Human Melanoma Metastases Express Functional CXCR4

Share Embed


Descrição do Produto

DOI:10.1158/1078-0432.CCR-05-1940

Human Melanoma Metastases Express Functional CXCR4 Stefania Scala, Paola Giuliano, Paolo A. Ascierto, et al. Clin Cancer Res 2006;12:2427-2433. Published online April 25, 2006.

Updated Version

Cited Articles Citing Articles

E-mail alerts Reprints and Subscriptions Permissions

Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-05-1940

This article cites 43 articles, 22 of which you can access for free at: http://clincancerres.aacrjournals.org/content/12/8/2427.full.html#ref-list-1 This article has been cited by 1 HighWire-hosted articles. Access the articles at: http://clincancerres.aacrjournals.org/content/12/8/2427.full.html#related-urls

Sign up to receive free email-alerts related to this article or journal. To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at [email protected]. To request permission to re-use all or part of this article, contact the AACR Publications Department at [email protected].

Downloaded from clincancerres.aacrjournals.org on March 9, 2013 Copyright © 2006 American Association for Cancer Research

DOI:10.1158/1078-0432.CCR-05-1940

Human Cancer Biology

Human Melanoma Metastases Express Functional CXCR4 Stefania Scala,1 Paola Giuliano,1 Paolo A. Ascierto,1 Caterina Ierano`,1 Renato Franco,3 Maria Napolitano,1 Alessandro Ottaiano,1 Maria L. Lombardi,2 Monica Luongo,2 Ester Simeone,1 Daniele Castiglia,6 Francesca Mauro,1 Ileana De Michele,1 Rosa Calemma,1 Gerardo Botti,3 Corrado Caraco`,4 Gianfranco Nicoletti,5 Rocco A. Satriano,5 and Giuseppe Castello1

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-1and -2 (Erk-1and -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 Amol/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.

The incidence and mortality rate of melanoma have increased in the last 30 years. The National Cancer Institute Surveillance, Epidemiology, and End Results database documents increases of 619% in annual diagnoses of cutaneous melanoma and of 165% in annual mortality from 1950 to 2000 (1). Metastatic spread may arise from very small tumor masses and in about two-thirds of all cases of malignant melanoma, spreading develop primarily as locoregional metastases. In about onethird of the cases, primary development of distant metastases is observed (2). The metastatic potential of primary melanoma is

Authors’ Affiliations: Departments of 1Clinical Immunology, 2Experimental Oncology, 3Pathology, and 4Surgical Oncology, National Cancer Institute, G. Pascale Foundation; 5Department of Dermatology, Second University of Naples, Naples; and 6Laboratory of Molecular and Cell Biology, IDI-IRCCS, Rome, Italy Received 9/6/05; revised 1/11/06; accepted 1/13/06. Grant support: Associazione Italiana per la Ricerca sul Cancro, Ministry of Health Research grant 2003, and Ministero dell’Istruzione, dell’Universita` e della Ricerca (D.D. 1105/2002 prog. no. 132). The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Requests for reprints: Stefania Scala, Department of Clinical Immunology, National Cancer Institute, G. Pascale Foundation, via Mariano Semmola, 80131 Naples, Italy. Phone: 39-81-590-3841; Fax: 39-81-590-3841; E-mail: scalaste@ unina.it. F 2006 American Association for Cancer Research. doi:10.1158/1078-0432.CCR-05-1940

www.aacrjournals.org

considerably higher than that of other primary solid tumors when comparing the size of primary lesion. The usual outcome for patients with distant metastases remains bleak, with median survival of 6 to 10 months and 5 years (1). Except for high-dose IFN as adjuvant therapy in stage III disease, little success has emerged over the last 20 years for metastatic melanoma (3). The underlying molecular events that explain malignant melanoma genesis and progression have been only partially characterized, and only a small number of genes have been identified as playing key roles in melanoma. Among these, some cell cycle regulators, apoptotic, signal transduction, cell adhesion, and matrix digestion genes have been shown to be deregulated in this neoplasm (4). Chemokines and their receptors have emerged as attractive targets regulating the migration of tumor cells in vivo. There is evidence that antigen-presenting cells such as dendritic cells, Langerhans cells, T cells, and natural killer cells bearing chemokine receptors migrate from the skin to the draining lymph node in response to specific chemotactic factors referred to as chemokines. Chemokines have been hypothesized to recruit solid tumor cells to lymph nodes. Expression of chemokine receptors on tumor cells and their involvement in metastases revealed that these receptors could indeed provide migratory directions to tumor cells (5 – 8). The most widely expressed chemokine receptor among cancers is likely to be CXCR4, described in breast cancer, glioblastoma, pancreatic, prostatic,

2427

Clin Cancer Res 2006;12(8) April 15, 2006

Downloaded from clincancerres.aacrjournals.org on March 9, 2013 Copyright © 2006 American Association for Cancer Research

DOI:10.1158/1078-0432.CCR-05-1940 Human Cancer Biology

colon, thyroid, and non – small cell lung cancers (6 – 20). In melanoma cells, several chemokine receptors have been described. CXCR4 and CXCR3 were originally reported in human melanoma cell lines (21). Further insight into the mechanism of CXCL12/SDF1-a-mediated cell invasion was described. SDF1-a promotes invasion across basement membranes through the induction of membrane type 1-matrix metalloproteinase. Moreover, SDF1-a triggered the activation of small GTPases such as RhoA and Rac1 (22). Other chemokine receptors were described on melanoma cells; expression of CCR7 (23) and CXCR3 (24) were mainly correlated to lymph node metastases, whereas the expression of CCR10 was mainly detected in skin metastases (25). The role of the CXCR4CXCL12 axis was also validated in in vivo studies. B16 melanoma cells transfected with CXCR4 produced an increased number of pulmonary nodules compared with the lung metastases induced by B16 cells transfected with mock vector (26). CXCR4-specific ligand, CXCL12, is frequently produced at sites of melanoma metastases. Cardones et al. showed that CXCR4 enhances adhesion of B16 tumor cells to endothelial cells in vitro and in vivo via h1 integrin through CXCL12; in vivo, metastases of CXCR4-B16 cells to murine lungs was strongly inhibited by anti-CXCL12 and two different antih(1)integrin monoclonal antibodies (27). Recently, it was shown that CXCR4 expression predicted prognosis in human primary melanoma (28). The aim of the study was to evaluate the role of CXCR4 in human melanoma metastases. CXCR4 expression was analyzed in a panel of 63 melanoma metastases and 8 metastases-derived human melanoma cell lines. CXCR4 was expressed on 52.4% of metastases; the CXCR4 receptor was active in the melanoma cell lines and was inhibited by the specific antagonist, AMD3100. Thus, strategies targeting the CXCR4 receptor may be of benefit in melanoma patients.

Materials and Methods Immunohistochemistry. Two serial 5-Am sections of formalin-fixed, paraffin-embedded cutaneous melanomas were stained; one for H&E and the other immunostained using the biotin-streptavidin-peroxidase method (YLEM). Deparaffinized sections were microwaved in 1 mmol/L EDTA (pH 8.0) for two cycles of 5 minutes each to unmask epitopes. After treatment with 1% hydrogen peroxidase for 30 minutes to block endogenous peroxide, the sections were subsequently incubated with monoclonal antibodies (anti-CXCR4, MAB 172, clone 44716; R&D Systems, Minneapolis, MN) for 2 hours at room temperature. The sections were then incubated with biotin-labeled secondary antibody (1:30) for 30 minutes and with streptavidin-peroxidase (1:30) for 10 minutes. Slides were stained for 10 minutes with AEC chromogen (DAKO Cytomation, Milan, Italy) and then counterstained with hematoxylin, washed and mounted in water fluid. The dilutions of the monoclonal antibody, biotin-labeled secondary antibody, and streptavidin-peroxidase were made with PBS (pH 7.4) containing 5% bovine serum albumin. CXCR4 staining was defined as positive when expression involved >80% of neoplastic cells. Moreover, membrane positivity, cytoplasmic positivity, or both membrane and cytoplasmic positivity and absolute percentage of positivity were scored. All series included positive controls of well-characterized sections (melanomas and breast cancer). Negative controls were obtained by substituting the primary antibody with a mouse myeloma protein of the same subclass, at the same concentration as the monoclonal antibody. All controls gave satisfactory results. Macrophage positivity was used as an adequate internal positive control for each case, in order to validate the technical

Clin Cancer Res 2006;12(8) April 15, 2006

procedures. Slides were evaluated by two blinded observers (G. Botti and R. Franco); discordant cases were discussed and concordance was then achieved. Cell lines. Melanoma metastases were obtained from patients undergoing surgery with curative intent. PES 41, PES 43, PES 47, Colo 38, Alo 39, Alo 40, COPA, and CIMA cells were cultivated in Iscove’s modified Dulbecco’s medium (Cambrex Bioscience, Verviers, Belgium) supplemented with heat-inactivated 10% fetal bovine serum, penicillin, and streptomycin (100 units/mL each). The cell lines PES 41, PES 43, and PES 47 were isolated from different metastases of the same patient (PES 41 and PES 47 from two s.c. metastases and PES 43 from a lung metastases). Also, Alo 39 and Alo 40 were derived from two lymph node metastases of the same patient. Flow cytometry. To evaluate the expression of CXCR4, adherent cancer cells at subconfluency (50-70% confluent) were detached with 2 mmol/L EDTA in PBS, washed, resuspended in ice-cold PBS, and incubated for 30 minutes at 4jC with anti-CXCR4 antibody PE conjugated (FAB 173P, clone 44717; R&D Systems) or mouse IgG2a PE conjugated as negative control. After three washes in PBS, the cells were analyzed by FACScan cytofluorometer (Becton Dickinson Immunocytometry Systems; Becton Dickinson, Mountain View, CA). Immunofluorescence. Cells were grown onto 15 mm glass coverslips and fixed with 4% paraformaldehyde but not permeabilized. The primary antibody anti-CXCR4 (10 Ag/mL, MAB 172; R&D Systems) was incubated overnight in 3% bovine serum albumin in PBS. TRITCconjugated secondary antibody (Jackson ImmunoResearch, West Grove, PA) was used at 1:100 dilution in 3% bovine serum albumin in PBS for 45 minutes. After washing in PBS, the images were analyzed with a confocal microscopy. RNA isolation and reverse transcriptase-PCR. Total cellular RNA from melanoma cell lines was extracted using TriPure reagent (Roche Diagnostics Corporation, Indianapolis, IN). Briefly, cells were homogenized and RNA was extracted using a monophasic solution of guanidine thiocyanate and phenol. The RNA was quantified and assessed for purity by UV spectrophotometry. The expression of mRNA for glyceraldehyde-3-phosphate dehydrogenase was assessed on all RNA samples as an internal control. Chemokine and chemokine receptors mRNA were detected by reverse transcriptase-PCR. DNase-treated RNA (2 Ag) was reversed transcribed with Superscript II RNase H-reverse transcriptase according to the manufacturer’s instructions (InvitrogenLife Technologies, Carlsbad, CA). Reverse transcriptase-PCR was carried out using 2 AL of cDNA in a 20 AL final reaction mixture. A Robocycler gradient 96 (Stratagene, La Jolla, CA) was used for the amplification. Cycling conditions of the respective PCR were as follows: initial denaturation (4 minutes at 94jC) followed by 32 cycles of denaturation (1 minute at 94jC), annealing (75 seconds at 56jC, CXCR4; 58jC, CXCL12; 60jC, CCR10; 62jC, CCR7), and elongation (3 minutes at 72jC). Ten microliters of the products were run on a 2% agarose gel and analyzed under UV light. The gene-specific primers used for the ampl ifi cation were as f ol lows: CXCR4, 5V-GG TGGTCTATGTTGGCGTCT-3V (forward) and 5V-TGGAGTGTGACAGCTTGGAG-3V (reverse); CXCL12, 5V-GGGCTCCTGGGTTTTGTATT-3V (forward) and 5V-GTCCTGAGAGTCCTTTTGCG-3V (reverse); CCR7, 5V-TCCTTCTCATCAGCAAGCTGTC-3V (forward) and 5V-GAGGCAGCCCAGGTCCTTGAA-3V(reverse); CCR10, 5V-GGGTTTCTCCTTCCACTCCT-3V(forward) and 5V-TATTCCCCACATCCTCCTTG-3V(reverse). Western blot. Cells were homogenized in lysis buffer [40 mmol/L Hepes (pH 7.5), 120 mmol/L NaCl, 5 mmol/L MgCl2, 1 mmol/L EGTA, 0.5 mmol/L EDTA, 1% Triton X-100] containing protease (Complete Tablets, EDTA-free; Roche) and phosphatase (20 mmol/L h-glycerolphosphate, 2.5 mmol/L Na-PPi) inhibitors. Total lysate was cleared by centrifugation at 15,000  g for 20 minutes. The protein concentration of the supernatant was measured using Bio-Rad protein assay. Extracts (50 Ag) were denatured and resolved by SDS-PAGE (12%) gel electrophoresis, transferred to a Hybond enhanced chemiluminescence nitrocellulose membrane (Amersham Biosciences, Freiburg, Germany) by electroblotting at 250 mA for 2 hours. The following primary

2428

Downloaded from clincancerres.aacrjournals.org on March 9, 2013 Copyright © 2006 American Association for Cancer Research

www.aacrjournals.org

DOI:10.1158/1078-0432.CCR-05-1940 CXCR4 in Melanoma Metastases antibodies were used: anti-CXCR4 (ProSci, Inc., Poway, CA), anti-P-ERK (sc 7383, Santa Cruz Biotechnology, Inc., Santa Cruz, CA), and antiERK2 (sc 154G, Santa Cruz Biotechnology). Anti-mouse, anti-rabbit, and anti-goat IgG coupled to peroxidase were used as secondary antibodies (Sigma-Aldrich, Corp., St. Louis, MO) and the signal was revealed through chemiluminescence detection kit (ECL detection kit, Amersham Biosciences). Growth curve viability and cell growth assay. Cells (20-50  104) were seeded in a six-multiwell system in a medium culture containing 10% fetal bovine serum. After 24 hours, the medium was replaced as indicated and SDF1-a (20 ng/mL, from Upstate, Lake Placid, NY) was added in the presence or absence of AMD3100 (Sigma-Aldrich). Cells were trypsinized each day and counted by using hemocytometer.

Results CXCR4 is expressed in human melanoma metastases. Recently, CXCR4 was identified as an independent predictor of poor prognosis in primary melanoma (28). To evaluate the role of CXCR4 in human melanoma metastases, CXCR4 expression was analyzed in a panel of 63 melanoma metastases from melanoma patients who had undergone curative intent surgery (29 s.c., 22 lymph node, and 12 visceral metastases). CXCR4 expression was detected in 33 out of 63 (52.4%) metastases from cutaneous melanomas (15 out of 29 s.c., 12 out of 22 lymph node, 6 out of 12 visceral metastases). Figure 1 shows some examples of CXCR4 staining of melanoma on a dermal infiltration by epidermotropic melanoma (A), in liver metastases with normal hepatocytes weakly expressing CXCR4 (B), in nodal metastases (C), and lung metastases (D). The staining was mostly cytoplasmic and few cases showed focal membrane positivity. CXCR4 detection on human metastatic melanoma cell lines. The CXCR4 cell surface was evaluated on cell lines derived from melanoma metastases. Figure 2 showed CXCR4 protein through flow cytometry, immunoblotting, and immunofluorescence (A, B, and C, respectively). 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

Fig. 1. CXCR4 expression in melanoma metastases. CXCR4 staining was detected in dermal infiltration by epidermotropic melanoma (A, 40), in liver metastases with normal hepatocytes weakly expressing CXCR4 (arrow, B, 25), in nodal metastases (C, 25), and lung metastases (D, 25).

www.aacrjournals.org

positive cells). In Fig. 2B, the metastatic melanoma cell lines expressed CXCR4 protein compared with Colo 38, human melanoma cell line, and to human primary melanocytes (MPR1). In Fig. 2C, immunofluorescent staining of CXCR4 was detectable in PES cells. PES 43 showed the most intense staining compared with PES 47 and PES 41 cells. In addition to the cell surface punctated staining, there is a clear cytoplasmic CXCR4 staining concentrated perinuclearly in all three lines tested. It might represent a functional status of the receptor because the binding to the specific ligand induces receptor internalization (29). Human metastatic melanoma cell lines expressed CXCR4 and other chemokine receptors. CXCR4 expression was then evaluated in eight metastatic melanoma cell lines by reverse transcription-PCR. CXCR4 expression was detected in PES 41, PES 43, PES 47, Colo 38, Alo 39, and Alo 40 cells with different amount of CXCR4 transcripts; in particular, PES 43 and PES 47 showed high CXCR4 expression compared with normal peripheral blood mononuclear cells, used as positive controls (Fig. 3). Consistent with previous reports (23, 24, 26), the expression of CCR7 and CCR10 was detected on melanoma cell lines, CCR7 was expressed mainly in PES 47 cells, whereas CCR10 was detectable in all tested cell lines (Fig. 3). CXCL12 increased the growth of human melanoma cells. CXCL12 production was described in human cancer cells such as ovary (18) and colon cancers (30). CXCL12 was not detectable in the human melanoma cell lines analyzed through ELISA and cytofluorometric evaluation (data not shown). mRNA for CXCL12 was detected in Alo 40, PES 41, and PES 47 cells (Fig. 3). The effect of CXCL12 on tumor cell proliferation was assessed. Under optimal culture conditions (in the presence of 10% serum), the addition of CXCL12 did not affect the cell growth of PES 41, PES 43, and PES 47, neither did AMD3100, a specific CXCR4 inhibitor (Fig. 4). Under suboptimal culture conditions (1% serum), CXCL12 increased the growth in PES 41, PES 43, and PES 47 cells. Treatment with AMD3100 (1 Amol/L) inhibited the spontaneous and CXCL12-induced proliferation in PES 43 and PES 41 human melanoma cells, less in PES 47 human melanoma cells (Fig. 4). Interestingly, 200 ng/mL of CXCL12 was weaker than 20 ng/mL in promoting cell growth, mainly in PES 43 cells in which CXCR4 receptor is overexpressed. Furthermore, in serum-free conditions, CXCL12 (20 ng/mL) induced cell growth and AMD3100 specifically inhibited the spontaneous and CXCL12-induced growth in PES 41, PES 43, and PES 47 cells (Fig. 4). Moreover, CXCL12 (20 or 100 ng/mL) was not able to rescue PES 43, PES 47, and Colo 38 cells from apoptosis (data not shown). AMD3100 inhibited the CXCL12-induced activation of extracellular signal-regulated kinases 1 and 2 in human melanoma cells. To study whether CXCL12 could activate a downstream pathway, we focused our attention on the activation of the mitogen-activated protein kinase, extracellular signal-regulated kinase 1 and 2 (ERK1/2) in human melanoma cells. PES 43 and PES 41 cells were serum-starved for 48 hours and then incubated with CXCL12 (20 ng/mL). Changes in the phosphorylation of p44/42 (Erk1/2) kinases were analyzed by Western blotting. In PES 43 and PES 41 cells, human melanoma cell lines derived, respectively, from a lung and s.c. metastases, CXCL12 induced rapid (2-minute) ERK1/2 activation. Notably, no Erk activation

2429

Clin Cancer Res 2006;12(8) April 15, 2006

Downloaded from clincancerres.aacrjournals.org on March 9, 2013 Copyright © 2006 American Association for Cancer Research

DOI:10.1158/1078-0432.CCR-05-1940 Human Cancer Biology

Fig. 2. Human metastatic melanoma cell lines express cell surface CXCR4. CXCR4 cell surface was evaluated on cell lines derived from melanoma metastases. CXCR4 level was evaluated through flow cytometry (A), immunoblotting (B), and immunofluorescence (C).

was induced in normal human melanocytes. Moreover, the addition of AMD3100 (1 Amol/L) inhibited the CXCL12induced activation of Erk (Fig. 5, top). ERK1/2 activation was also inhibited in PES 43 cells (Fig. 5, bottom) and in PES 41 human melanoma cells (data not shown) by T22, a specific peptide inhibiting CXCR4 receptor. The analysis of cell lysates for the total expression of mitogen-activated protein kinases ensured the equal loading of proteins in different lanes (Fig. 5, top and bottom). AKT phosphorylation was not induced in PES 43 and PES 41 cells following treatment with CXCL12 (20 and 200 ng/mL; data not shown). The CXCR4 receptor also induced the migration of PES cells toward CXCL12. PES 41, PES 43, and PES 47 migrated toward CXCL12 (20 ng/mL) and the migration was specifically inhibited by AMD3100 (data not shown).

Fig. 3. Melanoma cell lines express other chemokine receptors. CXCR4, CXCL12, CCR10, and CCR7 expression was evaluated by reverse transcription-PCR in eight metastatic melanoma cell lines. Positive controls were as follows: peripheral blood lymphocytes (CXCR4), ovarian cancer cell line (CXCL12), and lymphoblastic cell lines (CCR7 and CCR10).

Clin Cancer Res 2006;12(8) April 15, 2006

Discussion The role of CXCR4 in human melanoma metastases was investigated. CXCR4 expression was evaluated on 63 melanoma metastases and on 8 cell lines isolated from melanoma metastases. CXCR4 was detectable in 33 out of 63 melanoma metastases by immunohistochemistry (52.4%; 15 of 29 s.c., 12 of 22 lymph node, 6 of 12 visceral metastases). In cell lines isolated from melanoma metastases, CXCR4 receptor was expressed and functional because CXCL12, the specific CXCR4 ligand induced Erk1-2 activation, cell migration, and cell growth. Several chemokine receptors have been described on human melanoma cells such as CXCR4 (21, 31), CCR7 (23), CXCR3 (24), CCR10 (25), and CXCR2 (32). In this article, CXCR4 expression was described in eight metastatic melanoma cell lines with a wide range in the receptor level. Three cell lines derived from the same patient (PES 43, PES 41, and PES 47) expressed different levels and activity of the receptor. In agreement with Murakami et al. (26), PES 43 cell line derived from lung metastasis expressed high levels of CXCR4 receptor. PES 43 cells also expressed CCR10, a chemokine receptor mainly described in s.c. melanoma metastasis (25). The two cell lines, PES 41 and PES 47, originated from s.c. nodules, expressed the CCR7 chemokine receptor in addition to CXCR4. PES 47 cells expressed the mRNA for the CXCL12, the specific CXCR4 ligand, which is not otherwise detectable by ELISA and cytofluorimetric detection. These evidences suggest that other chemokine receptors and the production of ligand at the metastasization site may affect the efficacy of the process. The effect of the CXCR4-CXCL12 axis activation on the cell growth of melanoma cells is not dramatic, although present in stressed cell conditions. This result is in agreement with the intrinsic resistance of melanoma cells to conventional antineoplastic therapies (33). As assessed by the specific Erk1-2 activation, the receptor is functional in metastatic cell lines mainly in encouraging the cells’ motility. Erk phosphorylation induction, cell growth, and migration are successfully inhibited

2430

Downloaded from clincancerres.aacrjournals.org on March 9, 2013 Copyright © 2006 American Association for Cancer Research

www.aacrjournals.org

DOI:10.1158/1078-0432.CCR-05-1940 CXCR4 in Melanoma Metastases

Fig. 4. CXCL12 increases growth of human melanoma cells. The effect of CXCL12 on cell proliferation was assessed through hemocytometric cell count in different culture conditions as previously reported.

by AMD3100 and T22 inhibitory peptide; thus, inhibition of the pathway may be very helpful in an early step of metastasization. Malignant melanoma represents a peculiar neoplasm in which disease explosion follows a silent behavior. Previous results showed that the expression of CXCR4 correlated with a

worse prognosis in terms of disease-free survival and overall survival (28). Here, the analysis of the data through a KaplanMeier curve showed no statistically significant differences in terms of overall survival between patients with CXCR4 and CXCR4+ metastases (data not shown). This evidence may represent the inefficacy of a single factor to affect prognosis in

Fig. 5. AMD3100 inhibited the CXCL12induced activation of ERK1and ERK2 phosphorylation in human melanoma cells. Changes in the phosphorylation of p44/42 (Erk1/2) kinases were analyzed by Western blotting. PES 43 and PES 41cells were serum-starved and treated with CXCL12 (20 ng/mL) in the presence or absence of AMD3100 (1 Amol/L; top). ERK1/2 activation was also inhibited in PES 43 human melanoma cells byT22, a specific peptide inhibiting CXCR4 receptor (bottom). The analysis of cell lysates for the total expression of mitogen-activated protein kinases ensured the equal loading of proteins in the different lanes (top and bottom).

www.aacrjournals.org

2431

Clin Cancer Res 2006;12(8) April 15, 2006

Downloaded from clincancerres.aacrjournals.org on March 9, 2013 Copyright © 2006 American Association for Cancer Research

DOI:10.1158/1078-0432.CCR-05-1940 Human Cancer Biology

an advanced state of disease. Nevertheless, CXCR4 expression remains in 33 of 63 melanoma metastases, suggesting that the receptor is involved in the metastasization process and thus is a suitable target for therapy. The identification of new prognostic markers, such as CXCR4, features a subgroup of patients with a high risk of relapse, and are thus suitable for a tight follow up. Recently, the relationship between lymphocytes which expressed CC or CXC chemokine receptor and soluble chemokines derived from the host tissues in proximity to the tumor or the tumor itself was determined. Differential chemokine receptor expression by activated tumor-specific CD8+ T cells could be associated with divergent clinical outcomes, suggesting that they may serve as a biomarker of potential clinical responsiveness to immunotherapy. Expression of CXCR3 by CD8+ T cells was reported to be associated with survival in melanoma patients with stage III disease (34). We are currently evaluating the peripheral blood lymphocytes of melanoma patients for the distribution of specific chemokine receptors in CD8+ T cell subgroups. Moreover, we are tracing CXCR4 expression during the metastatic process through the detection of CXCR4-positive melanoma cells in biopsies of suspicious lymph nodes. The inhibition of CXCR4 has been previously described. AMD3100, the small-molecule byciclam, is the better studied CXCR4 inhibitor. This molecule was shown to be safe in healthy volunteers (35), and is an effective agent for the rapid mobilization of CD34+ cells in patients who have received

prior chemotherapy in multiple myeloma and non – Hodgkin lymphoma (36, 37). Recently, novel low-molecular weight CXCR4 antagonists were developed and aimed at an orally available molecule which was also described by Hatse et al. (38). Also, the orally available CXCR4 inhibitor, AMD3465, inhibited intracellular calcium signaling, chemotaxis, CXCR4 endocytosis, and mitogen-activated protein kinase phosphorylation induced by CXCL12. T22 is one of the several described inhibitory peptides (39 – 41). Some antagonist peptides in microcapsules have been tested in in vivo studies demonstrating antimetastatic activity in murine models (42). The identification of new peptides with stronger CXCR4 antagonistic activities have recently been described (43, 44). These results indicate that the CXCR4 receptor is frequently expressed in melanoma metastases, and is part of an active transduction pathway that is efficiently inhibited by the antagonist. Taken together, these evidences strongly support the value of CXCR4 inhibition in melanoma patients encouraging the use of active inhibitors such as AMD3100, or its derivatives, in human melanoma clinical trials.

Acknowledgments We thank Dr. Sam Hwang from the Dermatology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, for providing the T22 peptide. We also thank the family of Sandro Fabiani for supporting melanoma research.

References 1. Tsao H, Atkins MB, Sober AJ. Management of cutaneous melanoma. N Engl J Med 2004;351: 998 ^ 1012. 2. Leiter U, Meier F, Schittek B, Garbe C. The natural course of cutaneous melanoma. J Surg Oncol 2004; 86:172 ^ 8. 3. Ascierto PA, Scala S, OttaianoA, et al. Adjuvant treatment of malignant melanoma: where are we? Crit Rev Oncol Hematol 2006;57:45 ^ 52. 4. Alonso SR, Ortiz P, Pollan M, et al. Progression in cutaneous malignant melanoma is associated with distinct expression profiles: a tissue microarray-based study. Am J Pathol 2004;164:193 ^ 203. 5. Moser B, Wolf M, Walz A, Loetscher P. Chemokines: multiple levels of leukocyte migration control. Trends Immunol 2004;25:75 ^ 84. 6. Balkwill F. The significance of cancer cell expression of the chemokine receptor CXCR4. Semin Cancer Biol 2004;14:171 ^ 9. 7. Balkwill F. Cancer and the chemokine network. Nat Rev Cancer 2004;4:540 ^ 50. 8. Zlotnik A. Chemokines in neoplastic progression. Semin Cancer Biol 2004;14:181 ^ 5. 9. Epstein RJ. The CXCL12 ^ 4 chemotactic pathway as a target of adjuvant breast cancer therapies. Nat Rev Cancer 2004;4:901 ^ 9. 10. Castellone MD, Guarino V, De Falco V, et al. Functional expression of the CXCR4 chemokine receptor is induced by RET/PTC oncogenes and is a common event in human papillary thyroid carcinomas. Oncogene 2004;23:5958 ^ 67. 11. Libura J, Drukala J, Majka M, et al. CXCR4-SDF-1 signaling is active in rhabdomyosarcoma cells and regulates locomotion, chemotaxis, and adhesion. Blood 2002;100:2597 ^ 606. 12. Taichman RS, Cooper C, Keller ET, Pienta KJ, Taichman NS, McCauley LK. Use of the stromal cellderived factor-1/CXCR4 pathway in prostate cancer metastases to bone. Cancer Res 2002;62:1832 ^ 7. 13. Zeelenberg IS, Ruuls-Van Stalle L, Roos E. The chemokine receptor CXCR4 is required for outgrowth of

colon carcinoma micrometastases. Cancer Res 2003; 63:3833 ^ 9. 14. Kato M, Kitayama J, Kazama S, Nagawa H. Expression pattern of CXC chemokine receptor-4 is correlated with lymph node metastases in human invasive ductal carcinoma. Breast Cancer Res 2003;5: 144 ^ 50. 15. Spano JP, Andre F, Morat L, et al. Chemokine receptor CXCR4 and early-stage non-small cell lung cancer: pattern of expression and correlation with outcome. Ann Oncol 2004;15:613 ^ 7. 16. Jordan NJ, Kolios G, Abbot SE, et al. Expression of functional CXCR4 chemokine receptors on human colonic epithelial cells. J Clin Invest 1999;104: 1061 ^ 9. 17. Dwinell MB, Eckmann L, Leopard JD, Varki NM, Kagnoff MF. Chemokine receptor expression by human intestinal epithelial cells. Gastroenterology 1999; 117:359 ^ 67. 18. Scotton CJ, Wilson JL, Scott K, et al. Multiple actions of the chemokine CXCL12 on epithelial tumor cells in human ovarian cancer. Cancer Res 2002;62: 5930 ^ 8. 19. Mochizuki H, Matsubara A, Teishima J, et al. Interaction of ligand-receptor system between stromalcell-derived factor-1 and CXC chemokine receptor 4 in human prostate cancer: a possible predictor of metastases. Biochem Biophys Res Commun 2004;320: 656 ^ 63. 20. Marchesi F, Monti P, Leone BE, et al. Increased survival, proliferation, and migration in metastatic human pancreatic tumor cells expressing functional CXCR4. Cancer Res 2004;64:8420 ^ 7. 21. Robledo MM, Bartolome RA, Longo N, et al. Expression of functional chemokine receptors CXCR3 and CXCR4 on human melanoma cells. J Biol Chem 2001;276:45098 ^ 105. 22. Bartolome RA, Galvez BG, Longo N, et al. Stromal cell-derived factor-1a promotes melanoma cell invasion across basement membranes involving stimulation of membrane-type 1 matrix metalloproteinase

Clin Cancer Res 2006;12(8) April 15, 2006

2432

and Rho GTPase activities. Cancer Res 2004;64: 2534 ^ 43. 23. Takeuchi H, Fujimoto A,Tanaka M,YamanoT, Hsueh E, Hoon DSB. CCL21chemokine regulates chemokine receptor CCR7 bearing malignant melanoma cells. Clin Cancer Res 2004;10:2351 ^ 8. 24. Kawada K, Sonoshita M, Sakashita H, et al. Pivotal role of CXCR3 in melanoma cell metastases to lymph nodes. Cancer Res 2004;64:4010 ^ 7. 25. Murakami T, Cardones AR, Finkelstein SE, et al. Immune evasion by murine melanoma mediated through CC chemokine receptor-10. J Exp Med 2003;198: 1337 ^ 47. 26. MurakamiT, MakiW, Cardones AR, et al. Expression of CXC chemokine receptor-4 enhances the pulmonary metastatic potential of murine B16 melanoma cells. Cancer Res 2002;62:7328 ^ 34. 27. Cardones AR, Murakami T, Hwang ST. CXCR4 enhances adhesion of B16 tumor cells to endothelial cells in vitro and in vivo via h(1) integrin. Cancer Res 2003;63:6751 ^ 7. 28. Scala S, OttaianoA, Ascierto PA, et al. Expression of CXCR4 predicts poor prognosis in patients with malignant melanoma. Clin Cancer Res 2005;11:1835 ^ 41. 29. Roland J, Murphy BJ, Alu B, et al. Role of the intracellular domains of CXCR4 in SDF-1-mediated signaling. Blood 2003;101:399 ^ 406. 30. OttaianoA, di Palma A, Napolitano M, et al. Inhibitory effectsofanti-CXCR4antibodiesonhumancoloncancer cells. Cancer Immunol Immunother 2005;54:781 ^ 91. 31. Muller A, Homey B, Soto H, et al. Involvement of chemokine receptors in breast cancer metastases. Nature 2001;410:50 ^ 6. 32. Varney ML, Li A, Dave BJ, Bucana CD, Johansson SL, Singh RK. Expression of CXCR1 and CXCR2 receptors in malignant melanoma with different metastatic potential and their role in interleukin-8 (CXCL8)-mediated modulation of metastatic phenotype. Clin Exp Metastasis 2003;20:723 ^ 31. 33. Soengas MS, Lowe SW. Apoptosis and melanoma chemoresistance. Oncogene 2003;22:3138 ^ 51.

Downloaded from clincancerres.aacrjournals.org on March 9, 2013 Copyright © 2006 American Association for Cancer Research

www.aacrjournals.org

DOI:10.1158/1078-0432.CCR-05-1940 CXCR4 in Melanoma Metastases 34. Mullins IM, Slingluff CL, Lee JK, et al. CXC chemokine receptor 3 expression by activated CD8+ T cells is associated with survival in melanoma patients with stage III disease. Cancer Res 2004; 64:7697 ^ 701. 35. Liles WC, Broxmeyer HE, Rodger E, et al. Mobilization of hematopoietic progenitor cells in healthy volunteers byAMD3100, a CXCR4 antagonist. Blood 2003; 102:2728 ^ 30. 36. Devine SM, Flomenberg N, Vesole DH, et al. Rapid mobilization of CD34+ cells following administration of the CXCR4 antagonist AMD3100 to patients with multiple myeloma and non-Hodgkin’s lymphoma. J Clin Oncol 2004;22:1095 ^ 102. 37. Broxmeyer HE, Orschell CM, Clapp DW, et al. Rapid mobilization of murine and human hematopoietic stem

www.aacrjournals.org

and progenitor cells with AMD3100, a CXCR4 antagonist. J Exp Med 2005;201:1307 ^ 18. 38. Hatse S, Princen K, De Clercq E, et al. AMD3465, a monomacrocyclic CXCR4 antagonist and potent HIV entry inhibitor. Biochem Pharmacol 2005;70: 752 ^ 61. 39. Murakami T, Nakajima T, Koyanagi Y, et al. A small molecule CXCR4 inhibitor that blocksTcell line-tropic HIV-1infection. J Exp Med 1997;186:1389 ^ 93. 40. Arakaki R,Tamamura H, Premanathan M, et al. T134, a small-molecule CXCR4 inhibitor, has no cross-drug resistance with AMD3100, a CXCR4 antagonist with a different structure. J Virol 1999;73:1719 ^ 23. 41. Liang Z,WuT, Lou H, et al. Inhibition of breast cancer metastases by selective synthetic polypeptide against CXCR4. Cancer Res 2004;64:4302 ^ 8.

2433

42. Takenaga M,Tamamura H, Hiramatsu K, et al. A single treatment with microcapsules containing a CXCR4 antagonist suppresses pulmonary metastases of murine melanoma. Biochem Biophys Res Commun 2004;320:226 ^ 32. 43. Tamamura H, Araki T, Ueda S, et al. Identification of novel low molecular weight CXCR4 antagonists by structural tuning of cyclic tetrapeptide scaffolds. J Med Chem 2005;48:3280 ^ 9. 44. Tamamura H, Hiramatsu K, Ueda S, et al. Stereoselective synthesis of [L-Arg-L/D-3-(2-naphthyl)alanine]-type (E)-alkene dipeptide isosteres and its application to the synthesis and biological evaluation of pseudopeptide analogues of the CXCR4 antagonist FC131. J Med Chem 2005;48: 380 ^ 91.

Clin Cancer Res 2006;12(8) April 15, 2006

Downloaded from clincancerres.aacrjournals.org on March 9, 2013 Copyright © 2006 American Association for Cancer Research

Lihat lebih banyak...

Comentários

Copyright © 2017 DADOSPDF Inc.