1901
|
Shive HR, West RR, Embree LJ, Golden CD, Hickstein DD. BRCA2 and TP53 collaborate in tumorigenesis in zebrafish. PLoS One 2014; 9:e87177. [PMID: 24489863 PMCID: PMC3906131 DOI: 10.1371/journal.pone.0087177] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 12/23/2013] [Indexed: 12/02/2022] Open
Abstract
Germline mutations in the tumor suppressor genes BRCA2 and TP53 significantly influence human cancer risk, and cancers from humans who inherit one mutant allele for BRCA2 or TP53 often display loss of the wildtype allele. In addition, BRCA2-associated cancers often exhibit mutations in TP53. To determine the relationship between germline heterozygous mutation (haploinsufficiency) and somatic loss of heterozygosity (LOH) for BRCA2 and TP53 in carcinogenesis, we analyzed zebrafish with heritable mutations in these two genes. Tumor-bearing zebrafish were examined by histology, and normal and neoplastic tissues were collected by laser-capture microdissection for LOH analyses. Zebrafish on a heterozygous tp53M214K background had a high incidence of malignant tumors. The brca2Q658X mutation status determined both the incidence of LOH and the malignant tumor phenotype. LOH for tp53 occurred in the majority of malignant tumors from brca2 wildtype and heterozygous mutant zebrafish, and most of these were malignant peripheral nerve sheath tumors. Malignant tumors in zebrafish with heterozygous mutations in both brca2 and tp53 frequently displayed LOH for both genes. In contrast, LOH for tp53 was uncommon in malignant tumors from brca2 homozygotes, and these tumors were primarily undifferentiated sarcomas. Thus, carcinogenesis in zebrafish with combined mutations in tp53 and brca2 typically requires biallelic mutation or loss of at least one of these genes, and the specific combination of inherited mutations influences the development of LOH and the tumor phenotype. These results provide insight into cancer development associated with heritable BRCA2 and TP53 mutations.
Collapse
Affiliation(s)
- Heather R. Shive
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Robert R. West
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lisa J. Embree
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Champa D. Golden
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Dennis D. Hickstein
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
1902
|
Morandi A, Chiarugi P. Metabolic implication of tumor:stroma crosstalk in breast cancer. J Mol Med (Berl) 2014; 92:117-26. [PMID: 24458539 DOI: 10.1007/s00109-014-1124-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 01/10/2014] [Indexed: 12/14/2022]
Abstract
The metabolic properties of cancer cells significantly differ from those of normal cells. In particular, cancer cells are largely dependent on aerobic glycolysis, a phenomenon that has been exploited clinically by using labelled glucose for positron emission tomography imaging. Importantly, cancer-associated alterations in metabolism are not merely due to the resulting response to cell proliferation and survival. Indeed, direct metabolic regulation could be driven by tumor oncogenes and/or suppressors, as demonstrated in several solid tumors, including breast cancer. Despite the fact that most breast cancer studies have focused on the intrinsic characteristics of breast tumor cells, it is now widely accepted that tumor microenvironment plays an important role in defining and reprogramming cancer cell metabolism. Tumor:stroma crosstalk, as well as inflammatory cues, concurs to outlining the cancer metabolism, impact on cancer aggressiveness and ultimately on patient survival and therapeutic responses. The aim of this review is to (i) gather the most recent data regarding the metabolic alterations in breast cancer, (ii) describe the role of tumor microenvironment in breast cancer cell metabolic reprogramming, and (iii) contemplate how targeting metabolic pathways aberrantly activated in breast cancer could help current therapeutic regimens.
Collapse
Affiliation(s)
- Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, viale GB Morgagni 50, Florence, I-50134, Italy
| | | |
Collapse
|
1903
|
Lei T, Huang Z, Ohno N, Wu B, Sakoh T, Saitoh Y, Saiki I, Ohno S. Bioimaging of fluorescence-labeled mitochondria in subcutaneously grafted murine melanoma cells by the "in vivo cryotechnique". J Histochem Cytochem 2014; 62:251-64. [PMID: 24394469 DOI: 10.1369/0022155413520313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The microenvironments of organs with blood flow affect the metabolic profiles of cancer cells, which are influenced by mitochondrial functions. However, histopathological analyses of these aspects have been hampered by technical artifacts of conventional fixation and dehydration, including ischemia/anoxia. The purpose of this study was to combine the in vivo cryotechnique (IVCT) with fluorescent protein expression, and examine fluorescently labeled mitochondria in grafted melanoma tumors. The intensity of fluorescent proteins was maintained well in cultured B16-BL6 cells after cryotechniques followed by freeze-substitution (FS). In the subcutaneous tumors of mitochondria-targeted DsRed2 (mitoDsRed)-expressing cells, a higher number of cancer cells were found surrounding the widely opened blood vessels that contained numerous erythrocytes. Such blood vessels were immunostained positively for immunoglobulin M and ensheathed by basement membranes. MitoDsRed fluorescence was detected in scattering melanoma cells using the IVCT-FS method, and the total mitoDsRed volume in individual cancer cells was significantly decreased with the expression of markers of hypoxia. MitoDsRed was frequently distributed throughout the cytoplasm and in processes extending along basement membranes. IVCT combined with fluorescent protein expression is a useful tool to examine the behavior of fluorescently labeled cells and organelles. We propose that the mitochondrial volume is dynamically regulated in the hypoxic microenvironment and that mitochondrial distribution is modulated by cancer cell interactions with basement membranes.
Collapse
Affiliation(s)
- Ting Lei
- Department of Anatomy and Molecular Histology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo-city, Yamanashi, Japan (TL, ZH, NO, BW, TS, YS, SO)
| | | | | | | | | | | | | | | |
Collapse
|
1904
|
Menzies AM, Haydu LE, Carlino MS, Azer MWF, Carr PJA, Kefford RF, Long GV. Inter- and intra-patient heterogeneity of response and progression to targeted therapy in metastatic melanoma. PLoS One 2014; 9:e85004. [PMID: 24400126 PMCID: PMC3882277 DOI: 10.1371/journal.pone.0085004] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 11/27/2013] [Indexed: 12/22/2022] Open
Abstract
Background MAPK inhibitors (MAPKi) are active in BRAF-mutant metastatic melanoma patients, but the extent of response and progression-free survival (PFS) is variable, and complete responses are rare. We sought to examine the patterns of response and progression in patients treated with targeted therapy. Methods MAPKi-naïve patients treated with combined dabrafenib and trametinib had all metastases ≥5 mm (lymph nodes ≥15 mm in short axis) visible on computed tomography measured at baseline and throughout treatment. Results 24 patients had 135 measured metastases (median 4.5/patient, median diameter 16 mm). Time to best response (median 5.5 mo, range 1.7–20.1 mo), and the degree of best response (median −70%, range +9 to −100%) varied amongst patients. 17% of patients achieved complete response (CR), whereas 53% of metastases underwent CR, including 42% ≥10 mm. Metastases that underwent CR were smaller than non-CR metastases (median 11 vs 20 mm, P<0.001). PFS was variable among patients (median 8.2 mo, range 2.6–18.3 mo), and 50% of patients had disease progression in new metastases only. Only 1% (1/71) of CR-metastases subsequently progressed. Twelve-month overall survival was poorer in those with a more heterogeneous initial response to therapy than less heterogeneous (67% vs 93%, P = 0.009). Conclusion Melanoma response and progression with MAPKi displays marked inter- and intra-patient heterogeneity. Most metastases undergo complete response, yet only a small proportion of patients achieve an overall complete response. Similarly, disease progression often occurs only in a subset of the tumor burden, and often in new metastases alone. Clinical heterogeneity, likely reflecting molecular heterogeneity, remains a barrier to the effective treatment of melanoma patients.
Collapse
Affiliation(s)
- Alexander M. Menzies
- Melanoma Institute Australia, Sydney, Australia
- The University of Sydney, Sydney, Australia
- * E-mail:
| | - Lauren E. Haydu
- Melanoma Institute Australia, Sydney, Australia
- The University of Sydney, Sydney, Australia
| | - Matteo S. Carlino
- Melanoma Institute Australia, Sydney, Australia
- The University of Sydney, Sydney, Australia
- Westmead Hospital, Crown Princess Mary Cancer Centre, Sydney, Australia
- Westmead Institute for Cancer Research, Westmead, Australia
| | - Mary W. F. Azer
- Westmead Hospital, Crown Princess Mary Cancer Centre, Sydney, Australia
| | - Peter J. A. Carr
- The University of Sydney, Sydney, Australia
- Westmead Hospital, Department of Radiology, Sydney, Australia
| | - Richard F. Kefford
- Melanoma Institute Australia, Sydney, Australia
- The University of Sydney, Sydney, Australia
- Westmead Hospital, Crown Princess Mary Cancer Centre, Sydney, Australia
- Westmead Institute for Cancer Research, Westmead, Australia
| | - Georgina V. Long
- Melanoma Institute Australia, Sydney, Australia
- The University of Sydney, Sydney, Australia
| |
Collapse
|
1905
|
Van Overmeire E, Laoui D, Keirsse J, Van Ginderachter JA. Hypoxia and tumor-associated macrophages: A deadly alliance in support of tumor progression. Oncoimmunology 2014; 3:e27561. [PMID: 24744977 PMCID: PMC3989296 DOI: 10.4161/onci.27561] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 12/17/2013] [Indexed: 12/12/2022] Open
Abstract
Tumor-associated macrophages (TAMs) provide a significant contribution to tumor growth and metastasis. We demonstrated the existence of two main TAM subsets, differing in activation state and localization. Of these, M2-like TAMs reside in hypoxic regions of the tumor mass and can be used as targets for hypoxia tracers. This said, hypoxia does not regulate the differentiation of TAMs but finely tunes the activity of the M2-like population.
Collapse
Affiliation(s)
- Eva Van Overmeire
- Lab of Cellular and Molecular Immunology; Vrije Universiteit Brussel; Brussels, Belgium ; Myeloid Cell Immunology Lab; VIB; Brussels, Belgium
| | - Damya Laoui
- Lab of Cellular and Molecular Immunology; Vrije Universiteit Brussel; Brussels, Belgium ; Myeloid Cell Immunology Lab; VIB; Brussels, Belgium
| | - Jiri Keirsse
- Lab of Cellular and Molecular Immunology; Vrije Universiteit Brussel; Brussels, Belgium ; Myeloid Cell Immunology Lab; VIB; Brussels, Belgium
| | - Jo A Van Ginderachter
- Lab of Cellular and Molecular Immunology; Vrije Universiteit Brussel; Brussels, Belgium ; Myeloid Cell Immunology Lab; VIB; Brussels, Belgium
| |
Collapse
|
1906
|
Chemokine receptors in epithelial ovarian cancer. Int J Mol Sci 2013; 15:361-76. [PMID: 24384839 PMCID: PMC3907814 DOI: 10.3390/ijms15010361] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 12/17/2013] [Accepted: 12/19/2013] [Indexed: 01/03/2023] Open
Abstract
Ovarian carcinoma is the deadliest gynecologic malignancy with very poor rate of survival, and it is characterized by the presence of vast incurable peritoneal metastasis. Studies of the role of chemokine receptors, a family of proteins belonging to the group of G protein-coupled receptors, in ovarian carcinoma strongly placed this family of membrane receptors as major regulators of progression of this malignancy. In this review, we will discuss the roles that chemokine-receptor interactions play to support angiogenesis, cell proliferation, migration, adhesion, invasion, metastasis, and immune evasion in progression of ovarian carcinoma. Data regarding the role that the chemokine receptors play in the disease progression accumulated insofar strongly suggest that this family of proteins could be good therapeutic targets against ovarian carcinoma.
Collapse
|
1907
|
Ilyas SI, Gores GJ. Pathogenesis, diagnosis, and management of cholangiocarcinoma. Gastroenterology 2013; 145:1215-29. [PMID: 24140396 PMCID: PMC3862291 DOI: 10.1053/j.gastro.2013.10.013] [Citation(s) in RCA: 954] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 10/08/2013] [Accepted: 10/10/2013] [Indexed: 12/13/2022]
Abstract
Cholangiocarcinomas (CCAs) are hepatobiliary cancers with features of cholangiocyte differentiation; they can be classified anatomically as intrahepatic CCA (iCCA), perihilar CCA (pCCA), or distal CCA. These subtypes differ not only in their anatomic location, but in epidemiology, origin, etiology, pathogenesis, and treatment. The incidence and mortality of iCCA has been increasing over the past 3 decades, and only a low percentage of patients survive until 5 years after diagnosis. Geographic variations in the incidence of CCA are related to variations in risk factors. Changes in oncogene and inflammatory signaling pathways, as well as genetic and epigenetic alterations and chromosome aberrations, have been shown to contribute to the development of CCA. Furthermore, CCAs are surrounded by a dense stroma that contains many cancer-associated fibroblasts, which promotes their progression. We have gained a better understanding of the imaging characteristics of iCCAs and have developed advanced cytologic techniques to detect pCCAs. Patients with iCCAs usually are treated surgically, whereas liver transplantation after neoadjuvant chemoradiation is an option for a subset of patients with pCCAs. We review recent developments in our understanding of the epidemiology and pathogenesis of CCA, along with advances in classification, diagnosis, and treatment.
Collapse
Affiliation(s)
- Sumera I Ilyas
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
1908
|
Yan Y, Björnmalm M, Caruso F. Particle carriers for combating multidrug-resistant cancer. ACS NANO 2013; 7:9512-9517. [PMID: 24215340 DOI: 10.1021/nn405632s] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Multidrug resistance (MDR) in tumors accounts for significant treatment failure. Particle carriers offer potential benefits for treating cancer, including the ability to target tumors and to deliver multiple cargo, providing opportunities to overcome drug resistance. In this Perspective, we provide a brief introduction to the MDR mechanisms and implications of tumor heterogeneity that contribute to drug resistance. We also highlight recent advances in the design of particles aimed at treating resistant tumors through particle-based codelivery of therapeutics. Finally, we discuss future directions, where an increased understanding of the tumor biology can be leveraged to develop new and improved particle-based cancer therapies.
Collapse
Affiliation(s)
- Yan Yan
- Department of Chemical and Biomolecular Engineering, The University of Melbourne , Victoria 3010, Australia
| | | | | |
Collapse
|
1909
|
Abstract
Multiple subclonal populations of tumor cells can coexist within the same tumor. This intra-tumor heterogeneity will have clinical implications and it is therefore important to identify factors that drive or suppress such heterogeneous tumor progression. Evolutionary biology can provide important insights into this process. In particular, experimental evolution studies of microbial populations, which exist as clonal populations that can diversify into multiple subclones, have revealed important evolutionary processes driving heterogeneity within a population. There are transferrable lessons that can be learnt from these studies that will help us to understand the process of intra-tumor heterogeneity in the clinical setting. In this review, we summarize drivers of microbial diversity that have been identified, such as mutation rate and environmental influences, and discuss how knowledge gained from microbial experimental evolution studies may guide us to identify and understand important selective factors that promote intra-tumor heterogeneity. Furthermore, we discuss how these factors could be used to direct and optimize research efforts to improve patient care, focusing on therapeutic resistance. Finally, we emphasize the need for longitudinal studies to address the impact of these potential tumor heterogeneity-promoting factors on drug resistance, metastatic potential and clinical outcome.
Collapse
|
1910
|
Matsubara S, Ding Q, Miyazaki Y, Kuwahata T, Tsukasa K, Takao S. mTOR plays critical roles in pancreatic cancer stem cells through specific and stemness-related functions. Sci Rep 2013; 3:3230. [PMID: 24231729 PMCID: PMC3828572 DOI: 10.1038/srep03230] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 10/31/2013] [Indexed: 12/23/2022] Open
Abstract
Pancreatic cancer is characterized by near-universal mutations in KRAS. The mammalian target of rapamycin (mTOR), which functions downstream of RAS, has divergent effects on stem cells. In the present study, we investigated the significance of the mTOR pathway in maintaining the properties of pancreatic cancer stem cells. The mTOR inhibitor, rapamycin, reduced the viability of CD133+ pancreatic cancer cells and sphere formation which is an index of self-renewal of stem-like cells, indicating that the mTOR pathway functions to maintain cancer stem-like cells. Further, rapamycin had different effects on CD133+ cells compared to cyclopamine which is an inhibitor of the Hedgehog pathway. Thus, the mTOR pathway has a distinct role although both pathways maintain pancreatic cancer stem cells. Therefore, mTOR might be a promising target to eliminate pancreatic cancer stem cells.
Collapse
Affiliation(s)
- Shyuichiro Matsubara
- Cancer and Regenerative Medicine, Frontier Biomedical Science and Swine Research Center, Sakuragaoka, Kagoshima, 890-8520, Japan
| | | | | | | | | | | |
Collapse
|