, progression, and treatment resistance. However, it seems that there is still skepticism regarding the role of CSCs in NENs, even in light of studies that support the CSC model in these tumors and the therapeutic benefits of targeting them. For example, in lung neuroendocrine carcinoids, a high percentage of CSCs have been found in atypical carcinoids, suggesting the presence of CSCs in these cancers. In PanNENs, CSCs marked by aldehyde dehydrogenases or CD90 have been identified, and targeting CSCs with inhibitors of molecular pathways has shown therapeutic potential. Overall, while evidence exists for the presence of CSCs in NENs, either the CSC field has neglected NENs or the NEN field has not fully embraced the CSC model. Both might apply and/or may be a consequence of the fact that NENs are a relatively rare and heterogeneous tumor entity, with confusing histology and nomenclature to match. Regardless, this review intends to summarize our current knowledge of CSCs in NENs and highlight the importance of understanding the role of CSCs in the biology of these rare tumors, with a special focus on developing targeted therapies to improve patients' outcomes.
Collapse
Affiliation(s)
- Ignacio Ruz-Caracuel
- Pathology Department, Hospital
Universitario Ramón y Cajal, Madrid,
Spain
- Molecular Pathology of Cancer
Group, Area 3 Cancer, Instituto Ramón y Cajal de Investigación
Sanitaria (IRYCIS), Madrid,
Spain
- Centro de Investigación
Biomédica en Red, CIBERONC, ISCIII, Madrid,
Spain
| | - Sergio Pedraza-Arevalo
- Maimonides Biomedical Research
Institute of Córdoba (IMIBIC), Cordoba,
Spain
- Department of Cell Biology,
Physiology, and Immunology, University of Córdoba,
Cordoba, Spain
- Reina Sofía University
Hospital (HURS), Cordoba,
Spain
| | - Teresa Alonso-Gordoa
- Molecular Pathology of Cancer
Group, Area 3 Cancer, Instituto Ramón y Cajal de Investigación
Sanitaria (IRYCIS), Madrid,
Spain
- Medical Oncology Department,
Hospital Universitario Ramón y Cajal, Madrid,
Spain
| | | | - Julie Earl
- Centro de Investigación
Biomédica en Red, CIBERONC, ISCIII, Madrid,
Spain
- Biomarkers and Personalized
Approach to Cancer (BIOPAC) Group, Area 3 Cancer, Instituto Ramón y Cajal
de Investigación Sanitaria (IRYCIS), Madrid,
Spain
| | - Bruno Sainz
- Centro de Investigación
Biomédica en Red, CIBERONC, ISCIII, Madrid,
Spain
- Biomarkers and Personalized
Approach to Cancer (BIOPAC) Group, Area 3 Cancer, Instituto Ramón y Cajal
de Investigación Sanitaria (IRYCIS), Madrid,
Spain
- Department of Cancer, Instituto
de Investigaciones Biomédicas (IIBm) Sols-Morreale
(CSIC-UAM), Madrid, Spain
| |
Collapse
|
3
|
Wnt/β-catenin modulating drugs regulate somatostatin receptor expression and internalization of radiolabelled octreotide in neuroendocrine tumor cells. Nucl Med Commun 2023; 44:259-269. [PMID: 36804512 DOI: 10.1097/mnm.0000000000001666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
BACKGROUND Differentiated neuroendocrine tumors (NETs) express somatostatin receptors (SSTRs), targets for therapy with either unlabeled or radioactively labeled somatostatin analogs (SSA). Associated with worse prognosis, dedifferentiated NET loose SSTR expression, which may be linked to deregulation of Wnt/β-catenin signaling on an intracellular level. The aim of the present study was to investigate the effect of Wnt/β-catenin signaling pathway alterations on SSTR expression and its function in NET. METHODS The NET cell lines BON-1 and QGP-1 were incubated with the Wnt-inhibitors 5-aza-2'-deoxycytidine (5-aza-CdR), Quercetin, or Niclosamide, or the Wnt activator lithium chloride (LiCl). Expression of SSTR1, SSTR2, and SSTR5 was determined by quantitative RT-PCR (qRT-PCR), immunocytomicroscopy and western blot. Changes in the Wnt pathway were analyzed by qRT-PCR of selected target genes and the TaqMan Array Human WNT Pathway. Receptor-associated function was determined by measuring the cellular uptake of [125I-Tyr3] octreotide. RESULTS The mRNAs of SSTRs 1-5 were expressed in both cell lines. Wnt inhibitors caused downregulation of Wnt target genes, while 5-aza-CdR had the highest inhibitory effect. LiCl lead to an upregulation of Wnt genes, which was more marked in QGP-1 cells. SSTR expression increased in both cell lines upon Wnt inhibition. All three Wnt inhibitors lead to a marked increase in the specific uptake of [125I-Tyr3]octreotide, with 5-aza-CdR showing the greatest effect (increase by more than 50% in BON-1 cells), while a decreased uptake of [125I-Tyr3]octreotide was seen upon activation of Wnt signaling by LiCl. CONCLUSIONS We demonstrate here that Wnt signaling orchestrates SSTR expression and function in a preclinical NET model. Wnt inhibition increases [125I-Tyr3]octreotide uptake offering an opportunity to enhance the efficacy of SSTR-targeted theranostic approaches.
Collapse
|
4
|
Ungefroren H, Künstner A, Busch H, Franzenburg S, Luley K, Viol F, Schrader J, Konukiewitz B, Wellner UF, Meyhöfer SM, Keck T, Marquardt JU, Lehnert H. Differential Effects of Somatostatin, Octreotide, and Lanreotide on Neuroendocrine Differentiation and Proliferation in Established and Primary NET Cell Lines: Possible Crosstalk with TGF-β Signaling. Int J Mol Sci 2022; 23:ijms232415868. [PMID: 36555512 PMCID: PMC9781720 DOI: 10.3390/ijms232415868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
GEP-NETs are heterogeneous tumors originating from the pancreas (panNET) or the intestinal tract. Only a few patients with NETs are amenable to curative tumor resection, and for most patients, only palliative treatments to successfully control the disease or manage symptoms remain, such as with synthetic somatostatin (SST) analogs (SSAs), such as octreotide (OCT) or lanreotide (LAN). However, even cells expressing low levels of SST receptors (SSTRs) may exhibit significant responses to OCT, which suggests the possibility that SSAs signal through alternative mechanisms, e.g., transforming growth factor (TGF)-β. This signaling mode has been demonstrated in the established panNET line BON but not yet in other permanent (i.e., QGP) or primary (i.e., NT-3) panNET-derived cells. Here, we performed qPCR, immunoblot analyses, and cell counting assays to assess the effects of SST, OCT, LAN, and TGF-β1 on neuroendocrine marker expression and cell proliferation in NT-3, QGP, and BON cells. SST and SSAs were found to regulate a set of neuroendocrine genes in all three cell lines, with the effects of SST, mainly LAN, often differing from those of OCT. However, unlike NT-3 cells, BON cells failed to respond to OCT with growth arrest but paradoxically exhibited a growth-stimulatory effect after treatment with LAN. As previously shown for BON, NT-3 cells responded to TGF-β1 treatment with induction of expression of SST and SSTR2/5. Of note, the ability of NT-3 cells to respond to TGF-β1 with upregulation of the established TGF-β target gene SERPINE1 depended on cellular adherence to a collagen-coated matrix. Moreover, when applied to NT-3 cells for an extended period, i.e., 14 days, TGF-β1 induced growth suppression as shown earlier for BON cells. Finally, next-generation sequencing-based identification of microRNAs (miRNAs) in BON and NT-3 revealed that SST and OCT impact positively or negatively on the regulation of specific miRNAs. Our results suggest that primary panNET cells, such as NT-3, respond similarly as BON cells to SST, SSA, and TGF-β treatment and thus provide circumstantial evidence that crosstalk of SST and TGF-β signaling is not confined to BON cells but is a general feature of panNETs.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, D-23538 Lübeck, Germany
- Institute of Pathology, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, D-24105 Kiel, Germany
- Correspondence:
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, D-23538 Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, D-23538 Lübeck, Germany
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, D-23538 Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, D-23538 Lübeck, Germany
| | - Sören Franzenburg
- Institute for Clinical Molecular Biology, University of Kiel, D-24118 Kiel, Germany
| | - Kim Luley
- Clinic of Oncology, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, D-23538 Lübeck, Germany
| | - Fabrice Viol
- Medical Clinic and Policlinic, University Hospital Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Jörg Schrader
- Medical Clinic and Policlinic, University Hospital Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Björn Konukiewitz
- Institute of Pathology, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, D-24105 Kiel, Germany
| | - Ulrich F. Wellner
- Department of Surgery, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, D-23538 Lübeck, Germany
| | - Sebastian M. Meyhöfer
- Institute of Endocrinology and Diabetes, University of Lübeck, D-23538 Lübeck, Germany
- German Center of Diabetes Research, D-85764 Neuherberg, Germany
| | - Tobias Keck
- Department of Surgery, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, D-23538 Lübeck, Germany
| | - Jens-Uwe Marquardt
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, D-23538 Lübeck, Germany
| | | |
Collapse
|
5
|
Dai M, Mullins CS, Lu L, Alsfasser G, Linnebacher M. Recent advances in diagnosis and treatment of gastroenteropancreatic neuroendocrine neoplasms. World J Gastrointest Surg 2022; 14:383-396. [PMID: 35734622 PMCID: PMC9160679 DOI: 10.4240/wjgs.v14.i5.383] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/17/2022] [Accepted: 04/29/2022] [Indexed: 02/06/2023] Open
Abstract
Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) are a rare group of tumors originating from neuroendocrine cells of the digestive system. Their incidence has increased over the last decades. The specific pathogenetic mechanisms underlying GEP-NEN development have not been completely revealed. Unfunctional GEP-NENs are usually asymptomatic; some grow slowly and thus impede early diagnosis, which ultimately results in a high rate of misdiagnosis. Therefore, many GEP-NEN patients present with later staged tumors. Motivated hereby, research attention for diagnosis and treatment for GEP-NENs increased in recent years. The result of which is great progress in clinical diagnosis and treatment. According to the most recent clinical guidelines, improved grading standards can accurately define poorly differentiated grade 3 neuroendocrine tumors and neuroendocrine carcinomas (NECs), which are subclassified into large and small cell NECs. Combining different functional imaging methods facilitates precise diagnosis. The expression of somatostatin receptors helps to predict prognosis. Genetic analyses of mutations affecting death domain associated protein (DAXX), multiple endocrine neoplasia type 1 (MEN 1), alpha thalassemia/intellectual disability syndrome X-linked (ATRX), retinoblastoma transcriptional corepressor 1 (RB 1), and mothers against decapentaplegic homolog 4 (SMAD 4) help distinguishing grade 3 NENs from poorly differentiated NECs. The aim of this review is to summarize the latest research progress on diagnosis and treatment of GEP-NENs.
Collapse
Affiliation(s)
- Meng Dai
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Christina S Mullins
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Lili Lu
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Guido Alsfasser
- Clinic of General Surgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Michael Linnebacher
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
6
|
Feng LH, Su T, Lu Y, Ren S, Huang L, Qin X, Liao T. A model for predicting the overall survival of gastroenteropancreatic neuroendocrine neoplasms after surgery. Scand J Gastroenterol 2022; 57:581-588. [PMID: 35001789 DOI: 10.1080/00365521.2021.2024247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND The increase in the incidence of gastroenteropancreatic neuroendocrine tumors (GEP-NENs) and refined morphological imaging techniques have led to a rise in the number of patients undergoing surgery. However, there is still a paucity of objective, clinically reliable and personalized tools to evaluate patient prognosis. MATERIALS AND METHODS We identified patients from the Surveillance, Epidemiology, and End Results (SEER) database who underwent surgery for GEP-NEN from 1975 to 2018. The predictors associated with OS were investigated by Multivariate Cox proportional hazards (PHs) regression analysis in the primary cohort; a prognostic nomogram was then built based on the multivariate analysis results. The performance of the nomogram was assessed by Harrell's concordance index (C-index) and calibration curve and compared with the eighth edition of the American Joint Committee on Cancer (AJCC) staging system. RESULTS A total of 45,889 patients were enrolled in our study; 32,321 were included in the primary cohort, and 13,568 were included in the validation cohort. A nomogram incorporating Age, Differentiation, M staging, and AJCC staging was subsequently built based on the multivariate analysis. The C-index (0.833 for the primary cohort and 0.845 for the validation cohort) and calibration curves indicated good discriminative ability and calibration of the nomogram. Further analysis demonstrated that the nomogram had superior discriminatory ability than the AJCC staging system (C-index= 0.706). CONCLUSION The proposed nomogram showed excellent prediction with good calibration and discrimination, which can be used to make well-informed and individualized clinical decisions regarding the clinical management of GEP-NENs.
Collapse
Affiliation(s)
- Lu-Huai Feng
- Department of Comprehensive Internal Medicine, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Tingting Su
- Department of ECG Diagnostics, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yang Lu
- Department of Comprehensive Internal Medicine, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Shuang Ren
- Department of Comprehensive Internal Medicine, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Lina Huang
- Department of Comprehensive Internal Medicine, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Xiuyu Qin
- Department of Comprehensive Internal Medicine, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Tianbao Liao
- Department of President's Office, Youjiang Medical University for Nationalities, Baise, China.,Philippine Christian University Center for International Education, Manila City, Philippine
| |
Collapse
|
7
|
Krieg S, Roderburg C, Fung S, Luedde T, Knoefel WT, Krieg A. Nuclear survivin is a prognosticator in gastroenteropancreatic neuroendocrine neoplasms: a meta-analysis. J Cancer Res Clin Oncol 2022; 148:2235-2246. [PMID: 35428913 PMCID: PMC9349075 DOI: 10.1007/s00432-022-04013-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/03/2022] [Indexed: 11/28/2022]
Abstract
Abstract
Purpose
Gastroenteropancreatic neuroendocrine neosplasms (GEP-NEN) are biologically heterogenous tumors with an increasing incidence over the past decades. Although efforts have been made in the treatment of these tumors, survival rates in metastasized tumor stages remain frustrating. Thus, there is an urgent need to identify novel targets as alternative treatment options. In this regard, the inhibitor of apoptosis protein (IAP) family member survivin could be such an attractive target. Therefore, aim of our meta-analysis was to assess the role of survivin as a biomarker and predictor in GEP-NEN.
Methods
Medline, Web of Science and Scopus were screened for studies that fulfilled our selection criteria. Quality assessement of the studies was based on design, methodology, generalizability and results analysis. Meta-analyses were conducted using a random-effects model and effect size measures were expressed as pooled Hazard Ratio (HR) or Odds Ratio (OR) with 95% Confidence Interval (CI).
Results
Six eligible studies with 649 patients (range 77–132) assessed survivin expression in GEP-NEN by immunohistochemistry. High expression levels of nuclear survivin in GEP-NEN correlated with a shorter overall survival (HR 3.10; 95% CI 2.15–4.47; p < 0.0001). In contrast to cytoplasmic survivin (OR 1.24; CI 0.59–2.57; p = 0.57), nuclear survivin was also associated (OR 15.23; CI 3.61–64.23; p = 0.0002) with G3/poorly differentiated GEP-NEN.
Conclusion
Nuclear Survivin is highly expressed in more aggressive G3 GEP-NEN and correlates with a poor outcome. Survivin is therefore an interesting molecule for a targeted therapy, especially for patients with highly proliferative G3 GEP-NENs.
Collapse
Affiliation(s)
- Sarah Krieg
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University and University Hospital Duesseldorf, Duesseldorf, Germany
| | - Christoph Roderburg
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University and University Hospital Duesseldorf, Duesseldorf, Germany
| | - Stephen Fung
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Bldg. 12.46, 40225, Duesseldorf, Germany
| | - Tom Luedde
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University and University Hospital Duesseldorf, Duesseldorf, Germany
| | - Wolfram Trudo Knoefel
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Bldg. 12.46, 40225, Duesseldorf, Germany
| | - Andreas Krieg
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, Moorenstr. 5, Bldg. 12.46, 40225, Duesseldorf, Germany.
| |
Collapse
|
8
|
Liverani C, Spadazzi C, Ibrahim T, Pieri F, Foca F, Calabrese C, De Vita A, Miserocchi G, Cocchi C, Vanni S, Ercolani G, Cavaliere D, Ranallo N, Chiadini E, Prisinzano G, Severi S, Sansovini M, Martinelli G, Bongiovanni A, Mercatali L. HRAS overexpression predicts response to Lenvatinib treatment in gastroenteropancreatic neuroendocrine tumors. Front Endocrinol (Lausanne) 2022; 13:1045038. [PMID: 36743926 PMCID: PMC9895371 DOI: 10.3389/fendo.2022.1045038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/28/2022] [Indexed: 01/22/2023] Open
Abstract
INTRODUCTION Neuroendocrine neoplasms (NENs) are a rare group of tumors exceptionally heterogeneous, with clinical presentation ranging from well differentiated more indolent tumors to poorly differentiated very aggressive forms. Both are often diagnosed after the metastatic spread and require appropriate medical treatment. A high priority need in the management of this disease is the identification of effective therapeutic strategies for advanced and metastatic patients. The recent TALENT trial demonstrated the efficacy of lenvatinib, a multi-tyrosine kinase inhibitor, in patients with gastroenteropancreatic neuroendocrine tumors (GEP-NETs) with no other treatment indication. Further development of this drug in advanced NETs is warranted. METHODS We investigated potential clinical and molecular determinants of lenvatinib response in human primary cultures derived from patients with GEP-NET of different grades and sites of origin. We correlated response to treatment with patient clinical characteristics, with the mutational status of 161-cancer associated genes and with the expression levels of MKI-related genes. RESULTS Lenvatinib exerted a significant antitumor activity in primary GEP-NET cells, with median survival inhibitions similar or higher than those of standard frontline treatments. Of the 11 primary cultures analyzed in our case series, 6 were classified as responder showing a significant survival inhibition, and 5 as non-responder. We observed that the overexpression of HRAS in the original tumor tissue compared to the matched healthy tissue significantly correlated with responsiveness of primary cells to lenvatinib (p=.048). All 5 non-responder cultures showed normal HRAS expression, while of the 6 responder cultures, 4 had HRAS overexpression. Overexpression of HRAS was not associated with gene mutation. None of the other evaluated clinical variables (grade, Ki67, site of origin and syndromic disease) or molecular markers correlated with response. DISCUSSION Lenvatinib appears to be a highly effective drug for the treatment of NETs. The evaluation of HRAS expression in the tumor tissue might improve patient selection and optimize therapeutic outcome.
Collapse
Affiliation(s)
- Chiara Liverani
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Chiara Spadazzi
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- *Correspondence: Chiara Spadazzi,
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Federica Pieri
- Pathology Unit, “Morgagni-Pierantoni” Hospital, Forlì, Italy
| | - Flavia Foca
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Chiara Calabrese
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Alessandro De Vita
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giacomo Miserocchi
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Claudia Cocchi
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Silvia Vanni
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giorgio Ercolani
- General and Oncologic Surgery, “Morgagni-Pierantoni” Hospital, Forlì, Italy
| | - Davide Cavaliere
- General and Oncologic Surgery, “Morgagni-Pierantoni” Hospital, Forlì, Italy
| | - Nicoletta Ranallo
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Elisa Chiadini
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giovanna Prisinzano
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Stefano Severi
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Maddalena Sansovini
- Unit of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Giovanni Martinelli
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Laura Mercatali
- Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
9
|
Maharjan CK, Ear PH, Tran CG, Howe JR, Chandrasekharan C, Quelle DE. Pancreatic Neuroendocrine Tumors: Molecular Mechanisms and Therapeutic Targets. Cancers (Basel) 2021; 13:5117. [PMID: 34680266 PMCID: PMC8533967 DOI: 10.3390/cancers13205117] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/16/2022] Open
Abstract
Pancreatic neuroendocrine tumors (pNETs) are unique, slow-growing malignancies whose molecular pathogenesis is incompletely understood. With rising incidence of pNETs over the last four decades, larger and more comprehensive 'omic' analyses of patient tumors have led to a clearer picture of the pNET genomic landscape and transcriptional profiles for both primary and metastatic lesions. In pNET patients with advanced disease, those insights have guided the use of targeted therapies that inhibit activated mTOR and receptor tyrosine kinase (RTK) pathways or stimulate somatostatin receptor signaling. Such treatments have significantly benefited patients, but intrinsic or acquired drug resistance in the tumors remains a major problem that leaves few to no effective treatment options for advanced cases. This demands a better understanding of essential molecular and biological events underlying pNET growth, metastasis, and drug resistance. This review examines the known molecular alterations associated with pNET pathogenesis, identifying which changes may be drivers of the disease and, as such, relevant therapeutic targets. We also highlight areas that warrant further investigation at the biological level and discuss available model systems for pNET research. The paucity of pNET models has hampered research efforts over the years, although recently developed cell line, animal, patient-derived xenograft, and patient-derived organoid models have significantly expanded the available platforms for pNET investigations. Advancements in pNET research and understanding are expected to guide improved patient treatments.
Collapse
Affiliation(s)
- Chandra K. Maharjan
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
| | - Po Hien Ear
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (P.H.E.); (C.G.T.); (J.R.H.)
| | - Catherine G. Tran
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (P.H.E.); (C.G.T.); (J.R.H.)
| | - James R. Howe
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (P.H.E.); (C.G.T.); (J.R.H.)
| | - Chandrikha Chandrasekharan
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
| | - Dawn E. Quelle
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
10
|
Mpilla GB, Uddin MH, Al-Hallak MN, Aboukameel A, Li Y, Kim SH, Beydoun R, Dyson G, Baloglu E, Senapedis WT, Landesman Y, Wagner KU, Viola NT, El-Rayes BF, Philip PA, Mohammad RM, Azmi AS. PAK4-NAMPT Dual Inhibition Sensitizes Pancreatic Neuroendocrine Tumors to Everolimus. Mol Cancer Ther 2021; 20:1836-1845. [PMID: 34253597 PMCID: PMC8492493 DOI: 10.1158/1535-7163.mct-20-1105] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/12/2021] [Accepted: 07/08/2021] [Indexed: 11/16/2022]
Abstract
Metastatic pancreatic neuroendocrine tumors (PNET) remain an unmet clinical problem. Chronologic treatment in PNETs includes observation (watchful protocol), surgery, targeted therapy, and chemotherapy. However, increasing evidence illustrates that the outcomes of targeted therapeutic options for the treatment of advanced PNETs show minimal response. The FDA-approved mTOR inhibitor everolimus does not shrink these tumors. It only delays disease progression in a subset of patients, while a significant fraction acquires resistance and shows disease progression. Thus, there is a need for more effective targeted approaches to sensitize PNETs to everolimus for better treatment outcomes. Previously, we showed that mTOR regulator p21 activated kinase 4 (PAK4) and nicotinamide adenine dinucleotide biosynthesis enzyme nicotinamide phosphoribosyl transferase (NAMPT) were aberrantly expressed in PNET tissue and promoted everolimus resistance. In this report, we demonstrate that PAK4-NAMPT dual inhibitor KPT-9274 can synergize with everolimus (growth inhibition, colony suppression, and glucose uptake assays). KPT-9274-everolimus disrupted spheroid formation in multiple PNET models. Molecular analysis showed alteration of mTORC2 through downregulation of RICTOR as a mechanism supporting synergy with everolimus in vitro KPT-9274 suppressed β-catenin activity via inhibition of PAK4, highlighting the cross-talk between Rho GTPases and Wnt signaling in PNETs. KPT-9274, given at 150 mg/kg in combination with sub-MTD everolimus (2.5 mg/kg), significantly suppressed two PNET-derived xenografts. These studies bring forward a well-grounded strategy for advanced PNETs that fail to respond to single-agent everolimus.
Collapse
Affiliation(s)
- Gabriel B Mpilla
- Karmanos Cancer Institute, Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan
| | - Md Hafiz Uddin
- Karmanos Cancer Institute, Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan
| | - Mohammed N Al-Hallak
- Karmanos Cancer Institute, Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan
| | - Amro Aboukameel
- Karmanos Cancer Institute, Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan
| | - Yiwei Li
- Karmanos Cancer Institute, Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan
| | - Steve H Kim
- Karmanos Cancer Institute, Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan
| | - Rafic Beydoun
- Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan
| | - Gregory Dyson
- Karmanos Cancer Institute, Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan
| | | | | | | | - Kay-Uwe Wagner
- Karmanos Cancer Institute, Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan
| | - Nerissa T Viola
- Karmanos Cancer Institute, Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan
| | | | - Philip A Philip
- Karmanos Cancer Institute, Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan
| | - Ramzi M Mohammad
- Karmanos Cancer Institute, Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan
| | - Asfar S Azmi
- Karmanos Cancer Institute, Department of Oncology, School of Medicine, Wayne State University, Detroit, Michigan.
| |
Collapse
|
11
|
Rinke A, Auernhammer CJ, Bodei L, Kidd M, Krug S, Lawlor R, Marinoni I, Perren A, Scarpa A, Sorbye H, Pavel ME, Weber MM, Modlin I, Gress TM. Treatment of advanced gastroenteropancreatic neuroendocrine neoplasia, are we on the way to personalised medicine? Gut 2021; 70:1768-1781. [PMID: 33692095 DOI: 10.1136/gutjnl-2020-321300] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 02/03/2021] [Accepted: 02/08/2021] [Indexed: 12/14/2022]
Abstract
Gastroenteropancreatic neuroendocrine neoplasia (GEPNEN) comprises clinically as well as prognostically diverse tumour entities often diagnosed at late stage. Current classification provides a uniform terminology and a Ki67-based grading system, thereby facilitating management. Advances in the study of genomic and epigenetic landscapes have amplified knowledge of tumour biology and enhanced identification of prognostic and potentially predictive treatment subgroups. Translation of this genomic and mechanistic biology into advanced GEPNEN management is limited. 'Targeted' treatments such as somatostatin analogues, peptide receptor radiotherapy, tyrosine kinase inhibitors and mammalian target of rapamycin inhibitors are treatment options but predictive tools are lacking. The inability to identify clonal heterogeneity and define critical oncoregulatory pathways prior to therapy, restrict therapeutic efficacy as does the inability to monitor disease status in real time. Chemotherapy in the poor prognosis NEN G3 group, though associated with acceptable response rates, only leads to short-term tumour control and their molecular biology requires delineation to provide new and more specific treatment options.The future requires an exploration of the NEN tumour genome, its microenvironment and an identification of critical oncologic checkpoints for precise drug targeting. In the advance to personalised medical treatment of patients with GEPNEN, clinical trials need to be based on mechanistic and multidimensional characterisation of each tumour in order to identify the therapeutic agent effective for the individual tumour.This review surveys advances in NEN research and delineates the current status of translation with a view to laying the basis for a genome-based personalised medicine management of advanced GEPNEN.
Collapse
Affiliation(s)
- Anja Rinke
- Department of Gastroenterology, Endocrinology, Metabolism and Infectiology, University Hospital Marburg and Philipps University, Marburg, Germany
| | - Christoph J Auernhammer
- Department of Internal Medicine IV and Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Ludwig Maximilian University, LMU Klinikum, Munich, Germany
| | - Lisa Bodei
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mark Kidd
- Wren Laboratories, Branford, Connecticut, USA
| | - Sebastian Krug
- Clinic for Internal Medicine I, Martin Luther University, Halle, Germany
| | - Rita Lawlor
- Applied Research on Cancer Centre, Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Ilaria Marinoni
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Aurel Perren
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Aldo Scarpa
- Applied Research on Cancer Centre, Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Halfdan Sorbye
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Marianne Ellen Pavel
- Department of Internal Medicine I, Endocrinology, University of Erlangen, Erlangen, Germany
| | - Matthias M Weber
- Department of Internal Medicine I, Endocrinology, Johannes Gutenberg University Hospital Mainz, Mainz, Germany
| | - Irvin Modlin
- Gastroenterological and Endoscopic Surgery, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Thomas M Gress
- Department of Gastroenterology, Endocrinology, Metabolism and Infectiology, University Hospital Marburg and Philipps University, Marburg, Germany
| |
Collapse
|
12
|
Antitumoral Activity of the MEK Inhibitor Trametinib (TMT212) Alone and in Combination with the CDK4/6 Inhibitor Ribociclib (LEE011) in Neuroendocrine Tumor Cells In Vitro. Cancers (Basel) 2021; 13:cancers13061485. [PMID: 33807122 PMCID: PMC8004919 DOI: 10.3390/cancers13061485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES This study assessed the antitumoral activity of the MEK inhibitor trametinib (TMT212) and the ERK1/2 inhibitor SCH772984, alone and in combination with the CDK4/6 inhibitor ribociclib (LEE011) in human neuroendocrine tumor (NET) cell lines in vitro. METHODS Human NET cell lines BON1, QGP-1, and NCI-H727 were treated with trametinib or SCH772984, alone and in combination with ribociclib, to assess cell proliferation, cell cycle distribution, and protein signaling using cell proliferation, flow cytometry, and Western blot assays, respectively. RESULTS Trametinib and SCH772984, alone and in combination with ribociclib, significantly reduced NET cell viability and arrested NET cells at the G1 phase of the cell cycle in all three cell lines tested. In addition, trametinib also caused subG1 events and apoptotic PARP cleavage in QGP1 and NCI-H727 cells. A western blot analysis demonstrated the use of trametinib alone and trametinib in combination with ribociclib to decrease the expression of pERK, cMyc, Chk1, pChk2, pCDK1, CyclinD1, and c-myc in a time-dependent manner in NCI-H727 and QGP-1 cells. CONCLUSIONS MEK and ERK inhibition causes antiproliferative effects in human NET cell lines in vitro. The combination of the MEK inhibitor trametinib (TMT212) with the CDK4/6 inhibitor ribociclib (LEE011) causes additive antiproliferative effects. Future preclinical and clinical studies of MEK inhibition in NETs should be performed.
Collapse
|
13
|
Asa SL, La Rosa S, Basturk O, Adsay V, Minnetti M, Grossman AB. Molecular Pathology of Well-Differentiated Gastro-entero-pancreatic Neuroendocrine Tumors. Endocr Pathol 2021; 32:169-191. [PMID: 33459926 DOI: 10.1007/s12022-021-09662-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/03/2021] [Indexed: 12/17/2022]
Abstract
Well differentiated neuroendocrine tumors (NETs) arising in the gastrointestinal and pancreaticobiliary system are the most common neuroendocrine neoplasms. Studies of the molecular basis of these lesions have identified genetic mutations that predispose to familial endocrine neoplasia syndromes and occur both as germline events and in sporadic tumors. The mutations often involve epigenetic regulators rather than the oncogenes and tumor suppressors that are affected in other malignancies. Somatic copy number alterations and miRNAs have also been implicated in the development and progression of some of these tumors. The molecular profiles differ by location, but many are shared by tumors in other sites, including those outside the gastroenteropancreatic system. The approach to therapy relies on both the neuroendocrine nature of these tumors and the identification of specific alterations that can serve as targets for precision oncologic approaches.
Collapse
Affiliation(s)
- Sylvia L Asa
- Department of Pathology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, USA.
| | - Stefano La Rosa
- Institute of Pathology, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Olca Basturk
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Volkan Adsay
- Department of Pathology and Research Center for Translational Medicine (KUTTAM), Koç University Hospital, Istanbul, Turkey
| | - Marianna Minnetti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Ashley B Grossman
- Green Templeton College, University of Oxford and ENETS Centre of Excellence, Royal Free Hospital, London, UK
| |
Collapse
|
14
|
Wnt/β-Catenin Signaling Regulates CXCR4 Expression and [ 68Ga] Pentixafor Internalization in Neuroendocrine Tumor Cells. Diagnostics (Basel) 2021; 11:diagnostics11020367. [PMID: 33671498 PMCID: PMC7926465 DOI: 10.3390/diagnostics11020367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 02/08/2023] Open
Abstract
Loss of Somatostatin Receptor 2 (SSTR2) expression and rising CXC Chemokine Receptor Type 4 (CXCR4) expression are associated with dedifferentiation in neuroendocrine tumors (NET). In NET, CXCR4 expression is associated with enhanced metastatic and invasive potential and worse prognosis but might be a theragnostic target. Likewise, activation of Wnt/β-catenin signaling may promote a more aggressive phenotype in NET. We hypothesized an interaction of the Wnt/β-catenin pathway with CXCR4 expression and function in NET. The NET cell lines BON-1, QGP-1, and MS-18 were exposed to Wnt inhibitors (5-aza-CdR, quercetin, and niclosamide) or the Wnt activator LiCl. The expressions of Wnt pathway genes and of CXCR4 were studied by qRT-PCR, Western blot, and immunohistochemistry. The effects of Wnt modulators on uptake of the CXCR4 ligand [68Ga] Pentixafor were measured. The Wnt activator LiCl induced upregulation of CXCR4 and Wnt target gene expression. Treatment with the Wnt inhibitors had opposite effects. LiCl significantly increased [68Ga] Pentixafor uptake, while treatment with Wnt inhibitors decreased radiopeptide uptake. Wnt pathway modulation influences CXCR4 expression and function in NET cell lines. Wnt modulation might be a tool to enhance the efficacy of CXCR4-directed therapies in NET or to inhibit CXCR4-dependent proliferative signaling. The underlying mechanisms for the interaction of the Wnt pathway with CXCR4 expression and function have yet to be clarified.
Collapse
|
15
|
Xie S, Li L, Wang X, Li L. Development and validation of a nomogram for predicting the overall survival of patients with gastroenteropancreatic neuroendocrine neoplasms. Medicine (Baltimore) 2021; 100:e24223. [PMID: 33466202 PMCID: PMC7808509 DOI: 10.1097/md.0000000000024223] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 12/15/2020] [Indexed: 01/05/2023] Open
Abstract
Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) are increasing in incidence. Clinicians urgently need a method that can effectively predict the prognosis of GEP-NENs.A total of 14770 GEP-NENs patients with pathologically confirmed between 1975 and 2016 were obtained from the surveillance, epidemiology, and end results database. All the patients were divided into primary (n = 10377) and validation (n = 4393) cohorts based on the principle of random grouping. Multivariate Cox proportional hazards proportional hazards regression analysis was performed to evaluate predictors associated with overall survival, and a nomogram was constructed based on the primary cohort. An independent external validation cohort and comparison with the eighth edition American Joint Committee on Cancer TNM staging system were subsequently used to assess the predictive performance of the nomogram.The multivariate Cox model indicated that age, tumour differentiation, and distant metastases were independent predictors associated with overall survival. With respect to the primary cohort, the nomogram exhibited better discriminatory power than the TNM classification (C-index: 0.821 vs 0.738). Discrimination was also superior to that of TNM classification for the validation cohort (C-index: 0.823 vs 0.738). The calibrated nomogram predicted 3- and 5-years survival rate that closely corresponded to the actual survival rate.This study developed and validated a prognostic nomogram applied to patients with GEP-NENs, which may help clinicians make reasonable prognostic judgments and treatment plans to a certain extent.
Collapse
Affiliation(s)
- Si Xie
- Department of Hepatobiliary Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
- Department of Gastrointestinal Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lei Li
- Department of Gastrointestinal Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xiaotong Wang
- Department of Gastrointestinal Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lequn Li
- Department of Hepatobiliary Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
| |
Collapse
|
16
|
Kloker LD, Berchtold S, Smirnow I, Beil J, Krieg A, Sipos B, Lauer UM. Oncolytic vaccinia virus GLV-1h68 exhibits profound antitumoral activities in cell lines originating from neuroendocrine neoplasms. BMC Cancer 2020; 20:628. [PMID: 32631270 PMCID: PMC7339398 DOI: 10.1186/s12885-020-07121-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
Background Oncolytic virotherapy is an upcoming treatment option for many tumor entities. But so far, a first oncolytic virus only was approved for advanced stages of malignant melanomas. Neuroendocrine tumors (NETs) constitute a heterogenous group of tumors arising from the neuroendocrine system at diverse anatomic sites. Due to often slow growth rates and (in most cases) endocrine non-functionality, NETs are often detected only in a progressed metastatic situation, where therapy options are still severely limited. So far, immunotherapies and especially immunovirotherapies are not established as novel treatment modalities for NETs. Methods In this immunovirotherapy study, pancreatic NET (BON-1, QGP-1), lung NET (H727, UMC-11), as well as neuroendocrine carcinoma (NEC) cell lines (HROC-57, NEC-DUE1) were employed. The well characterized genetically engineered vaccinia virus GLV-1 h68, which has already been investigated in various clinical trials, was chosen as virotherapeutical treatment modality. Results Profound oncolytic efficiencies were found for NET/NEC tumor cells. Besides, NET/NEC tumor cell bound expression of GLV-1 h68-encoded marker genes was observed also. Furthermore, a highly efficient production of viral progenies was detected by sequential virus quantifications. Moreover, the mTOR inhibitor everolimus, licensed for treatment of metastatic NETs, was not found to interfere with GLV-1 h68 replication, making a combinatorial treatment of both feasible. Conclusions In summary, the oncolytic vaccinia virus GLV-1 h68 was found to exhibit promising antitumoral activities, replication capacities and a potential for future combinatorial approaches in cell lines originating from neuroendocrine neoplasms. Based on these preliminary findings, virotherapeutic effects now have to be further evaluated in animal models for treatment of Neuroendocrine neoplasms (NENs).
Collapse
Affiliation(s)
- Linus D Kloker
- Department of Internal Medicine VIII, Department of Medical Oncology and Pneumology, University Hospital Tuebingen, University of Tuebingen, Otfried-Mueller-Strasse 10, 72076, Tuebingen, Baden-Wuerttemberg, Germany
| | - Susanne Berchtold
- Department of Internal Medicine VIII, Department of Medical Oncology and Pneumology, University Hospital Tuebingen, University of Tuebingen, Otfried-Mueller-Strasse 10, 72076, Tuebingen, Baden-Wuerttemberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 72076, Tuebingen, Germany
| | - Irina Smirnow
- Department of Internal Medicine VIII, Department of Medical Oncology and Pneumology, University Hospital Tuebingen, University of Tuebingen, Otfried-Mueller-Strasse 10, 72076, Tuebingen, Baden-Wuerttemberg, Germany
| | - Julia Beil
- Department of Internal Medicine VIII, Department of Medical Oncology and Pneumology, University Hospital Tuebingen, University of Tuebingen, Otfried-Mueller-Strasse 10, 72076, Tuebingen, Baden-Wuerttemberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 72076, Tuebingen, Germany
| | - Andreas Krieg
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, 40225, Duesseldorf, Germany
| | - Bence Sipos
- Department of Internal Medicine VIII, Department of Medical Oncology and Pneumology, University Hospital Tuebingen, University of Tuebingen, Otfried-Mueller-Strasse 10, 72076, Tuebingen, Baden-Wuerttemberg, Germany
| | - Ulrich M Lauer
- Department of Internal Medicine VIII, Department of Medical Oncology and Pneumology, University Hospital Tuebingen, University of Tuebingen, Otfried-Mueller-Strasse 10, 72076, Tuebingen, Baden-Wuerttemberg, Germany. .,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 72076, Tuebingen, Germany.
| |
Collapse
|
17
|
Abstract
The increased incidence and prevalence of neuroendocrine tumors (NETs) over the past few decades has been accompanied by an improvement in overall survival. There are differences in the management of small bowel NETs versus pNETs. The management of all patients with NETs must be individualized based on patient characteristics as well tumor-related factors. This article reviews the role of somatostatin analogues, historical results with chemotherapy in gastroenteropancreatic NETs (GEPNETs), and more recent evidence for the use of cytotoxic chemotherapy in GEPNETs. The article also discusses molecular targeted therapies approved for use in GEPNETs and some ongoing clinical trials.
Collapse
Affiliation(s)
- Chandrikha Chandrasekharan
- Division of Medical Oncology, University of Iowa, 200 Hawkins Drive, C GH 32, Iowa City, Iowa 52242, USA.
| |
Collapse
|
18
|
Jin XF, Spöttl G, Maurer J, Nölting S, Auernhammer CJ. Inhibition of Wnt/β-Catenin Signaling in Neuroendocrine Tumors in vitro: Antitumoral Effects. Cancers (Basel) 2020; 12:cancers12020345. [PMID: 32033025 PMCID: PMC7072467 DOI: 10.3390/cancers12020345] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/18/2020] [Accepted: 01/30/2020] [Indexed: 12/17/2022] Open
Abstract
Background and aims: Inhibition of Wnt/β-catenin signaling by specific inhibitors is currently being investigated as an antitumoral strategy for various cancers. The role of Wnt/β-catenin signaling in neuroendocrine tumors still needs to be further investigated. Methods: This study investigated the antitumor activity of the porcupine (PORCN) inhibitor WNT974 and the β-catenin inhibitor PRI-724 in human neuroendocrine tumor (NET) cell lines BON1, QGP-1, and NCI-H727 in vitro. NET cells were treated with WNT974, PRI-724, or small interfering ribonucleic acids against β-catenin, and subsequent analyses included cell viability assays, flow cytometric cell cycle analysis, caspase3/7 assays and Western blot analysis. Results: Treatment of NET cells with WNT974 significantly reduced NET cell viability in a dose- and time-dependent manner by inducing NET cell cycle arrest at the G1 and G2/M phases without inducing apoptosis. WNT974 primarily blocked Wnt/β-catenin signaling by the dose- and time-dependent downregulation of low-density lipoprotein receptor-related protein 6 (LRP6) phosphorylation and non-phosphorylated β-catenin and total β-catenin, as well as the genes targeting the latter (c-Myc and cyclinD1). Furthermore, the WNT974-induced reduction of NET cell viability occurred through the inhibition of GSK-3-dependent or independent signaling (including pAKT/mTOR, pEGFR and pIGFR signaling). Similarly, treatment of NET cells with the β-catenin inhibitor PRI-724 caused significant growth inhibition, while the knockdown of β-catenin expression by siRNA reduced NET tumor cell viability of BON1 cells but not of NCI-H727 cells. Conclusions: The PORCN inhibitor WNT974 possesses antitumor properties in NET cell lines by inhibiting Wnt and related signaling. In addition, the β-catenin inhibitor PRI-724 possesses antitumor properties in NET cell lines. Future studies are needed to determine the role of Wnt/β-catenin signaling in NET as a potential therapeutic target.
Collapse
Affiliation(s)
- Xi-Feng Jin
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany; (X.-F.J.); (G.S.); (J.M.); (S.N.)
| | - Gerald Spöttl
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany; (X.-F.J.); (G.S.); (J.M.); (S.N.)
| | - Julian Maurer
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany; (X.-F.J.); (G.S.); (J.M.); (S.N.)
| | - Svenja Nölting
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany; (X.-F.J.); (G.S.); (J.M.); (S.N.)
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Marchioninistr. 15, 81377 Munich, Germany
| | - Christoph Josef Auernhammer
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany; (X.-F.J.); (G.S.); (J.M.); (S.N.)
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Marchioninistr. 15, 81377 Munich, Germany
- Correspondence:
| |
Collapse
|
19
|
Mpilla G, Aboukameel A, Muqbil I, Kim S, Beydoun R, Philip PA, Mohammad RM, Kamgar M, Shidham V, Senapedis W, Baloglu E, Li J, Dyson G, Xue Y, El-Rayes B, Azmi AS. PAK4-NAMPT Dual Inhibition as a Novel Strategy for Therapy Resistant Pancreatic Neuroendocrine Tumors. Cancers (Basel) 2019; 11:1902. [PMID: 31795447 PMCID: PMC6966587 DOI: 10.3390/cancers11121902] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/16/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022] Open
Abstract
Pancreatic neuroendocrine tumors (PNET) remain an unmet clinical need. In this study, we show that targeting both nicotinamide phosphoribosyltransferase (NAMPT) and p21-activated kinase 4 (PAK4) could become a synthetic lethal strategy for PNET. The expression of PAK4 and NAMPT was found to be higher in PNET tissue compared to normal cells. PAK4-NAMPT dual RNAi suppressed proliferation of PNET cell lines. Treatment with KPT-9274 (currently in a Phase I trial or analogs, PF3758309 (the PAK4 selective inhibitor) or FK866 (the NAMPT inhibitor)) suppressed the growth of PNET cell lines and synergized with the mammalian target of rapamycin (mTOR) inhibitors everolimus and INK-128. Molecular analysis of the combination treatment showed down-regulation of known everolimus resistance drivers. KPT-9274 suppressed NAD pool and ATP levels in PNET cell lines. Metabolomic profiling showed a statistically significant alteration in cellular energetic pathways. KPT-9274 given orally at 150 mg/kg 5 days/week for 4 weeks dramatically reduced PNET sub-cutaneous tumor growth. Residual tumor analysis demonstrated target engagement in vivo and recapitulated in vitro results. Our investigations demonstrate that PAK4 and NAMPT are two viable therapeutic targets in the difficult to treat PNET that warrant further clinical investigation.
Collapse
Affiliation(s)
- Gabriel Mpilla
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Amro Aboukameel
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Irfana Muqbil
- University of Detroit Mercy, Detroit, MI 48201, USA;
| | - Steve Kim
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Rafic Beydoun
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (R.B.); (V.S.)
| | - Philip A. Philip
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Ramzi M. Mohammad
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Mandana Kamgar
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Vinod Shidham
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (R.B.); (V.S.)
| | | | - Erkan Baloglu
- Karyopharm Therapeutics Inc., Newton, MA 02459, USA; (W.S.); (E.B.)
| | - Jing Li
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Gregory Dyson
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| | - Yue Xue
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA; (Y.X.); (B.E.-R.)
| | - Bassel El-Rayes
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA; (Y.X.); (B.E.-R.)
| | - Asfar S. Azmi
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (G.M.); (A.A.); (S.K.); (P.A.P.); (R.M.M.); (M.K.); (J.L.); (G.D.)
| |
Collapse
|
20
|
Abstract
Neuroendocrine tumors (NETs) originate from the neuroendocrine cell system in the bronchial and gastrointestinal tract and can produce hormones leading to distinct clinical syndromes. Systemic treatment of patients with unresectable NETs aims to control symptoms related to hormonal overproduction and tumor growth. In the last decades prognosis has improved as a result of increased detection of early stage disease and the introduction of somatostatin analogs (SSAs) as well as several new therapeutic options. SSAs are the first-line medical treatment of NETs and can control hormonal production and tumor growth. The development of next-generation multireceptor targeted and radiolabelled somatostatin analogs, as well as target-directed therapies (as second-line treatment options) further improve progression-free survival in NET patients. To date, however, a significant prolongation of overall survival with systemic treatment in NET has not been convincingly demonstrated. Several new medical options and treatment combinations will become available in the upcoming years, and although preliminary results of preclinical and clinical trials are encouraging, large, preferrably randomized clinical studies are required to provide definitive evidence of their effect on survival and symptom control.
Collapse
|
21
|
Romano D. Relevance of neuroendocrine tumours models assessed by kinomic profiling. ANNALES D'ENDOCRINOLOGIE 2019; 80:144-148. [PMID: 31054767 DOI: 10.1016/j.ando.2019.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although there is evidence of a significant rise of neuroendocrine tumours (NETs) incidence, current treatments are largely insufficient due to somewhat poor knowledge of these tumours. Despite many efforts achieved to expose driver oncogene mutations in NETs, the genetic landscape of NETs is characterized by relatively few mutations and chromosomal aberrations per tumour compared with other tumour types. In addition, NETs display few actionable mutations providing compelling rationale for targeted therapies. Recent works aiming at characterizing currently used NETs in vitro models at the genomic level raised concerns on their reliability as bona fide tools to study NETs biology. However, the lack of actionable mutation in NETs implies that sole use of genomic is not sufficient to describe these models and establish appropriate therapeutic strategies. Several kinases and kinase-involving signalling pathways have been demonstrated as abnormally regulated in NETs. Yet, kinases have only been investigated regardless of their involvement in large intracellular signalling networks. In order to assess the validity of in vitro NETs models to study NETs biology, "next-generation" high throughput functional technologies based on "kinome-wide activity" will demonstrate the similarities between signalling pathways in NETs models and patients' samples. These approaches will significantly assist in identifying actionable alterations in NETs signalling pathways and guide patient stratification into early-phase clinical trials based on kinase inhibition targeted therapies.
Collapse
Affiliation(s)
- David Romano
- Marseille Medical Genetics, MMG, U1251 Inserm, Aix-Marseille université, Marseille, France.
| |
Collapse
|
22
|
Mafficini A, Scarpa A. Genetics and Epigenetics of Gastroenteropancreatic Neuroendocrine Neoplasms. Endocr Rev 2019; 40:506-536. [PMID: 30657883 PMCID: PMC6534496 DOI: 10.1210/er.2018-00160] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 12/27/2018] [Indexed: 12/11/2022]
Abstract
Gastroenteropancreatic (GEP) neuroendocrine neoplasms (NENs) are heterogeneous regarding site of origin, biological behavior, and malignant potential. There has been a rapid increase in data publication during the last 10 years, mainly driven by high-throughput studies on pancreatic and small intestinal neuroendocrine tumors (NETs). This review summarizes the present knowledge on genetic and epigenetic alterations. We integrated the available information from each compartment to give a pathway-based overview. This provided a summary of the critical alterations sustaining neoplastic cells. It also highlighted similarities and differences across anatomical locations and points that need further investigation. GEP-NENs include well-differentiated NETs and poorly differentiated neuroendocrine carcinomas (NECs). NENs are graded as G1, G2, or G3 based on mitotic count and/or Ki-67 labeling index, NECs are G3 by definition. The distinction between NETs and NECs is also linked to their genetic background, as TP53 and RB1 inactivation in NECs set them apart from NETs. A large number of genetic and epigenetic alterations have been reported. Recurrent changes have been traced back to a reduced number of core pathways, including DNA damage repair, cell cycle regulation, and phosphatidylinositol 3-kinase/mammalian target of rapamycin signaling. In pancreatic tumors, chromatin remodeling/histone methylation and telomere alteration are also affected. However, also owing to the paucity of disease models, further research is necessary to fully integrate and functionalize data on deregulated pathways to recapitulate the large heterogeneity of behaviors displayed by these tumors. This is expected to impact diagnostics, prognostic stratification, and planning of personalized therapy.
Collapse
Affiliation(s)
- Andrea Mafficini
- ARC-Net Center for Applied Research on Cancer, University and Hospital Trust of Verona, Verona, Italy.,Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Scarpa
- ARC-Net Center for Applied Research on Cancer, University and Hospital Trust of Verona, Verona, Italy.,Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| |
Collapse
|
23
|
Jin XF, Auernhammer CJ, Ilhan H, Lindner S, Nölting S, Maurer J, Spöttl G, Orth M. Combination of 5-Fluorouracil with Epigenetic Modifiers Induces Radiosensitization, Somatostatin Receptor 2 Expression, and Radioligand Binding in Neuroendocrine Tumor Cells In Vitro. J Nucl Med 2019; 60:1240-1246. [DOI: 10.2967/jnumed.118.224048] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 02/06/2019] [Indexed: 02/06/2023] Open
|
24
|
Aristizabal Prada ET, Spöttl G, Maurer J, Lauseker M, Koziolek EJ, Schrader J, Grossman A, Pacak K, Beuschlein F, Auernhammer CJ, Nölting S. The role of GSK3 and its reversal with GSK3 antagonism in everolimus resistance. Endocr Relat Cancer 2018; 25:893-908. [PMID: 29895527 PMCID: PMC7439002 DOI: 10.1530/erc-18-0159] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 06/11/2018] [Indexed: 12/11/2022]
Abstract
Pancreatic neuroendocrine tumors (panNETs) are often inoperable at diagnosis. The mTORC1 inhibitor everolimus has been approved for the treatment of advanced NETs. However, the regular development of resistance to everolimus limits its clinical efficacy. We established two independent everolimus-resistant panNET (BON1) cell lines (BON1 RR1, BON1 RR2) to find potential mechanisms of resistance. After 24 weeks of permanent exposure to 10 nM everolimus, BON1 RR1 and BON1 RR2 showed stable resistance with cellular survival rates of 96.70% (IC50 = 5200 nM) and 92.30% (IC50 = 2500 nM), respectively. The control cell line showed sensitivity to 10 nM everolimus with cellular survival declining to 54.70% (IC50 = 34 nM). Both resistant cell lines did not regain sensitivity over time and showed persistent stable resistance after a drug holiday of 13 weeks. The mechanisms of resistance in our cell line model included morphological adaptations, G1 cell cycle arrest associated with reduced CDK1(cdc2) expression and decreased autophagy. Cellular migration potential was increased and indirectly linked to c-Met activation. GSK3 was over-activated in association with reduced baseline IRS-1 protein levels. Specific GSK3 inhibition strongly decreased BON1 RR1/RR2 cell survival. The combination of everolimus with the PI3Kα inhibitor BYL719 re-established everolimus sensitivity through GSK3 inhibition and restoration of autophagy. We suggest that GSK3 over-activation combined with decreased baseline IRS-1 protein levels and decreased autophagy may be a crucial feature of everolimus resistance, and hence, a possible therapeutic target.
Collapse
Affiliation(s)
- Elke Tatjana Aristizabal Prada
- Medizinische Klinik und Poliklinik IVInterdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München (KUM), Ludwig-Maximilians-University, Munich, Germany
| | - Gerald Spöttl
- Medizinische Klinik und Poliklinik IVInterdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München (KUM), Ludwig-Maximilians-University, Munich, Germany
| | - Julian Maurer
- Medizinische Klinik und Poliklinik IVInterdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München (KUM), Ludwig-Maximilians-University, Munich, Germany
| | - Michael Lauseker
- Institute for Medical Information SciencesBiometry, and Epidemiology, Campus Grosshadern, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Eva Jolanthe Koziolek
- Department of Nuclear MedicineUniversity Medical Center Charité, Berlin, Germany
- German Cancer Consortium (DKTK)Heidelberg, Germany
- German Cancer Research Center (DKFZ)Heidelberg, Germany
| | - Jörg Schrader
- I. Medizinische Klinik und PoliklinikUniversitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ashley Grossman
- Oxford Centre for DiabetesEndocrinology and Metabolism, University of Oxford, Oxford, UK
- Royal Free Hospital ENETS Centre of ExcellenceLondon, UK
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health, Bethesda, Maryland, USA
| | - Felix Beuschlein
- Medizinische Klinik und Poliklinik IVInterdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München (KUM), Ludwig-Maximilians-University, Munich, Germany
- Klinik für EndokrinologieDiabetologie und Klinische Ernährung, Universitätsspital Zürich, Zurich, Switzerland
| | - Christoph Joseph Auernhammer
- Medizinische Klinik und Poliklinik IVInterdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München (KUM), Ludwig-Maximilians-University, Munich, Germany
| | - Svenja Nölting
- Medizinische Klinik und Poliklinik IVInterdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universität München (KUM), Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|