1
|
Xiao Z, Xu H, Strosberg JR, Lu R, Zhu X, Deng S, Ding L, Ni Q, Warshaw AL, Yu X, Luo G. EGFR is a potential therapeutic target for highly glycosylated and aggressive pancreatic neuroendocrine neoplasms. Int J Cancer 2023; 153:164-172. [PMID: 36891979 DOI: 10.1002/ijc.34499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 10/22/2022] [Accepted: 11/04/2022] [Indexed: 03/10/2023]
Abstract
pNENs are relative indolent tumors with heterogeneous clinical presentation at diagnosis. It is important to establish aggressive subgroups of pNENs and identify potential therapeutic targets. Patients with pNEN (322 cases) were included to examine the association between glycosylation biomarkers and clinical/pathological traits. The molecular and metabolic features stratified by glycosylation status were assessed by RNA-seq/whole exome sequencing and immunohistochemistry. A considerable proportion of patients had elevated glycosylation biomarkers (carbohydrate antigen [CA] 19-9, 11.9%; CA125, 7.5%; carcinoembryonic antigen [CEA], 12.8%). CA19-9 (hazard ratio [HR] = 2.26, P = .019), CA125 (HR = 3.79, P = .004) and CEA (HR = 3.16, P = .002) were each independent prognostic variables for overall survival. High glycosylation group, defined as pNENs with elevated level of circulating CA19-9, CA125 or CEA, accounted for 23.4% of all pNENs. High glycosylation (HR = 3.14, P = .001) was an independent prognostic variable for overall survival and correlated with G3 grade (P < .001), poor differentiation (P = .001), perineural invasion (P = .004) and distant metastasis (P < .001). Epidermal growth factor receptor (EGFR) was enriched in high glycosylation pNENs using RNA-seq. EGFR was expressed in 21.2% of pNENs using immunohistochemistry and associated with poor overall survival (P = .020). A clinical trial focusing on EGFR expressed pNENs was initiated (NCT05316480). Thus, pNEN with aberrant glycosylation correlates with a dismal outcome and suggests potential therapeutic target of EGFR.
Collapse
Affiliation(s)
- Zhiwen Xiao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Huaxiang Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, Shanghai, China
| | | | - Renquan Lu
- Department of Laboratory, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xinzhe Zhu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Shengming Deng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Lei Ding
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Quanxing Ni
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Andrew L Warshaw
- Department of Surgery and the Warshaw Institute for Pancreatic Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Guopei Luo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai Pancreatic Cancer Institute, Shanghai, China
| |
Collapse
|
2
|
Wang Y, Wang F, Qin Y, Lou X, Ye Z, Zhang W, Gao H, Chen J, Xu X, Yu X, Ji S. Recent progress of experimental model in pancreatic neuroendocrine tumors: drawbacks and challenges. Endocrine 2023; 80:266-282. [PMID: 36648608 DOI: 10.1007/s12020-023-03299-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/31/2022] [Indexed: 01/18/2023]
Abstract
The neuroendocrine neoplasm, in general, refers to a heterogeneous group of all tumors originating from peptidergic neurons and neuroendocrine cells. Neuroendocrine neoplasms are divided into two histopathological subtypes: well-differentiated neuroendocrine tumors and poorly differentiated neuroendocrine carcinomas. Pancreatic neuroendocrine tumors account for more than 80% of pancreatic neuroendocrine neoplasms. Due to the greater proportion of pancreatic neuroendocrine tumors compared to pancreatic neuroendocrine carcinoma, this review will only focus on them. The worldwide incidence of pancreatic neuroendocrine tumors is rising year by year due to sensitive detection with an emphasis on medical examinations and the improvement of testing technology. Although the biological behavior of pancreatic neuroendocrine tumors tends to be inert, distant metastasis is common, often occurring very early. Because of the paucity of basic research on pancreatic neuroendocrine tumors, the mechanism of tumor development, metastasis, and recurrence are still unclear. In this context, the representative preclinical models simulating the tumor development process are becoming ever more widely appreciated to address the clinical problems of pancreatic neuroendocrine tumors. So far, there is no comprehensive report on the experimental model of pancreatic neuroendocrine tumors. This article systematically summarizes the characteristics of preclinical models, such as patient-derived cell lines, patient-derived xenografts, genetically engineered mouse models, and patient-derived organoids, and their advantages and disadvantages, to provide a reference for further studies of neuroendocrine tumors. We also highlight the method of establishment of liver metastasis mouse models.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Fei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xin Lou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Wuhu Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Heli Gao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Jie Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Bevere M, Gkountakos A, Martelli FM, Scarpa A, Luchini C, Simbolo M. An Insight on Functioning Pancreatic Neuroendocrine Neoplasms. Biomedicines 2023; 11:303. [PMID: 36830839 PMCID: PMC9953748 DOI: 10.3390/biomedicines11020303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Pancreatic neuroendocrine neoplasms (PanNENs) are rare neoplasms arising from islets of the Langerhans in the pancreas. They can be divided into two groups, based on peptide hormone secretion, functioning and nonfunctioning PanNENs. The first group is characterized by different secreted peptides causing specific syndromes and is further classified into subgroups: insulinoma, gastrinoma, glucagonoma, somatostatinoma, VIPoma and tumors producing serotonin and adrenocorticotrophic hormone. Conversely, the second group does not release peptides and is usually associated with a worse prognosis. Today, although the efforts to improve the therapeutic approaches, surgery remains the only curative treatment for patients with PanNENs. The development of high-throughput techniques has increased the molecular knowledge of PanNENs, thereby allowing us to understand better the molecular biology and potential therapeutic vulnerabilities of PanNENs. Although enormous advancements in therapeutic and molecular aspects of PanNENs have been achieved, there is poor knowledge about each subgroup of functioning PanNENs.Therefore, we believe that combining high-throughput platforms with new diagnostic tools will allow for the efficient characterization of the main differences among the subgroups of functioning PanNENs. In this narrative review, we summarize the current landscape regarding diagnosis, molecular profiling and treatment, and we discuss the future perspectives of functioning PanNENs.
Collapse
Affiliation(s)
- Michele Bevere
- Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy
- ARC-Net Applied Research on Cancer Centre, University and Hospital Trust of Verona, 37134 Verona, Italy
| | - Anastasios Gkountakos
- Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy
- ARC-Net Applied Research on Cancer Centre, University and Hospital Trust of Verona, 37134 Verona, Italy
| | - Filippo Maria Martelli
- Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy
- ARC-Net Applied Research on Cancer Centre, University and Hospital Trust of Verona, 37134 Verona, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy
| | - Michele Simbolo
- Department of Diagnostics and Public Health, Section of Anatomical Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy
| |
Collapse
|
4
|
Xiao Y, Xu G, Cloyd JM, Du S, Mao Y, Pawlik TM. Predicting Novel Drug Candidates for Pancreatic Neuroendocrine Tumors via Gene Signature Comparison and Connectivity Mapping. J Gastrointest Surg 2022; 26:1670-1678. [PMID: 35508682 DOI: 10.1007/s11605-022-05337-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/19/2022] [Indexed: 02/08/2023]
Abstract
INTRODUCTION There is a paucity of effective treatment options for advanced pancreatic neuroendocrine tumors (pNETs). Genome-wide analyses may allow for potential drugs to be identified based on differentially expressed genes (DEGs). METHODS Oligo microarray data of RNA expression profiling of pNETs and normal pancreas tissues were downloaded from the Gene Expression Omnibus. Functional and pathway enrichment information of the DEGs was obtained using the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes databases. Corresponding homologous proteins were analyzed and potential therapeutic drugs for pNETs were identified using the Connectivity Map and Drug-Gene Interaction Database. RESULTS Assessment of raw data from 12,610 pNET genes demonstrated that 1082 and 380 genes were upregulated and downregulated, respectively, compared with normal pancreas tissue. Upregulated pathways were associated with nitrogen metabolism (i.e., GABAergic synapse, and graft-versus-host disease), whereas downregulated pathways included C-type leptin receptor signaling pathway, pertussis and AMPK signaling pathway. In particular, the protein-protein interaction analysis revealed 10 upregulated hub genes (DYNLL1, GNB5, GNB2, GNG4, GNAI2, GNAI1, HIST2H2BE, NUP107, NUP133, and SNAP25) and 10 downregulated hub genes (CXCL8, F2, CXCL2, GCG, SST, INS, GALR3, CCL20, ADRA2B, and CXCL6). Using the Drug-Gene Interaction Database, candidate drugs for pNETs treatment included 3 EGFR inhibitors (canertinib, erlotinib, WZ-4-145), as well as other cell-signaling pathway inhibitors such as AG-592, acarbose, lonidamine, azacytidine, rottlerin, and HU-211. CONCLUSION Using available genetic atlas data, potential drug candidates for treatment of pNETs were identified based on differentially expressed genes. These results may help focus efforts on identifying targeted agents with therapeutic efficacy to treat patients with pNETs.
Collapse
Affiliation(s)
- Yao Xiao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Gang Xu
- Department of Liver Surgery and Liver Transplant Center, West China Hospital of Sichuan University, Chengdu, China
| | - Jordan M Cloyd
- Department of Surgery, The Ohio State University Wexner Medical Center and James Cancer Hospital and Solove Research Institute, 395 W. 12th Ave., Suite 670, Columbus, OH, USA
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Cancer Hospital and Solove Research Institute, 395 W. 12th Ave., Suite 670, Columbus, OH, USA.
| |
Collapse
|
5
|
Cuny T, van Koetsveld PM, Mondielli G, Dogan F, de Herder WW, Barlier A, Hofland LJ. Reciprocal Interactions between Fibroblast and Pancreatic Neuroendocrine Tumor Cells: Putative Impact of the Tumor Microenvironment. Cancers (Basel) 2022; 14:cancers14143481. [PMID: 35884539 PMCID: PMC9321816 DOI: 10.3390/cancers14143481] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/16/2022] [Indexed: 02/04/2023] Open
Abstract
Introduction: Pancreatic neuroendocrine neoplasms (PNENs) present with a fibrotic stroma that constitutes the tumor microenvironment (TME). The role played by stromal fibroblasts in the growth of PNENs and their sensitivity to the mTOR inhibitor RAD001 has not yet been established. Methods: We investigated reciprocal interactions between (1) human PNEN cell lines (BON-1/QGP-1) or primary cultures of human ileal neuroendocrine neoplasm (iNEN) or PNEN and (2) human fibroblast cell lines (HPF/HFL-1). Proliferation was assessed in transwell (tw) co-culture or in the presence of serum-free conditioned media (cm), with and without RAD001. Colony formation and migration of BON-1/QGP-1 were evaluated upon incubation with HPFcm. Results: Proliferation of BON-1 and QGP-1 increased in the presence of HFL-1cm, HPFcm, HFL-1tw and HPFtw (BON-1: +46−70% and QGP-1: +42−55%, p < 0.001 vs. controls) and HPFcm significantly increased the number of BON-1 or QGP-1 colonies (p < 0.05). This stimulatory effect was reversed in the presence of RAD001. Likewise, proliferation of human iNEN and PNEN primary cultures increased in the presence of HFL-1 or HPF. Reciprocally, BON-1cm and BONtw stimulated the proliferation of HPF (+90 ± 61% and +55 ± 47%, respectively, p < 0.001 vs. controls), an effect less pronounced with QGP-1cm or QGPtw (+19 to +27%, p < 0.05 vs. controls). Finally, a higher migration potential for BON-1 and QGP-1 was found in the presence of HPFcm (p < 0.001 vs. controls). Conclusions: Fibroblasts in the TME of PNENs represent a target of interest, the stimulatory effect of which over PNENs is mitigated by the mTOR inhibitor everolimus.
Collapse
Affiliation(s)
- Thomas Cuny
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (T.C.); (P.M.v.K.); (F.D.); (W.W.d.H.)
- Department of Endocrinology, Hôpital de la Conception, Centre de Référence des Maladies Rares Hypophysaires HYPO, Hôpitaux Universitaires de Marseille, 13005 Marseille, France;
- DiPNET Team, U1251, INSERM, Marseille Medical Genetics, Aix-Marseille Université, CEDEX 05, 13385 Marseille, France;
| | - Peter M. van Koetsveld
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (T.C.); (P.M.v.K.); (F.D.); (W.W.d.H.)
| | - Grégoire Mondielli
- DiPNET Team, U1251, INSERM, Marseille Medical Genetics, Aix-Marseille Université, CEDEX 05, 13385 Marseille, France;
| | - Fadime Dogan
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (T.C.); (P.M.v.K.); (F.D.); (W.W.d.H.)
| | - Wouter W. de Herder
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (T.C.); (P.M.v.K.); (F.D.); (W.W.d.H.)
| | - Anne Barlier
- Department of Endocrinology, Hôpital de la Conception, Centre de Référence des Maladies Rares Hypophysaires HYPO, Hôpitaux Universitaires de Marseille, 13005 Marseille, France;
- DiPNET Team, U1251, INSERM, Marseille Medical Genetics, Aix-Marseille Université, CEDEX 05, 13385 Marseille, France;
- Laboratory of Molecular Biology, Hôpital de la Conception, Hôpitaux Universitaires de Marseille, 13005 Marseille, France
| | - Leo J. Hofland
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (T.C.); (P.M.v.K.); (F.D.); (W.W.d.H.)
- Correspondence: ; Tel.: +31-10-703-46-33; Fax: +31-10-703-54-30
| |
Collapse
|
6
|
Cdk5 drives formation of heterogeneous pancreatic neuroendocrine tumors. Oncogenesis 2021; 10:83. [PMID: 34862365 PMCID: PMC8642406 DOI: 10.1038/s41389-021-00372-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/02/2021] [Accepted: 11/11/2021] [Indexed: 01/07/2023] Open
Abstract
Pancreatic neuroendocrine tumors (PanNETs) are a heterogeneous population of neoplasms that arise from hormone-secreting islet cells of the pancreas and have increased markedly in incidence over the past four decades. Non-functional PanNETs, which occur more frequently than hormone-secreting tumors, are often not diagnosed until later stages of tumor development and have poorer prognoses. Development of successful therapeutics for PanNETs has been slow, partially due to a lack of diverse animal models for pre-clinical testing. Here, we report development of an inducible, conditional mouse model of PanNETs by using a bi-transgenic system for regulated expression of the aberrant activator of Cdk5, p25, specifically in β-islet cells. This model produces a heterogeneous population of PanNETs that includes a subgroup of well-differentiated, non-functional tumors. Production of these tumors demonstrates the causative potential of aberrantly active Cdk5 for generation of PanNETs. Further, we show that human PanNETs express Cdk5 pathway components, are dependent on Cdk5 for growth, and share genetic and transcriptional overlap with the INS-p25OE model. The utility of this model is enhanced by the ability to form tumor-derived allografts. This new model of PanNETs will facilitate molecular delineation of Cdk5-dependent PanNETs and the development of new targeted therapeutics.
Collapse
|
7
|
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
|
8
|
Carpizo DR, Harris CR. Genetic Drivers of Ileal Neuroendocrine Tumors. Cancers (Basel) 2021; 13:cancers13205070. [PMID: 34680217 PMCID: PMC8533727 DOI: 10.3390/cancers13205070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Although ileal neuroendocrine tumors are the most common tumors of the small intestine, they are not well-defined at the genetic level. Unlike most cancers, they have an unusually low number of mutations, and also lack recurrently mutated genes. Moreover ileal NETs have been difficult to study in the laboratory because there were no animal models and because cell lines were generally unavailable. But recent advances, including the first ileal NET mouse model as well as methods for culturing patient tumor samples, have been described and have already helped to identify IGF2 and CDK4 as two of the genetic drivers for this tumor type. These advances may help in the development of new treatments for patients. Abstract The genetic causes of ileal neuroendocrine tumors (ileal NETs, or I-NETs) have been a mystery. For most types of tumors, key genes were revealed by large scale genomic sequencing that demonstrated recurrent mutations of specific oncogenes or tumor suppressors. In contrast, genomic sequencing of ileal NETs demonstrated a distinct lack of recurrently mutated genes, suggesting that the mechanisms that drive the formation of I-NETs may be quite different than the cell-intrinsic mutations that drive the formation of other tumor types. However, recent mouse studies have identified the IGF2 and RB1 pathways in the formation of ileal NETs, which is supported by the subsequent analysis of patient samples. Thus, ileal NETs no longer appear to be a cancer without genetic causes.
Collapse
|
9
|
Luo J, Lu C, Feng M, Dai L, Wang M, Qiu Y, Zheng H, Liu Y, Li L, Tang B, Xu C, Wang Y, Yang X. Cooperation between liver-specific mutations of pten and tp53 genetically induces hepatocarcinogenesis in zebrafish. J Exp Clin Cancer Res 2021; 40:262. [PMID: 34416907 PMCID: PMC8377946 DOI: 10.1186/s13046-021-02061-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 08/05/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Liver cancer, mainly hepatocellular carcinoma, is one of the deadliest cancers worldwide and has a poor prognosis due to insufficient understanding of hepatocarcinogenesis. Previous studies have revealed that the mutations in PTEN and TP53 are the two most common genetic events in hepatocarcinogenesis. Here, we illustrated the crosstalk between aberrant Pten and Tp53 pathways during hepatocarcinogenesis in zebrafish. METHODS We used the CRISPR/Cas9 system to establish several transgenic zebrafish lines with single or double tissue-specific mutations of pten and tp53 to genetically induce liver tumorigenesis. Next, the morphological and histological determination were performed to investigate the roles of Pten and Tp53 signalling pathways in hepatocarcinogenesis in zebrafish. RESULTS We demonstrated that Pten loss alone induces hepatocarcinogenesis with only low efficiency, whereas single mutation of tp53 failed to induce tumour formation in liver tissue in zebrafish. Moreover, zebrafish with double mutations of pten and tp53 exhibits a much higher tumour incidence, higher-grade histology, and a shorter survival time than single-mutant zebrafish, indicating that these two signalling pathways play important roles in dynamic biological events critical for the initiation and progression of hepatocarcinogenesis in zebrafish. Further histological and pathological analyses showed significant similarity between the tumours generated from liver tissues of zebrafish and humans. Furthermore, the treatment with MK-2206, a specific Akt inhibitor, effectively suppressed hepatocarcinogenesis in zebrafish. CONCLUSION Our findings will offer a preclinical animal model for genetically investigating hepatocarcinogenesis and provide a useful platform for high-throughput anticancer drug screening.
Collapse
Affiliation(s)
- Juanjuan Luo
- Key laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
- Shantou University Medical College, Shantou, China
| | - Chunjiao Lu
- Shantou University Medical College, Shantou, China
| | - Meilan Feng
- Key laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Lu Dai
- Key laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Maya Wang
- Key laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Yang Qiu
- Key laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Huilu Zheng
- Key laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Yao Liu
- Shantou University Medical College, Shantou, China
| | - Li Li
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Key Laboratory of Aquatic Science of Chongqing, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Chongqing, China
| | - Bo Tang
- Department of Hepatobiliary Surgery, The first Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chuan Xu
- Integrative Cancer Center & Cancer Clinical Research Center, Cancer Center, Sichuan Cancer Hospital & Institute Sichuan, School of Medicine University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Yajun Wang
- Key laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China.
| | - Xiaojun Yang
- Shantou University Medical College, Shantou, China.
| |
Collapse
|
10
|
RABL6A Promotes Pancreatic Neuroendocrine Tumor Angiogenesis and Progression In Vivo. Biomedicines 2021; 9:biomedicines9060633. [PMID: 34199469 PMCID: PMC8228095 DOI: 10.3390/biomedicines9060633] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic neuroendocrine tumors (pNETs) are difficult-to-treat neoplasms whose incidence is rising. Greater understanding of pNET pathogenesis is needed to identify new biomarkers and targets for improved therapy. RABL6A, a novel oncogenic GTPase, is highly expressed in patient pNETs and required for pNET cell proliferation and survival in vitro. Here, we investigated the role of RABL6A in pNET progression in vivo using a well-established model of the disease. RIP-Tag2 (RT2) mice develop functional pNETs (insulinomas) due to SV40 large T-antigen expression in pancreatic islet β cells. RABL6A loss in RT2 mice significantly delayed pancreatic tumor formation, reduced tumor angiogenesis and mitoses, and extended survival. Those effects correlated with upregulation of anti-angiogenic p19ARF and downregulation of proangiogenic c-Myc in RABL6A-deficient islets and tumors. Our findings demonstrate that RABL6A is a bona fide oncogenic driver of pNET angiogenesis and development in vivo.
Collapse
|
11
|
Targeted Cancer Therapy: What's New in the Field of Neuroendocrine Neoplasms? Cancers (Basel) 2021; 13:cancers13071701. [PMID: 33916707 PMCID: PMC8038369 DOI: 10.3390/cancers13071701] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/18/2022] Open
Abstract
Neuroendocrine tumors (NETs) are a heterogeneous family of neoplasms of increasing incidence and high prevalence due to their relatively indolent nature. Their wide anatomic distribution and their characteristic ability to secrete hormonally active substances pose unique challenges for clinical management. They are also characterized by the common expression of somatostatin receptors, a target that has been extremely useful for diagnosis and treatment (i.e., somatostatin analogues (SSAs) and peptide-receptor radionuclide therapy (PRRT)). Chemotherapy is of limited use for NETs of non-pancreatic origin, and the only approved targeted agents for advanced progressive NETs are sunitinib for those of pancreatic origin, and everolimus for lung, gastrointestinal and pancreatic primaries. Despite recent therapeutic achievements, thus, systemic treatment options remain limited. In this review we will discuss the state-of-the-art targeted therapies in the field of NETs, and also future perspectives of novel therapeutic drugs or strategies in clinical development, including recently presented results from randomized trials of yet unapproved antiangiogenic agents (i.e., pazopanib, surufatinib and axitinib), PRRT including both approved radiopharmaceuticals (177Lu-Oxodotreotide) and others in development (177Lu-Edotreotide, 177Lu-Satoreotide Tetraxetan), immunotherapy and other innovative targeted strategies (antibody-drug conjugates, bites,…) that shall soon improve the landscape of personalized treatment options in NET patients.
Collapse
|
12
|
Detjen K, Hammerich L, Özdirik B, Demir M, Wiedenmann B, Tacke F, Jann H, Roderburg C. Models of Gastroenteropancreatic Neuroendocrine Neoplasms: Current Status and Future Directions. Neuroendocrinology 2021; 111:217-236. [PMID: 32615560 DOI: 10.1159/000509864] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/23/2020] [Indexed: 11/19/2022]
Abstract
Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) are a rare, heterogeneous group of tumors that originate from the endocrine system of the gastrointestinal tract and pancreas. GEP-NENs are subdivided according to their differentiation into well-differentiated neuroendocrine tumors (NETs) and poorly differentiated neuroendocrine carcinomas (NECs). Since GEP-NENs represent rare diseases, only limited data from large prospective, randomized clinical trials are available, and recommendations for treatment of GEP-NEN are in part based on data from retrospective analyses or case series. In this context, tractable disease models that reflect the situation in humans and that allow to recapitulate the different clinical aspects and disease stages of GEP-NET or GEP-NEC are urgently needed. In this review, we highlight available data on mouse models for GEP-NEN. We discuss how these models reflect tumor biology of human disease and whether these models could serve as a tool for understanding the pathogenesis of GEP-NEN and for disease modeling and pharmacosensitivity assays, facilitating prediction of treatment response in patients. In addition, open issues applicable for future developments will be discussed.
Collapse
Affiliation(s)
- Katharina Detjen
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Linda Hammerich
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Burcin Özdirik
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Münevver Demir
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Bertram Wiedenmann
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Henning Jann
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Christoph Roderburg
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany,
| |
Collapse
|
13
|
Yamauchi Y, Kodama Y, Shiokawa M, Kakiuchi N, Marui S, Kuwada T, Sogabe Y, Tomono T, Mima A, Morita T, Matsumori T, Ueda T, Tsuda M, Nishikawa Y, Kuriyama K, Sakuma Y, Ota Y, Maruno T, Uza N, Masuda A, Tatsuoka H, Yabe D, Minamiguchi S, Masui T, Inagaki N, Uemoto S, Chiba T, Seno H. Rb and p53 Execute Distinct Roles in the Development of Pancreatic Neuroendocrine Tumors. Cancer Res 2020; 80:3620-3630. [PMID: 32591410 DOI: 10.1158/0008-5472.can-19-2232] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/26/2020] [Accepted: 06/23/2020] [Indexed: 11/16/2022]
Abstract
Pancreatic neuroendocrine tumors (PanNET) were classified into grades (G) 1 to 3 by the World Health Organization in 2017, but the precise mechanisms of PanNET initiation and progression have remained unclear. In this study, we used a genetically engineered mouse model to investigate the mechanisms of PanNET formation. Although pancreas-specific deletion of the Rb gene (Pdx1-Cre;Rbf/f ) in mice did not affect pancreatic exocrine cells, the α-cell/β-cell ratio of islet cells was decreased at 8 months of age. During long-term observation (18-20 months), mice formed well-differentiated PanNET with a Ki67-labeling index of 2.7%. In contrast, pancreas-specific induction of a p53 mutation (Pdx1-Cre;Trp53R172H ) had no effect on pancreatic exocrine and endocrine tissues, but simultaneous induction of a p53 mutation with Rb gene deletion (Pdx1-Cre;Trp53R172H;Rb f/f ) resulted in the formation of aggressive PanNET with a Ki67-labeling index of 24.7% over the short-term (4 months). In Pdx1-Cre;Trp53R172H;Rbf/f mice, mRNA expression of Pten and Tsc2, negative regulators of the mTOR pathway, significantly decreased in the islet cells, and activation of the mTOR pathway was confirmed in subsequently formed PanNET. Thus, by manipulating Rb and p53 genes, we established a multistep progression model from dysplastic islet to indolent PanNET and aggressive metastatic PanNET in mice. These observations suggest that Rb and p53 have distinct roles in the development of PanNET. SIGNIFICANCE: Pancreas-specific manipulation of Rb and p53 genes induced malignant transformation of islet cells, reproducing stepwise progression from microadenomas to indolent (grade 1) and subsequent aggressive PanNETs (grade 2-3).
Collapse
Affiliation(s)
- Yuki Yamauchi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Yuzo Kodama
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan. .,Department of Gastroenterology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, Japan
| | - Masahiro Shiokawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Nobuyuki Kakiuchi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Saiko Marui
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Takeshi Kuwada
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Yuko Sogabe
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Teruko Tomono
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Atsushi Mima
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Toshihiro Morita
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Tomoaki Matsumori
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Tatsuki Ueda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Motoyuki Tsuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Yoshihiro Nishikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Katsutoshi Kuriyama
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Yojiro Sakuma
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Yuji Ota
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Takahisa Maruno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Atsuhiro Masuda
- Department of Gastroenterology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, Japan
| | - Hisato Tatsuoka
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Daisuke Yabe
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Sachiko Minamiguchi
- Department of Diagnostic Pathology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Toshihiko Masui
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Kyoto University, Shogoin-Kawahara, Sakyo-ku, Kyoto, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Shinji Uemoto
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Kyoto University, Shogoin-Kawahara, Sakyo-ku, Kyoto, Japan
| | - Tsutomu Chiba
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan.,Kansai Electric Power Hospital, Fukushima-ku, Osaka, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Shogoin, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
14
|
Wong C, Tang LH, Davidson C, Vosburgh E, Chen W, Foran DJ, Notterman DA, Levine AJ, Xu EY. Two well-differentiated pancreatic neuroendocrine tumor mouse models. Cell Death Differ 2020; 27:269-283. [PMID: 31160716 PMCID: PMC7206057 DOI: 10.1038/s41418-019-0355-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/26/2019] [Accepted: 05/07/2019] [Indexed: 02/08/2023] Open
Abstract
Multiple endocrine neoplasia type 1 (MEN1) is a genetic syndrome in which patients develop neuroendocrine tumors (NETs), including pancreatic neuroendocrine tumors (PanNETs). The prolonged latency of tumor development in MEN1 patients suggests a likelihood that other mutations cooperate with Men1 to induce PanNETs. We propose that Pten loss combined with Men1 loss accelerates tumorigenesis. To test this, we developed two genetically engineered mouse models (GEMMs)-MPR (Men1flox/flox Ptenflox/flox RIP-Cre) and MPM (Men1flox/flox Ptenflox/flox MIP-Cre) using the Cre-LoxP system with insulin-specific biallelic inactivation of Men1 and Pten. Cre in the MPR mouse model was driven by the transgenic rat insulin 2 promoter while in the MPM mouse model was driven by the knock-in mouse insulin 1 promoter. Both mouse models developed well-differentiated (WD) G1/G2 PanNETs at a much shorter latency than Men1 or Pten single deletion alone and exhibited histopathology of human MEN1-like tumor. The MPR model, additionally, developed pituitary neuroendocrine tumors (PitNETs) in the same mouse at a much shorter latency than Men1 or Pten single deletion alone as well. Our data also demonstrate that Pten plays a role in NE tumorigenesis in pancreas and pituitary. Treatment with the mTOR inhibitor rapamycin delayed the growth of PanNETs in both MPR and MPM mice, as well as the growth of PitNETs, resulting in prolonged survival in MPR mice. Our MPR and MPM mouse models are the first to underscore the cooperative roles of Men1 and Pten in cancer, particularly neuroendocrine cancer. The early onset of WD PanNETs mimicking the human counterpart in MPR and MPM mice at 7 weeks provides an effective platform for evaluating therapeutic opportunities for NETs through targeting the MENIN-mediated and PI3K/AKT/mTOR signaling pathways.
Collapse
Affiliation(s)
- Chung Wong
- Raymond and Beverly Sackler Foundation Laboratory, New Brunswick, NJ, 08901, USA
- Regeneron Inc., Tarrytown, NY, 10591, USA
| | - Laura H Tang
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Christian Davidson
- Department of Pathology, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, 84112, USA
| | - Evan Vosburgh
- Raymond and Beverly Sackler Foundation Laboratory, New Brunswick, NJ, 08901, USA
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08903, USA
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08901, USA
- Department of Medicine, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Wenjin Chen
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08903, USA
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - David J Foran
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08903, USA
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Daniel A Notterman
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Arnold J Levine
- School of Natural Sciences, Institute for Advanced Study, Princeton, NJ, 08540, USA
| | - Eugenia Y Xu
- Raymond and Beverly Sackler Foundation Laboratory, New Brunswick, NJ, 08901, USA.
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08903, USA.
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08901, USA.
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
15
|
Kim S. Pathway Interactions Based on Drug-Induced Datasets. Cancer Inform 2019; 18:1176935119851518. [PMID: 31205412 PMCID: PMC6535899 DOI: 10.1177/1176935119851518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 04/23/2019] [Indexed: 11/16/2022] Open
Abstract
In this study, we identified enrichment pathway connections from MCF7 breast cancer epithelial cells that were treated with 87 drugs. We extracted drug-treated samples, where the sample size was greater than or equal to 5. The drugs included 17-allylamino-geldanamycin, LY294002, trichostatin A, valproic acid, sirolimus, and wortmannin, which had sample sizes of 11, 8, 7, 7, 7, and 5, respectively. We found meaningful pathways using gene set enrichment analysis and identified intradrug and interdrug pathway interactions, which implied the influence of drug combination. Among the top 20 enrichment pathways that were wortmannin induced, there were a total of 37 intradrug pathway interactions via common genes. Thirty-seven pathway interactions were induced by valproic acid, 11 induced by trichostatin A, 20 induced by LY294002, and 59 induced by sirolimus, all via common genes. The number of interdrug-induced pathway interactions ranged from one pair of pathways to 23. The pair of ERBB_SIGNALING and INSULIN_SIGNALING pathways showed the highest score from a pair of 2 individual drugs. The highest number of pathway interactions was observed between the drugs 17-allylamino-geldanamycin and LY294002.
Collapse
Affiliation(s)
- Shinuk Kim
- Department of Civil Engineering, Sangmyung University, Cheonan, Republic of Korea
| |
Collapse
|
16
|
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: 3.8] [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
|
17
|
Alleles of Insm1 determine whether RIP1-Tag2 mice produce insulinomas or nonfunctioning pancreatic neuroendocrine tumors. Oncogenesis 2019; 8:16. [PMID: 30796198 PMCID: PMC6386750 DOI: 10.1038/s41389-019-0127-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/31/2019] [Accepted: 02/08/2019] [Indexed: 01/22/2023] Open
Abstract
The two most common types of pancreatic neuroendocrine tumors (PanNETs) are insulinomas and nonfunctioning PanNETs (NF-PanNETs). Insulinomas are small, rarely metastatic tumors that secrete high amounts of insulin, and nonfunctioning PanNETs are larger tumors that are frequently metastatic but that do not secrete hormones. Insulinomas are modeled by the highly studied RIP1-Tag2 (RT2) transgenic mice when bred into a C57Bl/6 (B6) genetic background (also known as RT2 B6 mice). But there has been a need for an animal model of nonfunctioning PanNETs, which in the clinic are a more common and severe disease. Here we show that when bred into a hybrid AB6F1 genetic background, RT2 mice make nonfunctioning PanNETs. Compared to insulinomas produced by RT2 B6 mice, the tumors produced by RT2 AB6F1 mice were larger and more metastatic, and the animals did not suffer from hypoglycemia or hyperinsulinemia. Genetic crosses revealed that a locus in mouse chromosome 2qG1 was linked to liver metastasis and to lack of insulin production. This locus was tightly linked to the gene encoding Insm1, a beta cell transcription factor that was highly expressed in human insulinomas but unexpressed in other types of PanNETs due to promoter hypermethylation. Insm1-deficient human cell lines expressed stem cell markers, were more invasive in vitro, and metastasized at higher rates in vivo when compared to isogenic Insm1-expressing cell lines. These data demonstrate that expression of Insm1 can determine whether a PanNET is a localized insulinoma or a metastatic nonfunctioning tumor.
Collapse
|
18
|
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
|
19
|
Gahete MD, Jimenez-Vacas JM, Alors-Perez E, Herrero-Aguayo V, Fuentes-Fayos AC, Pedraza-Arevalo S, Castaño JP, Luque RM. Mouse models in endocrine tumors. J Endocrinol 2018; 240:JOE-18-0571.R1. [PMID: 30475226 DOI: 10.1530/joe-18-0571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022]
Abstract
Endocrine and neuroendocrine tumors comprise a highly heterogeneous group of neoplasms that can arise from (neuro)endocrine cells, either from endocrine glands or from the widespread diffuse neuroendocrine system, and, consequently, are widely distributed throughout the body. Due to their diversity, heterogeneity and limited incidence, studying in detail the molecular and genetic alterations that underlie their development and progression is still a highly elusive task. This, in turn, hinders the discovery of novel therapeutic options for these tumors. To circumvent these limitations, numerous mouse models of endocrine and neuroendocrine tumors have been developed, characterized and used in pre-clinical, co-clinical (implemented in mouse models and patients simultaneously) and post-clinical studies, for they represent powerful and necessary tools in basic and translational tumor biology research. Indeed, different in vivo mouse models, including cell line-based xenografts (CDXs), patient-derived xenografts (PDXs) and genetically engineered mouse models (GEMs), have been used to delineate the development, progression and behavior of human tumors. Results gained with these in vivo models have facilitated the clinical application in patients of diverse breakthrough discoveries made in this field. Herein, we review the generation, characterization and translatability of the most prominent mouse models of endocrine and neuroendocrine tumors reported to date, as well as the most relevant clinical implications obtained for each endocrine and neuroendocrine tumor type.
Collapse
Affiliation(s)
- Manuel D Gahete
- M Gahete, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, 14011, Spain
| | - Juan M Jimenez-Vacas
- J Jimenez-Vacas, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Emilia Alors-Perez
- E Alors-Perez, Department of Cell Biology, Physiology and Inmunology, Maimonides Institute for Biomedical Research of Cordoba (IMIBIC) / University of Cordoba, Cordoba, Spain
| | - Vicente Herrero-Aguayo
- V Herrero-Aguayo, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- A Fuentes-Fayos, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Sergio Pedraza-Arevalo
- S Pedraza-Arevalo, Department of Cell Biology, Physiology and Immunology, University of Córdoba, Cordoba, Spain
| | - Justo P Castaño
- J Castaño, Dpt. of Cell Biology-University of Córdoba, IMIBIC-Maimonides Biomedical Research Institute of Cordoba, Cordoba, E-14004, Spain
| | - Raul M Luque
- R Luque, Dept of Cell Biology, Phisiology and Inmunology, Section of Cell Biology, University of Cordoba, Cordoba, Spain, Cordoba, 14014, Spain
| |
Collapse
|
20
|
Nowak-Sliwinska P, Alitalo K, Allen E, Anisimov A, Aplin AC, Auerbach R, Augustin HG, Bates DO, van Beijnum JR, Bender RHF, Bergers G, Bikfalvi A, Bischoff J, Böck BC, Brooks PC, Bussolino F, Cakir B, Carmeliet P, Castranova D, Cimpean AM, Cleaver O, Coukos G, Davis GE, De Palma M, Dimberg A, Dings RPM, Djonov V, Dudley AC, Dufton NP, Fendt SM, Ferrara N, Fruttiger M, Fukumura D, Ghesquière B, Gong Y, Griffin RJ, Harris AL, Hughes CCW, Hultgren NW, Iruela-Arispe ML, Irving M, Jain RK, Kalluri R, Kalucka J, Kerbel RS, Kitajewski J, Klaassen I, Kleinmann HK, Koolwijk P, Kuczynski E, Kwak BR, Marien K, Melero-Martin JM, Munn LL, Nicosia RF, Noel A, Nurro J, Olsson AK, Petrova TV, Pietras K, Pili R, Pollard JW, Post MJ, Quax PHA, Rabinovich GA, Raica M, Randi AM, Ribatti D, Ruegg C, Schlingemann RO, Schulte-Merker S, Smith LEH, Song JW, Stacker SA, Stalin J, Stratman AN, Van de Velde M, van Hinsbergh VWM, Vermeulen PB, Waltenberger J, Weinstein BM, Xin H, Yetkin-Arik B, Yla-Herttuala S, Yoder MC, Griffioen AW. Consensus guidelines for the use and interpretation of angiogenesis assays. Angiogenesis 2018; 21:425-532. [PMID: 29766399 PMCID: PMC6237663 DOI: 10.1007/s10456-018-9613-x] [Citation(s) in RCA: 455] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.
Collapse
Affiliation(s)
- Patrycja Nowak-Sliwinska
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, Faculty of Sciences, University of Geneva, University of Lausanne, Rue Michel-Servet 1, CMU, 1211, Geneva 4, Switzerland.
- Translational Research Center in Oncohaematology, University of Geneva, Geneva, Switzerland.
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Elizabeth Allen
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Louvain, Belgium
| | - Andrey Anisimov
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Alfred C Aplin
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - Hellmut G Augustin
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - David O Bates
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - R Hugh F Bender
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance, Department of Oncology, VIB-Center for Cancer Biology, KU Leuven, Louvain, Belgium
- Department of Neurological Surgery, Brain Tumor Research Center, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Andreas Bikfalvi
- Angiogenesis and Tumor Microenvironment Laboratory (INSERM U1029), University Bordeaux, Pessac, France
| | - Joyce Bischoff
- Vascular Biology Program and Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Barbara C Böck
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - Peter C Brooks
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Federico Bussolino
- Department of Oncology, University of Torino, Turin, Italy
- Candiolo Cancer Institute-FPO-IRCCS, 10060, Candiolo, Italy
| | - Bertan Cakir
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Anca M Cimpean
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Ondine Cleaver
- Department of Molecular Biology, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - George Coukos
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - George E Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, School of Medicine and Dalton Cardiovascular Center, Columbia, MO, USA
| | - Michele De Palma
- School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Andrew C Dudley
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Emily Couric Cancer Center, The University of Virginia, Charlottesville, VA, USA
| | - Neil P Dufton
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute, Leuven, Belgium
| | | | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London, UK
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bart Ghesquière
- Metabolomics Expertise Center, VIB Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, Metabolomics Expertise Center, KU Leuven, Leuven, Belgium
| | - Yan Gong
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Adrian L Harris
- Molecular Oncology Laboratories, Oxford University Department of Oncology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Nan W Hultgren
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | | | - Melita Irving
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joanna Kalucka
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Robert S Kerbel
- Department of Medical Biophysics, Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Jan Kitajewski
- Department of Physiology and Biophysics, University of Illinois, Chicago, IL, USA
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hynda K Kleinmann
- The George Washington University School of Medicine, Washington, DC, USA
| | - Pieter Koolwijk
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Elisabeth Kuczynski
- Department of Medical Biophysics, Biological Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | | | - Juan M Melero-Martin
- Department of Cardiac Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Roberto F Nicosia
- Department of Pathology, University of Washington, Seattle, WA, USA
- Pathology and Laboratory Medicine Service, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Agnes Noel
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Jussi Nurro
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Tatiana V Petrova
- Department of oncology UNIL-CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund, Sweden
| | - Roberto Pili
- Genitourinary Program, Indiana University-Simon Cancer Center, Indianapolis, IN, USA
| | - Jeffrey W Pollard
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Mark J Post
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Paul H A Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Department Surgery, LUMC, Leiden, The Netherlands
| | - Gabriel A Rabinovich
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine, National Council of Scientific and Technical Investigations (CONICET), Buenos Aires, Argentina
| | - Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Anna M Randi
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
- National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Curzio Ruegg
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Stefan Schulte-Merker
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU, Münster, Germany
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Steven A Stacker
- Tumour Angiogenesis and Microenvironment Program, Peter MacCallum Cancer Centre and The Sir Peter MacCallum, Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Jimmy Stalin
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU, Münster, Germany
| | - Amber N Stratman
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Maureen Van de Velde
- Laboratory of Tumor and Developmental Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Victor W M van Hinsbergh
- Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Peter B Vermeulen
- HistoGeneX, Antwerp, Belgium
- Translational Cancer Research Unit, GZA Hospitals, Sint-Augustinus & University of Antwerp, Antwerp, Belgium
| | - Johannes Waltenberger
- Medical Faculty, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Hong Xin
- University of California, San Diego, La Jolla, CA, USA
| | - Bahar Yetkin-Arik
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Seppo Yla-Herttuala
- Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
21
|
Downregulation of GEP100 Improved the Growth Inhibition Effect of Erlotinib Through Modulating Mesenchymal Epithelial Transition Process in Pancreatic Cancer. Pancreas 2018; 47:732-737. [PMID: 29851753 DOI: 10.1097/mpa.0000000000001076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVE The epidermal growth factor receptor is overexpressed in the majority of pancreatic cancer. Epidermal growth factor receptor tyrosine kinase inhibitor erlotinib was approved to treat patients combining with gemcitabine. However, the sensitivity is low. Here, we try to reveal the regulatory role of guanine nucleotide exchange protein 100 (GEP100) in erlotinib sensitivity. METHODS We investigated the correlation between GEP100 expression and sensitivity to erlotinib in different pancreatic cancer cell lines, followed by examination of the effect of GEP100 on erlotinib sensitivity by establishing the stable knocked-down cell line. The expression level of epithelial mesenchymal transition-related protein was examined by Western blot, and the regulatory mechanism was investigated by short hairpin RNA. Xenograft experiment was also performed in nude mice. RESULTS We identified a significant correlation between sensitivity to erlotinib and expression of GEP100. GEP100 downregulation increased its sensitivity to erlotinib. E-cadherin short hairpin RNA treatment inhibited this sensitivity. Immunohistochemical staining showed a mutual exclusive expression pattern of GEP100 and E-cadherin in human pancreatic cancer tissues. Xenograft showed that downregulation of GEP100 enhanced the growth inhibition of erlotinib in nude mice. CONCLUSIONS Our results suggested that GEP100 and E-cadherin have the predictive value for responsiveness to erlotinib in pancreatic cancer.
Collapse
|
22
|
Lamberti G, Brighi N, Maggio I, Manuzzi L, Peterle C, Ambrosini V, Ricci C, Casadei R, Campana D. The Role of mTOR in Neuroendocrine Tumors: Future Cornerstone of a Winning Strategy? Int J Mol Sci 2018; 19:ijms19030747. [PMID: 29509701 PMCID: PMC5877608 DOI: 10.3390/ijms19030747] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 02/28/2018] [Accepted: 03/05/2018] [Indexed: 12/13/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) is part of the phosphoinositide-3-kinase (PI3K)/protein kinase B (AkT)/mTOR pathway and owes its name to the inhibitory effect of rapamycin. The mTOR has a central converging role for many cell functions, serving as a sensor for extracellular signals from energy status and nutrients availability, growth factors, oxygen and stress. Thus, it also modulates switch to anabolic processes (protein and lipid synthesis) and autophagy, in order to regulate cell growth and proliferation. Given its functions in the cell, its deregulation is implicated in many human diseases, including cancer. Its predominant role in tumorigenesis and progression of neuroendocrine tumors (NETs), in particular, has been demonstrated in preclinical studies and late clinical trials. mTOR inhibition by everolimus is an established therapeutic target in NETs, but there are no identified predictive or prognostic factors. This review is focused on the role of mTOR and everolimus in NETs, from preclinical studies to major clinical trials, and future perspectives involving mTOR in the treatment of NETs.
Collapse
Affiliation(s)
- Giuseppe Lamberti
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi University Hospital, 40138 Bologna, Italy.
| | - Nicole Brighi
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi University Hospital, 40138 Bologna, Italy.
| | - Ilaria Maggio
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi University Hospital, 40138 Bologna, Italy.
| | - Lisa Manuzzi
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi University Hospital, 40138 Bologna, Italy.
| | - Chiara Peterle
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi University Hospital, 40138 Bologna, Italy.
| | - Valentina Ambrosini
- Nuclear Medicine Unit, Medicina Nucleare Metropolitana, S.Orsola-Malpighi University Hospital, 40138 Bologna, Italy.
| | - Claudio Ricci
- Department of Medical and Surgical Sciences, S.Orsola-Malpighi University Hospital, 40138 Bologna, Italy.
| | - Riccardo Casadei
- Department of Medical and Surgical Sciences, S.Orsola-Malpighi University Hospital, 40138 Bologna, Italy.
| | - Davide Campana
- Department of Medical and Surgical Sciences, S.Orsola-Malpighi University Hospital, 40138 Bologna, Italy.
| |
Collapse
|
23
|
Wolff RA, Wang-Gillam A, Alvarez H, Tiriac H, Engle D, Hou S, Groff AF, San Lucas A, Bernard V, Allenson K, Castillo J, Kim D, Mulu F, Huang J, Stephens B, Wistuba II, Katz M, Varadhachary G, Park Y, Hicks J, Chinnaiyan A, Scampavia L, Spicer T, Gerhardinger C, Maitra A, Tuveson D, Rinn J, Lizee G, Yee C, Levine AJ. Dynamic changes during the treatment of pancreatic cancer. Oncotarget 2018; 9:14764-14790. [PMID: 29599906 PMCID: PMC5871077 DOI: 10.18632/oncotarget.24483] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/01/2018] [Indexed: 01/17/2023] Open
Abstract
This manuscript follows a single patient with pancreatic adenocarcinoma for a five year period, detailing the clinical record, pathology, the dynamic evolution of molecular and cellular alterations as well as the responses to treatments with chemotherapies, targeted therapies and immunotherapies. DNA and RNA samples from biopsies and blood identified a dynamic set of changes in allelic imbalances and copy number variations in response to therapies. Organoid cultures established from biopsies over time were employed for extensive drug testing to determine if this approach was feasible for treatments. When an unusual drug response was detected, an extensive RNA sequencing analysis was employed to establish novel mechanisms of action of this drug. Organoid cell cultures were employed to identify possible antigens associated with the tumor and the patient's T-cells were expanded against one of these antigens. Similar and identical T-cell receptor sequences were observed in the initial biopsy and the expanded T-cell population. Immunotherapy treatment failed to shrink the tumor, which had undergone an epithelial to mesenchymal transition prior to therapy. A warm autopsy of the metastatic lung tumor permitted an extensive analysis of tumor heterogeneity over five years of treatment and surgery. This detailed analysis of the clinical descriptions, imaging, pathology, molecular and cellular evolution of the tumors, treatments, and responses to chemotherapy, targeted therapies, and immunotherapies, as well as attempts at the development of personalized medical treatments for a single patient should provide a valuable guide to future directions in cancer treatment.
Collapse
Affiliation(s)
- Robert A Wolff
- Department of Gastrointestinal (GI) Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Hector Alvarez
- Department of Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Hervé Tiriac
- Cold Spring Harbor Laboratory, New York, NY, USA
| | | | - Shurong Hou
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, FL, USA
| | - Abigail F Groff
- Department of Molecular and Cellular Biology, Harvard University, The Broad Institute, Cambridge, MA, USA
| | - Anthony San Lucas
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Vincent Bernard
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Kelvin Allenson
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Jonathan Castillo
- Department of Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Dong Kim
- Department of Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Feven Mulu
- Department of Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Jonathan Huang
- Department of Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Bret Stephens
- Department of Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew Katz
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Gauri Varadhachary
- Department of Gastrointestinal (GI) Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | | | - James Hicks
- Cold Spring Harbor Laboratory, New York, NY, USA
| | - Arul Chinnaiyan
- Center for Translational Pathology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Louis Scampavia
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, FL, USA
| | - Timothy Spicer
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, FL, USA
| | - Chiara Gerhardinger
- Department of Molecular and Cellular Biology, Harvard University, The Broad Institute, Cambridge, MA, USA
| | - Anirban Maitra
- Department of Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | | | - John Rinn
- Department of Molecular and Cellular Biology, Harvard University, The Broad Institute, Cambridge, MA, USA.,Current address: University of Colorado Boulder, BioFrontiers Institute, Boulder, CO, USA
| | - Gregory Lizee
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Cassian Yee
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Arnold J Levine
- Simons Center for Systems Biology, Institute for Advanced Study, Princeton, NJ, USA
| |
Collapse
|
24
|
Aristizabal Prada ET, Auernhammer CJ. Targeted therapy of gastroenteropancreatic neuroendocrine tumours: preclinical strategies and future targets. Endocr Connect 2018; 7:R1-R25. [PMID: 29146887 PMCID: PMC5754510 DOI: 10.1530/ec-17-0286] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 11/16/2017] [Indexed: 12/12/2022]
Abstract
Molecular targeted therapy of advanced neuroendocrine tumours (NETs) of the gastroenteropancreatic (GEP) system currently encompasses approved therapy with the mammalian target of rapamycin (mTOR) inhibitor everolimus and the multi-tyrosinkinase inhibitor sunitinib. However, clinical efficacy of these treatment strategies is limited by low objective response rates and limited progression-free survival due to tumour resistance. Further novel strategies for molecular targeted therapy of NETs of the GEP system are needed. This paper reviews preclinical research models and signalling pathways in NETs of the GEP system. Preclinical and early clinical data on putative novel targets for molecular targeted therapy of NETs of the GEP system are discussed, including PI3K, Akt, mTORC1/mTORC2, GSK3, c-Met, Ras-Raf-MEK-ERK, embryogenic pathways (Hedgehog, Notch, Wnt/beta-catenin, TGF-beta signalling and SMAD proteins), tumour suppressors and cell cycle regulators (p53, cyclin-dependent kinases (CDKs) CDK4/6, CDK inhibitor p27, retinoblastoma protein (Rb)), heat shock protein HSP90, Aurora kinase, Src kinase family, focal adhesion kinase and epigenetic modulation by histone deacetylase inhibitors.
Collapse
Affiliation(s)
- E T Aristizabal Prada
- Department of Internal Medicine IVCampus Grosshadern, University-Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - C J Auernhammer
- Department of Internal Medicine IVCampus Grosshadern, University-Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
25
|
Contractor T, Kobayashi S, da Silva E, Clausen R, Chan C, Vosburgh E, Tang LH, Levine AJ, Harris CR. Sexual dimorphism of liver metastasis by murine pancreatic neuroendocrine tumors is affected by expression of complement C5. Oncotarget 2017; 7:30585-96. [PMID: 27105526 PMCID: PMC5058703 DOI: 10.18632/oncotarget.8874] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 03/31/2016] [Indexed: 01/04/2023] Open
Abstract
In a mouse model for neuroendocrine tumors of the pancreas (PanNETs), liver metastasis occurred at a higher frequency in males. Male mice also had higher serum and intratumoral levels of the innate immunity protein complement C5. In mice that lost the ability to express complement C5, there was a lower frequency of metastasis, and males no longer had a higher frequency of metastasis than females. Treatment with PMX53, a small molecule antagonist of C5aR1/CD88, the receptor for complement C5a, also reduced metastasis. Mice lacking a functional gene for complement C5 had smaller primary tumors, which were less invasive and lacked the CD68+ macrophages that have previously been associated with metastasis in this type of tumor. This is the first report of a gene that causes sexual dimorphism of metastasis in a mouse model. In the human disease, which also shows sexual dimorphism for metastasis, clinically advanced tumors expressed more complement C5 than less advanced tumors.
Collapse
Affiliation(s)
| | | | - Edaise da Silva
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Richard Clausen
- Raymond and Beverly Sackler Foundation Laboratory, New Brunswick, NJ, USA
| | - Chang Chan
- Rutgers University Cancer Institute of New Jersey and Department of Pediatrics, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Evan Vosburgh
- Raymond and Beverly Sackler Foundation Laboratory, New Brunswick, NJ, USA
| | - Laura H Tang
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Arnold J Levine
- Institute for Advanced Study, Princeton, NJ, USA.,Rutgers University Cancer Institute of New Jersey and Department of Pediatrics, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Chris R Harris
- Raymond and Beverly Sackler Foundation Laboratory, New Brunswick, NJ, USA.,Rutgers University Cancer Institute of New Jersey and Department of Pediatrics, Robert Wood Johnson Medical School, New Brunswick, NJ, USA.,Department of Pediatrics, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
26
|
Prognostic and predictive role of the PI3K-AKT-mTOR pathway in neuroendocrine neoplasms. Clin Transl Oncol 2017; 20:561-569. [PMID: 29124519 DOI: 10.1007/s12094-017-1758-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 09/30/2017] [Indexed: 12/20/2022]
Abstract
Neuroendocrine neoplasms (NENs) are considered a heterogeneous and rare entity. Its natural history is influenced by multiple clinicopathological characteristics, which guide the management of these patients. The development of molecular biology reveals that the PI3K-AKT-mTOR pathway plays a relevant role in tumorigenesis and progression of NENs. Mammalian target of rapamycin (mTOR) inhibitors, targeted agents that block this pathway, has improved outcomes in neuroendocrine tumors (NETs). Different therapeutic approaches, such as somatostatin analogs, chemotherapy, peptide receptor radionuclide therapy, and targeted agents, have shown benefits in the treatment of NETs. However, there are not any established prognostic or predictive biomarkers to select the best therapy option to individualize treatment. Although a relation between alterations in the PI3K-AKT-mTOR pathway and clinical outcomes has not been found, these anomalies are considered attractive biomarkers. Additional molecular analysis should be integrated in future clinical trials' design to identify potential predictive or prognostic biomarkers.
Collapse
|
27
|
Orr-Asman MA, Chu Z, Jiang M, Worley M, LaSance K, Koch SE, Carreira VS, Dahche HM, Plas DR, Komurov K, Qi X, Mercer CA, Anthony LB, Rubinstein J, Thomas HE. mTOR Kinase Inhibition Effectively Decreases Progression of a Subset of Neuroendocrine Tumors that Progress on Rapalog Therapy and Delays Cardiac Impairment. Mol Cancer Ther 2017; 16:2432-2441. [PMID: 28864682 DOI: 10.1158/1535-7163.mct-17-0058] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 06/01/2017] [Accepted: 08/22/2017] [Indexed: 11/16/2022]
Abstract
Inhibition of mTOR signaling using the rapalog everolimus is an FDA-approved targeted therapy for patients with lung and gastroenteropancreatic neuroendocrine tumors (NET). However, patients eventually progress on treatment, highlighting the need for additional therapies. We focused on pancreatic NETs (pNET) and reasoned that treatment of these tumors upon progression on rapalog therapy, with an mTOR kinase inhibitor (mTORKi), such as CC-223, could overcome a number of resistance mechanisms in tumors and delay cardiac carcinoid disease. We performed preclinical studies using human pNET cells in vitro and injected them subcutaneously or orthotopically to determine tumor progression and cardiac function in mice treated with either rapamycin alone or switched to CC-223 upon progression. Detailed signaling and RNA sequencing analyses were performed on tumors that were sensitive or progressed on mTOR treatment. Approximately 57% of mice bearing pNET tumors that progressed on rapalog therapy showed a significant decrease in tumor volume upon a switch to CC-223. Moreover, mice treated with an mTORKi exhibited decreased cardiac dilation and thickening of heart valves than those treated with placebo or rapamycin alone. In conclusion, in the majority of pNETs that progress on rapalogs, it is possible to reduce disease progression using an mTORKi, such as CC-223. Moreover, CC-223 had an additional transient cardiac benefit on valvular fibrosis compared with placebo- or rapalog-treated mice. These results provide the preclinical rationale to further develop mTORKi clinically upon progression on rapalog therapy and to further test their long-term cardioprotective benefit in those NET patients prone to carcinoid syndrome. Mol Cancer Ther; 16(11); 2432-41. ©2017 AACR.
Collapse
Affiliation(s)
- Melissa A Orr-Asman
- Division of Hematology and Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Zhengtao Chu
- Division of Hematology and Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Min Jiang
- Division of Cardiovascular Diseases, University of Cincinnati, Cincinnati, Ohio
| | - Mariah Worley
- Division of Cardiovascular Diseases, University of Cincinnati, Cincinnati, Ohio
| | - Kathleen LaSance
- Department of Radiology, University of Cincinnati, Cincinnati, Ohio
| | - Sheryl E Koch
- Division of Cardiovascular Diseases, University of Cincinnati, Cincinnati, Ohio
| | - Vinicius S Carreira
- Department of Environmental Health and Center for Environmental Genetics, University of Cincinnati, Cincinnati, Ohio
| | - Hanan M Dahche
- Division of Hematology and Oncology, University of Cincinnati, Cincinnati, Ohio
| | - David R Plas
- Department of Cancer Biology, University of Cincinnati, Cincinnati, Ohio
| | - Kakajan Komurov
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Ohio
| | - Xiaoyang Qi
- Division of Hematology and Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Carol A Mercer
- Division of Hematology and Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Lowell B Anthony
- Department of Medicine, University of Kentucky, Lexington, Kentucky
| | - Jack Rubinstein
- Division of Cardiovascular Diseases, University of Cincinnati, Cincinnati, Ohio
| | - Hala E Thomas
- Division of Hematology and Oncology, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
28
|
Pavel ME, Sers C. WOMEN IN CANCER THEMATIC REVIEW: Systemic therapies in neuroendocrine tumors and novel approaches toward personalized medicine. Endocr Relat Cancer 2016; 23:T135-T154. [PMID: 27649723 DOI: 10.1530/erc-16-0370] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 09/20/2016] [Indexed: 12/19/2022]
Abstract
Neuroendocrine tumors (NETs) are a group of heterogenous neoplasms. Evidence-based treatment options for antiproliferative therapy include somatostatin analogues, the mTOR inhibitor everolimus, the multiple tyrosine kinase inhibitor sunitinib and peptide receptor radionuclide therapy with 177-Lu-octreotate. In the absence of definite predictive markers, therapeutic decision making follows clinical and pathological criteria. As objective response rates with targeted drugs are rather low, and response duration is limited in most patients, numerous combination therapies targeting multiple pathways have been explored in the field. Upfront combination of drugs, however, is associated with increasing toxicity and has shown little benefit. Major advancements in the molecular understanding of NET based on genomic, epigenomic and transcriptomic analysis have been achieved with prognostic and therapeutic impact. New insight into molecular alterations has paved the way to biomarker-driven clinical trials and may facilitate treatment stratification toward personalized medicine in the near future. However, an improved understanding of the complexity of pathway interactions is required for successful treatment. A systems biology approach is one of the tools that may help to achieve this endeavor.
Collapse
Affiliation(s)
- Marianne E Pavel
- Medical DepartmentDivision of Hepatology and Gastroenterology including Metabolic Diseases, Campus Virchow Klinikum, Charité University Medicine, Berlin, Germany
| | - Christine Sers
- Institute of PathologyCharité University Medicine, Berlin, Germany
| |
Collapse
|
29
|
Soler A, Figueiredo AM, Castel P, Martin L, Monelli E, Angulo-Urarte A, Milà-Guasch M, Viñals F, Baselga J, Casanovas O, Graupera M. Therapeutic Benefit of Selective Inhibition of p110α PI3-Kinase in Pancreatic Neuroendocrine Tumors. Clin Cancer Res 2016; 22:5805-5817. [PMID: 27225693 DOI: 10.1158/1078-0432.ccr-15-3051] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 04/15/2016] [Accepted: 05/16/2016] [Indexed: 01/12/2023]
Abstract
PURPOSE Mutations in the PI3K pathway occur in 16% of patients with pancreatic neuroendocrine tumors (PanNETs), which suggests that these tumors are an exciting setting for PI3K/AKT/mTOR pharmacologic intervention. Everolimus, an mTOR inhibitor, is being used to treat patients with advanced PanNETs. However, resistance to mTOR-targeted therapy is emerging partially due to the loss of mTOR-dependent feedback inhibition of AKT. In contrast, the response to PI3K inhibitors in PanNETs is unknown. EXPERIMENTAL DESIGN In the current study, we assessed the frequency of PI3K pathway activation in human PanNETs and in RIP1-Tag2 mice, a preclinical tumor model of PanNETs, and we investigated the therapeutic efficacy of inhibiting PI3K in RIP1-Tag2 mice using a combination of pan (GDC-0941) and p110α-selective (GDC-0326) inhibitors and isoform-specific PI3K kinase-dead-mutant mice. RESULTS Human and mouse PanNETs showed enhanced pAKT, pPRAS40, and pS6 positivity compared with normal tissue. Although treatment of RIP1-Tag2 mice with GDC-0941 led to reduced tumor growth with no impact on tumor vessels, the selective inactivation of the p110α PI3K isoform, either genetically or pharmacologically, reduced tumor growth as well as vascular area. Furthermore, GDC-0326 reduced the incidence of liver and lymph node metastasis compared with vehicle-treated mice. We also demonstrated that tumor and stromal cells are implicated in the antitumor activity of GDC-0326 in RIP1-Tag2 tumors. CONCLUSIONS Our data provide a rationale for p110α-selective intervention in PanNETs and unravel a new function of this kinase in cancer biology through its role in promoting metastasis. Clin Cancer Res; 22(23); 5805-17. ©2016 AACR.
Collapse
Affiliation(s)
- Adriana Soler
- Vascular Signaling Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ana M Figueiredo
- Vascular Signaling Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Pau Castel
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laura Martin
- Translational Research Laboratory, Catalan Institute of Oncology, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Erika Monelli
- Vascular Signaling Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ana Angulo-Urarte
- Vascular Signaling Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Maria Milà-Guasch
- Vascular Signaling Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Francesc Viñals
- Translational Research Laboratory, Catalan Institute of Oncology, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.,Departament de Ciències Fisiològiques II, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jose Baselga
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Oriol Casanovas
- Translational Research Laboratory, Catalan Institute of Oncology, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Mariona Graupera
- Vascular Signaling Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
30
|
Allen E, Miéville P, Warren CM, Saghafinia S, Li L, Peng MW, Hanahan D. Metabolic Symbiosis Enables Adaptive Resistance to Anti-angiogenic Therapy that Is Dependent on mTOR Signaling. Cell Rep 2016; 15:1144-60. [PMID: 27134166 PMCID: PMC4872464 DOI: 10.1016/j.celrep.2016.04.029] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 01/28/2016] [Accepted: 04/07/2016] [Indexed: 01/01/2023] Open
Abstract
Therapeutic targeting of tumor angiogenesis with VEGF inhibitors results in demonstrable, but transitory efficacy in certain human tumors and mouse models of cancer, limited by unconventional forms of adaptive/evasive resistance. In one such mouse model, potent angiogenesis inhibitors elicit compartmental reorganization of cancer cells around remaining blood vessels. The glucose and lactate transporters GLUT1 and MCT4 are induced in distal hypoxic cells in a HIF1α-dependent fashion, indicative of glycolysis. Tumor cells proximal to blood vessels instead express the lactate transporter MCT1, and p-S6, the latter reflecting mTOR signaling. Normoxic cancer cells import and metabolize lactate, resulting in upregulation of mTOR signaling via glutamine metabolism enhanced by lactate catabolism. Thus, metabolic symbiosis is established in the face of angiogenesis inhibition, whereby hypoxic cancer cells import glucose and export lactate, while normoxic cells import and catabolize lactate. mTOR signaling inhibition disrupts this metabolic symbiosis, associated with upregulation of the glucose transporter GLUT2. Angiogenesis inhibitors causing acute hypoxia elicit metabolic compartmentalization Hypoxic cancer cells import and metabolize glucose, secreting lactate Normoxic vessel-proximal cancer cells import and metabolize lactate, involving mTOR Co-inhibiting mTOR disrupts the metabolic symbiosis
Collapse
Affiliation(s)
- Elizabeth Allen
- The Swiss Institute for Experimental Cancer Research (ISREC), EPFL SV ISREC, Station 19, 1015 Lausanne, Switzerland
| | - Pascal Miéville
- The Institute of Chemical Sciences and Engineering (ISIC-SB-EPFL), Ecole Polytechnique Fédérale de Lausanne, EPFL SB ISIC-Direction, CH A3 398 Station 6, 1015 Lausanne, Switzerland
| | - Carmen M Warren
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA 90095, USA
| | - Sadegh Saghafinia
- The Swiss Institute for Experimental Cancer Research (ISREC), EPFL SV ISREC, Station 19, 1015 Lausanne, Switzerland
| | - Leanne Li
- The Swiss Institute for Experimental Cancer Research (ISREC), EPFL SV ISREC, Station 19, 1015 Lausanne, Switzerland
| | - Mei-Wen Peng
- The Swiss Institute for Experimental Cancer Research (ISREC), EPFL SV ISREC, Station 19, 1015 Lausanne, Switzerland
| | - Douglas Hanahan
- The Swiss Institute for Experimental Cancer Research (ISREC), EPFL SV ISREC, Station 19, 1015 Lausanne, Switzerland; The Swiss Cancer Center Lausanne (SCCL), Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
31
|
Faggiano A, Malandrino P, Modica R, Agrimi D, Aversano M, Bassi V, Giordano EA, Guarnotta V, Logoluso FA, Messina E, Nicastro V, Nuzzo V, Sciaraffia M, Colao A. Efficacy and Safety of Everolimus in Extrapancreatic Neuroendocrine Tumor: A Comprehensive Review of Literature. Oncologist 2016; 21:875-86. [PMID: 27053503 DOI: 10.1634/theoncologist.2015-0420] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 02/08/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Everolimus, an oral mTOR (mammalian target of rapamycin) inhibitor, is currently approved for the treatment of progressive pancreatic neuroendocrine tumors (NETs). Although promising, only scattered data, often from nondedicated studies, are available for extrapancreatic NETs. PATIENTS AND METHODS A systematic review of the published data was performed concerning the use of everolimus in extrapancreatic NET, with the aim of summarizing the current knowledge on its efficacy and tolerability. Moreover, the usefulness of everolimus was evaluated according to the different sites of the primary. RESULTS The present study included 22 different publications, including 874 patients and 456 extrapancreatic NETs treated with everolimus. Nine different primary sites of extrapancreatic NETs were found. The median progression-free survival ranged from 12.0 to 29.9 months. The median time to progression was not reached in a phase II prospective study, and the interval to progression ranged from 12 to 36 months in 5 clinical cases. Objective responses were observed in 7 prospective studies, 2 retrospective studies, and 2 case reports. Stabilization of the disease was obtained in a high rate of patients, ranging from 67.4% to 100%. The toxicity of everolimus in extrapancreatic NETs is consistent with the known safety profile of the drug. Most adverse events were either grade 1 or 2 and easy manageable with a dose reduction or temporary interruption and only rarely requiring discontinuation. CONCLUSION Treatment with everolimus in patients with extrapancreatic NETs appears to be a promising strategy that is safe and well tolerated. The use of this emerging opportunity needs to be validated with clinical trials specifically designed on this topic. IMPLICATIONS FOR PRACTICE The present study reviewed all the available published data concerning the use of everolimus in 456 extrapancreatic neuroendocrine tumors (NETs) and summarized the current knowledge on the efficacy and safety of this drug, not yet approved except for pancreatic NETs. The progression-free survival rates and some objective responses seem promising and support the extension of the use of this drug. The site-by-site analysis seems to suggest that some subtypes of NETs, such as colorectal, could be more sensitive to everolimus than other primary NETs. No severe adverse events were usually reported and discontinuation was rarely required; thus, everolimus should be considered a valid therapeutic option for extrapancreatic NETs.
Collapse
Affiliation(s)
- Antongiulio Faggiano
- Thyroid and Parathyroid Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | | | - Roberta Modica
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Daniela Agrimi
- District Hospital, Azienda Sanitaria Locale, Brindisi, Italy
| | - Maurizio Aversano
- Endocrinology Unit, Azienda Sanitaria Locale Napoli 3, Naples, Italy
| | - Vincenzo Bassi
- Unit of Internal Medicine, San Giovanni Bosco Hospital, Naples, Italy
| | - Ernesto A Giordano
- Endocrinology Unit, Azienda Sanitaria Provinciale di Calabria, Reggio Calabria, Italy
| | - Valentina Guarnotta
- Biomedical Department of Internal and Specialist Medicine, Section of Endocrinology, University of Palermo, Palermo, Italy
| | - Francesco A Logoluso
- Endocrinology Unit, Azienda Ospedaliero-Universitaria Consorziale Policlinico, Bari, Italy
| | - Erika Messina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | | | - Vincenzo Nuzzo
- Unit of Internal Medicine, San Gennaro Hospital, Naples, Italy
| | | | - Annamaria Colao
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| |
Collapse
|
32
|
Zatelli MC, Fanciulli G, Malandrino P, Ramundo V, Faggiano A, Colao A. Predictive factors of response to mTOR inhibitors in neuroendocrine tumours. Endocr Relat Cancer 2016; 23:R173-83. [PMID: 26666705 DOI: 10.1530/erc-15-0413] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 12/14/2015] [Indexed: 12/13/2022]
Abstract
Medical treatment of neuroendocrine tumours (NETs) has drawn a lot of attention due to the recent demonstration of efficacy of several drugs on progression-free survival, including somatostatin analogs, small tyrosine kinase inhibitors and mTOR inhibitors (or rapalogs). The latter are approved as therapeutic agents in advanced pancreatic NETs and have been demonstrated to be effective in different types of NETs, with variable efficacy due to the development of resistance to treatment. Early detection of patients that may benefit from rapalogs treatment is of paramount importance in order to select the better treatment and avoid ineffective and expensive treatments. Predictive markers for therapeutic response are under intensive investigation, aiming at a tailored patient management and more appropriate resource utilization. This review summarizes the available data on the tissue, circulating and imaging markers that are potentially predictive of rapalog efficacy in NETs.
Collapse
Affiliation(s)
- Maria Chiara Zatelli
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Giuseppe Fanciulli
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Pasqualino Malandrino
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Valeria Ramundo
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Antongiulio Faggiano
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | - Annamaria Colao
- Section of Endocrinology and Internal MedicineDepartment of Medical Sciences, University of Ferrara, Via Aldo Moro 8, 44124 Cona - Ferrara, ItalyNeuroendocrine Tumours UnitDepartment of Clinical and Experimental Medicine, University of Sassari - AOU Sassari, Sassari, ItalyEndocrinology UnitGaribaldi Nesima Medical Center, Catania, ItalyDepartment of Clinical Medicine and Surgery"Federico II" University of Naples, Naples, ItalyThyroid and Parathyroid Surgery UnitIstituto Nazionale per lo studio e la cura dei tumori "Fondazione G. Pascale" - IRCCS, Naples, Italy
| | | |
Collapse
|
33
|
Day CP, Merlino G, Van Dyke T. Preclinical mouse cancer models: a maze of opportunities and challenges. Cell 2015; 163:39-53. [PMID: 26406370 DOI: 10.1016/j.cell.2015.08.068] [Citation(s) in RCA: 453] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Indexed: 12/20/2022]
Abstract
Significant advances have been made in developing novel therapeutics for cancer treatment, and targeted therapies have revolutionized the treatment of some cancers. Despite the promise, only about five percent of new cancer drugs are approved, and most fail due to lack of efficacy. The indication is that current preclinical methods are limited in predicting successful outcomes. Such failure exacts enormous cost, both financial and in the quality of human life. This Primer explores the current status, promise, and challenges of preclinical evaluation in advanced mouse cancer models and briefly addresses emerging models for early-stage preclinical development.
Collapse
Affiliation(s)
- Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| | - Terry Van Dyke
- Center for Advanced Preclinical Research, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
34
|
Sadanandam A, Wullschleger S, Lyssiotis CA, Grötzinger C, Barbi S, Bersani S, Körner J, Wafy I, Mafficini A, Lawlor RT, Simbolo M, Asara JM, Bläker H, Cantley LC, Wiedenmann B, Scarpa A, Hanahan D. A Cross-Species Analysis in Pancreatic Neuroendocrine Tumors Reveals Molecular Subtypes with Distinctive Clinical, Metastatic, Developmental, and Metabolic Characteristics. Cancer Discov 2015; 5:1296-313. [PMID: 26446169 DOI: 10.1158/2159-8290.cd-15-0068] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 10/05/2015] [Indexed: 12/14/2022]
Abstract
UNLABELLED Seeking to assess the representative and instructive value of an engineered mouse model of pancreatic neuroendocrine tumors (PanNET) for its cognate human cancer, we profiled and compared mRNA and miRNA transcriptomes of tumors from both. Mouse PanNET tumors could be classified into two distinctive subtypes, well-differentiated islet/insulinoma tumors (IT) and poorly differentiated tumors associated with liver metastases, dubbed metastasis-like primary (MLP). Human PanNETs were independently classified into these same two subtypes, along with a third, specific gene mutation-enriched subtype. The MLP subtypes in human and mouse were similar to liver metastases in terms of miRNA and mRNA transcriptome profiles and signature genes. The human/mouse MLP subtypes also similarly expressed genes known to regulate early pancreas development, whereas the IT subtypes expressed genes characteristic of mature islet cells, suggesting different tumorigenesis pathways. In addition, these subtypes exhibit distinct metabolic profiles marked by differential pyruvate metabolism, substantiating the significance of their separate identities. SIGNIFICANCE This study involves a comprehensive cross-species integrated analysis of multi-omics profiles and histology to stratify PanNETs into subtypes with distinctive characteristics. We provide support for the RIP1-TAG2 mouse model as representative of its cognate human cancer with prospects to better understand PanNET heterogeneity and consider future applications of personalized cancer therapy.
Collapse
Affiliation(s)
- Anguraj Sadanandam
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland. Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Lausanne (EPFL), Lausanne, Switzerland. Division of Molecular Pathology, Institute of Cancer Research (ICR), London, United Kingdom.
| | - Stephan Wullschleger
- Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Lausanne (EPFL), Lausanne, Switzerland
| | | | - Carsten Grötzinger
- Department of Hepatology and Gastroenterology, Charite, Campus Virchow-Klinikum, University Medicine Berlin, Berlin, Germany
| | - Stefano Barbi
- ARC-Net Research Centre and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Samantha Bersani
- ARC-Net Research Centre and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Jan Körner
- Department of Hepatology and Gastroenterology, Charite, Campus Virchow-Klinikum, University Medicine Berlin, Berlin, Germany
| | - Ismael Wafy
- Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Lausanne (EPFL), Lausanne, Switzerland
| | - Andrea Mafficini
- ARC-Net Research Centre and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Rita T Lawlor
- ARC-Net Research Centre and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Michele Simbolo
- ARC-Net Research Centre and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - John M Asara
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Hendrik Bläker
- Institut für Pathologie, Charite, Campus Virchow-Klinikum, University Medicine, Berlin, Germany
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medical College, New York, New York
| | - Bertram Wiedenmann
- Department of Hepatology and Gastroenterology, Charite, Campus Virchow-Klinikum, University Medicine Berlin, Berlin, Germany
| | - Aldo Scarpa
- ARC-Net Research Centre and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy.
| | - Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
35
|
Zhi X, Chen W, Xue F, Liang C, Chen BW, Zhou Y, Wen L, Hu L, Shen J, Bai X, Liang T. OSI-027 inhibits pancreatic ductal adenocarcinoma cell proliferation and enhances the therapeutic effect of gemcitabine both in vitro and in vivo. Oncotarget 2015; 6:26230-26241. [PMID: 26213847 PMCID: PMC4694897 DOI: 10.18632/oncotarget.4579] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 07/10/2015] [Indexed: 02/06/2023] Open
Abstract
Despite its relative rarity, pancreatic ductal adenocarcinoma (PDAC) accounts for a large percentage of cancer deaths. In this study, we investigated the in vitro efficacy of OSI-027, a selective inhibitor of mammalian target of rapamycin complex 1 (mTORC1) and mTORC2, to treat PDAC cell lines alone, and in combination with gemcitabine (GEM). Similarly, we tested the efficacy of these two compounds in a xenograft mouse model of PDAC. OSI-027 significantly arrested cell cycle in G0/G1 phase, inhibited the proliferation of Panc-1, BxPC-3, and CFPAC-1 cells, and downregulated mTORC1, mTORC2, phospho-Akt, phospho-p70S6K, phospho-4E-BP1, cyclin D1, and cyclin-dependent kinase 4 (CDK4) in these cells. Moreover, OSI-027 also downregulated multidrug resistance (MDR)-1, which has been implicated in chemotherapy resistance in PDAC cells and enhanced apoptosis induced by GEM in the three PDAC cell lines. When combined, OSI-027 with GEM showed synergistic cytotoxic effects both in vitro and in vivo. This is the first evidence of the efficacy of OSI-027 in PDAC and may provide the groundwork for a new clinical PDAC therapy.
Collapse
MESH Headings
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Apoptosis/drug effects
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Cycle Checkpoints
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Dose-Response Relationship, Drug
- Drug Synergism
- Gene Expression Regulation, Neoplastic
- Humans
- Imidazoles/pharmacology
- Inhibitory Concentration 50
- Mechanistic Target of Rapamycin Complex 1
- Mechanistic Target of Rapamycin Complex 2
- Mice, Inbred BALB C
- Mice, Nude
- Multiprotein Complexes/antagonists & inhibitors
- Multiprotein Complexes/genetics
- Multiprotein Complexes/metabolism
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Protein Kinase Inhibitors/pharmacology
- RNA Interference
- Signal Transduction/drug effects
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- Time Factors
- Transfection
- Triazines/pharmacology
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
- Gemcitabine
Collapse
Affiliation(s)
- Xiao Zhi
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Wei Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Fei Xue
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Chao Liang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Bryan Wei Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Yue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Liang Wen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Liqiang Hu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Jian Shen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
- Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R.China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, P.R.China
- Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R.China
- Collaborative Innovation Center for Cancer Medicine, Zhejiang University, Hangzhou, P.R.China
| |
Collapse
|
36
|
Bill R, Fagiani E, Zumsteg A, Antoniadis H, Johansson D, Haefliger S, Albrecht I, Hilberg F, Christofori G. Nintedanib Is a Highly Effective Therapeutic for Neuroendocrine Carcinoma of the Pancreas (PNET) in the Rip1Tag2 Transgenic Mouse Model. Clin Cancer Res 2015. [DOI: 10.1158/1078-0432.ccr-14-3036] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
37
|
Alonso-Gordoa T, Díez JJ, Molina J, Reguera P, Martínez-Sáez O, Grande E. An Overview on the Sequential Treatment of Pancreatic Neuroendocrine Tumors (pNETs). ACTA ACUST UNITED AC 2015; 3:13-33. [PMID: 27182476 PMCID: PMC4837935 DOI: 10.1007/s40487-015-0007-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Indexed: 01/10/2023]
Abstract
Patients suffering from pancreatic neuroendocrine tumors (pNETs) are now candidates to receive novel approved drugs that have demonstrated benefit in disease control rate and delay the time taken for tumor progression in Phase III clinical trials; for example, sunitinib, everolimus and lanreotide. Though pNETs represent a rare and heterogeneous disease, recent approaches are being taken to better understand the molecular pathways involved in carcinogenesis. Consequently, new treatment strategies are now available and others still under investigation show promising results. However, some questions around how to approach patients with pNETs are still unresolved, such as what the best sequence of treatments we can offer to each of our patients in the clinic at any time of their disease would be. Therapeutic decisions are, at the moment, guided by clinical judgment, based on different parameters coming from retrospective analysis and non-randomized clinical trials. However, advances in genomic research would lead to a more precise approach using therapeutic targets that would also allow the development of new agents, prognostic or predictive biomarkers and a better understanding of resistance mechanisms. The following article is a comprehensive review of the approved and investigational drugs in pNET, and highlights the current concerns about treatment sequencing, but also provides an update of some of the present and future efforts for an improvement in the therapeutic algorithm of the disease.
Collapse
Affiliation(s)
- Teresa Alonso-Gordoa
- Medical Oncology Department, Ramón y Cajal University Hospital, Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain
| | - Juan José Díez
- Endocrinology and Nutrition Department, Ramón y Cajal University Hospital, Madrid, Spain
| | - Javier Molina
- Medical Oncology Department, Ramón y Cajal University Hospital, Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain
| | - Pablo Reguera
- Medical Oncology Department, Ramón y Cajal University Hospital, Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain
| | - Olga Martínez-Sáez
- Medical Oncology Department, Ramón y Cajal University Hospital, Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain
| | - Enrique Grande
- Medical Oncology Department, Ramón y Cajal University Hospital, Ctra. Colmenar Viejo km 9100, 28034 Madrid, Spain
| |
Collapse
|
38
|
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
|
39
|
Cook N, Hansen AR, Siu LL, Abdul Razak AR. Early phase clinical trials to identify optimal dosing and safety. Mol Oncol 2015; 9:997-1007. [PMID: 25160636 PMCID: PMC4329110 DOI: 10.1016/j.molonc.2014.07.025] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 07/30/2014] [Indexed: 12/13/2022] Open
Abstract
The purpose of early stage clinical trials is to determine the recommended dose and toxicity profile of an investigational agent or multi-drug combination. Molecularly targeted agents (MTAs) and immunotherapies have distinct toxicities from chemotherapies that are often not dose dependent and can lead to chronic and sometimes unpredictable side effects. Therefore utilizing a dose escalation method that has toxicity based endpoints may not be as appropriate for determination of recommended dose, and alternative parameters such as pharmacokinetic or pharmacodynamic outcomes are potentially appealing options. Approaches to enhance safety and optimize dosing include improved preclinical models and assessment, innovative model based design and dose escalation strategies, patient selection, the use of expansion cohorts and extended toxicity assessments. Tailoring the design of phase I trials by adopting new strategies to address the different properties of MTAs is required to enhance the development of these agents. This review will focus on the limitations to safety and dose determination that have occurred in the development of MTAs and immunotherapies. In addition, strategies are proposed to overcome these challenges to develop phase I trials that can more accurately define the recommended dose and identify adverse events.
Collapse
Affiliation(s)
- Natalie Cook
- Princess Margaret Cancer Centre, University Health Network, Division of Medical Oncology and Hematology, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Aaron R Hansen
- Princess Margaret Cancer Centre, University Health Network, Division of Medical Oncology and Hematology, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Lillian L Siu
- Princess Margaret Cancer Centre, University Health Network, Division of Medical Oncology and Hematology, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada.
| | - Albiruni R Abdul Razak
- Princess Margaret Cancer Centre, University Health Network, Division of Medical Oncology and Hematology, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
40
|
De Dosso S, Grande E, Barriuso J, Castellano D, Tabernero J, Capdevila J. The targeted therapy revolution in neuroendocrine tumors: in search of biomarkers for patient selection and response evaluation. Cancer Metastasis Rev 2014; 32:465-77. [PMID: 23589060 DOI: 10.1007/s10555-013-9421-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The molecular events of tumorigenesis in neuroendocrine tumors are poorly understood. Understanding of the molecular alterations will lead to the identification of molecular markers, providing new targets for therapeutics. The purpose of this review was to critically analyze the genetic abnormalities in neuroendocrine tumors, with the aim of identifying biomarkers that indicate a response to agents developed against these targets and to serve as an understanding for the combinations of different active compounds. Human epidermal growth factor receptor 1/2 (EGFR and HER2), vascular endothelial growth factor receptors, hepatocyte growth factor receptor (c-Met), platelet-derived growth factor receptor, insulin-like growth factor, phosphatidylinositol 3-kinase-Akt-mammalian target of rapamycin pathway, and heat shock proteins are all interesting candidate biomarkers with involvement in carcinogenesis and tumor evolution of several neoplasms, including neuroendocrine tumors. Some of them have already been evaluated both as targets and also as biomarkers in clinical trials conducted in advanced neuroendocrine tumor settings, and others should encourage further investigations into innovative therapeutic opportunities.
Collapse
Affiliation(s)
- Sara De Dosso
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | | | | | | | | | | |
Collapse
|
41
|
Meeker A, Heaphy C. Gastroenteropancreatic endocrine tumors. Mol Cell Endocrinol 2014; 386:101-20. [PMID: 23906538 DOI: 10.1016/j.mce.2013.07.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 07/19/2013] [Accepted: 07/22/2013] [Indexed: 02/06/2023]
Abstract
Gastroenteropancreatic endocrine tumors (GEP-NETs) are relatively uncommon; comprising approximately 0.5% of all human cancers. Although they often exhibit relatively indolent clinical courses, GEP-NETs have the potential for lethal progression. Due to their scarcity and various technical challenges, GEP-NETs have been understudied. As a consequence, we have few diagnostic, prognostic and predictive biomarkers for these tumors. Early detection and surgical removal is currently the only reliable curative treatment for GEP-NET patients; many of whom, unfortunately, present with advanced disease. Here, we review the genetics and epigenetics of GEP-NETs. The last few years have witnessed unprecedented technological advances in these fields, and their application to GEP-NETS has already led to important new information on the molecular abnormalities underlying them. As outlined here, we expect that "omics" studies will provide us with new diagnostic and prognostic biomarkers, inform the development of improved pre-clinical models, and identify novel therapeutic targets for GEP-NET patients.
Collapse
Affiliation(s)
- Alan Meeker
- The Johns Hopkins University School of Medicine, Department of Pathology, Bond Street Research Annex Bldg., Room B300, 411 North Caroline Street, Baltimore, MD 21231, United States.
| | - Christopher Heaphy
- The Johns Hopkins University School of Medicine, Department of Pathology, Bond Street Research Annex Bldg., Room B300, 411 North Caroline Street, Baltimore, MD 21231, United States
| |
Collapse
|
42
|
Colvin EK, Scarlett CJ. A historical perspective of pancreatic cancer mouse models. Semin Cell Dev Biol 2014; 27:96-105. [PMID: 24685616 DOI: 10.1016/j.semcdb.2014.03.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 03/20/2014] [Accepted: 03/21/2014] [Indexed: 12/22/2022]
Abstract
Pancreatic cancer is an inherently aggressive disease with an extremely poor prognosis and lack of effective treatments. Over the past few decades, much has been uncovered regarding the pathogenesis of pancreatic cancer and the underlying genetic alterations necessary for tumour initiation and progression. Much of what we know about pancreatic cancer has come from mouse models of this disease. This review focusses on the development of genetically engineered mouse models that phenotypically and genetically recapitulate human pancreatic cancer, as well as the increasing use of patient-derived xenografts for preclinical studies and the development of personalised medicine strategies.
Collapse
Affiliation(s)
- Emily K Colvin
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, Australia.
| | - Christopher J Scarlett
- Pancreatic Cancer Research, Nutrition, Food and Health Research Group, School of Environmental and Life Sciences, University of Newcastle, Ourimbah, NSW, Australia.
| |
Collapse
|
43
|
Colvin EK, Weir C, Ikin RJ, Hudson AL. SV40 TAg mouse models of cancer. Semin Cell Dev Biol 2014; 27:61-73. [PMID: 24583142 DOI: 10.1016/j.semcdb.2014.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 02/03/2014] [Accepted: 02/05/2014] [Indexed: 02/09/2023]
Abstract
The discovery of a number of viruses with the ability to induce tumours in animals and transform human cells has vastly impacted cancer research. Much of what is known about tumorigenesis today regarding tumour drivers and tumour suppressors has been discovered through experiments using viruses. The SV40 virus has proven extremely successful in generating transgenic models of many human cancer types and this review provides an overview of these models and seeks to give evidence as to their relevance in this modern era of personalised medicine and technological advancements.
Collapse
Affiliation(s)
- Emily K Colvin
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia.
| | - Chris Weir
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia.
| | - Rowan J Ikin
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia.
| | - Amanda L Hudson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia.
| |
Collapse
|
44
|
Raymond E, García-Carbonero R, Wiedenmann B, Grande E, Pavel M. Systemic therapeutic strategies for GEP-NETS: what can we expect in the future? Cancer Metastasis Rev 2013; 33:367-72. [DOI: 10.1007/s10555-013-9467-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
45
|
Ryu K, Kim TI. Therapeutic gene delivery using bioreducible polymers. Arch Pharm Res 2013; 37:31-42. [DOI: 10.1007/s12272-013-0275-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 10/22/2013] [Indexed: 12/14/2022]
|
46
|
Liu E, Marincola P, Öberg K. Everolimus in the treatment of patients with advanced pancreatic neuroendocrine tumors: latest findings and interpretations. Therap Adv Gastroenterol 2013; 6:412-9. [PMID: 24003341 PMCID: PMC3756638 DOI: 10.1177/1756283x13496970] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Pancreatic neuroendocrine tumors (pNETs) are a heterogeneous group of neoplasms with various clinical presentations. More than half of patients present with so-called nonfunctioning tumors with no hormone-related symptoms, whereas other tumors produce symptoms like gastric problems, ulcers, hypoglycemia, skin rash and diarrhea related to hormone production. The traditional treatment for pNETs over the last three decades has been cytotoxic agents, mainly streptozotocin plus 5-fluorouracil or doxorubicin. Most recently two new compounds have been registered worldwide for the treatment of pNETs, the mammalian target of rapamycin (mTOR) inhibitor everolimus and the tyrosine kinase inhibitor sunitinib. This paper concentrates on the use of mTOR inhibitors and the mechanisms of action. The mTOR pathway is altered in a number of pNETs. Everolimus (RAD001) is an orally active rapamycin analog and mTOR inhibitor. It blocks activity of the mTOR pathway by binding with high affinity to the cytoplasmic protein FKBP-12. The efficacy of everolimus in pNETs has been demonstrated in two multicenter studies (RADIANT 1 and 3). The RADIANT 3 study was a randomized controlled study in pNETs of everolimus 10 mg/day versus placebo, showing an increased progression-free survival (11.7 months versus 4.6 months) and hazard ratio of 0.35 (p < 0.001). Current studies indicate that there is strong evidence to support the antitumor effect of rapalogs in pNETs. However, significant tumor reduction is very rarely obtained, usually in less than 10% of treated patients. Therefore, these drugs may be more effective in combination with other anticancer agents, including chemotherapy, targeted therapies as well as peptide receptor radiotherapy.
Collapse
Affiliation(s)
- Eric Liu
- Department of Surgery, Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Paula Marincola
- Department of Surgery, Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kjell Öberg
- Department of Medical Sciences, Uppsala University, Entrance 40, 5th floor, SE-751 85 Uppsala, Sweden
| |
Collapse
|
47
|
Zhang J, Francois R, Iyer R, Seshadri M, Zajac-Kaye M, Hochwald SN. Current understanding of the molecular biology of pancreatic neuroendocrine tumors. J Natl Cancer Inst 2013; 105:1005-17. [PMID: 23840053 PMCID: PMC6281020 DOI: 10.1093/jnci/djt135] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 04/05/2013] [Accepted: 04/06/2013] [Indexed: 12/11/2022] Open
Abstract
Pancreatic neuroendocrine tumors (PanNETs) are complicated and often deadly neoplasms. A recent increased understanding of their molecular biology has contributed to expanded treatment options. DNA sequencing of samples derived from patients with PanNETs and rare genetic syndromes such as multiple endocrine neoplasia type 1 (MEN1) and Von Hippel-Lindau (VHL) syndrome reveals the involvement of MEN1, DAXX/ATRX, and the mammalian target of rapamycin (mTOR) pathways in PanNET tumorigenesis. Gene knock-out/knock-in studies indicate that inactivation of factors including MEN1 and abnormal PI3K/mTOR signaling uncouples endocrine cell cycle progression from the control of environmental cues such as glucose, leading to islet cell overgrowth. In addition, accumulating evidence suggests that further impairment of endothelial-endocrine cell interactions contributes to tumor invasion and metastasis. Recent phase III clinical trials have shown that therapeutic interventions, such as sunitinib and everolimus, targeting those signal transduction pathways improve disease-free survival rates. Yet, cure in the setting of advanced disease remains elusive. Further advances in our understanding of the molecular mechanisms of PanNETs and improved preclinical models will assist in developing personalized therapy utilizing novel drugs to provide prolonged control or even cure the disease.
Collapse
Affiliation(s)
- Jianliang Zhang
- Affiliations of authors:Department of Surgical Oncology (JZ, SNH), Department of Medical Oncology (RI), and Department of Pharmacology and Therapeutics (MS), Roswell Park Cancer Institute, Buffalo, NY; Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL (RF, MZ-K)
| | - Rony Francois
- Affiliations of authors:Department of Surgical Oncology (JZ, SNH), Department of Medical Oncology (RI), and Department of Pharmacology and Therapeutics (MS), Roswell Park Cancer Institute, Buffalo, NY; Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL (RF, MZ-K)
| | - Renuka Iyer
- Affiliations of authors:Department of Surgical Oncology (JZ, SNH), Department of Medical Oncology (RI), and Department of Pharmacology and Therapeutics (MS), Roswell Park Cancer Institute, Buffalo, NY; Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL (RF, MZ-K)
| | - Mukund Seshadri
- Affiliations of authors:Department of Surgical Oncology (JZ, SNH), Department of Medical Oncology (RI), and Department of Pharmacology and Therapeutics (MS), Roswell Park Cancer Institute, Buffalo, NY; Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL (RF, MZ-K)
| | - Maria Zajac-Kaye
- Affiliations of authors:Department of Surgical Oncology (JZ, SNH), Department of Medical Oncology (RI), and Department of Pharmacology and Therapeutics (MS), Roswell Park Cancer Institute, Buffalo, NY; Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL (RF, MZ-K)
| | - Steven N. Hochwald
- Affiliations of authors:Department of Surgical Oncology (JZ, SNH), Department of Medical Oncology (RI), and Department of Pharmacology and Therapeutics (MS), Roswell Park Cancer Institute, Buffalo, NY; Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL (RF, MZ-K)
| |
Collapse
|
48
|
Yin Q, Shen J, Zhang Z, Yu H, Chen L, Gu W, Li Y. Multifunctional Nanoparticles Improve Therapeutic Effect for Breast Cancer by Simultaneously Antagonizing Multiple Mechanisms of Multidrug Resistance. Biomacromolecules 2013; 14:2242-52. [DOI: 10.1021/bm400378x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Qi Yin
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai
201203, China
| | - Jianan Shen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai
201203, China
| | - Zhiwen Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai
201203, China
| | - Haijun Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai
201203, China
| | - Lingli Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai
201203, China
| | - Wangwen Gu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai
201203, China
| | - Yaping Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai
201203, China
| |
Collapse
|
49
|
Identification and characterization of poorly differentiated invasive carcinomas in a mouse model of pancreatic neuroendocrine tumorigenesis. PLoS One 2013; 8:e64472. [PMID: 23691228 PMCID: PMC3653861 DOI: 10.1371/journal.pone.0064472] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 04/15/2013] [Indexed: 11/19/2022] Open
Abstract
Pancreatic neuroendocrine tumors (PanNETs) are a relatively rare but clinically challenging tumor type. In particular, high grade, poorly-differentiated PanNETs have the worst patient prognosis, and the underlying mechanisms of disease are poorly understood. In this study we have identified and characterized a previously undescribed class of poorly differentiated PanNETs in the RIP1-Tag2 mouse model. We found that while the majority of tumors in the RIP1-Tag2 model are well-differentiated insulinomas, a subset of tumors had lost multiple markers of beta-cell differentiation and were highly invasive, leading us to term them poorly differentiated invasive carcinomas (PDICs). In addition, we found that these tumors exhibited a high mitotic index, resembling poorly differentiated (PD)-PanNETs in human patients. Interestingly, we identified expression of Id1, an inhibitor of DNA binding gene, and a regulator of differentiation, specifically in PDIC tumor cells by histological analysis. The identification of PDICs in this mouse model provides a unique opportunity to study the pathology and molecular characteristics of PD-PanNETs.
Collapse
|
50
|
Mankal P, O'Reilly E. Sunitinib malate for the treatment of pancreas malignancies--where does it fit? Expert Opin Pharmacother 2013; 14:783-92. [PMID: 23458511 DOI: 10.1517/14656566.2013.776540] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Sunitinib , a broad-spectrum multikinase inhibitor, was recently approved for use in progressive, well-differentiated pancreatic neuroendocrine tumors (pNETs). Its mechanism of action affects various signaling cascades involving antiangiogenesis and tumor proliferation, including vascular endothelial growth factors and platelet-derived growth factors. AREAS COVERED In this article, we review sunitinib's mechanism of action at a molecular level and review key preclinical and clinical studies for pNETs and more limited data regarding sunitinib's evaluation in pancreas adenocarcinoma. The data for sunitinib in pNETs are placed in the context of the changing landscape of therapeutic options for this cancer, and relevant ongoing clinical trials and future directions are highlighted. EXPERT OPINION Sunitinib malate has become integrated into routine clinical management for pNETs; however, its role in pancreas adenocarcinoma is not established.
Collapse
Affiliation(s)
- Pavan Mankal
- Columbia University College of Physicians and Surgeons, St. Luke's-Roosevelt Hospital Center, Department of Medicine, New York, USA
| | | |
Collapse
|