201
|
Liang Z, Wu H, Reddy S, Zhu A, Wang S, Blevins D, Yoon Y, Zhang Y, Shim H. Blockade of invasion and metastasis of breast cancer cells via targeting CXCR4 with an artificial microRNA. Biochem Biophys Res Commun 2007; 363:542-6. [PMID: 17889832 DOI: 10.1016/j.bbrc.2007.09.007] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Accepted: 09/01/2007] [Indexed: 11/29/2022]
Abstract
miRNAs have been shown to function as regulatory molecules and to play an important role in cancer progression. Very little is currently known about the increasing invasion and metastasis of breast cancer due to the loss of expressive levels of certain miRNAs in breast tumor cells. In order to determine whether the CXCR4/SDF-1 pathway is regulated by expression of miRNAs, we designed and synthesized pre-miRNA against CXCR4. This double-stranded miRNA gene was ligated with a miR-155-based Block-iT Pol II miR RNAi Expression Vector (Invitrogen). Expression levels of CXCR4 in CXCR4-miRNA-transfected breast tumor cells had significantly declined. These cells exhibited reduced migration and invasion in vitro. Furthermore, they formed fewer lung metastases in vivo compared to ctrl-miRNA-transfected cells. These data support the conclusion that miRNA against CXCR4 can serve as an alterative means of therapy to lower CXCR4 expression and to block the invasion and metastasis of breast cancer cells.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/therapy
- Cell Line, Tumor
- Chemokine CXCL12/metabolism
- Chemokine CXCL12/physiology
- Down-Regulation
- Humans
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
- Mice
- MicroRNAs/genetics
- Neoplasm Invasiveness
- Neoplasm Metastasis
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-akt/physiology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Receptors, CXCR4/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction/genetics
- Signal Transduction/physiology
- Transfection
- Xenograft Model Antitumor Assays/methods
Collapse
Affiliation(s)
- Zhongxing Liang
- Department of Radiology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
202
|
Yoon Y, Liang Z, Zhang X, Choe M, Zhu A, Cho HT, Shin DM, Goodman MM, Chen ZG, Shim H. CXC chemokine receptor-4 antagonist blocks both growth of primary tumor and metastasis of head and neck cancer in xenograft mouse models. Cancer Res 2007; 67:7518-24. [PMID: 17671223 DOI: 10.1158/0008-5472.can-06-2263] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Squamous cell carcinoma of the head and neck (SCCHN) metastasizes to the lymph nodes and lungs. We have generated previously an orthotopic mouse model for head and neck metastasis and did in vivo selection of SCCHN cells through four rounds of serial metastases. A subpopulation of 686LN cells with high metastatic potential (686LN-Ms) was isolated. When the highly metastatic cells were compared with their low metastatic parental cells (686LN-Ps), we found that CXC chemokine receptor-4 (CXCR4) mRNA levels were significantly higher in the 686LN-Ms cells than the 686LN-Ps cells. Interestingly, the metastatic subclones had lost epithelial morphology and acquired mesenchymal features, which were maintained during cell expansion in vitro. This was featured by decreased E-cadherin and involucrin and increased vimentin and integrin beta(1). These results imply that CXCR4 and epithelial-mesenchymal transition markers can be potential biomarkers to identify the subpopulation of cells with high metastatic potential. Using the orthotopic SCCHN animal model, we showed that anti-CXCR4 treatment suppressed primary tumor growth by inhibiting tumor angiogenesis and prevented lung metastasis. Because the reduction of metastasis seen in the treated group could have resulted from 2-fold reduction in primary tumor size compared with that in the control group, we examined the effects of the CXCR4 antagonist in an experimental metastatic animal model in which 686LN-Ms cells were i.v. injected. 686LN-Ms cells failed to metastasize in the CXCR4 antagonist-treated group, whereas they metastasized to the lungs in the control group. Our data indicate that CXCR4 is an important target to inhibit tumor progression in SCCHN.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Northern
- Blotting, Western
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/prevention & control
- Carcinoma, Squamous Cell/secondary
- Cell Division/physiology
- Female
- Head and Neck Neoplasms/metabolism
- Head and Neck Neoplasms/pathology
- Head and Neck Neoplasms/prevention & control
- Humans
- Lung Neoplasms/metabolism
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Mice
- Mice, Nude
- Molecular Sequence Data
- Neovascularization, Pathologic/prevention & control
- Oligonucleotide Array Sequence Analysis
- Oligopeptides/therapeutic use
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, CXCR4/antagonists & inhibitors
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Transduction, Genetic
Collapse
Affiliation(s)
- Younghyoun Yoon
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Dittmar T, Heyder C, Gloria-Maercker E, Hatzmann W, Zänker KS. Adhesion molecules and chemokines: the navigation system for circulating tumor (stem) cells to metastasize in an organ-specific manner. Clin Exp Metastasis 2007; 25:11-32. [PMID: 17828597 DOI: 10.1007/s10585-007-9095-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2007] [Accepted: 08/19/2007] [Indexed: 12/15/2022]
Abstract
To date, cancer is still the second most prevalent cause of death after cardiovascular diseases in the industrialized word, whereby the primary cause of cancer is not attributed to primary tumor formation, but rather to the growth of metastases at distant organ sites. For several years it was considered that the well-known phenomenon of organ-specific spreading of tumor cells is mostly a mechanical process either directed passively due to size constraints (mechanical trapping theory) or due to a fertile environment provided by the organ in which tumor cells can proliferate (seed and soil hypothesis). Both mechanisms strongly depend on the adhesive properties of tumor cells either to endothelial cells and/or cancer cells, which are facilitated by a variety of cell adhesion molecules including carbohydrates and integrins. Within the past years it became evident that the organ-specific metastatic spreading of tumor cells does not only rely on heterotypic and homotypic adhesive interactions, but also on the interplay of chemokines and their appropriate receptors. Moreover, the identification of cancer stem cells in various tumor tissues has opened new questions. Cancer stem cells possess self-renewal, differentiation, and tumor-initiating capacities. Thus these cells are ideal candidates to be the seed of a secondary tumor. In the present review we will give a brief overview about the complex process of organ-specific metastasis formation depending on the interplay of adhesion molecules, chemokines, and the putative role of cancer stem cells in metastasis formation.
Collapse
Affiliation(s)
- Thomas Dittmar
- Institute of Immunology, Witten/Herdecke University, Stockumer Str. 10, 58448, Witten, Germany.
| | | | | | | | | |
Collapse
|
204
|
Bian XW, Yang SX, Chen JH, Ping YF, Zhou XD, Wang QL, Jiang XF, Gong W, Xiao HL, Du LL, Chen ZQ, Zhao W, Shi JQ, Wang JM. PREFERENTIAL EXPRESSION OF CHEMOKINE RECEPTOR CXCR4 BY HIGHLY MALIGNANT HUMAN GLIOMAS AND ITS ASSOCIATION WITH POOR PATIENT SURVIVAL. Neurosurgery 2007; 61:570-8; discussion 578-9. [PMID: 17881971 DOI: 10.1227/01.neu.0000290905.53685.a2] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE CXCR4 is implicated in the growth, metastasis, and angiogenesis of malignant tumors. We investigated the potential role of CXCR4 in human gliomas. METHODS The expression of CXCR4 messenger ribonucleic acid and protein by human glioma cell lines was examined by reverse-transcriptase polymerase chain reaction and immunocytochemistry analysis. Tumor cell chemotaxis and production of vascular endothelial growth factor induced by the CXCR4 ligand SDF-1beta were measured. Xenograft models were used for evaluation of glioma cell tumorigenesis. CXCR4 expression by xenografted tumors and primary human glioma specimens were evaluated for CXCR4 protein expression. The relationship between CXCR4 expression and patient survival was analyzed. A synthetic lipoxygenase inhibitor, Nordy, was tested for its effects on glioma cell expression and function of CXCR4, as well as on glioma cell tumorigenicity. RESULTS CXCR4 expression correlated directly with the degree of malignancy of the human glioma cell lines and primary tumors. Activation of CXCR4 induced tumor cell chemotaxis and increased production of vascular endothelial growth factor. Glioma cells expressing higher levels of CXCR4 formed more rapidly growing and lethal tumors in nude mice. Primary human glioma specimens expressing CXCR4 contained high-density microvessels. Patients with CXCR4-positive gliomas had poorer prognosis after surgery. The lipoxygenase inhibitor Nordy diminished CXCR4 expression by glioma cell lines in vitro and reduced their tumorigenicity in nude mice. CONCLUSION The level of CXCR4 expression seems to correlate with the degree of malignancy of human gliomas and may contribute to their rapid growth.
Collapse
Affiliation(s)
- Xiu-wu Bian
- Institute of Pathology, Southwest Hospital, Third Military Medical University, Chongqing, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
205
|
Zhao M, Mueller BM, DiScipio RG, Schraufstatter IU. Akt plays an important role in breast cancer cell chemotaxis to CXCL12. Breast Cancer Res Treat 2007; 110:211-22. [PMID: 17687643 DOI: 10.1007/s10549-007-9712-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2007] [Accepted: 07/23/2007] [Indexed: 01/22/2023]
Abstract
The chemokine receptor CXCR4 is functionally expressed on the cell surface of various cancer cells, and plays a role in cell proliferation and migration of these cells. Specifically, in breast cancer cells the CXCR4/CXCL12 axis has been implicated in cell migration in vitro and in metastasis in vivo, but the underlying signaling mechanisms are incompletely understood. The xenograft-derived MDA-MB-231 breast cancer cell line (231mfp), which was shown previously to grow more aggressively than the parent cells, showed increased CXCR4 expression at the mRNA, total protein and cell surface expression level. This correlated with an enhanced response to CXCL12, specifically in augmented and prolonged Akt activation in a G(i), Src family kinase and PI-3 kinase dependent fashion. 231mfp cells migrated towards CXCL12--in contrast to the parent cell line--and this chemotaxis was blocked by inhibition of G(i), Src family kinases, PI-3 kinase and interestingly, Akt itself, as could be shown with two pharmacological inhibitors, a dominant negative Akt construct and with Akt shRNA. Collectively, we have demonstrated that prolonged Akt activation is an important signaling pathway for breast cancer cells expressing CXCR4 and is necessary for CXCL12-dependent cell migration.
Collapse
Affiliation(s)
- Ming Zhao
- Division of Cancer Biology, La Jolla Institute for Molecular Medicine, San Diego, CA 92121, USA.
| | | | | | | |
Collapse
|
206
|
Ruffini PA, Morandi P, Cabioglu N, Altundag K, Cristofanilli M. Manipulating the chemokine-chemokine receptor network to treat cancer. Cancer 2007; 109:2392-404. [PMID: 17503430 DOI: 10.1002/cncr.22706] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Chemokines are chemoattractant cytokines that regulate the trafficking and activation of leukocytes and other cell types under a variety of inflammatory and noninflammatory conditions. Over the past few years, studies have increasingly shown that chemokines play an important role in several aspects of tumor progression. Tumor cells express functional chemokine receptors, which can sustain proliferation, angiogenesis, and survival and promote organ-specific localization of distant metastases. Chemokine expression in human malignancies is associated with a leukocyte infiltration favoring the establishment of immune escape mechanisms. A literature review of relevant publications on preclinical testing of cancer therapies based on interference with the cancer chemokine network was performed. The feasibility, potential advantages, and limitations of the clinical translation of the results of such studies in treatment of different tumor types and settings are discussed. The chemokine network is a key player in the establishment of metastases. In the preclinical setting, blocking agents and antibodies directed against CXCR4 prevent metastasis of different cancers. In mouse models, overexpression of selected chemokines causes tumor infiltration by distinct leukocyte subsets, resulting in tumor regression and tumor-specific immunity generation. Researchers have also successfully used chemokines as carriers and/or adjuvants for cancer vaccines. The cancer chemokine network is a multifaceted therapeutic target.
Collapse
Affiliation(s)
- Pier A Ruffini
- Divisione Oncologia Medica Falck, Ospedale Niguarda Ca' Granda, Milano, Italy.
| | | | | | | | | |
Collapse
|
207
|
Liang Z, Brooks J, Willard M, Liang K, Yoon Y, Kang S, Shim H. CXCR4/CXCL12 axis promotes VEGF-mediated tumor angiogenesis through Akt signaling pathway. Biochem Biophys Res Commun 2007; 359:716-22. [PMID: 17559806 PMCID: PMC1986788 DOI: 10.1016/j.bbrc.2007.05.182] [Citation(s) in RCA: 238] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2007] [Accepted: 05/24/2007] [Indexed: 01/08/2023]
Abstract
CXC chemokine receptor 4 (CXCR4) has been shown to play a critical role in chemotaxis and homing, which are key steps in cancer metastasis. There is also increasing evidence that links this receptor to angiogenesis; however, its molecular basis remains elusive. Vascular endothelial growth factor (VEGF), one of the major angiogenic factors, promotes the formation of leaky tumor vasculatures that are the hallmarks of tumor progression. Here, we investigated whether CXCR4 induces the expression of VEGF through the PI3K/Akt pathway. Our results showed that CXCR4/CXCL12 induced Akt phosphorylation, which resulted in upregulation of VEGF at both the mRNA and protein levels. Conversely, blocking the activation of Akt signaling led to a decrease in VEGF protein levels; blocking CXCR4/CXCL12 interaction with a CXCR4 antagonist suppressed tumor angiogenesis and growth in vivo. Furthermore, VEGF mRNA levels correlated well with CXCR4 mRNA levels in patient tumor samples. In summary, our study demonstrates that the CXCR4/CXCL12 signaling axis can induce angiogenesis and progression of tumors by increasing expression of VEGF through the activation of PI3K/Akt pathway. Our findings suggest that targeting CXCR4 could provide a potential new anti-angiogenic therapy to suppress the formation of both primary and metastatic tumors.
Collapse
Affiliation(s)
- Zhongxing Liang
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgina 30322
| | - Joann Brooks
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgina 30322
| | - Margaret Willard
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgina 30322
| | - Ke Liang
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgina 30322
| | - Younghyoun Yoon
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgina 30322
| | - Seunghee Kang
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgina 30322
| | - Hyunsuk Shim
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgina 30322
- Department of Radiology, Emory University, Atlanta, Georgina 30322
- *To whom requests for reprints should be addressed, at Winship Cancer Institute, 1365C Clifton Road, N.E., Clinic C5008, Atlanta, GA 30322. Phone: 404-778-4564, Fax: 404-778-5550, E-mail:
| |
Collapse
|
208
|
Abstract
The importance of cancer stem cells (CSCs) in tumor-initiation has been firmly established in leukemia and recently reported for a variety of solid tumors. However, the role of CSCs in multistage cancer progression, particularly with respect to metastasis, has not been well-defined. Cancer metastasis requires the seeding and successful colonization of specialized CSCs at distant organs. The biology of normal stem cells and CSCs share remarkable similarities and may have important implications when applied to the study of cancer metastasis. Furthermore, overlapping sets of molecules and pathways have recently been identified to regulate both stem cell migration and cancer metastasis. These molecules constitute a complex network of cellular interactions that facilitate both the initiation of the pre-metastasis niche by the primary tumor and the formation of a nurturing organ microenvironment for migrating CSCs. In this review, we surveyed the recent advances in this dynamic field and propose a unified model of cancer progression in which CSCs assume a central role in both tumorigenesis and metastasis. Better understanding of CSCs as a fundamental component of the metastatic cascade will lead to novel therapeutic strategies against metastatic cancer.
Collapse
Affiliation(s)
- Feng Li
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | | | | | | |
Collapse
|
209
|
Harvey JR, Mellor P, Eldaly H, Lennard TWJ, Kirby JA, Ali S. Inhibition of CXCR4-mediated breast cancer metastasis: a potential role for heparinoids? Clin Cancer Res 2007; 13:1562-70. [PMID: 17332302 DOI: 10.1158/1078-0432.ccr-06-1987] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The pattern of breast cancer metastasis may be determined by interactions between CXCR4 on breast cancer cells and CXCL12 within normal tissues. Glycosaminoglycans bind chemokines for presentation to responsive cells. This study was designed to test the hypothesis that soluble heparinoid glycosaminoglycan molecules can disrupt the normal response to CXCL12, thereby reducing the metastasis of CXCR4-expressing cancer cells. EXPERIMENTAL DESIGN Inhibition of the response of CXCR4-expressing Chinese hamster ovary cells to CXCL12 was assessed by measurement of calcium flux and chemotaxis. Radioligand binding was also assessed to quantify the potential of soluble heparinoids to prevent specific receptor ligation. The human breast cancer cell line MDA-MB-231 and a range of sublines were assessed for their sensitivity to heparinoid-mediated inhibition of chemotaxis. A model of hematogenous breast cancer metastasis was established, and the potential of clinically relevant doses of heparinoids to inhibit CXCL12 presentation and metastatic disease was assessed. RESULTS Unfractionated heparin and the low-molecular-weight heparin tinzaparin inhibited receptor ligation and the response of CXCR4-expressing Chinese hamster ovary cells and human breast cancer cell lines to CXCL12. Heparin also removed CXCL12 from its normal site of expression on the surface of parenchymal cells in the murine lung. Both heparin and two clinically relevant dose regimens of tinzaparin reduced hematogenous metastatic spread of human breast cancer cells to the lung in a murine model. CONCLUSIONS Clinically relevant concentrations of tinzaparin inhibit the interaction between CXCL12 and CXCR4 and may be useful to prevent chemokine-driven breast cancer metastasis.
Collapse
Affiliation(s)
- James R Harvey
- Breast Research Group, School of Surgical and Reproductive Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | | | | | | |
Collapse
|
210
|
Abstract
The process of cancer metastasis is sequential and selective and contains stochastic elements. The growth of metastases represents the endpoint of many lethal events that few tumor cells can survive. Primary tumors consist of multiple subpopulations of cells with heterogeneous metastatic properties, and the outcome of metastasis depends on the interplay of tumor cells with various host factors. The findings that different metastases can originate from different progenitor cells account for the biological diversity that exists among various metastases. Even within a solitary metastasis of proven clonal origin, however, heterogeneity of biological characteristics can develop rapidly. The pathogenesis of metastasis depends on multiple interactions of metastatic cells with favorable host homeostatic mechanisms. Interruption of one or more of these interactions can lead to the inhibition or eradication of cancer metastasis. For many years, all of our efforts to treat cancer have concentrated on the inhibition or destruction of tumor cells. Strategies both to treat tumor cells (such as chemotherapy and immunotherapy) and to modulate the host microenvironment (including the tumor vasculature) should offer additional approaches for cancer treatment. The recent advances in our understanding of the biological basis of cancer metastasis present unprecedented possibilities for translating basic research to the clinical reality of cancer treatment.
Collapse
Affiliation(s)
- Robert R Langley
- Department of Cancer Biology, Unit 173, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA.
| | | |
Collapse
|
211
|
Xu J, Mora A, Shim H, Stecenko A, Brigham KL, Rojas M. Role of the SDF-1/CXCR4 axis in the pathogenesis of lung injury and fibrosis. Am J Respir Cell Mol Biol 2007; 37:291-9. [PMID: 17463394 PMCID: PMC1994230 DOI: 10.1165/rcmb.2006-0187oc] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Stromal cell-derived factor-1 (SDF-1) participates in mobilizing bone marrow-derived stem cells, via its receptor CXCR4. We studied the role of the SDF-1/CXCR4 axis in a rodent model of bleomycin-induced lung injury in C57BL/6 wild-type and matrix metalloproteinase (MMP)-9 knockout mice. After intratracheal instillation of bleomycin, SDF-1 levels in serum and bronchial alveolar lavage fluid increased. These changes were accompanied by increased numbers of CXCR4(+) cells in the lung and a decrease in a population of CXCR4(+) cells in the bone marrow that did not occur in MMP-9(-)/(-) mice. Both SDF-1 and lung lysates from bleomycin-treated mice induced migration of bone marrow-derived stem cells in vitro that was blocked by a CXCR4 antagonist, TN14003. Treatment of mice with TN14003 with bleomycin-induced lung injury significantly attenuated lung fibrosis. Lung tissue from patients with idiopathic pulmonary fibrosis had higher numbers of cells expressing both SDF-1 and CXCR4 than did normal lungs. Our data suggest that the SDF-1/CXCR4 axis is important in the complex sequence of events triggered by bleomycin exposure that eventuates in lung repair. SDF-1 participates in mobilizing bone marrow-derived stem cells, via its receptor CXCR4.
Collapse
Affiliation(s)
- Jianguo Xu
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|
212
|
Siclari VA, Guise TA, Chirgwin JM. Molecular interactions between breast cancer cells and the bone microenvironment drive skeletal metastases. Cancer Metastasis Rev 2007; 25:621-33. [PMID: 17165131 DOI: 10.1007/s10555-006-9023-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Breast cancer cells preferentially spread to bone. Bone metastases are currently incurable and therefore better treatments need to be developed. Metastasis is an inefficient, multi-step process. Specific aspects of both breast cancer cells and the bone microenvironment contribute to the development of bone metastases. Breast cancers express chemokine receptors, integrins, cadherins, and bone-resorbing and bone-forming factors that contribute to the successful and preferential spread of tumor to bone. Bone is rich in growth factors and cell types that make it a hospitable environment for breast cancer growth. Once breast cancer cells enter the bone, a highly complex vicious cycle develops, in which breast cancer cells secrete factors that act on bone cells and other cells within the bone (stem cells, T cells, platelets, adipocytes, fibroblasts, and endothelial cells), causing them to secrete factors that act on adjacent cancer cells. The steps in the metastatic cascade and the vicious cycle within bone offer unique targets for adjuvant treatments to treat and cure bone metastases.
Collapse
Affiliation(s)
- V A Siclari
- University of Virginia, Charlottesville, VA, USA.
| | | | | |
Collapse
|
213
|
Abstract
Tumor cells are known to adapt to and utilize existing physiological mechanisms to promote survival and metastasis. The role of the microenvironment in the establishment of a metastatic lesion has become increasingly important as several factors secreted by stromal cells regulate metastatic pattern in a variety of tumor types. Tumor cells interact with osteoblasts, osteoclasts and bone matrix to form a vicious cycle that is essential for successful metastases. Here we review the current concepts regarding the role of an important chemokine/chemokine receptor (SDF-1 or CXCL12/CXCR4) pathway in tumor development and metastasis. CXCL12 secretion by stromal cells is known to attract cancer cells via stimulation of the CXCR4 receptor that is up regulated by tumor cells. CXCL12/CXCR4 activation regulates the pattern of metastatic spread with organs expressing high levels of CXCL12 developing secondary tumors (i.e., the bone marrow compartment). CXCL12 has a wide range of effects in regards to tumor development but the primary role of CXCL12 appears to be the mobilization of hematopoietic stem cells and the establishment of the cancer stem-like cell niche where high levels of CXCL12 recruit a highly tumorigenic population of tumor cells and promotes cell survival, proliferation, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Jianhua Wang
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Avenue, Ann Arbor, MI 48109-1078, USA
| | | | | |
Collapse
|
214
|
Abstract
Tumor development and progression are multifactorial processes, regulated by a large variety of intrinsic and microenvironmental factors. A key role in cancer is played by members of the chemokine superfamily. Chemokines and their receptors are expressed by tumor cells and by host cells, in primary tumors and in specific metastatic loci. The effects of chemokines on tumorigenesis are diverse: While some members of the superfamily significantly support this process, others inhibit fundamental events required for tumor establishment and metastasis. The current review describes the multifaceted roles of chemokines in malignancy, addressing four major aspects of their activities: (1) inducing leukocyte infiltration to tumors and regulating immune functions, with emphasis on tumor-associated macrophages (and the chemokines CCL2, CCL5), T cells (and the chemokines CXCL9, CXCL10) and dendritic cells (and the chemokines CCL19, CCL20, CCL21); (2) directing the homing of tumor cells to specific metastatic sites (the CXCL12-CXCR4 axis); (3) regulating angiogenic processes (mainly the ELR(+)-CXC and non-ELR-CXC chemokines); (4) acting directly on the tumor cells to control their malignancy-related functions. Together, these different chemokine functions establish a net of interactions between the tumor cells and their microenvironment, and partly dictate the fate of the malignancy cascade.
Collapse
Affiliation(s)
- A Ben-Baruch
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
215
|
Abstract
Metastatic spread of cancer cells is the main cause of death of breast cancer patients, and elucidation of the molecular mechanisms underlying this process is a major focus in cancer research. The identification of appropriate therapeutic targets and proof-of-concept experimentation involves an increasing number of experimental mouse models, including spontaneous and chemically induced carcinogenesis, tumor transplantation, and transgenic and/or knockout mice. Here we give a progress report on how mouse models have contributed to our understanding of the molecular processes underlying breast cancer metastasis and on how such experimentation can open new avenues to the development of innovative cancer therapy.
Collapse
Affiliation(s)
- Anna Fantozzi
- Institute of Biochemistry and Genetics, Department of Clinical-Biological Sciences (DKBW), Center of Biomedicine, University of Basel, Mattenstrasse 28, CH-4058 Basel, Switzerland
| | - Gerhard Christofori
- Institute of Biochemistry and Genetics, Department of Clinical-Biological Sciences (DKBW), Center of Biomedicine, University of Basel, Mattenstrasse 28, CH-4058 Basel, Switzerland
| |
Collapse
|
216
|
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: 1.0] [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
|
217
|
Sun YX, Fang M, Wang J, Cooper CR, Pienta KJ, Taichman RS. Expression and activation of alpha v beta 3 integrins by SDF-1/CXC12 increases the aggressiveness of prostate cancer cells. Prostate 2007; 67:61-73. [PMID: 17034033 DOI: 10.1002/pros.20500] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Stromal cell-derived factor-1 (SDF-1 or CXCL12) and CXCR4 are key elements in the metastasis of prostate cancer cells to bone--but the mechanisms as to how it localizes to the marrow remains unclear. METHODS Prostate cancer cell lines were stimulated with SDF-1 and evaluated for alterations in the expression of adhesion molecules using microarrays, FACs, and Western blotting to identify alpha(v)beta(3) receptors. Cell-cell adhesion and invasion assays were used to verify that activation of the receptor is responsive to SDF-1. RESULTS We demonstrate that SDF-1 transiently regulates the number and affinity of alpha(v)beta(3) receptors by prostate cancer cells to enhance their metastatic behavior by increasing adhesiveness and invasiveness. SDF-1 transiently increased the expression of beta(3) receptor subunit and increased its phosphorylation in metastatic but not nonmetastatic cells. CONCLUSIONS The transition from a locally invasive phenotype to a metastatic phenotype may be primed by the elevated expression of alpha(v)beta(3) receptors. Activation and increased expression of alpha(v)beta(3) within SDF-1-rich organs may participate in metastatic localization.
Collapse
Affiliation(s)
- Yan-Xi Sun
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, USA
| | | | | | | | | | | |
Collapse
|
218
|
Moskovits N, Kalinkovich A, Bar J, Lapidot T, Oren M. p53 Attenuates cancer cell migration and invasion through repression of SDF-1/CXCL12 expression in stromal fibroblasts. Cancer Res 2006; 66:10671-6. [PMID: 17108103 DOI: 10.1158/0008-5472.can-06-2323] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The p53 tumor suppressor acts as a major barrier against cancer. To a large extent, this is due to its ability to maintain genome stability and to eliminate cancer cells from the replicative pool through cell-autonomous mechanisms. However, in addition to its well-documented functions within the malignant cancer cell, p53 can also exert non-cell-autonomous effects that contribute to tumor suppression. We now report that p53 can suppress the production of the chemokine SDF-1 in cultured fibroblasts of both human and mouse origin. This is due to a p53-mediated down-regulation of SDF-1 mRNA, which can be exacerbated on activation of p53 by the drug Nutlin-3. SDF-1 promotes the migration and invasiveness of cells that express its cognate receptor CXCR4. Indeed, medium conditioned by p53-deficient fibroblasts induces cancer cells towards increased directional migration and invasiveness, which are largely reversed by CXCR4 antagonist peptides. Because SDF-1 produced by stromal fibroblasts plays an important role in cancer progression and metastasis, our findings suggest that the ability of p53 to suppress stromal SDF-1 production may be an important mechanism whereby it does its non-cell-autonomous tumor suppressor function.
Collapse
Affiliation(s)
- Neta Moskovits
- Departments of Molecular Cell Biology and Immunology, The Weizmann Institute of Science, Rehovot, Israel and Department of Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | | | | | | | | |
Collapse
|
219
|
Rahman KMW, Sarkar FH, Banerjee S, Wang Z, Liao DJ, Hong X, Sarkar NH. Therapeutic intervention of experimental breast cancer bone metastasis by indole-3-carbinol in SCID-human mouse model. Mol Cancer Ther 2006; 5:2747-56. [PMID: 17121921 DOI: 10.1158/1535-7163.mct-06-0221] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Several lines of experimental evidence have suggested that chemokine receptor CXCR4, a metastasis-promoting molecule, may play important roles in breast cancer bone metastasis. There is emerging evidence linking CXCR4 to matrix metalloproteinases (MMP) as well as their regulator nuclear factor-kappaB (NF-kappaB), a key transcription factor, which is known to activate metastasis-promoting molecules for many types of malignancies, including breast cancer. A recent study also showed that promoter region of CXCR4 has several NF-kappaB-binding sites, suggesting that there may be a cross-talk between CXCR4 and NF-kappaB. We have shown previously that indole-3-carbinol (I3C), a natural compound present in vegetables of the genus Brassica, can inhibit NF-kappaB in breast cancer cells. However, there are no reports in the literature showing any effect of I3C on CXCR4 expression in vitro and in vivo. We therefore examined whether I3C could inhibit bone metastasis of breast cancer by inhibiting CXCR4 and MMP-9 expression mediated via the inhibition of the NF-kappaB signaling pathway. Here, we have modified the severe combined immunodeficient (SCID)-human mouse model of experimental bone metastasis for use with the MDA-MB-231 breast cancer cell line. In this animal model, we found that I3C significantly inhibited MDA-MB-231 bone tumor growth, and our results were correlated with the down-regulation of NF-kappaB. Moreover, we found that I3C significantly inhibited the expression of multiple genes involved in the control of metastasis and invasion in vitro and in vivo, especially the expression of CXCR4 and MMP-9 along with pro-MMP-9, with concomitant decrease in Bcl-2 and increase in the proapoptotic protein Bax. From these results, we conclude that the CXCR4/NF-kappaB pathway is critical during I3C-induced inhibition of experimental breast cancer bone metastasis. These results also suggest that I3C could be a promising agent for the prevention and/or treatment of breast cancer bone metastasis in the future.
Collapse
Affiliation(s)
- K M Wahidur Rahman
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, 9374 Scott Hall, 540 East Canfield, Detroit, MI 48201, USA.
| | | | | | | | | | | | | |
Collapse
|
220
|
Abstract
PURPOSE OF REVIEW Transplantation of hematopoietic stem cells is dependent upon the successful homing, engraftment and repopulation of stem cells in the bone marrow. Stem cell homing through the circulation to the bone marrow is the critical first step in this process. This review discusses the latest progress in defining the molecular processes underlying stem cell homing and the specialized niches where stem cells reside. RECENT FINDINGS Over the past decade, remarkable advances have been made in characterizing the complex sequence of events involved in stem cell homing to the bone marrow. Specifically, the molecular basis of stem cell adhesion and rolling along bone marrow sinusoidal endothelial cells has been defined, and mechanisms underlying endothelial transmigration and enlodgement in bone marrow niches have now been identified. The processes governing hematopoietic stem cell homing to the bone marrow also regulate hematopoietic stem cell migration to extramedullary tissues and the metastasis of cancer stem cells. Improved understanding of these processes has catalyzed the development of therapies to facilitate stem cell mobilization for clinical purposes. SUMMARY Several components of the essential process of stem cell homing have now been characterized. Cell adhesion molecules and their ligands, extracellular matrix components, chemokines, and specialized bone marrow niches all participate in the precise regulation of this process.
Collapse
Affiliation(s)
- John P Chute
- Division of Cellular Therapy, Duke University Medical Center, Durham, North Carolina 27710, USA.
| |
Collapse
|
221
|
Scala S, Giuliano P, Ascierto PA, Ieranò C, Franco R, Napolitano M, Ottaiano A, Lombardi ML, Luongo M, Simeone E, Castiglia D, Mauro F, De Michele I, Calemma R, Botti G, Caracò C, Nicoletti G, Satriano RA, Castello G. Human melanoma metastases express functional CXCR4. Clin Cancer Res 2006; 12:2427-33. [PMID: 16638848 DOI: 10.1158/1078-0432.ccr-05-1940] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The chemokine receptor CXCR4 was identified as an independent predictor of poor prognosis in primary melanoma. The aim of the study was to investigate the role of CXCR4 in human melanoma metastases. EXPERIMENTAL DESIGN CXCR4 expression was evaluated in melanoma metastases and in metastatic cell lines through immunohistochemistry, immunoblotting, immunofluorescence, and reverse transcription-PCR. The function of CXCR4 was tested in the presence of the ligand, CXCL12, through induction of extracellular signal-regulated kinase-1 and -2 (Erk-1 and -2) phosphorylation, proliferation, apoptosis, and migration capabilities. RESULTS CXCR4 expression was detected in 33 out of 63 (52.4%) metastases from cutaneous melanomas. Metastatic melanoma cell lines expressed cell surface CXCR4; PES 43, Alo 40, and COPA cell lines showed the highest levels of CXCR4 (>90% of positive cells); PES 41, Alo 39, PES 47, POAG, and CIMA cell lines showed low to moderate degrees of expression (5-65% of positive cells). Other chemokine receptors, CCR7 and CCR10, were detected on the melanoma cell lines; CXCL12 activated Erk-1 and Erk-2, the whose induction was specifically inhibited by AMD3100 treatment. CXCL12 increased the growth in PES 41, PES 43, and PES 47 cells under suboptimal (1% serum) and serum-free culture conditions; AMD3100 (1 mumol/L) inhibited the spontaneous and CXCL12-induced proliferation. No rescue from apoptosis was shown but PES 41, PES 43, and PES 47 cells migrate toward CXCL12. CONCLUSIONS These findings indicate that CXCR4 is expressed and active in human melanoma metastases, suggesting that active inhibitors such as AMD3100 may be experienced in human melanoma.
Collapse
Affiliation(s)
- Stefania Scala
- Department of Clinical Immunology, National Cancer Institute, G. Pascale Foundation, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
222
|
Slettenaar VIF, Wilson JL. The chemokine network: a target in cancer biology? Adv Drug Deliv Rev 2006; 58:962-74. [PMID: 16996642 DOI: 10.1016/j.addr.2006.03.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Accepted: 07/10/2006] [Indexed: 01/30/2023]
Abstract
Chemokine gradients are central to the movement of cells in both homeostatic and pathological processes. Most cancers express a complex array of chemokines that influence the local microenvironment through recruitment of stromal cells and by stimulating angiogenesis. Recently, the discovery of chemokine receptors on tumor cells has led to speculation that the chemokine system may be involved in cancer cell growth and survival, and possibly the development of site-specific spread. Understanding the networks of chemokines and their receptors in cancer will enable manipulation of this system. Both chemokines and their receptors represent targets for therapeutic intervention either with antibodies or small molecule antagonists. However, due to the complexity of the system, and the number of chemokines and receptors that are also expressed by normal cells, issues remain concerning whether systemic or local drug delivery are preferable and whether the redundancy of the system will compensate if one chemokine or receptor is targeted. Nevertheless, efficacy has been demonstrated in a number of experimental models. By targeting this network, it may be possible to generate anti-tumor immune responses by altering the chemokine and/or leukocyte balance in tumors; alternatively, chemokine/chemokine receptor-expressing cancer cells could be directly targeted.
Collapse
Affiliation(s)
- Violet I F Slettenaar
- Centre for Translational Oncology, Institute of Cancer and the CR-UK Clinical Centre, Barts and The London Queen Mary's School of Medicine and Dentistry, 3rd Floor, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | | |
Collapse
|
223
|
Kryczek I, Wei S, Keller E, Liu R, Zou W. Stroma-derived factor (SDF-1/CXCL12) and human tumor pathogenesis. Am J Physiol Cell Physiol 2006; 292:C987-95. [PMID: 16943240 DOI: 10.1152/ajpcell.00406.2006] [Citation(s) in RCA: 234] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The chemokine stroma-derived factor (SDF-1/CXCL12) plays multiple roles in tumor pathogenesis. It has been demonstrated that CXCL12 promotes tumor growth and malignancy, enhances tumor angiogenesis, participates in tumor metastasis, and contributes to immunosuppressive networks within the tumor microenvironment. Therefore, it stands to reason that the CXCL12/CXCR4 pathway is an important target for the development of novel anti-cancer therapies. In this review, we consider the pathological nature and characteristics of the CXCL12/CXCR4 pathway in the tumor microenvironment. Strategies for therapeutically targeting the CXCL12/CXCR4 axis also are discussed.
Collapse
Affiliation(s)
- Ilona Kryczek
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109-0669, USA
| | | | | | | | | |
Collapse
|
224
|
Ratajczak MZ, Zuba-Surma E, Kucia M, Reca R, Wojakowski W, Ratajczak J. The pleiotropic effects of the SDF-1–CXCR4 axis in organogenesis, regeneration and tumorigenesis. Leukemia 2006; 20:1915-24. [PMID: 16900209 DOI: 10.1038/sj.leu.2404357] [Citation(s) in RCA: 311] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Proper response of normal stem cells (NSC) to motomorphogens and chemoattractants plays a pivotal role in organ development and renewal/regeneration of damaged tissues. Similar chemoattractants may also regulate metastasis of cancer stem cells (CSC). Growing experimental evidence indicates that both NSC and CSC express G-protein-coupled seven-transmembrane span receptor CXCR4 and respond to its specific ligand alpha-chemokine stromal derived factor-1 (SDF-1), which is expressed by stroma cells from different tissues. In addition, a population of very small embryonic-like (VSEL) stem cells that express CXCR4 and respond robustly to an SDF-1 gradient was recently identified in adult tissues. VSELs express several markers of embryonic and primordial germ cells. It is proposed that these cells are deposited early in the development as a dormant pool of embryonic/pluripotent NSC. Expression of both CXCR4 and SDF-1 is upregulated in response to tissue hypoxia and damage signal attracting circulating NSC and CSC. Thus, pharmacological modulation of the SDF-1-CXCR4 axis may lead to the development of new therapeutic strategies to enhance mobilization of CXCR4+ NSC and their homing to damaged organs as well as inhibition of the metastasis of CXCR4+ cancer cells.
Collapse
Affiliation(s)
- M Z Ratajczak
- Stem Cell Biology Program at James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA.
| | | | | | | | | | | |
Collapse
|
225
|
Shim H, Lau SK, Devi S, Yoon Y, Cho HT, Liang Z. Lower expression of CXCR4 in lymph node metastases than in primary breast cancers: potential regulation by ligand-dependent degradation and HIF-1alpha. Biochem Biophys Res Commun 2006; 346:252-8. [PMID: 16756955 DOI: 10.1016/j.bbrc.2006.05.110] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2006] [Accepted: 05/17/2006] [Indexed: 01/08/2023]
Abstract
Stromal-derived factor-1 (SDF-1) is a unique ligand of the CXC chemokine receptor 4 (CXCR4), which is critically involved in the metastasis of breast cancer. High levels of SDF-1 in the common destination organs of metastasis, such as the lymph nodes, lungs, liver, and bones, attract CXCR4-positive tumor cells. The interaction between SDF-1 and CXCR4 leads to the activation of specific signaling pathways, allowing for homing and metastatic progression. However, regulation of CXCR4 expression at the metastatic organ site is not well-documented. We detected the expression of CXCR4 and hypoxia inducible factor (HIF)-1alpha in breast tumor tissues by immunohistochemical staining and analyzed SDF-1 in primary tumors and lymph nodes using real-time RT-PCR. Compared to the corresponding metastasized tumors in the lymph nodes, primary invasive carcinomas showed more intense staining for CXCR4, particularly on the cellular membrane. Both primary tumors and lymph node metastases exhibited higher levels of CXCR4 expression compared to non-neoplastic breast tissues. Therefore, we hypothesized that the tumor environment in the lymph nodes may cause the reduction of CXCR4 levels in the metastatic tumor cells because of: (1) high SDF-1 levels and (2) lower levels of HIF-1alpha. Our in vitro data demonstrated that high levels of SDF-1 can induce the internalization and degradation of CXCR4 through the lysosome pathway. In addition, lower levels of HIF-1alpha in the lymph node metastases, probably induced by the less hypoxic environment, further lowered CXCR4 levels. These results indicate that ligand-dependent degradation and lower HIF-1alpha levels may be potential causes of lowered levels of CXCR4 in the lymph nodes compared to the primary tumors. Our study suggests that CXCR4 levels in tumor cells are regulated by its microenvironment. These findings may enhance our ability to understand the biological behavior of breast cancers.
Collapse
Affiliation(s)
- Hyunsuk Shim
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|
226
|
Yip S, Sabetrasekh R, Sidman RL, Snyder EY. Neural stem cells as novel cancer therapeutic vehicles. Eur J Cancer 2006; 42:1298-308. [PMID: 16697638 DOI: 10.1016/j.ejca.2006.01.046] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2006] [Accepted: 01/23/2006] [Indexed: 01/14/2023]
Abstract
The startling resemblance of many of the behaviours of brain tumours to the intrinsic properties of the neural stem/progenitor cell has triggered a recent dual interest in arming stem cells to track and help eradicate tumours and in viewing stem cell biology as somehow integral to the emergence and/or propagation of the neoplasm itself. These aspects are reviewed and discussed here.
Collapse
Affiliation(s)
- Stephen Yip
- Department of Pathology & Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
| | | | | | | |
Collapse
|
227
|
Jiang YP, Wu XH, Shi B, Wu WX, Yin GR. Expression of chemokine CXCL12 and its receptor CXCR4 in human epithelial ovarian cancer: an independent prognostic factor for tumor progression. Gynecol Oncol 2006; 103:226-33. [PMID: 16631235 DOI: 10.1016/j.ygyno.2006.02.036] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2005] [Revised: 02/16/2006] [Accepted: 02/23/2006] [Indexed: 01/10/2023]
Abstract
OBJECTIVES Chemokine CXCL12 and its unique receptor CXCR4 have been recently implicated in cancer metastasis. Our goal was to explore expression of CXCL12 and CXCR4 protein in normal ovarian surface epithelium, primary tumors and paired metastases of epithelial ovarian cancer as well as its association with clinicopathological features. We also wanted to test if expression of CXCR4 has prognostic value in epithelial ovarian cancer patients. METHOD Sections from 6 normal ovarian surface epithelium, 44 primary epithelial ovarian tumors and 30 paired metastatic tumors in omentum were evaluated for CXCL12 and CXCR4 expression using immunohistochemistry (IHC). RESULTS All samples of normal ovarian surface epithelium were negative for CXCL12 and CXCR4 protein. Ovarian cancer cells mainly showed cytoplasmic staining of CXCL12 and CXCR4. CXCL12 and CXCR4 staining were detected in 40/44 (91%) and 26/44 (59%) patients with primary epithelial ovarian tumors respectively. CXCR4 expression in primary tumors had no significant correlation with lymph nodes metastasis. However, if we combined CXCR4 expression in primary tumors with metastatic tumors, a significant correlation with lymph nodes metastasis was found (P = 0.018). The intensity of CXCL12 staining correlated with ascites (P = 0.014). The rate of CXCR4 expression in refractory and recurrent group (81% versus 28%, P = 0.0008) was significantly higher than that in no-recurrent group. After a median follow-up of 37 months, CXCR4 expression was found associated with an unfavorable prognosis with significantly reduced median disease progression-free survival and overall survival of 15 and 27 months (P = 0.0004, P = 0.017) respectively. Median time-to-event was not reached in patients with negative CXCR4 staining. In multivariate analysis, CXCR4 expression and residual tumor size emerged as independent prognostic factors in epithelial ovarian cancer patients. CONCLUSIONS This article provides the first evidence that CXCR4 expression could be an independent prognostic factor for epithelial ovarian cancer patients.
Collapse
Affiliation(s)
- Yu-Ping Jiang
- Department of Obstetrics and Gynecology, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | | | | | | | | |
Collapse
|
228
|
Hanaoka H, Mukai T, Tamamura H, Mori T, Ishino S, Ogawa K, Iida Y, Doi R, Fujii N, Saji H. Development of a 111In-labeled peptide derivative targeting a chemokine receptor, CXCR4, for imaging tumors. Nucl Med Biol 2006; 33:489-94. [PMID: 16720240 DOI: 10.1016/j.nucmedbio.2006.01.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2005] [Revised: 01/12/2006] [Accepted: 01/12/2006] [Indexed: 11/24/2022]
Abstract
The chemokine receptor CXCR4 is highly expressed in tumor cells and plays an important role in tumor metastasis. The aim of this study was to develop a radiopharmaceutical for the imaging of CXCR4-expressing tumors in vivo. Based on structure-activity relationships, we designed a 14-residue peptidic CXCR4 inhibitor, Ac-TZ14011, as a precursor for radiolabeled peptides. For 111In-labeling, diethylenetriaminepentaacetic acid (DTPA) was attached to the side chain of d-Lys(8) which is distant from the residues indispensable for the antagonistic activity. In-DTPA-Ac-TZ14011 inhibited the binding of a natural ligand, stromal cell-derived factor-1alpha, to CXCR4 in a concentration-dependent manner with an IC50 of 7.9 nM (Ac-TZ14011: 1.2 nM). In biodistribution experiments, more 111In-DTPA-Ac-TZ14011 accumulated in the CXCR4-expressing tumor than in blood or muscle. Furthermore, the tumor-to-blood and tumor-to-muscle ratios were significantly reduced by coinjection of Ac-TZ14011, indicating a CXCR4-mediated accumulation in tumor. These findings suggested that 111In-DTPA-Ac-TZ14011 would be a potential agent for the imaging of CXCR4 expression in metastatic tumors in vivo.
Collapse
Affiliation(s)
- Hirofumi Hanaoka
- Graduate School of Pharmaceutical Sciences, Kyoto University, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
Bartolomé RA, Molina-Ortiz I, Samaniego R, Sánchez-Mateos P, Bustelo XR, Teixidó J. Activation of Vav/Rho GTPase signaling by CXCL12 controls membrane-type matrix metalloproteinase-dependent melanoma cell invasion. Cancer Res 2006; 66:248-58. [PMID: 16397238 PMCID: PMC1952211 DOI: 10.1158/0008-5472.can-05-2489] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Melanoma cells express the chemokine receptor CXCR4, which confers invasive signals on binding to its ligand CXCL12. We show here that knocking down membrane-type matrix metalloproteinase (MT1-MMP) expression translates into a blockade of invasion across reconstituted basement membranes and type I collagen gels in response to CXCL12, which is the result of lack of MMP-2 activation. Interference with MMP-2 expression further confirms its important role during this invasion. Vav proteins are guanine-nucleotide exchange factors for Rho GTPases that regulate actin dynamics and gene expression. We show that melanoma cells express Vav1 and Vav2, which are activated by CXCL12 involving Jak activity. Blocking Vav expression by RNA interference results in impaired activation of Rac and Rho by CXCL12 and in a remarkable inhibition of CXCL12-promoted invasion. Importantly, up-regulation of MT1-MMP expression by CXCL12, a mechanism contributing to melanoma cell invasion, is blocked by knocking down Vav expression or by inhibiting Jak. Together, these data indicate that activation of Jak/Vav/Rho GTPase pathway by CXCL12 is a key signaling event for MT1-MMP/MMP-2-dependent melanoma cell invasion.
Collapse
Affiliation(s)
- Rubén A. Bartolomé
- Department of Immunology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas
| | - Isabel Molina-Ortiz
- Department of Immunology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas
| | - Rafael Samaniego
- Servicio de Inmuno-Oncología, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Paloma Sánchez-Mateos
- Servicio de Inmuno-Oncología, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Xosé R. Bustelo
- Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas, Salamanca, Spain
| | - Joaquin Teixidó
- Department of Immunology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas
| |
Collapse
|
230
|
Abstract
BACKGROUND Expression of the inflammatory chemokine CCL5 (RANTES) by tumor cells is thought to correlate with the progression of several cancers. CCL5 was shown to induce breast cancer cell migration, mediated by the receptor CCR5. A CCR5 antagonist was demonstrated to inhibit experimental breast tumor growth. Recently, CCL5 and CCR5 mRNA expression was reported in prostate cancer (PCa) tissues. Herein, we characterized CCL5 and CCR5 expression in cultures of PCa cells and explored possible functions of CCL5 in PCa progression. METHODS Quantitative RT-PCR, ELISA, and immunohistochemical staining were performed to examine CCL5 expression in prostate cell lines. CCR5 expression was measured by flow cytometry. Proliferation and invasion assays were performed to determine potential functions of CCL5 and CCR5 in PCa. RESULTS Expression of CCL5 mRNA and protein was found in human PCa cell lines (PC-3; DU-145; LNCaP) and primary prostate adenocarcinoma cells. CCL5 and CCR5 were also detected in human PCa tissues. CCR5 expression was demonstrated on the cell surface of PCa cells, as well as in intracellular pools. Incubation with CCL5 (10-100 ng/ml) induced PCa cell proliferation, and the CCR5 antagonist TAK-779 inhibited CCL5-induced proliferation. CCL5 was found to stimulate PCa cell invasion, and TAK-779 blocked the effects of CCL5. CONCLUSIONS In light of evidence that inflammation influences the pathogenesis of PCa, these results suggest that inflammatory chemokines, such as CCL5, expressed by prostate cells may act directly on the growth and survival of PCa cells. Chemokine receptor antagonists may thus block autocrine mechanisms of PCa progression.
Collapse
MESH Headings
- Adenocarcinoma/chemistry
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Amides/pharmacology
- Autocrine Communication
- Cell Line, Tumor
- Cell Membrane/metabolism
- Cell Proliferation/drug effects
- Chemokine CCL5
- Chemokines, CC/analysis
- Chemokines, CC/antagonists & inhibitors
- Chemokines, CC/genetics
- Chemokines, CC/physiology
- Disease Progression
- Flow Cytometry
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Male
- Neoplasm Invasiveness/physiopathology
- Prostatic Neoplasms/chemistry
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Quaternary Ammonium Compounds/pharmacology
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- Receptors, CCR5/analysis
- Receptors, CCR5/genetics
- Receptors, CCR5/physiology
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Gayle G Vaday
- Department of Research, Northport Veterans Affairs Medical Center, Northport, New York 11768, USA.
| | | | | | | |
Collapse
|
231
|
Li HC, Stoicov C, Rogers AB, Houghton J. Stem cells and cancer: Evidence for bone marrow stem cells in epithelial cancers. World J Gastroenterol 2006; 12:363-71. [PMID: 16489634 PMCID: PMC4066053 DOI: 10.3748/wjg.v12.i3.363] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer commonly arises at the sites of chronic inflammation and infection. Although this association has long been recognized, the reason has remained unclear. Within the gastrointestinal tract, there are many examples of inflammatory conditions associated with cancer, and these include reflux disease and Barrett’s adenocarcinoma of the esophagus, Helicobacter infection and gastric cancer, inflammatory bowel disease and colorectal cancer and viral hepatitis leading to hepatocellular carcinoma. There are several mechanisms by which chronic inflammation has been postulated to lead to cancer which includes enhanced proliferation in an endless attempt to heal damage, the presence of a persistent inflammatory environment creating a pro-carcinogenic environment and more recently a role for engraftment of circulating marrow-derived stem cells which may contribute to the stromal components of the tumor as well as the tumor mass itself. Here we review the recent advances in our understanding of the contributions of circulating bone marrow-derived stem cells to the formation of tumors in animal models as well as in human beings.
Collapse
Affiliation(s)
- Han-Chen Li
- Department of Medicine, University of Massachusetts Medical School, LRB-Second Floor, Room 209, 364 Plantation Street, Worcester, MA 01605-2324, United States
| | | | | | | |
Collapse
|
232
|
Shibata MA, Ito Y, Morimoto J, Kusakabe K, Yoshinaka R, Otsuki Y. In vivo electrogene transfer ofinterleukin-12 inhibits tumor growth and lymph node and lung metastases in mouse mammary carcinomas. J Gene Med 2006; 8:335-52. [PMID: 16345101 DOI: 10.1002/jgm.854] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Human breast cancer metastasizes mainly to lymph nodes, lungs, liver, and bone; in the majority of cases, it is the development of metastases which leads to death. In order to suppress mammary cancer metastasis, we applied in vivo electrogene transfer (non-viral method) as a means of interleukin-12 (IL-12) gene therapy on highly metastatic murine mammary cancer model. METHODS Metastatic mammary tumors induced by inoculation in BALB/c female mice were treated by intratumoral injections of either a plasmid vector containing IL-12 or empty vector and then subjected to in vivo electrogene transfer once a week for 8 weeks. RESULTS Treatment with IL-12 resulted in elevation of both IL-12 and IFNgamma levels in mammary tumors and in serum and intratumoral levels of CD4 and CD8 proteins were also increased. Tumor volumes and lymphatic and pulmonary metastases were significantly reduced. The histopathological changes induced by IL-12 characteristically included marked inflammation, increased apoptosis, decreased DNA synthesis, peripheral influx of significantly greater numbers of active macrophages, and reduced blood microvessel density, and apoptotic vascular endothelial cells were frequently seen. Western blotting showed decreases in VEGFR-3 of tumors exposed to IL-12 gene therapy. In adjuvant immunofluorescence studies, the CD31-positive endothelial cells of microvessels showed decreased VEGFR-3 expression in IL-12-treated tumors. However, apparent alterations in VEGFR-3 expression of podoplanin-positive lymphatic endothelial cells were not observed in IL-12-treated tumors. Although recombinant IL-12 did not inhibit tubular formation of human umbilical vein endothelial cells in a Matrigel assay, recombinant IFNgamma did completely suppress the tubular formation. CONCLUSIONS In vivo electrogene transfer of IL-12 exerts strong anti-tumorigenic and anti-metastatic effects likely due to T-cell-mediated immune responses as well as anti-angiogenic action.
Collapse
Affiliation(s)
- Masa-Aki Shibata
- Department of Anatomy and Biology, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | | | | | | | | | | |
Collapse
|
233
|
Uchida H, Iwashita Y, Sasaki A, Shibata K, Matsumoto T, Ohta M, Kitano S. Chemokine receptor CCR6 as a prognostic factor after hepatic resection for hepatocellular carcinoma. J Gastroenterol Hepatol 2006; 21:161-8. [PMID: 16706828 DOI: 10.1111/j.1440-1746.2005.04157.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIMS Chemokines and their receptors have recently been shown to have major roles in cancer metastasis. The aim of this study was to determine whether the interaction between chemokine receptor 6 (CCR6) and its ligand, macrophage inflammatory protein-3 alpha (MIP-3alpha), correlates with metastasis of hepatocellular carcinoma (HCC). METHODS To observe the reaction of CCR6 expressed cancer cells to MIP-3alpha stimulation, chemotactic and actin polymerization assays for both CCR6 high cells (HepG2) and CCR6 low cells (MCF-7) were performed. CCR6 mRNA levels in tumor specimens from 30 HCC patients were quantified by real-time polymerase chain reaction. Patients were classified into two groups, high (>or= 20 copies; n=10) CCR6 and low (<20 copies; n=20) CCR6 on the basis of CCR6 expression, and the groups were compared with respect to clinicopathological features. RESULTS When HepG2 cells (CCR6 high) were stimulated with MIP-3alpha, they migrated in a dose-dependent manner, and formation of pseudopodia was observed. These phenomena were not observed in the CCR6 low cells. The incidence of intrahepatic metastasis was higher in the high CCR6 expression group than in the low CCR6 expression group (P<0.05). Disease-free survival was significantly poorer in the high CCR6 expression group than in the low CCR6 expression group (P<0.05). CONCLUSIONS It was indicated that CCR6 might be associated with intrahepatic metastasis of HCC and might be able to become one of the prognostic factor after hepatic resection for HCC.
Collapse
MESH Headings
- Aged
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/surgery
- Cell Line, Tumor
- Chemotaxis
- Cytoskeleton/drug effects
- Disease-Free Survival
- Dose-Response Relationship, Drug
- Female
- Hepatectomy
- Humans
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/surgery
- Macrophage Inflammatory Proteins/pharmacology
- Male
- Middle Aged
- Neoplasm Metastasis
- Pseudopodia
- RNA, Messenger/metabolism
- Receptors, CCR6
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
Collapse
Affiliation(s)
- Hiroki Uchida
- Department of Surgery I, Oita University Faculty of Medicine, Oita, Japan.
| | | | | | | | | | | | | |
Collapse
|
234
|
Kaifi JT, Yekebas EF, Schurr P, Obonyo D, Wachowiak R, Busch P, Heinecke A, Pantel K, Izbicki JR. Tumor-cell homing to lymph nodes and bone marrow and CXCR4 expression in esophageal cancer. J Natl Cancer Inst 2005; 97:1840-7. [PMID: 16368946 DOI: 10.1093/jnci/dji431] [Citation(s) in RCA: 169] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The chemokine and bone marrow-homing receptor CXCR4 has been implicated in metastatic dissemination of various cancers. We investigated CXCR4 expression in esophageal cancer specimens and its association with survival, lymph node microinvolvement, and bone marrow micrometastasis. METHODS We analyzed frozen tumor specimens from 136 patients with completely resected esophageal cancer for CXCR4 expression by immunohistochemistry. Lymph node microinvolvement and bone marrow micrometastasis were assessed by immunohistochemistry with monoclonal antibodies Ber-EP4 (against epithelial cell adhesion molecule) and pancytokeratin A45-B/B3 (against several cytokeratins), respectively. Associations between CXCR4 expression and clinicopathologic features, including tumor stage, histologic grade, lymph node metastasis and microinvolvement, bone marrow micrometastasis, and survival, were investigated with Fisher's test, log-rank test, and Cox multivariable analysis. All statistical tests were two-sided. RESULTS CXCR4 protein was expressed in 75 (55%) of 136 esophageal tumors examined. CXCR4 expression was statistically significantly associated with reduced median overall and disease-specific survival, compared with CXCR4 nonexpression (P < .001; log-rank test). The median overall survival of patients with CXCR4-positive tumors was 20 months and with CXCR4-negative tumors, 76 months (difference = 56 months, 95% confidence interval [CI] = 4 to 108 months; P < .001). The median disease-specific survival of patients with CXCR4-positive tumors was 25 months and with CXCR4-negative tumors was 97 months (difference = 72 months, 95% CI = 34 to 110 months; P < .001). CXCR4 expression was statistically significantly associated with increased lymph node microinvolvement (P < .001) and with increased bone marrow micrometastasis (P < .001). In multivariable analysis, CXCR4 expression, compared with its nonexpression, was identified as the independent variable that was most strongly associated with reduced disease-specific survival (relative risk [RR] of death = 2.03, 95% CI = 1.20 to 3.41; P = .008) and overall survival (RR of death = 2.18, 95% CI = 1.33 to 3.59; P = .002). CONCLUSION CXCR4 expression was associated with poor clinical outcome in esophageal cancer patients. CXCR4 may have a role in early metastatic spread because its expression was associated with micrometastases to both the lymph nodes and bone marrow. Thus, CXCR4 should be explored further as a target for adjuvant therapy for micrometastatic disease.
Collapse
Affiliation(s)
- Jussuf T Kaifi
- Klinik fuer Allgemein-, Viszeral- und Thoraxchirurgie, Universitaetsklinikum Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Cabioglu N, Yazici MS, Arun B, Broglio KR, Hortobagyi GN, Price JE, Sahin A. CCR7 and CXCR4 as novel biomarkers predicting axillary lymph node metastasis in T1 breast cancer. Clin Cancer Res 2005; 11:5686-93. [PMID: 16115904 DOI: 10.1158/1078-0432.ccr-05-0014] [Citation(s) in RCA: 183] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The chemokine receptors CCR7 and CXCR4 have been shown to play an important role in cancer metastasis. We therefore studied the differential expression of CCR7 and CXCR4, along with that of the biomarker HER2-neu, to evaluate whether these biomarkers could predict axillary lymph node metastasis in breast cancer. EXPERIMENTAL DESIGN Biomarker expression levels were evaluated using paraffin-embedded tissue sections of lymph node-negative (n = 99) and lymph node-positive (n = 98) T1 breast cancer by immunohistochemical staining. RESULTS Lymph node-positive tumors showed higher rates of high cytoplasmic CCR7 staining (21.5% versus 8.5%, P = 0.013) and HER2-neu overexpression (21.5% versus 9.3%, P = 0.019) than did lymph node-negative tumors. Similarly, high cytoplasmic CXCR4 expression occurred more commonly in lymph node-positive tumors (11.2% versus 5.1%, P = 0.113). In contrast, predominantly nuclear CXCR4 staining was more likely to be found in lymph node-negative tumors (54.5% versus 37.8%, P = 0.018). Furthermore, cytoplasmic CXCR4 coexpressed with HER2-neu was the only factor associated with involvement of four or more lymph nodes (16.7% versus 1.2%, P = 0.04) among lymph node-positive tumors. When all three biomarkers (CCR7, CXCR4, HER2-neu) were utilized together, 50.0% of lymph node-positive tumors highly expressed one of these biomarkers compared with 18.8% of the lymph node-negative tumors (P < 0.0001). CONCLUSIONS Our results suggest that the chemokine receptor CCR7 is a novel biomarker that can predict lymph node metastases in breast cancer. Utilization of additional markers, such as CXCR4 and HER2-neu, further improves the prediction of the presence and extent of lymph node involvement.
Collapse
Affiliation(s)
- Neslihan Cabioglu
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|
236
|
Salvucci O, Bouchard A, Baccarelli A, Deschênes J, Sauter G, Simon R, Bianchi R, Basik M. The role of CXCR4 receptor expression in breast cancer: a large tissue microarray study. Breast Cancer Res Treat 2005; 97:275-83. [PMID: 16344916 DOI: 10.1007/s10549-005-9121-8] [Citation(s) in RCA: 165] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2005] [Accepted: 11/10/2005] [Indexed: 12/20/2022]
Abstract
The chemokine receptor CXCR4 is an important factor in the migration, invasiveness, metastasis and proliferation of breast cancer cells. We have retrospectively analyzed the levels of expression of CXCR4 in a large cohort of breast cancers and pre-invasive breast samples linked to clinical data. A total of 1808 invasive breast carcinomas and 214 pre-invasive breast samples could be analyzed in correlation with basic clinico-pathological data such as hormone receptor status, HER2 status and tumor grade. The majority of breast cancers expressed either nuclear or cytoplasmic staining or both. CXCR4 cytoplasmic expression was associated with parameters of tumor aggressivity (tumor grade and lymph node status) and had prognostic value (age-adjusted hazard ratio=1.73; Confidence Interval: 1.07-2.77) with respect to disease-specific survival. CXCR4 positivity in the cytoplasm but not the nucleus was associated with HER2 expression and amplification as well as with hormone receptor negativity (both ER and PR). The percentage of nuclear staining increased from normal breast tissue (20%) to ductal carcinoma-in-situ DCIS (43%) to invasive cancer (67%) while CXCR4 was expressed in the cytoplasm of 67% of (DCIS) cases (double that in normal breast samples), suggesting an important role in breast tumor progression. The CXCR4 receptor is expressed in many breast cancers, justifying its development as a therapeutic target in breast cancer patients. Its cytoplasmic expression is associated with breast tumor progression, suggesting potential value as a diagnostic marker.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/metabolism
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/mortality
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Cell Nucleus/metabolism
- Cytoplasm/metabolism
- Disease Progression
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Lymphatic Metastasis
- Middle Aged
- Neoplasm Invasiveness
- Precancerous Conditions/metabolism
- Precancerous Conditions/mortality
- Precancerous Conditions/pathology
- Prognosis
- Receptor, ErbB-2/metabolism
- Receptors, CXCR4/metabolism
- Retrospective Studies
- Survival Analysis
- Tissue Array Analysis
Collapse
Affiliation(s)
- Ombretta Salvucci
- Department of Biomedical Sciences and Biotechnology, Division of Human Anatomy, University of Brescia, Brescia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
237
|
Abstract
Cells derived from bone marrow are pluripotent, with the ability to differentiate into multiple cell types. Environmental cues dictate differentiation decisions. It should not be surprising then, that abnormal cell environments lead to abnormal differentiation of these cells, and in some cases, malignant transformation. Identifying a role for bone marrow-derived cells in the initiation and progression of cancer allows a dramatic change in the way in which cancer is viewed. Identifying the cell responsible for initiating a tumor offers the exciting possibility of specifically targeting unique aspects of these cells and altering signaling properties for more effective therapeutic approaches.
Collapse
Affiliation(s)
- Calin Stoicov
- University of Massachusetts Medical School, Department of Medicine, Division of Gasteroenterology, Department of Cancer Biology, Worcester MA 01605, USA
| | | | | | | |
Collapse
|
238
|
Matteucci E, Locati M, Desiderio MA. Hepatocyte growth factor enhances CXCR4 expression favoring breast cancer cell invasiveness. Exp Cell Res 2005; 310:176-85. [PMID: 16112111 DOI: 10.1016/j.yexcr.2005.07.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2005] [Revised: 07/15/2005] [Accepted: 07/17/2005] [Indexed: 01/02/2023]
Abstract
Microenvironmental factors affect different aspects of tumor cell biology, including cell survival, invasion, and metastasis. Here, we report that hepatocyte growth factor and hypoxia may contribute to breast carcinoma cell invasiveness by inducing the chemokine receptor CXCR4. Hepatocyte growth factor enhanced CXCR4 mRNA and protein expression exclusively in MCF-7 (low invasive) carcinoma cells, while in response to hypoxia, CXCR4 induction was observed in both MCF-7 and MDA-MB 231 (highly invasive) carcinoma cells. The receptor induction had a functional role in cancer cells, as demonstrated by the fact that hepatocyte growth factor pretreatment promoted MCF-7 cell migration toward the CXCR4-specific ligand CXCL12. Extracellular signal-regulated protein kinase 1/2 (ERK1/2) and phosphoinositide-3-kinase (PI3K) transduction pathways seemed to be differently implicated in the early induction of CXCR4 by hepatocyte growth factor or hypoxia in the two breast carcinoma cells examined.
Collapse
Affiliation(s)
- Emanuela Matteucci
- Institute of General Pathology, School of Medicine, University of Milan, via Luigi Mangiagalli, 31-20133 Milan, Italy
| | | | | |
Collapse
|
239
|
Kang H, Watkins G, Douglas-Jones A, Mansel RE, Jiang WG. The elevated level of CXCR4 is correlated with nodal metastasis of human breast cancer. Breast 2005; 14:360-7. [PMID: 16216737 DOI: 10.1016/j.breast.2004.12.007] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2004] [Revised: 12/02/2004] [Accepted: 12/21/2004] [Indexed: 01/08/2023] Open
Abstract
CXCR4, the receptor for stromal cell-derived factor-1(SDF-1), belongs to the chemokine receptor family and has been shown to play an important role in regulating the directional migration of breast cancer cells to sites of metastasis. In the present study, we evaluated the expression of CXCR4 and its association with pathological features and clinical outcome in human breast cancer. Expression of CXCR4 in eight breast cancer cell lines and breast cancer tissues was investigated using conventional PCR. Levels of CXCR4 transcript and protein were examined in human breast cancer tissues (n=120) and corresponding normal tissues (n=32) using real-time quantitative PCR and immunohistochemistry, respectively. The level of CXCR4 expression was analyzed against tumour types, grade, nodal status, recurrence, metastasis, and survival over a median 120 month follow-up period. The expression of CXCR4 was detected in all breast cancer cell lines examined, as well as in breast cancer tissues and breast normal tissues. Breast cancer tissues highly expressed CXCR4 compared with corresponding normal tissues (P=0.029). The level of CXCR4 expression showed a significant difference between node-positive group and node-negative group (19+/-13 vs. 49.7+/-9, respectively, P=0.03). The level of CXCR4 expression was marginal, yet statistically insignificant, higher in tumours from patients with metastatic disease compared with those who remained disease free. No correlation was seen between levels of CXCR4 and the overall survival, although at higher levels of CXCR4 linked to shorter disease free survival (113.0 vs. 136.7 months in patients with low CXCR4, P=0.14, Cox proportional test). The level of CXCR4 expression is significantly correlated with lymph node metastasis. The elevated levels of CXCR4 suggest that the patient has high possibility of lymph node metastasis. CXCR4 may be a useful prognostic indicator and a potential therapeutic target in cancer therapies in patients with breast cancer.
Collapse
Affiliation(s)
- Hua Kang
- University Department of Surgery, Wales College of Medicine, Metastasis and Angiogenesis Research Group, Cardiff University, Heath Park, Cardiff CF4 4XN, UK
| | | | | | | | | |
Collapse
|
240
|
Yang SX, Chen JH, Jiang XF, Jiang XF, Wang QL, Chen ZQ, Zhao W, Feng YH, Xin R, Shi JQ, Bian XW. Activation of chemokine receptor CXCR4 in malignant glioma cells promotes the production of vascular endothelial growth factor. Biochem Biophys Res Commun 2005; 335:523-8. [PMID: 16084492 DOI: 10.1016/j.bbrc.2005.07.113] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2005] [Accepted: 07/20/2005] [Indexed: 11/30/2022]
Abstract
Numerous studies have showed that chemokine receptors, such as CXCR4, contribute to the growth and metastasis of a variety of malignant tumors. In this study, we investigated the role of CXCR4 in the production of angiogenic factor, vascular endothelial growth factor (VEGF), in various human glioma cells from astrocytic origin. The expression of CXCR4 mRNA and protein in three glioma cell lines, U87-MG, SHG-44, and CHG-5, was determined by RT-PCR and immunocytochemistry, respectively. The malignancies of three gliomas were evaluated by expression of glial fibrillary acidic protein and vimentin, the differentiation markers of astrocytic cells. The role of functional CXCR4 in tumor cell migration was studied with chemotaxis assay. Ca2+ mobilization and VEGF production were measured in the cells after stimulation with CXCR4 ligand, SDF1beta. The results showed that the levels of functional CXCR4 expression at both mRNA and protein levels by several human glioma cell lines were correlated with the degree of differentiation of the tumor cells. Activation of CXCR4 induced glioma cell chemotaxis and could trigger the increase of intracellular [Ca2+]i. Such an activation could result in the increased production of VEGF by the stimulated tumor cells. Our results suggest that CXCR4 may contribute to the high level of VEGF produced by malignant glioma cells and thus constitute a therapeutic target for antiangiogenesis strategy.
Collapse
Affiliation(s)
- Shi-xin Yang
- Institute of Pathology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Luker KE, Luker GD. Functions of CXCL12 and CXCR4 in breast cancer. Cancer Lett 2005; 238:30-41. [PMID: 16046252 DOI: 10.1016/j.canlet.2005.06.021] [Citation(s) in RCA: 203] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2005] [Accepted: 06/15/2005] [Indexed: 12/12/2022]
Abstract
The chemokine CXCL12 (SDF-1) and its cognate receptor CXCR4 were first identified in the context of trafficking and homeostasis of immune cells, such as T lymphocytes. Subsequently, it has been determined that CXCR4 regulates several key processes in a wide variety of cancers. Functions of CXCL12 and CXCR4 in cancer first were described in metastatic breast cancer, and more recent studies also have identified roles for this signaling pathway in primary breast tumors. This review focuses on functions of CXCR4 and CXCL12 in primary and metastatic breast cancer, including molecular mechanisms of action and relationships of this pathway to other key regulators of breast cancer progression. We also describe pre-clinical studies indicating the potential to exploit CXCR4 as a new molecular target for diagnosis and treatment of breast cancer in patients.
Collapse
Affiliation(s)
- Kathryn E Luker
- Department of Radiology, University of Michigan Medical School, 1150 West Medical Center Dr, 9301 MSRB III, Ann Arbor, MI 48109-0648, USA
| | | |
Collapse
|
242
|
Scala S, Ottaiano A, Ascierto PA, Cavalli M, Simeone E, Giuliano P, Napolitano M, Franco R, Botti G, Castello G. Expression of CXCR4 predicts poor prognosis in patients with malignant melanoma. Clin Cancer Res 2005; 11:1835-41. [PMID: 15756007 DOI: 10.1158/1078-0432.ccr-04-1887] [Citation(s) in RCA: 210] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE CXCR4 receptor and its unique ligand, the CXCL12 chemokine, have been recently implicated in cancer metastasis. Evidence about the role of CXCR4/CXCL12 axis has been reported in several cancers including melanoma. Our goal was to investigate if CXCR4 expression has a prognostic value in malignant melanoma. EXPERIMENTAL DESIGN Immunohistochemical expression of CXCR4 was evaluated on 71 specimens of primary cutaneous melanoma with a Breslow tumor thickness of >1 mm after radical resection. Associations between baseline patient features and tumors were analyzed by chi(2) test. The prognostic value of CXCR4 expression was evaluated by univariate and multivariate analyses adjusted by age, sex, Breslow tumor thickness, presence of ulceration, and sentinel lymph node metastases. RESULTS CXCR4 expression was detected in 31 of 71 (43.6%) primary cutaneous melanomas. Membrane or cytoplasmic staining for CXCR4 protein was absent in 56% of the tumors. The positive cases were divided into three score classes according to their staining: low in 15 cases (21%), moderate in 10 (14%), and high in 6 (8%). After a median follow-up of 38 months, 26 patients progressed (16 of 26 expressed CXCR4) and 19 died (12 of 19 expressed CXCR4). The CXCR4 expression on tumor cells was correlated with an unfavorable prognosis with a median disease-free and overall survival of 22 and 35 months, respectively. The hazard ratios of relapse and death, compared with patients with CXCR4-negative tumors, were 2.5 (95% confidence interval, 1.2-6.1) and 3.1 (95% confidence interval, 1.1-7.2), respectively. Median time-to-event (progression and survival) was not reached in patients with CXCR4-negative tumors. In the multivariate analysis, CXCR4 expression, presence of ulceration, and sentinel lymph node status emerged as independent prognostic factors. CONCLUSIONS This article provides the first evidence that CXCR4 expression could be an independent and powerful prognostic marker in primary cutaneous malignant melanomas.
Collapse
Affiliation(s)
- Stefania Scala
- Department of Clinical Immunology, National Cancer Institute, G. Pascale Foundation, via Mariano Semmola, 80131 Naples, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Sood AK, Coleman RL, Wolf JK, Gershenson DM. Selected highlights from the 5th International Conference on Ovarian Cancer. Houston, TX, USA, 1-4 December 2004. Expert Opin Pharmacother 2005; 6:1269-75. [PMID: 15957979 DOI: 10.1517/14656566.6.7.1269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Anil K Sood
- Department of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, PO Box 301439, Houston, TX 77230-1439, USA.
| | | | | | | |
Collapse
|
244
|
Abstract
In the vast majority of cases, cancer continues to be an incurable disease when it has spread beyond the primary organ. Most cancer research and therapy design to date has focused on chemotherapy directed at killing the replicating tumor cells. Little attention has been placed on targeting the microenvironments of the primary tumor site, the circulating tumor cells, or the metastatic or secondary (target) tumor site and how cancer cells move among them. To develop these targets, a better understanding of metastasis and the mechanisms underlying the spread of tumors is required. This review describes the steps of metastasis using a paradigm of emigration to migration to immigration, with prostate cancer as a model system.
Collapse
Affiliation(s)
- Kenneth J Pienta
- University of Michigan Urology Center, Ann Arbor, MI 48109-0946, USA.
| | | |
Collapse
|
245
|
Kim J, Takeuchi H, Lam ST, Turner RR, Wang HJ, Kuo C, Foshag L, Bilchik AJ, Hoon DSB. Chemokine receptor CXCR4 expression in colorectal cancer patients increases the risk for recurrence and for poor survival. J Clin Oncol 2005; 23:2744-53. [PMID: 15837989 DOI: 10.1200/jco.2005.07.078] [Citation(s) in RCA: 289] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
PURPOSE Liver metastasis is the predominant cause of colorectal cancer (CRC) related mortality. Chemokines, soluble factors that orchestrate hematopoetic cell movement, have been implicated in directing cancer metastasis, although their clinical relevance in CRC has not been defined. Our hypothesis was that the chemokine receptor CXCR4 expressed by CRC is a prognostic factor for poor disease outcome. METHODS CRC cell lines (n = 6) and tumor specimens (n = 139) from patients with different American Joint Committee on Cancer (AJCC) stages of CRC were assessed. Microarray screening of select specimens and cell lines identified CXCR4 as a prominent chemokine receptor. CXCR4 expression in tumor and benign specimens was assessed by quantitative real-time reverse transcription polymerase chain reaction and correlated with disease recurrence and overall survival. RESULTS High CXCR4 expression in tumor specimens (n = 57) from AJCC stage I/II patients was associated with increased risk for local recurrence and/or distant metastasis (risk ratio, 1.35; 95% CI, 1.09 to 1.68; P = .0065). High CXCR4 expression in primary tumor specimens (n = 35) from AJCC stage IV patients correlated with worse overall median survival (9 months v 23 months; RR, 2.53; 95% CI, 1.19 to 5.40; P = .016). CXCR4 expression was significantly higher in liver metastases (n = 39) compared with primary CRC tumors (n = 100; P < .0001). CONCLUSION CXCR4, a well-characterized chemokine receptor for T-cells, is differentially expressed in CRC. CXCR4 gene expression in primary CRC demonstrated significant associations with recurrence, survival, and liver metastasis. The CXCR4-CXCL12 signaling mechanism may be clinically relevant for patients with CRC and represents a potential novel target for disease-directed therapy.
Collapse
Affiliation(s)
- Joseph Kim
- Department of Molecular Oncology, John Wayne Cancer Institute, 2200 Santa Monica Blvd., Santa Monica, CA 90404, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Laverdiere C, Hoang BH, Yang R, Sowers R, Qin J, Meyers PA, Huvos AG, Healey JH, Gorlick R. Messenger RNA Expression Levels of CXCR4 Correlate with Metastatic Behavior and Outcome in Patients with Osteosarcoma. Clin Cancer Res 2005; 11:2561-7. [PMID: 15814634 DOI: 10.1158/1078-0432.ccr-04-1089] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To determine if osteosarcoma cells express chemokine receptors and if their presence or absence relates to clinical features. EXPERIMENTAL DESIGN Using fluorescent quantitative real-time PCR, the pattern of 17 chemokine receptors in 3 osteosarcoma cell lines and 68 osteosarcoma patient samples was analyzed. RESULTS The expression of the chemokine receptors was generally low among the cell lines. In the high-grade osteosarcoma patient samples (n = 47), CXCR4 was the most commonly expressed (63%) and its expression level was inversely correlated to overall survival (P < 0.0001), event-free survival (P < 0.001), and metastasis-free survival (MFS; P = 0.002). There was also a correlation between the expression level of CXCR4 and the presence of metastasis at diagnosis (P = 0.002). CCR7 was expressed in 43% of the samples and its expression level was inversely correlated with overall survival (P = 0.03) and MFS (P = 0.007). CCR10 mRNA expression level was inversely correlated with MFS (P = 0.009). There was no association between the expression of CXCR4, CCR7, and CCR10. Of the 26 samples studied for stromal cell-derived factor-1 expression, 77% expressed it, but there was no correlation with the clinical variables or CXCR4 expression. Multivariate analysis revealed that mRNA expression level of CXCR4 was the only significant variable for overall survival (P = 0.0006), event-free survival (P = 0.004), and MFS (P = 0.025). CONCLUSIONS These data suggest that CXCR4 could be useful as a prognostic factor and as a predictor of potential metastatic development in osteosarcoma. If further studies confirm that it is relevant to metastases in this disease, it could represent a new therapeutic target.
Collapse
Affiliation(s)
- Caroline Laverdiere
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Sun YX, Schneider A, Jung Y, Wang J, Dai J, Wang J, Cook K, Osman NI, Koh-Paige AJ, Shim H, Pienta KJ, Keller ET, McCauley LK, Taichman RS. Skeletal localization and neutralization of the SDF-1(CXCL12)/CXCR4 axis blocks prostate cancer metastasis and growth in osseous sites in vivo. J Bone Miner Res 2005; 20:318-29. [PMID: 15647826 DOI: 10.1359/jbmr.041109] [Citation(s) in RCA: 274] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2004] [Revised: 08/02/2004] [Accepted: 09/08/2004] [Indexed: 11/18/2022]
Abstract
UNLABELLED To delineate the role of SDF-1 and CXCR4 in metastatic prostate cancer (CaP), positive correlations were established between SDF-1 levels and tumor metastasis. Neutralization of CXCR4 limited the number and the growth of intraosseous metastasis in vivo. Together, these in vivo metastasis data provide critical support that SDF-1/CXCR4 plays a role in skeletal metastasis. INTRODUCTION Previously we determined that the stromal-derived factor-1 (SDF-1)/CXCR4 chemokine axis is activated in prostate cancer (CaP) metastasis to bone. To delineate the role of SDF-1/CXCR4 in CaP, we evaluated SDF-1 levels in a variety of tissues and whether neutralization of SDF-1 prevented metastasis and/or intraosseous growth of CaPs. MATERIALS AND METHODS SDF-1 levels were established in various mouse tissues by ELISA, immunohistochemistry, and in situ hybridization. To assess the role of SDF-1/CXCR4 in metastasis, bone metastases were established by administering CaP cells into the left cardiac ventricle of nude animals in the presence or absence of neutralizing CXCR4 antibody. The effect of SDF-1 on intraosseous growth of CaP cells was determined using intratibial injections and anti-CXCR4 antibodies and peptides. RESULTS There was a positive correlation between the levels of SDF-1 and tissues in which metastatic CaP lesions were observed. SDF-1 levels were highest in the pelvis, tibia, femur, liver, and adrenal/kidneys compared with the lungs, tongue, and eye, suggesting a selective effect. SDF-1 staining was generally low or undetectable in the center of the marrow and in the diaphysis. SDF-1 mRNA was localized to the metaphysis of the long bones nearest to the growth plate where intense expression was observed near the endosteal surfaces covered by osteoblastic and lining cells. Antibody to CXCR4 significantly reduced the total metastatic load compared with IgG control-treated animals. Direct intratibial injection of tumor cells followed by neutralizing CXCR4 antibody or a specific peptide that blocks CXCR4 also decreased the size of the tumors compared with controls. CONCLUSIONS These data provide critical support for a role of SDF-1/CXCR4 in skeletal metastasis. Importantly, these data show that SDF-1/CXCR4 participate in localizing tumors to the bone marrow for prostate cancer.
Collapse
Affiliation(s)
- Yan-Xi Sun
- Department of Periodontics, Prevention, Geriatrics, University of Michigan School of Dentistry, Ann Arbor, MI 48109-1078, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Liang Z, Yoon Y, Votaw J, Goodman MM, Williams L, Shim H. Silencing of CXCR4 Blocks Breast Cancer Metastasis. Cancer Res 2005. [DOI: 10.1158/0008-5472.967.65.3] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
RNA interference technology, silencing targeted genes in mammalian cells, has become a powerful tool for studying gene function. For the first time in cancer research, we show that direct injection of a pool of naked small interfering RNA (siRNA) duplexes can prevent tumorigenesis in an animal model, suggesting a novel preventive and therapeutic strategy for cancer management. As a model system, we used siRNA duplexes of CXCR4 to block breast cancer metastasis. Here, we show that blocking CXCR4 expression at the mRNA level by a combination of two siRNAs impairs invasion of breast cancer cells in Matrigel invasion assay and inhibits breast cancer metastasis in an animal model. Targeting more than one site of the target gene may be important to overcome the functional redundancy of other variants of a single gene, especially in in vivo experiments. Moreover, our studies confirm the necessity of CXCR4 in breast cancer metastasis.
Collapse
Affiliation(s)
- Zhongxing Liang
- 1Department of Hematology/Oncology, Winship Cancer Institute and
| | - Younghyoun Yoon
- 1Department of Hematology/Oncology, Winship Cancer Institute and
| | - John Votaw
- 2Department of Radiology, Emory University, Atlanta, Georgia
| | - Mark M. Goodman
- 2Department of Radiology, Emory University, Atlanta, Georgia
| | - Larry Williams
- 2Department of Radiology, Emory University, Atlanta, Georgia
| | - Hyunsuk Shim
- 1Department of Hematology/Oncology, Winship Cancer Institute and
- 2Department of Radiology, Emory University, Atlanta, Georgia
| |
Collapse
|
249
|
Smith MCP, Luker KE, Garbow JR, Prior JL, Jackson E, Piwnica-Worms D, Luker GD. CXCR4 regulates growth of both primary and metastatic breast cancer. Cancer Res 2005; 64:8604-12. [PMID: 15574767 DOI: 10.1158/0008-5472.can-04-1844] [Citation(s) in RCA: 535] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The chemokine receptor CXCR4 and its cognate ligand CXCL12 recently have been proposed to regulate the directional trafficking and invasion of breast cancer cells to sites of metastases. However, effects of CXCR4 on the growth of primary breast cancer tumors and established metastases and survival have not been determined. We used stable RNAi to reduce expression of CXCR4 in murine 4T1 cells, a highly metastatic mammary cancer cell line that is a model for stage IV human breast cancer. Using noninvasive bioluminescence and magnetic resonance imaging, we showed that knockdown of CXCR4 significantly limited the growth of orthotopically transplanted breast cancer cells. Mice in which parental 4T1 cells were implanted had progressively enlarging tumors that spontaneously metastasized, and these animals all died from metastatic disease. Remarkably, RNAi of CXCR4 prevented primary tumor formation in some mice, and all mice transplanted with CXCR RNAi cells survived without developing macroscopic metastases. To analyze effects of CXCR4 on metastases to the lung, an organ commonly affected by metastatic breast cancer, we injected tumor cells intravenously and monitored cell growth with bioluminescence imaging. Inhibiting CXCR4 with RNAi, or the specific antagonist AMD3100, substantially delayed the growth of 4T1 cells in the lung, although neither RNAi nor AMD3100 prolonged overall survival in mice with experimental lung metastases. These data indicate that CXCR4 is required to initiate proliferation and/or promote survival of breast cancer cells in vivo and suggest that CXCR4 inhibitors will improve treatment of patients with primary and metastatic breast cancer.
Collapse
MESH Headings
- Animals
- Benzylamines
- Cell Growth Processes/physiology
- Cell Line, Tumor
- Cyclams
- Disease Models, Animal
- Female
- Heterocyclic Compounds/pharmacology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/secondary
- Magnetic Resonance Imaging
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Neoplasm Metastasis
- Neoplasm Transplantation
- RNA Interference
- Receptors, CXCR4/antagonists & inhibitors
- Receptors, CXCR4/biosynthesis
- Receptors, CXCR4/genetics
- Receptors, CXCR4/physiology
- Transfection
Collapse
Affiliation(s)
- Matthew C P Smith
- Molecular Imaging Center, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | | | | | |
Collapse
|
250
|
Lee AV. Location, location, location: regulation of breast cancer progression by the microenvironment. Breast Cancer Res 2004; 6:279-80. [PMID: 15535860 PMCID: PMC1064092 DOI: 10.1186/bcr957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Adrian V Lee
- Breast Center, Baylor College of Medicine and the Methodist Hospital, Houston, Texas, USA.
| |
Collapse
|