1
|
Harihar S, Ray S, Narayanan S, Santhoshkumar A, Ly T, Welch DR. Role of the tumor microenvironment in regulating the anti-metastatic effect of KISS1. Clin Exp Metastasis 2020; 37:209-223. [PMID: 32088827 PMCID: PMC7339126 DOI: 10.1007/s10585-020-10030-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 02/19/2020] [Indexed: 12/29/2022]
Abstract
KISS1, a metastasis suppressor gene, has been shown to block metastasis without affecting primary tumor formation. Loss of KISS1 leads to invasion and metastasis in multiple cancers, which is the leading cause of cancer morbidity and mortality. The discovery of KISS1 has provided a ray of hope for early clinical diagnosis and for designing effective treatments targeting metastatic cancer. However, this goal requires greater holistic understanding of its mechanism of action. In this review, we go back into history and highlight some key developments, from the discovery of KISS1 to its role in regulating multiple physiological processes including cancer. We discuss key emerging roles for KISS1, specifically interactions with tissue microenvironment to promote dormancy and regulation of tumor cell metabolism, acknowledged as some of the key players in tumor progression and metastasis. We finally discuss strategies whereby KISS1 might be exploited clinically to treat metastasis.
Collapse
Affiliation(s)
- Sitaram Harihar
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| | - Srijit Ray
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Samyukta Narayanan
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Anirudh Santhoshkumar
- Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Thuc Ly
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
- The University Kansas Cancer Center, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
| | - Danny R Welch
- Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
- The University Kansas Cancer Center, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, 66160, USA
| |
Collapse
|
2
|
Dutta R, Liba O, SoRelle ED, Winetraub Y, Ramani VC, Jeffrey SS, Sledge GW, de la Zerda A. Real-Time Detection of Circulating Tumor Cells in Living Animals Using Functionalized Large Gold Nanorods. NANO LETTERS 2019; 19:2334-2342. [PMID: 30895796 DOI: 10.1021/acs.nanolett.8b05005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Optical coherence tomography (OCT) can be utilized with significant speckle reduction techniques and highly scattering contrast agents for non-invasive, contrast-enhanced imaging of living tissues at the cellular scale. The advantages of reduced speckle noise and improved targeted contrast can be harnessed to track objects as small as 2 μm in vivo, which enables applications for cell tracking and quantification in living subjects. Here we demonstrate the use of large gold nanorods as contrast agents for detecting individual micron-sized polystyrene beads and single myeloma cells in blood circulation using speckle-modulating OCT. This report marks the first time that OCT has been used to detect individual cells within blood in vivo. This technical capability unlocks exciting opportunities for dynamic detection and quantification of tumor cells circulating in living subjects.
Collapse
Affiliation(s)
- Rebecca Dutta
- Department of Structural Biology , Stanford University , Stanford , California 94305 , United States
- Molecular Imaging Program and the Bio-X Program , Stanford University , Stanford , California 94305 , United States
| | - Orly Liba
- Department of Structural Biology , Stanford University , Stanford , California 94305 , United States
- Molecular Imaging Program and the Bio-X Program , Stanford University , Stanford , California 94305 , United States
- Electrical Engineering , Stanford University , Stanford , California 94305 , United States
| | - Elliott D SoRelle
- Department of Structural Biology , Stanford University , Stanford , California 94305 , United States
- Molecular Imaging Program and the Bio-X Program , Stanford University , Stanford , California 94305 , United States
- Biophysics Program , Stanford University , Stanford , California 94305 , United States
| | - Yonatan Winetraub
- Department of Structural Biology , Stanford University , Stanford , California 94305 , United States
- Molecular Imaging Program and the Bio-X Program , Stanford University , Stanford , California 94305 , United States
- Biophysics Program , Stanford University , Stanford , California 94305 , United States
| | - Vishnu C Ramani
- Department of Surgery , Stanford University School of Medicine , Stanford , California 94305 , United States
| | - Stefanie S Jeffrey
- Department of Surgery , Stanford University School of Medicine , Stanford , California 94305 , United States
| | - George W Sledge
- Department of Medicine , Stanford University School of Medicine , Stanford , California 94305 , United States
| | - Adam de la Zerda
- Department of Structural Biology , Stanford University , Stanford , California 94305 , United States
- Molecular Imaging Program and the Bio-X Program , Stanford University , Stanford , California 94305 , United States
- Electrical Engineering , Stanford University , Stanford , California 94305 , United States
- Biophysics Program , Stanford University , Stanford , California 94305 , United States
- The Chan Zuckerberg Biohub , San Francisco , California 94158 , United States
| |
Collapse
|
3
|
Automated quantitative image analysis for ex vivo metastasis assays reveals differing lung composition requirements for metastasis suppression by KISS1. Clin Exp Metastasis 2018; 35:77-86. [PMID: 29582202 DOI: 10.1007/s10585-018-9882-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 02/19/2018] [Indexed: 01/09/2023]
Abstract
Imaging is broadly used in biomedical research, but signal variation complicates automated analysis. Using the Pulmonary Metastasis Assay (PuMA) to study metastatic colonization by the metastasis suppressor KISS1, we cultured GFP-expressing melanoma cells in living mouse lung ex vivo for 3 weeks. Epifluorescence images of cells were used to measure growth, creating large datasets which were time consuming and challenging to quantify manually due to scattering of light from outside the focal plane. To address these challenges, we developed an automated workflow to standardize the measurement of disseminated cancer cell growth by applying statistical quality control to remove unanalyzable images followed and a filtering algorithm to quantify only in-focus cells. Using this tool, we demonstrate that expression of the metastasis suppressor KISS1 does not suppress growth of melanoma cells in the PuMA, in contrast to the robust suppression of lung metastasis observed in vivo. This result may suggest that a factor required for metastasis suppression is present in vivo but absent in the PuMA, or that KISS1 suppresses lung metastasis at a step in the metastatic cascade not tested by the PuMA. Together, these data provide a new tool for quantification of metastasis assays and further insight into the mechanism of KISS1 mediated metastasis suppression in the lung.
Collapse
|
4
|
Speak AO, Swiatkowska A, Karp NA, Arends MJ, Adams DJ, van der Weyden L. A high-throughput in vivo screening method in the mouse for identifying regulators of metastatic colonization. Nat Protoc 2017; 12:2465-2477. [PMID: 29095442 DOI: 10.1038/nprot.2017.118] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
We describe a sensitive, robust, high-throughput method for quantifying the ability of metastatic tumor cells to colonize a secondary organ. Metastasis is the leading cause of death in cancer patients, and successful colonization of the secondary organ is the rate-limiting step in the metastatic process; thus, experimental methods that can be used to interrogate the key factors required for this critical step are of great importance. The experimental metastasis assay we detail here includes tail-vein injection of cancer cells into the mouse and determination of the resulting secondary organ colonization, primarily in the lung, 10 d post dosing. This assay can be used to investigate factors that regulate metastatic colonization both at the tumor-cell-intrinsic level (via manipulation of the tumor cells before injection) and at the tumor-cell-extrinsic level (such as the tissue microenvironment, via the use of genetically modified (GM) mice or agents such as antibodies or drugs). Using this method, we have robustly screened more than 950 GM mouse lines to identify novel microenvironmental regulators of metastatic colonization. The experimental details discussed here include choosing of appropriate cell numbers, handling of the cells, selection of recipient animals and injection techniques. Furthermore, we discuss key experimental design considerations, including the choice of the method used to determine metastatic burden and statistical analysis of the results, as well as provide troubleshooting tips and identification of factors that contribute to experimental variability.
Collapse
Affiliation(s)
- Anneliese O Speak
- Experimental Cancer Genetics Group, Wellcome Trust Sanger Institute, Hinxton, UK
| | | | - Natasha A Karp
- Experimental Cancer Genetics Group, Wellcome Trust Sanger Institute, Hinxton, UK
- Quantitative Biology, Innovative Medicines and Early Development (IMED), AstraZeneca, Cambridge, UK
| | - Mark J Arends
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, UK
| | - David J Adams
- Experimental Cancer Genetics Group, Wellcome Trust Sanger Institute, Hinxton, UK
| | | |
Collapse
|
5
|
Amanatullah DF, Tamaresis JS, Chu P, Bachmann MH, Hoang NM, Collyar D, Mayer AT, West RB, Maloney WJ, Contag CH, King BL. Local estrogen axis in the human bone microenvironment regulates estrogen receptor-positive breast cancer cells. Breast Cancer Res 2017; 19:121. [PMID: 29141657 PMCID: PMC5688761 DOI: 10.1186/s13058-017-0910-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 10/16/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Approximately 70% of all breast cancers express the estrogen receptor, and are regulated by estrogen. While the ovaries are the primary source of estrogen in premenopausal women, most breast cancer is diagnosed following menopause, when systemic levels of this hormone decline. Estrogen production from androgen precursors is catalyzed by the aromatase enzyme. Although aromatase expression and local estrogen production in breast adipose tissue have been implicated in the development of primary breast cancer, the source of estrogen involved in the regulation of estrogen receptor-positive (ER+) metastatic breast cancer progression is less clear. METHODS Bone is the most common distant site of breast cancer metastasis, particularly for ER+ breast cancers. We employed a co-culture model using trabecular bone tissues obtained from total hip replacement (THR) surgery specimens to study ER+ and estrogen receptor-negative (ER-) breast cancer cells within the human bone microenvironment. Luciferase-expressing ER+ (MCF-7, T-47D, ZR-75) and ER- (SK-BR-3, MDA-MB-231, MCF-10A) breast cancer cells were cultured directly on bone tissue fragments or in bone tissue-conditioned media, and monitored over time with bioluminescence imaging (BLI). Bone tissue-conditioned media were generated in the presence vs. absence of aromatase inhibitors, and testosterone. Bone tissue fragments were analyzed for aromatase expression by immunohistochemistry. RESULTS ER+ breast cancer cells were preferentially sustained in co-cultures with bone tissues and bone tissue-conditioned media relative to ER- cells. Bone fragments analyzed by immunohistochemistry revealed expression of the aromatase enzyme. Bone tissue-conditioned media generated in the presence of testosterone had increased estrogen levels and heightened capacity to stimulate ER+ breast cancer cell proliferation. Pretreatment of cultured bone tissues with aromatase inhibitors, which inhibited estrogen production, reduced the capacity of conditioned media to stimulate ER+ cell proliferation. CONCLUSIONS These results suggest that a local estrogen signaling axis regulates ER+ breast cancer cell viability and proliferation within the bone metastatic niche, and that aromatase inhibitors modulate this axis. Although endocrine therapies are highly effective in the treatment of ER+ breast cancer, resistance to these treatments reduces their efficacy. Characterization of estrogen signaling networks within the bone microenvironment will identify new strategies for combating metastatic progression and endocrine resistance.
Collapse
Affiliation(s)
- Derek F. Amanatullah
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Pavilion C, 4th Floor, Redwood City, CA 94063-6342 USA
| | - John S. Tamaresis
- Department of Biomedical Data Science, Stanford University School of Medicine, Redwood Building, Room T101F (MC 5405), Stanford, CA 94305 USA
| | - Pauline Chu
- Department of Pathology, Stanford University School of Medicine, Edwards, Room 264, 1291 Welch Road, Stanford, CA 94305-5324 USA
| | - Michael H. Bachmann
- Department of Pediatrics, Stanford University School of Medicine, 150E Clark Center, 318 Campus Drive, Stanford, CA 94305-5427 USA
- Present address: Departments of Biomedical Engineering, and Microbiology & Molecular Genetics, Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Dr, East Lansing, MI 44823 USA
| | - Nhat M. Hoang
- Research IT, Stanford University School of Medicine, 3172 Porter Drive, Palo Alto, CA 94304 USA
| | - Deborah Collyar
- Patient Advocates in Research (PAIR), Danville, CA 94506 USA
| | - Aaron T. Mayer
- Department of Bioengineering, Stanford University School of Medicine, 153E Clark Center, 318 Campus Drive, Stanford, CA 94305 USA
| | - Robert B. West
- Department of Pathology, Stanford University School of Medicine, Edwards, Room 264, 1291 Welch Road, Stanford, CA 94305-5324 USA
| | - William J. Maloney
- Department of Orthopaedic Surgery, Stanford University School of Medicine, 450 Broadway Street, Pavilion C, 4th Floor, Redwood City, CA 94063-6342 USA
| | - Christopher H. Contag
- Department of Pediatrics, Stanford University School of Medicine, 150E Clark Center, 318 Campus Drive, Stanford, CA 94305-5427 USA
- Present address: Departments of Biomedical Engineering, and Microbiology & Molecular Genetics, Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Dr, East Lansing, MI 44823 USA
| | - Bonnie L. King
- Department of Pediatrics, Stanford University School of Medicine, 150E Clark Center, 318 Campus Drive, Stanford, CA 94305-5427 USA
| |
Collapse
|
6
|
Parris GE. Cell-Cell Fusion, Chemotaxis and Metastasis. INTERCELLULAR COMMUNICATION IN CANCER 2015:227-254. [DOI: 10.1007/978-94-017-7380-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
7
|
Beaty BT, Wang Y, Bravo-Cordero JJ, Sharma VP, Miskolci V, Hodgson L, Condeelis J. Talin regulates moesin-NHE-1 recruitment to invadopodia and promotes mammary tumor metastasis. J Cell Biol 2014; 205:737-51. [PMID: 24891603 PMCID: PMC4050723 DOI: 10.1083/jcb.201312046] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 04/28/2014] [Indexed: 02/08/2023] Open
Abstract
Invadopodia are actin-rich protrusions that degrade the extracellular matrix and are required for stromal invasion, intravasation, and metastasis. The role of the focal adhesion protein talin in regulating these structures is not known. Here, we demonstrate that talin is required for invadopodial matrix degradation and three-dimensional extracellular matrix invasion in metastatic breast cancer cells. The sodium/hydrogen exchanger 1 (NHE-1) is linked to the cytoskeleton by ezrin/radixin/moesin family proteins and is known to regulate invadopodium-mediated matrix degradation. We show that the talin C terminus binds directly to the moesin band 4.1 ERM (FERM) domain to recruit a moesin-NHE-1 complex to invadopodia. Silencing talin resulted in a decrease in cytosolic pH at invadopodia and blocked cofilin-dependent actin polymerization, leading to impaired invadopodium stability and matrix degradation. Furthermore, talin is required for mammary tumor cell motility, intravasation, and spontaneous lung metastasis in vivo. Thus, our findings provide a novel understanding of how intracellular pH is regulated and a molecular mechanism by which talin enhances tumor cell invasion and metastasis.
Collapse
Affiliation(s)
- Brian T Beaty
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - Yarong Wang
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - Jose Javier Bravo-Cordero
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - Ved P Sharma
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - Veronika Miskolci
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - Louis Hodgson
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| | - John Condeelis
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461
| |
Collapse
|
8
|
Mehta RS, Liman AD, Passero VA, Liman AK. Lung cancer with gastrointestinal metastasis - review of theories of metastasis with three rare case descriptions. CANCER MICROENVIRONMENT 2013; 6:203-11. [PMID: 23963996 DOI: 10.1007/s12307-013-0135-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 08/08/2013] [Indexed: 12/27/2022]
Abstract
Approximately 1 in 14 men and women during their lifetime will be diagnosed with lung cancer, which is the leading cause of cancer-related mortality in the world. As of January 1, 2008, there were about 373,500 men and women living with lung cancer in the United States. Fewer than 60,000 of these are estimated to be alive by January 2013, reflecting a poor overall 5-year relative survival rate of under 16 %. With metastatic cancer, the overall 5-year survival is meager 4 %. On the other hand, the overall five-year survival is over 50 % when the cancer is still in the localized stage. However, unfortunately, more than half of cases of lung cancer are diagnosed at an advanced stage Howlader et al. (2010). Cancer metastasis, the single most critical prognostic factor, is still poorly understood and a highly complex phenomenon. The most common sites of lung cancer metastasis are the lymph nodes, liver, adrenals, brain and bones. The gastrointestinal (GI) tract is an exceptionally rare site of metastasis; with only a handful of cases reported in the literature Centeno et al. (Lung Cancer, 18: 101-105, 1997); Hirasaki et al. (World J Gastroenterol, 14: 5481-5483, 2008); Carr and Boulos (Br J Surg, 83: 647, 1996); Otera et al. (Eur Respir Rev, 19: 248-252, 2010); Antler et al. (Cancer, 49: 170-172, 1982); Fujiwara et al. (Gen Thorac Cardiovasc Surg, 59: 748-752, 2011); Stinchcombe et al. (J Clin Oncol, 24: 4939-4940, 2006); John et al. (J Postgrad Med, 48: 199-200, 2002); Carroll and Rajesh (Eur J Cardiothorac Surg, 19: 719-720, 2001); Brown et al. (Dis Colon Rectum, 23: 343-345, 1980). We report three cases of non-small cell (squamous cell) lung cancer with GI tract metastasis-two in the colon and one in the jejunum. Then we present a review of literature exploring various theories of metastasis, as an attempt to understand the reason of preferential tumor metastasis.
Collapse
|
9
|
Insights into the regulation of tumor dormancy by angiogenesis in experimental tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 734:37-52. [PMID: 23143974 DOI: 10.1007/978-1-4614-1445-2_3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
While it is well established that an angiogenic switch marks escape from tumor dormancy in xenograft models, the molecular pathways involved in the control of tumor cell proliferation or survival by angiogenesis remain substantially uncharted. We recently demonstrated that signals stemming from angiogenic endothelial cells (EC) regulate the behavior of dormant cancer cells. Specifically, we observed that the Notch ligand Dll4, induced by angiogenic factors in EC, triggers Notch3 activation in neighboring tumor cells and promotes a tumorigenic phenotype. Evidence that Notch signaling is involved in tumor dormancy was further strengthened by the observation that MKP-1 levels-a broadly expressed phosphatase-are controlled by Notch3 by regulation of protein ubiquitination and stability. Notch3 and MKP-1 levels are consistently low in dormant tumors, and this is accompanied by relatively high levels of phosphorylated p38, a canonical MKP-1 target previously associated with maintenance of tumor dormancy. These results elucidate a novel angiogenesis-driven mechanism involving the Notch and MAPK pathways that controls tumor dormancy. More in general, angiogenic EC could form part of the vascular niche, a specialized microenvironment which appears to regulate metastatic outgrowth and future studies are needed to clarify the contribution of EC in the regulation of cancer stem cell behavior in the niche.The notion that EC could communicate signals to tumor cells raises questions about the possibility of achieving tumor dormancy by counteracting angiogenesis. In experimental tumors, anti-VEGF drugs typically prune the newly formed vasculature, thus reducing microvessel density, blood flow, and perfusion. These drugs eventually increase hypoxia and cause tumor necrosis but dormancy is rarely observed. Our group recently reported that anti-VEGF therapy causes a dramatic depletion of glucose and an exhaustion of ATP levels in tumors. Moreover, we found that the central metabolic checkpoint LKB1/AMPK-a cellular sensor of ATP levels that supports cell viability in response to energy stress-is activated by anti-VEGF therapy in experimental tumors and it has a key role in induction of sustained tumor regression. These functional links between activation of the LKB1/AMPK by anti-angiogenic therapy and tumor dormancy suggest a role for metabolism in the regulation of this phenomenon.
Collapse
|
10
|
Chen H, Ko JMY, Wong VCL, Hyytiainen M, Keski-Oja J, Chua D, Nicholls JM, Cheung FMF, Lee AWM, Kwong DLW, Chiu PM, Zabarovsky ER, Tsao SW, Tao Q, Kan R, Chan SHK, Stanbridge EJ, Lung ML. LTBP-2 confers pleiotropic suppression and promotes dormancy in a growth factor permissive microenvironment in nasopharyngeal carcinoma. Cancer Lett 2012; 325:89-98. [PMID: 22743615 DOI: 10.1016/j.canlet.2012.06.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 06/11/2012] [Accepted: 06/13/2012] [Indexed: 01/03/2023]
Abstract
This study identified LTBP-2 as a pleiotropic tumor suppressor in nasopharyngeal carcinoma, which safeguards against critical malignant behaviors of tumor cells. LTBP-2 expression was significantly decreased or lost in up to 100% of NPC cell lines (7/7) and 80% of biopsies (24/30). Promoter hypermethylation was found to be involved in LTBP-2 silencing. Using a tetracycline-regulated inducible expression system, we unveiled functional roles of LTBP-2 in suppressing colony formation, anchorage-independent growth, cell migration, angiogenesis, VEGF secretion, and tumorigenicity. Three-dimensional culture studies suggested the involvement of LTBP-2 in maintenance of tumor cell dormancy in a growth factor favorable microenvironment.
Collapse
Affiliation(s)
- Han Chen
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong Special Administrative Region
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Knopeke MT, Ritschdorff ET, Clark R, Vander Griend DJ, Khan S, Thobe M, Shear JB, Rinker-Schaeffer CW. Building on the foundation of daring hypotheses: using the MKK4 metastasis suppressor to develop models of dormancy and metastatic colonization. FEBS Lett 2011; 585:3159-65. [PMID: 21925502 DOI: 10.1016/j.febslet.2011.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 09/07/2011] [Indexed: 01/04/2023]
Abstract
The identification of a novel metastasis suppressor function for the MAP Kinase Kinase 4 protein established a role for the stress-activated kinases in regulating the growth of disseminated cancer cells. In this review, we describe MKK4's biological mechanism of action and how this information is being used to guide the development of new models to study cancer cell dormancy and metastatic colonization. Specifically, we describe the novel application of microvolume structures, which can be modified to represent characteristics similar to those that cancer cells experience at metastatic sites. Although MKK4 is currently one of many known metastasis suppressors, this field of research started with a single daring hypothesis, which revolutionized our understanding of metastasis, and opened up new areas of exploration for basic research. The combination of our increasing knowledge of metastasis suppressors and such novel technologies provide hope for possible clinical interventions to prevent suffering from the burden of metastatic disease.
Collapse
Affiliation(s)
- Matthew T Knopeke
- The Section of Urology, Department of Surgery, The University of Chicago, Chicago, IL 60637, United States
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Metastatic tumor dormancy in cutaneous melanoma: does surgery induce escape? Cancers (Basel) 2011; 3:730-46. [PMID: 24212638 PMCID: PMC3756387 DOI: 10.3390/cancers3010730] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 01/28/2011] [Accepted: 02/11/2011] [Indexed: 01/05/2023] Open
Abstract
According to the concept of tumor dormancy, tumor cells may exist as single cells or microscopic clusters of cells that are clinically undetectable, but remain viable and have the potential for malignant outgrowth. At metastatic sites, escape from tumor dormancy under more favorable local microenvironmental conditions or through other, yet undefined stimuli, may account for distant recurrence after supposed "cure" following surgical treatment of the primary tumor. The vast majority of evidence to date in support of the concept of tumor dormancy originates from animal studies; however, extensive epidemiologic data from breast cancer strongly suggests that this process does occur in human disease. In this review, we aim to demonstrate that metastatic tumor dormancy does exist in cutaneous melanoma based on evidence from mouse models and clinical observations of late recurrence and occult transmission by organ transplantation. Experimental data underscores the critical role of impaired angiogenesis and immune regulation as major mechanisms for maintenance of tumor dormancy. Finally, we examine evidence for the role of surgery in promoting escape from tumor dormancy at metastatic sites in cutaneous melanoma.
Collapse
|
13
|
Barkan D, Green JE, Chambers AF. Extracellular matrix: a gatekeeper in the transition from dormancy to metastatic growth. Eur J Cancer 2010; 46:1181-8. [PMID: 20304630 PMCID: PMC2856784 DOI: 10.1016/j.ejca.2010.02.027] [Citation(s) in RCA: 277] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 02/16/2010] [Indexed: 12/21/2022]
Abstract
Metastases can develop after apparently successful treatment of a primary tumour, sometimes following a period of tumour dormancy that can last for years. However, factors that regulate metastatic tumour dormancy remain poorly understood. Here we review the potential contribution of interactions between tumour cells and the microenvironment in metastatic sites, in regulating tumour dormancy vs. metastatic growth. We focus particularly on the potential role of the extracellular matrix (ECM) in regulating maintenance and release from dormancy. Tumour cells that fail to properly adhere to the ECM may enter a state of dormancy. The molecular and physical composition of the ECM can be affected by tumour cells themselves, as well as multiple stromal cell types. The roles of integrins, fibronectin, and collagen are discussed, as are factors that can change the ECM. A better understanding of the molecular details of the crosstalk between tumour cells and the ECM in secondary sites, and how these regulate the dormant state, may lead to improved therapeutic strategies to induce or maintain disseminated tumour cells in a dormant state, or alternatively to successfully eradicate dormant cells.
Collapse
Affiliation(s)
- Dalit Barkan
- Department of Biology, Faculty of Sciences, Haifa University, Haifa, Israel.
| | | | | |
Collapse
|
14
|
Abstract
Metastasis--the spread of cancer to distant organs--is responsible for most cancer deaths. Current adjuvant therapy is based on prognostic indicators that stratify patients into defined risk groups. However, some patients believed to have a good prognosis nonetheless develop metastases, in some cases many years after apparently successful treatment of their primary cancer. This period of clinical dormancy leads to many questions about how best to manage patients, including how to better assign risk of late recurrence, how long to monitor patients, and whether some patients will benefit from extended therapy to prevent late recurrences. The development of targeted therapies with fewer side effects is leading to clinical trials aimed at determining the effectiveness of such long-term therapy. However, much remains to be learned about tumor dormancy. Experimental studies are shedding light on biological and molecular mechanisms potentially responsible for tumor dormancy. Emerging research into tumor initiating cells, immunotherapy, and metastasis suppressor genes, may lead to new approaches for targeted antimetastatic therapy to prolong tumor dormancy. An improved understanding of tumor dormancy is needed for better management of patients at risk for late-developing metastases.
Collapse
Affiliation(s)
- Benjamin D Hedley
- Division of Hematology, London Health Sciences Centre, London, Ontario, Canada
| | | |
Collapse
|
15
|
Pozdnyakova O, Hoang MMP, Dresser KA, Mahalingam M. Prognostic Value of E-Cadherin, β-Catenin, CD44v6, and HER2/neu in Metastatic Cutaneous Adenocarcinoma. Arch Pathol Lab Med 2009; 133:1285-90. [PMID: 19653727 DOI: 10.5858/133.8.1285] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2008] [Indexed: 11/06/2022]
Abstract
Abstract
Context.—Our recent experience with a patient developing cutaneous metastases within 3 months of diagnosis of esophageal adenocarcinoma suggests that altered expression of the cellular adhesion molecules, E-cadherin and CD44v6, may have had a role to play in the rapid onset of metastases.
Objective.—To corroborate these findings, we designed a cross-sectional study to investigate the expression of select molecules involved in the metastatic cascade.
Design.—E-cadherin, β-catenin, CD44v6, and HER2/neu immunohistochemical stains were performed on archival materials of metastatic adenocarcinoma to the skin from 27 patients and the available corresponding primary tumors in 10 patients. The primary sites included breast (n = 10; 37%), gastrointestinal tract (n = 10; 37%), ovary (n = 1; 4%), thyroid (n = 2; 7%), lung (n = 1; 4%), and unknown primary (n = 3; 11%).
Results.—Expression of all markers was noted with the most significant increases observed in β-catenin (26 of 27 cases; 96%), followed by CD44v6 (24 of 27 cases; 89%), E-cadherin (22 of 27 cases; 82%), and HER2/neu (11 of 27 cases; 41%). Contrasting expression of these molecules in the primary versus the metastatic tumors, enhanced expression of CD44v6 was observed in the cutaneous metastases relative to the primary in 6 of 10 (60%) cases. Of interest, 2 of these 6 cases (33%) also showed reduction in E-cadherin—a member of the cadherin family functioning as an invasion suppressor molecule.
Conclusions.—These findings reinforce the complexities of the metastatic cascade and imply that the variation in adhesive properties of tumor cells is, perhaps, a consequence of the difference in density of the molecules mediating this process.
Collapse
Affiliation(s)
- Olga Pozdnyakova
- From the Department of Pathology, University of Massachusetts Medical School, Worcester (Dr Pozdnyakova and Ms Dresser); the Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston (Dr Hoang); and the Dermatopathology Section, Department of Dermatology, Boston University School of Medicine, Boston, Massachusetts (Dr Mahalingam)
| | - Mai M. P. Hoang
- From the Department of Pathology, University of Massachusetts Medical School, Worcester (Dr Pozdnyakova and Ms Dresser); the Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston (Dr Hoang); and the Dermatopathology Section, Department of Dermatology, Boston University School of Medicine, Boston, Massachusetts (Dr Mahalingam)
| | - Karen A. Dresser
- From the Department of Pathology, University of Massachusetts Medical School, Worcester (Dr Pozdnyakova and Ms Dresser); the Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston (Dr Hoang); and the Dermatopathology Section, Department of Dermatology, Boston University School of Medicine, Boston, Massachusetts (Dr Mahalingam)
| | - Meera Mahalingam
- From the Department of Pathology, University of Massachusetts Medical School, Worcester (Dr Pozdnyakova and Ms Dresser); the Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston (Dr Hoang); and the Dermatopathology Section, Department of Dermatology, Boston University School of Medicine, Boston, Massachusetts (Dr Mahalingam)
| |
Collapse
|
16
|
Shin R, Welch DR, Mishra VK, Nash KT, Hurst DR, Krishna NR. Nuclear magnetic resonance and circular dichroism study of metastin (Kisspeptin-54) structure in solution. Clin Exp Metastasis 2009; 26:527-33. [PMID: 19308666 PMCID: PMC2756240 DOI: 10.1007/s10585-009-9252-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Accepted: 03/09/2009] [Indexed: 12/12/2022]
Abstract
KISS1 was first discovered as a metastasis suppressor, but also plays crucial roles in the onset of puberty. The KISS1 gene encodes a secreted protein of 145 amino acids that exhibits no sequence similarity with any known proteins. KISS1 protein is proteolytically processed to generate a number of so-called kisspeptins (KP), the most well characterized is known as KP-54 or metastin. KP-54 is carboxy-terminally amidated and binds to and activates the KISS1 receptor (KISS1R). The current studies were undertaken in order to determine structure of KP-54 using nuclear magnetic resonance and circular dichroism. KP-54 is mostly disordered both in water and in trifluoroethanol/water mixed solvent, with no structural motifs. In sodium dodecyl sulfate micelles, KP-54 remains mostly disordered except for a small increase in helical propensity (from 3.7% in water to 9.9% in micelles). Despite this apparent lack of structure, KP-54 is biologically active. The intrinsic disorder of KP-54 may confer advantages in its ability to recognize and bind a wide range of target proteins.
Collapse
Affiliation(s)
- Ronald Shin
- Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Danny R. Welch
- Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA, Departments of Pathology, The University of Alabama at Birmingham, 1670 University Blvd. Room VH-G019, Birmingham, AL 35294-00194, USA , Department of Cell Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA, Department of Pharmacology & Toxicology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA, National Foundation for Cancer Research – Center for Metastasis Research, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Vinod K. Mishra
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kevin T. Nash
- Departments of Pathology, The University of Alabama at Birmingham, 1670 University Blvd. Room VH-G019, Birmingham, AL 35294-00194, USA
| | - Douglas R. Hurst
- Departments of Pathology, The University of Alabama at Birmingham, 1670 University Blvd. Room VH-G019, Birmingham, AL 35294-00194, USA, National Foundation for Cancer Research – Center for Metastasis Research, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - N. Rama Krishna
- Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA, Department of Biochemistry & Molecular Genetics, The University of Alabama at Birmingham, Community Health Services Building B31, Birmingham, AL 35294-2041, USA
| |
Collapse
|
17
|
Favaro E, Amadori A, Indraccolo S. Cellular interactions in the vascular niche: implications in the regulation of tumor dormancy. APMIS 2008; 116:648-59. [PMID: 18834409 DOI: 10.1111/j.1600-0463.2008.01025.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Angiogenesis plays an established role in the promotion of growth of dormant micrometastasis, because blood vessels deliver oxygen and nutrients to the tumor microenvironment. In addition to this feeding function, however, there is accumulating evidence suggesting that endothelial cells-and perhaps other cellular components of the microenvironment--could communicate both positive and negative signals to tumor cells. This cross-talk between heterogeneous cell types could turn out to be important in the regulation of cancer cell behavior. Normal cells recruited during the angiogenic process, or attracted to future sites of metastasis by soluble products released by cancer cells, have been shown to create a niche favorable to tumor cell proliferation and survival. In addition, following an exogenous angiogenic spike, as may occur during inflammation, the same mechanisms could lead to re-activation of poorly angiogenic tumor cells seeded into tissues. In this review, we discuss the possible implications of this hypothesis for our understanding of the phenomenon of tumor dormancy.
Collapse
Affiliation(s)
- Elena Favaro
- Department of Oncology and Surgical Sciences, Oncology Section, University of Padova, Padova, Italy
| | | | | |
Collapse
|
18
|
Hoffman RM. Imaging cancer dynamics in vivo at the tumor and cellular level with fluorescent proteins. Clin Exp Metastasis 2008; 26:345-55. [PMID: 18787963 DOI: 10.1007/s10585-008-9205-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Accepted: 08/03/2008] [Indexed: 02/06/2023]
Abstract
Whole-body imaging with fluorescent proteins has been shown to be a powerful technology to follow the dynamics of metastatic cancer. Whole-body imaging of fluorescent protein-expressing-cancer cells enables the facile determination of efficacy of candidate anti-tumor and anti-metastatic agents in mouse models. GFP-expressing transgenic mice transplanted with the RFP-expressing cancer cells enable the distinction of cancer and host cells and the efficacy of drugs on each type of cell. This is particularly useful for imaging tumor angiogenesis. Cancer-cell trafficking through the cardiovascular and lymphatic systems is the critical means of spread of cancer. The use of fluorescent proteins to differentially label cancer calls in the nucleus and cytoplasm and high-powered imaging technology are used to visualize the nuclear-cytoplasmic dynamics of cancer-cell trafficking in both blood vessels and lymphatic vessels in the live animal. This technology has furthered our understanding of the spread of cancer at the subcellular level in the live mouse. Fluorescent proteins thus enable both macro and micro imaging technology and thereby provide the basis for the new field of in vivo cell biology.
Collapse
Affiliation(s)
- Robert M Hoffman
- AntiCancer, Inc., 7917 Ostrow Street, San Diego, CA 92111-3604, USA.
| |
Collapse
|
19
|
Barkan D, Kleinman H, Simmons JL, Asmussen H, Kamaraju AK, Hoenorhoff MJ, Liu ZY, Costes SV, Cho EH, Lockett S, Khanna C, Chambers AF, Green JE. Inhibition of metastatic outgrowth from single dormant tumor cells by targeting the cytoskeleton. Cancer Res 2008; 68:6241-50. [PMID: 18676848 PMCID: PMC2561279 DOI: 10.1158/0008-5472.can-07-6849] [Citation(s) in RCA: 316] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Metastatic breast cancer may emerge from latent tumor cells that remain dormant at disseminated sites for many years. Identifying mechanisms regulating the switch from dormancy to proliferative metastatic growth has been elusive due to the lack of experimental models of tumor cell dormancy. We characterized the in vitro growth characteristics of cells that exhibit either dormant (D2.0R, MCF-7, and K7M2AS1.46) or proliferative (D2A1, MDA-MB-231, and K7M2) metastatic behavior in vivo. Although these cells proliferate readily in two-dimensional culture, we show that when grown in three-dimensional matrix, distinct growth properties of the cells were revealed that correlate to their dormant or proliferative behavior at metastatic sites in vivo. In three-dimensional culture, cells with dormant behavior in vivo remained cell cycle arrested with elevated nuclear expression of p16 and p27. The transition from quiescence to proliferation of D2A1 cells was dependent on fibronectin production and signaling through integrin beta1, leading to cytoskeletal reorganization with filamentous actin (F-actin) stress fiber formation. We show that phosphorylation of myosin light chain (MLC) by MLC kinase (MLCK) through integrin beta1 is required for actin stress fiber formation and proliferative growth. Inhibition of integrin beta1 or MLCK prevents transition from a quiescent to proliferative state in vitro. Inhibition of MLCK significantly reduces metastatic outgrowth in vivo. These studies show that the switch from dormancy to metastatic growth may be regulated, in part, through epigenetic signaling from the microenvironment, leading to changes in the cytoskeletal architecture of dormant cells. Targeting this process may provide therapeutic strategies for inhibition of the dormant-to-proliferative metastatic switch.
Collapse
Affiliation(s)
- Dalit Barkan
- Laboratory of Cell Biology and Genetics, National Cancer Institute, NIH, Bethesda MD 20892 USA
| | - Hynda Kleinman
- Craniofacial Developmental Biology and Regeneration Branch, National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD USA 20892
| | - Justin L. Simmons
- Laboratory of Cell Biology and Genetics, National Cancer Institute, NIH, Bethesda MD 20892 USA
| | - Holly Asmussen
- Laboratory of Cell Biology and Genetics, National Cancer Institute, NIH, Bethesda MD 20892 USA
| | - Anil K. Kamaraju
- Laboratory of Cell Biology and Genetics, National Cancer Institute, NIH, Bethesda MD 20892 USA
| | - Mark J. Hoenorhoff
- Laboratory of Cell Biology and Genetics, National Cancer Institute, NIH, Bethesda MD 20892 USA
| | - Zi-yao Liu
- Laboratory of Cell Biology and Genetics, National Cancer Institute, NIH, Bethesda MD 20892 USA
| | - Sylvain V. Costes
- Cancer Biology Department, Lawrence Berkeley National Laboratory, Cyclotron Road, Berkeley, CA 94720 USA
| | - Edward H. Cho
- Image Analysis Laboratory, SAIC-Frederick, National Cancer Institute at Fredrick, P.O. Box B, Fredrick, MD 21702 USA
| | - Stephen Lockett
- Image Analysis Laboratory, SAIC-Frederick, National Cancer Institute at Fredrick, P.O. Box B, Fredrick, MD 21702 USA
| | - Chand Khanna
- Pediatric Oncology Branch, National Cancer Institute, NIH Bethesda MD 20892 USA
| | - Ann F. Chambers
- London Regional Cancer Program, 790 Commissioners Road East, London, Ontario, N6A 4L6 Canada
| | - Jeffrey E. Green
- Laboratory of Cell Biology and Genetics, National Cancer Institute, NIH, Bethesda MD 20892 USA
| |
Collapse
|
20
|
Liang S, Dong C. Integrin VLA-4 enhances sialyl-Lewisx/a-negative melanoma adhesion to and extravasation through the endothelium under low flow conditions. Am J Physiol Cell Physiol 2008; 295:C701-7. [PMID: 18632734 DOI: 10.1152/ajpcell.00245.2008] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
During their passage through the circulatory system, tumor cells undergo extensive interactions with various host cells including endothelial cells. The capacity of tumor cells to form metastasis is related to their ability to interact with and extravasate through endothelial cell layers, which involves multiple adhesive interactions between tumor cells and endothelium (EC). Thus it is essential to identify the adhesive receptors on the endothelial and melanoma surface that mediate those specific adhesive interactions. P-selectin and E-selectin have been reported as adhesion molecules that mediate the cell-cell interaction of endothelial cells and melanoma cells. However, not all melanoma cells express ligands for selectins. In this study, we elucidated the molecular constituents involved in the endothelial adhesion and extravasation of sialyl-Lewis(x/a)-negative melanoma cell lines under flow in the presence and absence of polymorphonuclear neutrophils (PMNs). Results show the interactions of alpha(4)beta(1) (VLA-4) on sialyl-Lewis(x/a)-negative melanoma cells and vascular adhesion molecule (VCAM-1) on inflamed EC supported melanoma adhesion to and subsequent extravasation through the EC in low shear flow. These findings provide clear evidence for a direct role of the VLA-4/VCAM-1 pathway in melanoma cell adhesion to and extravasation through the vascular endothelium in a shear flow. PMNs facilitated melanoma cell extravasation under both low and high shear conditions via the involvement of distinct molecular mechanisms. In the low shear regime, beta(2)-integrins were sufficient to enhance melanoma cell extravasation, whereas in the high shear regime, selectin ligands and beta(2)-integrins on PMNs were necessary for facilitating the melanoma extravasation process.
Collapse
Affiliation(s)
- Shile Liang
- Department of Bioengineering, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | | |
Collapse
|
21
|
BRMS1 suppresses breast cancer metastasis in multiple experimental models of metastasis by reducing solitary cell survival and inhibiting growth initiation. Clin Exp Metastasis 2008; 25:727-40. [DOI: 10.1007/s10585-008-9184-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Accepted: 05/16/2008] [Indexed: 10/25/2022]
|
22
|
Single-cell tumor dormancy model of uveal melanoma. Clin Exp Metastasis 2008; 25:509-16. [DOI: 10.1007/s10585-008-9158-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Accepted: 02/26/2008] [Indexed: 10/22/2022]
|
23
|
Phadke PA, Vaidya KS, Nash KT, Hurst DR, Welch DR. BRMS1 suppresses breast cancer experimental metastasis to multiple organs by inhibiting several steps of the metastatic process. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:809-17. [PMID: 18276787 DOI: 10.2353/ajpath.2008.070772] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Breast cancer metastasis suppressor 1 (BRMS1) inhibits formation of macroscopic lung metastases in breast, ovary, and melanoma xenograft models. Because it is unclear which step(s) of the metastatic cascade are affected by BRMS1, the major aim of this study was to determine when and how BRMS1 acts to suppress metastasis. We also examined whether BRMS1 expression globally blocks metastasis or selectively inhibits metastatic outgrowths in specific tissues. Metastatic human breast carcinoma cell lines MDA-MB-231 and -435 expressing enhanced green fluorescent protein (GFP; 231 GFP and 435 GFP) and cell lines transduced with the BRMS1 gene (231 GFP-BRMS1 and 435 GFP-BRMS1) were injected into the left cardiac ventricle to achieve the widest possible cellular distribution, by minimizing first-pass clearance in the lungs. Compared with parental cells, BRMS1-expressing clones formed significantly fewer metastases in all organs tested. When cells were injected directly into the vasculature, fewer of the BRMS1-expressing cells reached lungs or bone compared with parental cells, suggesting that restoration of BRMS1 expression increased cell death during transit. Susceptibility to anoikis was verified in vitro by demonstrating decreased survival on poly-hydroxyethyl methacrylate-coated dishes. Most of the BRMS1-expressing cells reaching secondary sites failed to proliferate, suggesting that BRMS1 also inhibits colonization. Coupled with previous reports showing modest effects of BRMS1 on adhesion and invasion, our results indicate that BRMS1 inhibits metastases in multiple organs by blocking several steps in the metastatic cascade.
Collapse
Affiliation(s)
- Pushkar A Phadke
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | | | | | | | | |
Collapse
|
24
|
Stafford LJ, Vaidya KS, Welch DR. Metastasis suppressors genes in cancer. Int J Biochem Cell Biol 2008; 40:874-91. [DOI: 10.1016/j.biocel.2007.12.016] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2007] [Revised: 12/17/2007] [Accepted: 12/18/2007] [Indexed: 01/31/2023]
|
25
|
Abstract
Metastasis remains the most deadly aspect of cancer and still evades direct treatment. Clinically and experimentally, primary tumor development and metastasis are distinct processes-locally growing tumors can progress without the development of metastases. The discovery of endogenous molecules that exclusively inhibit metastasis suggests that metastasis is an amenable therapeutic target. By definition, metastasis suppressors inhibit metastasis without inhibiting tumorigenicity and are thus distinct from tumor suppressors. As the biology underlying functional mechanisms of metastasis suppressors becomes clearer, it is evident that metastasis suppressors could be harnessed as direct drug targets, prognostic markers, and to understand the fundamental biology of the metastatic process. Metastasis suppressors vary widely in their cellular localization: they are found in every cellular compartment and some are secreted. In general, metastasis suppressors appear to regulate selectively how cells respond to exogenous signals, by affecting signaling cascades which regulate downstream gene expression. This review briefly summarizes current functional and biochemical data on metastasis suppressors implicated in breast cancer. We also present a schematic integrating known mechanisms for these metastasis suppressors highlighting potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Kedar S Vaidya
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
26
|
Abstract
Metastasis is the most lethal attribute of cancer cells and clinical decisions regarding treatment are based largely upon the likelihood of developing metastases. However, improvements in detection as well as recent experimental data have raised questions about the most appropriate definition of a metastasis, especially whether the mere presence of cells at secondary sites constitute a metastatic lesion. After reviewing the experimental basis of metastasis, a definition of metastasis is proffered along with a proposal to consider regarding modification of staging parameters.
Collapse
Affiliation(s)
- Danny R Welch
- Department of Pathology, Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA.
| |
Collapse
|
27
|
Nash KT, Phadke PA, Navenot JM, Hurst DR, Accavitti-Loper MA, Sztul E, Vaidya KS, Frost AR, Kappes JC, Peiper SC, Welch DR. Requirement of KISS1 secretion for multiple organ metastasis suppression and maintenance of tumor dormancy. J Natl Cancer Inst 2007; 99:309-21. [PMID: 17312308 PMCID: PMC1820615 DOI: 10.1093/jnci/djk053] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The KISS1 protein suppresses metastasis of several tumor models without blocking orthotopic tumor growth, but the mechanism remains elusive. For its role in human sexual maturation, KISS1 protein is secreted and processed to kisspeptins, which bind to the G protein-coupled receptor GPR54. We tested the hypothesis that KISS1 secretion is required for metastasis suppression via GPR54. METHODS KISS1 containing an internal FLAG epitope with (KFM) or without (KFMdeltaSS) a signal sequence was transfected into C8161.9 human melanoma cells, which do not express endogenous KISS1. Whole-cell lysates and conditioned medium from C8161.9(KFM) and C8161.9(KFMdeltaSS) cells were collected and analyzed for kisspeptins by immunoprecipitation and enzyme-linked immunosorbent assay. GPR54 levels were measured using real-time reverse transcription-polymerase chain reaction. The ability of conditioned medium from C8161.9(KFM) and C8161.9(KFMdeltaSS) cells to stimulate calcium mobilization in GPR54-expressing Chinese hamster ovary cells (CHO-G) and in C8161.9 cells was evaluated. Metastasis was monitored in athymic mice (groups of 10 per experiment) that were injected with C8161.9(KFM) or C8161.9(KFMdeltaSS) cells labeled with enhanced green fluorescent protein. Survival of mice injected with C8161.9 or C8161.9(KFM) cells was analyzed by Kaplan-Meier methods. RESULTS Full-length KFM and KFMdeltaSS were detected in whole-cell lysates of C8161.9(KFM) and C8161.9(KFMdeltaSS) cells, respectively, but kisspeptins were detected only in conditioned medium of C8161.9(KFM) cells. In vivo, C8161.9(KFM), but not C8161.9(KFMdeltaSS), cells were suppressed for metastasis to lung, eye, kidney, and bone, with corresponding differences in mouse survival (median > 120 versus 42 days). C8161.9(KFM) cells seeded mouse lungs but did not form macroscopic metastases. Conditioned medium from C8161.9(KFM), but not C8161.9(KFMdeltaSS), cells stimulated calcium mobilization in CHO-G cells. GPR54 expression was low in C8161.9 cells, which were not stimulated by conditioned medium from C8161.9(KFM) cells. CONCLUSIONS KISS1 secretion was required for multiple organ metastasis suppression and for maintenance of disseminated cells in a dormant state. The absence of GPR54 expression in C8161.9 cells (whose metastatic spread was suppressed by KFM) suggests that metastasis suppression is not mediated through this receptor. The results imply the existence of another KISS1 receptor and/or paracrine signaling. The findings raise the possibility that soluble KISS1, kisspeptins, or mimetics could be used to maintain tumor dormancy, rendering treatment of already disseminated tumor cells (i.e., micrometastases) a legitimate target.
Collapse
Affiliation(s)
- Kevin T Nash
- Department of Pathology, University of Alabama, Birmingham, AL, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hedley BD, Allan AL, Chambers AF. Tumor dormancy and the role of metastasis suppressor genes in regulating ectopic growth. Future Oncol 2007; 2:627-41. [PMID: 17026454 DOI: 10.2217/14796694.2.5.627] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Metastasis, or tumor growth in an ectopic site, may occur several years after apparently successful treatment of the primary malignancy. Clinical dormancy is seen in a large number of cancer patients, but once growth in an ectopic site initiates, current adjuvant therapies are inadequate and the majority of patients with metastatic disease will die. Many genes may regulate ectopic growth in a secondary site, including a small subset, termed the metastasis suppressor genes. Investigation into this class of genes holds promise in terms of gaining a greater understanding of tumor dormancy and how the process of metastasis may be naturally inhibited. This review will focus on the role of metastasis suppressor genes in tumor dormancy. Insights into the metastatic process from studies of metastasis suppressor genes may lead to novel targets for antimetastatic therapy through drug-induced reactivation of one or more of these genes and/or their respective signaling pathways.
Collapse
Affiliation(s)
- Benjamin D Hedley
- University of Western Ontario, Schulich School of Medicine & Dentistry, London Regional Cancer Program, Department of Oncology, London, Ontario, Canada.
| | | | | |
Collapse
|
29
|
Affiliation(s)
- Patricia S Steeg
- Women's Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute Building 37, Room 1122, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
30
|
Suzuki M, Mose ES, Montel V, Tarin D. Dormant cancer cells retrieved from metastasis-free organs regain tumorigenic and metastatic potency. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:673-81. [PMID: 16877365 PMCID: PMC1698784 DOI: 10.2353/ajpath.2006.060053] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
This study shows that solitary, dormant human cancer cells, retrieved from metastasis-free organs of animals carrying spontaneously metastatic primary tumors, can reactivate their tumorigenic and metastatic potency. The tumors were produced by MDA-MB-435 CL16 breast cancer cells permanently labeled with green fluorescent protein and the neomycin resistance gene. This enabled unequivocal identification of tumor cells emerging from organ explants cultured in neomycin to eliminate nonneoplastic host cells. Rescued cells resumed proliferation and generated lines that were tumorigenic and metastatic in fresh animals. All resulting primary and secondary tumors were uniformly labeled. Cells recovered from bone marrows and spleens, where there were no metastases, were as tumorigenic and metastatic as cells recovered from lungs and lymph nodes, which are the preferred sites of colonization for this tumor line. This evidence that malignant growth of disseminated cancer cells is suspended indefinitely by microenvironmental conditions in metastasis-free organs, although it is still active in others of the same host, shows that neoplastic progression can be arrested and has far-reaching biological and clinical implications. Specifically, it predicts the existence of natural, nonimmune host mechanisms that stimulate or inactivate tumor growth in different anatomical sites, which may be exploitable for therapeutic benefit.
Collapse
Affiliation(s)
- Mika Suzuki
- Rebecca and John Moores Comprehensive Cancer Center and Department of Pathology, University of California, San Diego, 3855 Health Sciences Dr. MC0803, La Jolla, CA 92093-0803, USA
| | | | | | | |
Collapse
|
31
|
Lugassy C, Vernon SE, Busam K, Engbring JA, Welch DR, Poulos EG, Kleinman HK, Barnhill RL. Angiotropism of Human Melanoma: Studies Involving In Transit and Other Cutaneous Metastases and the Chicken Chorioallantoic Membrane. Am J Dermatopathol 2006; 28:187-93. [PMID: 16778482 PMCID: PMC1524855 DOI: 10.1097/00000372-200606000-00001] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Melanoma cell migration along the outside of vessels has been termed "extravascular migratory metastasis" (EVMM), as distinct from intravascular dissemination. Previous studies in both human and experimental melanoma models have shown angiotropism of melanoma cells, suggesting EVMM. Our objectives are to study the mechanism of dissemination of human melanoma cells in the chick chorioallantoic membrane (CAM) and to compare the histopathology in the CAM with that of patients with in transit and other cutaneous melanoma metastases. Human and murine melanoma cells were inoculated onto the CAM and observed over a 10-day period for tumor dissemination. Both human melanoma specimens from 26 patients and melanoma cells growing on the CAM showed the presence of tumor cell angiotropism at the invasive front of the tumor and at some distance from the tumor mass. In addition, a clear progression of melanoma cells spreading on the CAM was observed along the abluminal surface of vessels, where they occupied a perivascular location. By day 10 after injection, small micrometastases had developed along vessels, in a pattern similar to that in transit and other cutaneous melanoma metastases. In addition, the results suggested that the number of micrometastases directly correlated with increasing tumor volume. Taken together, these data suggest that the CAM is a relevant model for studying tumor cell dissemination, and that EVMM may be a mechanism by which some melanoma cells spread to nearby and even distant sites.
Collapse
Affiliation(s)
- Claire Lugassy
- Department of Patholog University of Miami School of Medicine/Jackson Memorial Hospital, Miami, FL
| | - Stephen E. Vernon
- Department of Patholog University of Miami School of Medicine/Jackson Memorial Hospital, Miami, FL
| | - Klaus Busam
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
| | - Jean A. Engbring
- National Institute of Dental and Craniofacial Research (HKK, JAE) National Institutes of Health, Bethesda, MD
| | - Danny R. Welch
- Department of Pathology and Comprehensive Cancer Center (DRW), University of Alabama, Birmingham, AL
| | - Evangelos G. Poulos
- Department z Dermatology, University of Miami School of Medicine/Jackson Memorial Hospital, Miami, FL
| | - Hynda K. Kleinman
- National Institute of Dental and Craniofacial Research (HKK, JAE) National Institutes of Health, Bethesda, MD
| | - Raymond L. Barnhill
- Department of Patholog University of Miami School of Medicine/Jackson Memorial Hospital, Miami, FL
- Department z Dermatology, University of Miami School of Medicine/Jackson Memorial Hospital, Miami, FL
| |
Collapse
|
32
|
Lee HJ, Lee EK, Lee KJ, Hong SW, Yoon Y, Kim JS. Ectopic expression of neutrophil gelatinase-associated lipocalin suppresses the invasion and liver metastasis of colon cancer cells. Int J Cancer 2006; 118:2490-7. [PMID: 16381001 DOI: 10.1002/ijc.21657] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL), also known as lipocalin 2, is a 25-kDa lipocalin initially purified from neutrophil granules. It is thought to play a role in regulating cellular growth since its expression is highly upregulated in a variety of proliferative cells such as cancer cells. However, experimental evidence showing a clear causal relationship between NGAL expression and the proliferation of tumor cells is lacking. Here, we found NGAL expression in highly and poorly metastatic colon cancer cell lines of the same genetic origin correlated inversely with the metastatic potential of these cells, which suggests NGAL participates in the metastatic process. To explore the role NGAL plays in tumor growth and metastasis, the KM12SM human colon cancer cell line, which is highly metastatic while showing decreased NGAL expression, was genetically manipulated to overexpress NGAL. The effects of this on tumor growth and liver metastasis were then analyzed using experimental animal models established by injecting BALB/c nude mice with tumor cells subcutaneously or intrasplenically. Ectopic expression of NGAL in the colon cancer cells had little effect on the growth and viability of the tumor cells both in vitro and in vivo. However, NGAL expression not only suppressed the ability of the colon carcinoma cells to invade Matrigel in vitro, it also substantially inhibited liver metastasis in an experimental animal model. Collectively, these results indicate that NGAL may be a candidate metastasis suppressor in colon cancer cells.
Collapse
Affiliation(s)
- Ho-Jeong Lee
- New Biologics Team, Mogam Biotechnology Research Institute, Yongin-city, Republic of Korea
| | | | | | | | | | | |
Collapse
|
33
|
Phadke PA, Mercer RR, Harms JF, Jia Y, Frost AR, Jewell JL, Bussard KM, Nelson S, Moore C, Kappes JC, Gay CV, Mastro AM, Welch DR. Kinetics of metastatic breast cancer cell trafficking in bone. Clin Cancer Res 2006; 12:1431-40. [PMID: 16533765 PMCID: PMC1523260 DOI: 10.1158/1078-0432.ccr-05-1806] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE In vivo studies have focused on the latter stages of the bone metastatic process (osteolysis), whereas little is known about earlier events, e.g., arrival, localization, and initial colonization. Defining these initial steps may potentially identify the critical points susceptible to therapeutic intervention. EXPERIMENTAL DESIGN MDA-MB-435 human breast cancer cells engineered with green fluorescent protein were injected into the cardiac left ventricle of athymic mice. Femurs were analyzed by fluorescence microscopy, immunohistochemistry, real-time PCR, flow cytometry, and histomorphometry at times ranging from 1 hour to 6 weeks. RESULTS Single cells were found in distal metaphyses at 1 hour postinjection and remained as single cells up to 72 hours. Diaphyseal arrest occurred rarely and few cells remained there after 24 hours. At 1 week, numerous foci (2-10 cells) were observed, mostly adjacent to osteoblast-like cells. By 2 weeks, fewer but larger foci (> or =50 cells) were seen. Most bones had a single large mass at 4 weeks (originating from a colony or coalescing foci) which extended into the diaphysis by 4 to 6 weeks. Little change (<20%) in osteoblast or osteoclast numbers was observed at 2 weeks, but at 4 to 6 weeks, osteoblasts were dramatically reduced (8% of control), whereas osteoclasts were reduced modestly (to approximately 60% of control). CONCLUSIONS Early arrest in metaphysis and minimal retention in diaphysis highlight the importance of the local milieu in determining metastatic potential. These results extend the Seed and Soil hypothesis by demonstrating both intertissue and intratissue differences governing metastatic location. Ours is the first in vivo evidence that tumor cells influence not only osteoclasts, as widely believed, but also eliminate functional osteoblasts, thereby restructuring the bone microenvironment to favor osteolysis. The data may also explain why patients receiving bisphosphonates fail to heal bone despite inhibiting resorption, implying that concurrent strategies that restore osteoblast function are needed to effectively treat osteolytic bone metastases.
Collapse
Affiliation(s)
| | - Robyn R. Mercer
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | | | | | - Andra R. Frost
- Departments of Pathology
- Comprehensive Cancer Center
- NFCR-Center for Metastasis Research, University of Alabama at Birmingham, Birmingham, AL and
| | - Jennifer L. Jewell
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | - Karen M. Bussard
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | - Shakira Nelson
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | | | | | - Carol V. Gay
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | - Andrea M. Mastro
- NFCR-Center for Metastasis Research, University of Alabama at Birmingham, Birmingham, AL and
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA
| | - Danny R. Welch
- Departments of Pathology
- Comprehensive Cancer Center
- Center for Metabolic Bone Disease
- NFCR-Center for Metastasis Research, University of Alabama at Birmingham, Birmingham, AL and
- Requests for reprints: Danny R. Welch, Ph.D. Department of Pathology, University of Alabama at Birmingham; 1670 University Blvd.; Volker Hall G-019A; Birmingham, AL 35294-0019; Phone: +1-205-934-2961 Fax: +1-205-975-1126; ; or, Andrea M. Mastro, Ph.D., Department of Biochemistry and Molecular Biology, 231 South Frear, Pennsylvania State University, University Park, PA 16802; Phone: +1-814-863-0152; Fax: +1-814-863-7024;
| |
Collapse
|
34
|
Jiang Y, Berk M, Singh LS, Tan H, Yin L, Powell CT, Xu Y. KiSS1 suppresses metastasis in human ovarian cancer via inhibition of protein kinase C alpha. Clin Exp Metastasis 2006; 22:369-76. [PMID: 16283480 DOI: 10.1007/s10585-005-8186-4] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2005] [Accepted: 05/31/2005] [Indexed: 01/02/2023]
Abstract
Metastasis is a vital target for cancer treatment, since the majority of cancer patients die from metastatic, rather than the primary disease. KiSS1 has been identified as a metastasis suppressor gene in melanoma and breast carcinomas. We show here that KiSS1 is also a metastasis suppressor in human ovarian cancer. Overexpression of KiSS1 in ovarian cancer cells inhibits cell migration induced by serum or lysophosphatidic acid (LPA), and colonization in soft agar, but not cell proliferation, representing the characteristics of a metastasis suppressor gene. Furthermore, using an experimental metastatic mouse model, we show that expression of KiSS1 in SKOV3 ovarian cancer cells suppresses >50% metastatic colonization in mice (P < 0.0001). We find that activating protein kinase C (PKC) reverses about 80% of the inhibited cell migration induced by KiSS1, while down-regulation of PKCalpha with shRNA restores KiSS1 effect, providing evidence that inhibiting PKCalpha may be an important mechanism of the effect of KiSS1. These results suggest that KiSS1 is a metastasis suppressor of ovarian cancer and may be a potential molecular target for the treatment.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Cancer Biology, The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Nash KT, Welch DR. The KISS1 metastasis suppressor: mechanistic insights and clinical utility. FRONT BIOSCI-LANDMRK 2006; 11:647-59. [PMID: 16146758 PMCID: PMC1343480 DOI: 10.2741/1824] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Melanoma is a highly metastatic cancer that accounts for the majority of skin cancer deaths. Unfortunately, very few improvements have been made during the last 20 years in the management of melanoma metastases, which is the major cause of melanoma deaths. Therefore, identification of molecular targets that can be exploited in the clinic to treat metastatic disease is desperately needed. The KISS1 metastasis suppressor gene has emerged as a promising molecular target for the management of metastatic disease. This review compiles data regarding the molecular and biochemical properties of KISS1 and its cognate receptor, focusing on the properties believed to be most pertinent to the use of KISS1 in the clinical setting. In addition, clinical data that supports KISS1 as having a dual role as a prognostic indicator and a therapeutic target for the management of metastatic disease will be highlighted.
Collapse
Affiliation(s)
| | - Danny R. Welch
- Send correspondence to: Dr. Danny R. Welch, Department for Pathology, University of Alabama at Birmingham, 1670 University Blvd., VH-G019A, Birmingham, Al 35294-0019, Tel: 205-934-2956, Fax: 205-975-1126, E-mail:
| |
Collapse
|
36
|
Abstract
Naturally fluorescent proteins have revolutionized biology by enabling what was formerly invisible to be seen clearly. These proteins have allowed us to visualize, in real time, important aspects of cancer in living animals, including tumour cell mobility, invasion, metastasis and angiogenesis. These multicoloured proteins have allowed the colour-coding of cancer cells growing in vivo and enabled the distinction of host from tumour with single-cell resolution. Visualization of many aspects of cancer initiation and progression in vivo should be possible with fluorescent proteins.
Collapse
Affiliation(s)
- Robert M Hoffman
- AntiCancer Inc. and Department of Surgery, University of California, San Diego, 7917 Ostrow Street, San Diego, California 92111, USA.
| |
Collapse
|
37
|
Abstract
This chapter describes a new cell biology where the behavior of individual cells can be visualized in the living animal. Previously it has been demonstrated that fluorescent proteins can be used for whole-body imaging of metastatic tumor growth, bacterial infection, and gene expression. An example of the new cell biology is dual-color fluorescence imaging using red fluorescent protein (RFP)-expressing tumors transplanted in green fluorescent protein (GFP)-expressing transgenic mice. These models show with great clarity the details of tumor-stroma interactions and especially tumor-induced angiogenesis, tumor-infiltrating lymphocytes, stromal fibroblasts, and macrophages. Another example is the color coding of cells with RFP or GFP such that both cell types can be simultaneously visualized in vivo. Stem cells can also be visualized and tracked in vivo. Mice in which the regulatory elements of the stem cell marker nestin drive GFP expression enable nascent vasculature to be visualized interacting with transplanted RFP-expressing cancer cells. Nestin-driven GFP expression can also be used to visualize hair follicle stem cells. Dual-color cells expressing GFP in the nucleus and RFP in the cytoplasm enable real-time visualization of nuclear-cytoplasm dynamics including cell cycle events and apoptosis. Highly elongated cancer cells in capillaries in living mice were observed within skin flaps. The migration velocities of the cancer cells in the capillaries were measured by capturing images of the dual-color fluorescent cells over time. The cells in the capillaries elongated to fit the width of these vessels. The use of the dual-color cancer cells differentially labeled in the cytoplasm and nucleus and associated fluorescent imaging provide a powerful tool to understand the mechanism of cancer cell migration and deformation in small vessels.
Collapse
|
38
|
Berger JC, Vander Griend D, Stadler WM, Rinker-Schaeffer C. Metastasis suppressor genes: signal transduction, cross-talk and the potential for modulating the behavior of metastatic cells. Anticancer Drugs 2004; 15:559-68. [PMID: 15205597 DOI: 10.1097/01.cad.0000132233.36512.fa] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In the past decade, research from various disciplines has stimulated a re-evaluation of our ideas of how cancers metastasize. Two important findings have been fundamental to this re-evaluation: that cancer cells are subject to growth regulation at the secondary site and that a specific class of proteins suppresses the metastatic phenotype. These proteins are encoded by metastasis suppressor genes, which are operationally defined as genes that suppress in vivo metastasis without inhibiting primary tumor growth when transfected into metastatic cell lines and injected into experimental animals. Recent biochemical studies have shown that certain metastasis suppressor proteins participate in highly conserved signal transduction cascades that mediate cellular responses to growth factors, cytokines and cellular stresses. Further elucidation of the biochemical foundations of these pathways coupled with strong in vivo studies should give us insight into the mechanisms of cancer metastasis, and may hold important implications for the future of cancer staging and therapy, using both existing and novel modalities.
Collapse
Affiliation(s)
- Jonathan C Berger
- Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | | | | | | |
Collapse
|
39
|
Lugassy C, Kleinman HK, Engbring JA, Welch DR, Harms JF, Rufner R, Ghanem G, Patierno SR, Barnhill RL. Pericyte-like location of GFP-tagged melanoma cells: ex vivo and in vivo studies of extravascular migratory metastasis. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:1191-8. [PMID: 15039208 PMCID: PMC1615331 DOI: 10.1016/s0002-9440(10)63207-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Previous studies have demonstrated that some tumor cells occupy a pericyte-like location in melanoma, forming angio-tumoral complexes. We hypothesized that these tumor cells are migrating along the abluminal surface of the endothelium, a mechanism termed "extravascular migratory metastasis." In the present study, we have used human and murine melanoma cells that stably express enhanced green fluorescence protein (GFP) to examine, in an ex vivo co-culture model, melanoma cell interactions with vessels that have sprouted from rat aortic rings. We also used in vivo tumor growth on the chick chorioallantoic membrane (CAM) to observe the dissemination pathway of melanoma cells. In the ex vivo rat aorta system, we observed a pericyte-like location of tumor cells that were spreading along the vascular channels. For examination of the CAM in vivo, we have used the Lugassy preparation, allowing one to obtain striking images of the relationship between fluorescent GFP cells and microvessels. Melanoma cells were found cuffing the outside of vessels around the tumor. Tumor cells were observed along the vessels several centimeters from the tumor. Confocal microscopy and histopathology confirmed the pericyte-like location of tumor cells, without any observable intravasation. The results indicate that melanoma cells can migrate along the abluminal surface of vessels. This study also demonstrates that these models can provide quantitation analysis that may prove useful in elucidating the molecular interactions involved in extravascular migratory metastasis.
Collapse
Affiliation(s)
- Claire Lugassy
- Department of Dermatology, The George Washington University Medical Center, Washington, District of Columbia, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Affiliation(s)
- Patricia S Steeg
- Women's Cancers Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
41
|
Affiliation(s)
- Victoria L Robinson
- Section of Urology, Department of Surgery, The University of Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
42
|
Jaeger EB, Chekmareva MA, Tennant TR, Luu HH, Hickson JA, Chen SL, Samant RS, Sokoloff MH, Rinker-Schaeffer CW. Inhibition of prostate cancer metastatic colonization by approximately 4.2 Mb of human chromosome 12. Int J Cancer 2003; 108:15-22. [PMID: 14618610 DOI: 10.1002/ijc.11483] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Our previous studies demonstrate that introduction of a approximately 70 cM region (now estimated at 63.75 Mb by the Human Genome Project) of human chromosome 12 into the highly metastatic Dunning rat prostate cancer cell line AT6.1 results in >30-fold (>/=90%) reduction in the number of overt metastases in spontaneous metastasis assays. We report the further localization and biological characterization of the metastasis-suppressor activity encoded by a reduced region of chromosome 12. To localize this metastasis-suppressor activity, a panel of AT6.1 microcell hybrids that retain varying portions of human chromosome 12 was constructed and subjected to sequence-tagged site (STS)-based PCR analysis and assessment of in vivo metastatic ability. Data from these complementary approaches localized the metastasis-suppressor activity to a approximately 4.2 Mb portion of human chromosome 12q24.3 comprised of 3 separate regions. Reverse transcriptase-polymerase chain reaction (RT-PCR) and immunoblotting were used for differential expression analyses to identify which characterized genes, predicted gene sequences and expressed sequence tags (ESTs) within this region could be responsible for the observed metastasis suppression. Comprehensive in vivo studies showed that suppressed AT6.1-12 hybrids that retain the metastasis-suppressor region on 12q24.3 are capable of arriving at the secondary site, but are not able to persist there. Thus, unlike other metastasis-suppressor genes characterized to date, the metastasis-suppressor gene encoded by this region appears to utilize a different biologic mechanism to suppress the growth of overt metastases at the secondary site.
Collapse
Affiliation(s)
- Erich B Jaeger
- Section of Urology, Department of Surgery, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Welch DR. Microarrays bring new insights into understanding of breast cancer metastasis to bone. Breast Cancer Res 2003; 6:61-4. [PMID: 14979907 PMCID: PMC400646 DOI: 10.1186/bcr736] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Using a microarray approach, Kang and colleagues identified several genes involved in the generation of breast cancer metastasis in bone and demonstrated their roles in bone colonization in vivo. Their findings and interpretations are reviewed in the context of recent array studies that compared gene expression in primary tumors and metastases. RNA expression array results have already demonstrated value in predicting whether metastases will develop in patients. They have also shown that expression patterns are similar in primary tumors and metastases. The latter data have invited re-examination of long-held notions related to mechanisms of metastasis. While the arrays show promise for improving diagnostic capability in breast cancers, ascribing mechanistic interpretations to correlative data should be done with extreme caution. Kang and colleagues' paper in Cancer Cell elegantly reinforces the concepts that efficiency of the metastatic process is dependent on the coordinated expression of multiple genes and that the expression of metastasis-associated genes is sometimes dependent on the microenvironment in which cells find themselves.
Collapse
Affiliation(s)
- Danny R Welch
- Department of Pathology, Comprehensive Cancer Center, National Foundation for Cancer Research Center for Metastasis Research, University of Alabama at Birmingham, USA.
| |
Collapse
|
44
|
Abstract
Breast cancer metastasizes to bone with high frequency and incidence. However, studies of breast cancer metastasis to bone have been limited by two factors. First, the number of models that colonize bone are limited. Second, detection of bone metastases is too insensitive or too laborious for routine, large-scale studies or for studying the earliest steps in bone colonization. To partially alleviate these problems, the highly metastatic MDA-MB-435 (435) human breast carcinoma cell line was engineered to constitutively express enhanced green fluorescent protein (GFP). While 435GFP cells did not form femoral metastases following orthotopic or intravenous injections, they produced widespread osteolytic skeletal metastases following injection into the left ventricle of the heart. All mice developed at least one femur metastasis as well as a mandibular metastasis. As in humans, osseous metastases localized predominantly to trabecular regions, especially proximal and distal femur, proximal tibia, proximal humerus and lumbar vertebrae. 435GFP cells also developed metastases in adrenal glands, brain and ovary following intracardiac injection, suggesting that this model may also be useful for studying organotropism to other tissues as well. Additionally, GFP-tagging permitted detection of single cells and microscopic metastases in bone at early time points following arrival and at stages of proliferation prior to coalescence of individual metastases.
Collapse
Affiliation(s)
- John F Harms
- Jake Gittlen Cancer Research Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | | |
Collapse
|
45
|
Abstract
A greater understanding of the processes of tumor invasion and metastasis, the principal cause of death in cancer patients, is essential to determine newer therapeutic targets. Metastasis suppressor genes, by definition, suppress metastasis without affecting tumorigenicity and, hence, present attractive targets as prognostic or therapeutic markers. This short review focuses on those twelve metastasis suppressor genes for which functional data exist. We also outline newly identified genes that bear promising traits of having metastasis suppressor activity, but for which functional data have not been completed. We also summarize the biochemical mechanism(s) of action (where known), and present a working model assembling potential metastasis suppression pathways.
Collapse
Affiliation(s)
- Lalita A Shevde
- Department of Pathology, 1670 University Boulevard, Volker Hall-G-038, The University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | | |
Collapse
|
46
|
Gross SC, Goodarzi G, Watabe M, Bandyopadhyay S, Pai SK, Watabe K. Antineoplastic activity of Solidago virgaurea on prostatic tumor cells in an SCID mouse model. Nutr Cancer 2003; 43:76-81. [PMID: 12467138 DOI: 10.1207/s15327914nc431_9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Solidago virgaurea (goldenrod) has traditionally been used as an anti-inflammatory herbal medicine for the treatment of various symptoms, including prostatic diseases. The plant has also been reported to have antibacterial, spasmolytic, and carminative properties. During the course of our screening for antineoplastic activities in various herbal plants, we found that the extract of S. virgaurea exhibits strong cytotoxic activities on various tumor cell lines. The active component mostly resides in the leaves of the plant and is soluble in water. When the extract was fractionated by a Sephadex G-100 column, the active fraction corresponded to a molecular weight of approximately 40,000. This cytotoxic activity is effective on various tumor cell lines, including human prostate (PC3), breast (MDA435), melanoma (C8161), and small cell lung carcinoma (H520). To examine the effect of the cytotoxic activity on tumor cells in vivo, we used the rat prostate cell line (AT6.1) and an SCID mouse model. AT6.1 cells were injected into the flank of SCID mice, and then the G-100 fraction of S. virgaurea was administered intraperitoneally or subcutaneously every 3 days. The size of the tumor was measured for up to 25 days. The growth of the tumor was significantly suppressed by the G-100 fraction at 5 mg/kg without any apparent side effects. Therefore, S. virgaurea is considered to be promising as an antineoplastic medicine with minimal toxicities.
Collapse
Affiliation(s)
- Steven C Gross
- Department of Medical Microbiology and Immunology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | | | | | | | | | | |
Collapse
|
47
|
Steeg PS, Ouatas T, Halverson D, Palmieri D, Salerno M. Metastasis suppressor genes: basic biology and potential clinical use. Clin Breast Cancer 2003; 4:51-62. [PMID: 12744759 DOI: 10.3816/cbc.2003.n.012] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Metastatic disease remains a significant contributor to morbidity and mortality in patients with breast cancer. An improved molecular and biochemical understanding of the metastatic process is expected to fuel the development of new therapeutic approaches. The suppression of tumor metastasis, despite tumor cell expression of oncogenes and metastasis-promoting events, has become a diverse and fruitful field of investigation. Although many genetic events promote metastasis, several genes show relatively reduced expression levels in metastatic tumor cells in mouse model systems and in aggressive human tumors. Re-expression of a metastasis-suppressor gene in a metastatic tumor cell line results in a significant reduction in metastatic behavior in vivo with no effect on tumorigenicity. The known metastasis-suppressor gene products nm23, KAI1, mitogen-activated protein kinase kinase 4, breast cancer metastasis suppressor-1, KiSS1, RHOGDI2, CRSP3, and vitamin D3-upregulated protein/thioredoxin interacting protein exhibit unexpected biochemical functions that have shed new light on signaling events that are important in metastasis. Most metastasis suppressors function at the translationally important stage of outgrowth of micrometastatic tumor cells at a distant site. We hypothesize that elevation of metastasis suppressor gene expression in micrometastatic tumor cells in the adjuvant high-risk population of patients with breast cancer will halt metastatic colonization and have a clinical benefit. DNA methylation inhibitors have shown limited promise in increasing metastasis-suppressor gene expression, and ligands of the nuclear hormone receptor family are currently under investigation in vitro and in vivo. Clinical testing of agents that increase metastasis-suppressor gene expression is expected to require tailored trial designs.
Collapse
Affiliation(s)
- Patricia S Steeg
- Women's Cancers Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA.
| | | | | | | | | |
Collapse
|
48
|
Abstract
Metastatic disease is the most critical impediment to cancer patient survival. However, comparatively little is known concerning the intricate pathways which govern the complex phenotypes associated with metastasis. The KISS1 metastasis suppressor gene inhibits metastasis in both in vivo melanoma and breast carcinoma models. Despite its clear physiological activity, the mechanism of KISS1 remains unclear. Recent identification of a 54 amino acid peptide of KISS1, termed metastin or kisspeptin-54, and its cognate G-protein coupled receptor (hOT7T175, AXOR12, GPR54) have provided additional clues and avenues of research. While studies have attributed KISS1 with modulation of NFkappaB regulation, experiments with metastin and its receptor implicate MAP kinase pathways and also suggest the potential of autocrine, paracrine and endocrine roles. Impacts on motility, chemotaxis, adhesion and invasion have each been documented in disparate cell lines and conflicting observations require resolution. Nevertheless, mounting clinical evidence, particularly the loss of KISS1 in metastases, correlates KISS1 and metastin receptor expression with human tumor progression. Together, the data substantiate roles for these molecules in metastasis regulation.
Collapse
Affiliation(s)
- John F Harms
- Jake Gittlen Cancer Research Institute, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | | | | |
Collapse
|
49
|
Kauffman EC, Robinson VL, Stadler WM, Sokoloff MH, Rinker-Schaeffer CW. Metastasis suppression: the evolving role of metastasis suppressor genes for regulating cancer cell growth at the secondary site. J Urol 2003; 169:1122-33. [PMID: 12576866 DOI: 10.1097/01.ju.0000051580.89109.4b] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE The prevention and treatment of prostate cancer metastasis continue to provide a significant clinical challenge. Identification of the rate limiting steps of metastasis and their underlying molecular mechanisms may lead to new therapeutic targets and also allow more accurate risk stratification for clinical metastases. We review the literature supporting growth of disseminated tumor cells at the secondary site as a key rate limiting step in metastasis. We also reviewed the definition, identification and characterization of metastasis suppressor genes, and discuss their evolving role in regulating this step. MATERIALS AND METHODS We performed MEDLINE searches and manual bibliographic reviews on the specific steps of metastasis, including growth at the secondary site. In addition, we performed a comprehensive literature review to identify genes fitting the classic definition of a metastasis suppressor gene. The literature was also searched to assess the status of each gene in clinical cancer and evaluate functional support for the potential involvement of each gene in regulating growth at the secondary site. RESULTS Clinical studies in prostate cancer and other cancer types suggest that dissemination to the secondary site is often an early clinical event. However, not all patients with tumor cells at the secondary site have overt metastatic lesions even in the absence of therapy, suggesting that growth at the secondary site may be highly inefficient. Complimentary approaches have allowed researchers to document and quantify the inefficiency of cancer cell growth at the secondary site. Regarding the mechanism of growth control, many studies support a role for the interaction of a cancer cell and the microenvironment at the secondary site influencing whether growth into metastasis may occur. The 7 genes that suppress metastasis without affecting primary tumor growth that have been identified are KAI1, CD44, mitogen activated protein kinase (MAPK) kinase 4, nm23-H1, nm23-H2, KiSS1 and BrMS1. Three of these genes (KAI1, CD44 and MAPK kinase 4) act as metastasis suppressor genes of prostate cancer, while the remainder have yet to be tested in this cancer type. Loss of expression has been demonstrated for most of these genes during the clinical progression of prostate cancer to metastasis. MAPK kinase 4 and KiSS1 appear to suppress metastasis by inhibiting cancer cell growth at the secondary site. Interestingly many metastasis suppressor genes have common roles in growth control, adhesion and cytoskeletal reorganization, suggesting a common mechanism of metastasis suppression. Proposed candidate pathways include signaling through Src kinase and Rac GTPase. CONCLUSIONS The findings discussed support growth at the secondary site as a clinical target for metastasis treatment and prevention. Metastasis suppressor genes may offer valuable mechanistic insight for guiding specific therapeutic strategies, which may include drug induced reactivation of metastasis suppressor genes and their signaling pathways. Clinical assessment of metastasis suppressor gene product status in disseminated cancer cells may improve the accuracy of predicting the prognosis in patients with clinically localized disease.
Collapse
Affiliation(s)
- Eric C Kauffman
- Section of Urology, Department of Surgery, University of Chicago Comprehensive Cancer Research Center, Illinois 60637, USA
| | | | | | | | | |
Collapse
|
50
|
Abstract
The central role of sequential accumulation of genetic alterations during the development of cancer has been firmly established since the pioneering cytogenetic studies successfully defined recurrent chromosome changes in specific types of tumor. In the course of carcinogenesis, cells experience several genetic alterations that are associated with the transition from a preneoplastic lesion to an invasive tumor and finally to the metastatic state. Tumor progression is characterized by stepwise accumulation of genetic alterations. So does the dominant metastatic clone. Modern molecular genetic analyses have clarified that genomic changes accumulate during the development and progression of cancers. In comparison with the corresponding primary tumor, additional events of chromosomal aberrations (including gains or allelic losses) are frequently found in metastases, and the incidence of combined chromosomal alterations in the primary tumor, plus the occurrence of additional aberrations in the distant metastases, correlated significantly with decreased postmetastatic survival. The deletions at 3p, 4p, 6q, 8p, 10q, 11p, 11q, 12p, 13q, 16q, 17p, 18q, 21q, and 22q, as well as the over-representations at 1q, 8q, 9q, 14q and 15q, have been found to associate preferentially with the metastatic phenotype of human cancers. Among of them, the deletions on chromosomes 8p, 17p, 11p and 13p seem to be more significant, and more detail fine regions of them, including 8p11, 8p21-12, 8p22, 8p23, 17p13.3, 11p15.5, and 13q12-13 have been suggested harboring metastasis-suppressor genes. During the past decade, several human chromosomes have been functionally tested through the use of microcell-mediated chromosome transfer (MMCT), and metastasis-suppressor activities have been reported on chromosomes 1, 6, 7, 8, 10, 11, 12, 16, and 17. However, it is not actually known at what stage of the metastatic cascade these alterations have occurred. There is still controversial with the association between the chromosomal aberrations and the metastatic phenotype of cancer. As the progression of human genome project and the establishment of more and more new techniques, it is hopeful to make clear the genetic mechanisms involved in the tumor metastasis in a not very long future, and provide new clues to predicting and controlling the metastasis.
Collapse
Affiliation(s)
- Lun-Xiu Qin
- Liver Cancer Institute Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|