Accepted Manuscript The role of CXCL12 in tumor microenvironment

Wenfang Meng, Shihang Xue, Ye Chen PII: DOI: Reference:

S0378-1119(17)30828-4 doi:10.1016/j.gene.2017.10.015 GENE 42234

To appear in:

Gene

Received date: Accepted date:

10 February 2017 6 October 2017

Please cite this article as: Wenfang Meng, Shihang Xue, Ye Chen , The role of CXCL12 in tumor microenvironment. The address for the corresponding author was captured as affiliation for all authors. Please check if appropriate. Gene(2017), doi:10.1016/ j.gene.2017.10.015

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Meng W et al. 2017

The Role of CXCL12 in Tumor Microenvironment Wenfang Meng1,2, Shihang Xue3, Ye Chen1,2*

Division of Clinical Genetics and Genomics, The Children's Hospital, Zhejiang University School of

PT

1

RI

Medicine, Hangzhou, Zhejiang, China

Institute of Genetics, Zhejiang University, Hangzhou, Zhejiang, China.

3

Ningbo No.4 People’s Hospital, Ningbo, Zhejiang, China.

D

MA

NU

SC

2

PT E

*Corresponding author:

Ye Chen, Ph.D,

CE

Institute of Genetics, Zhejiang University,

AC

866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China

Tel: +86-571-88206760; Fax: +86-571-88206497

Email: [email protected]

ACCEPTED MANUSCRIPT Meng W et al. 2017

Abstract The chemokine ligand C-X-C motif chemokine ligand 12(CXCL12) is a kind of small molecules of cytokines that widely expressed in diversified tissues. Recent evidence suggests that CXCL12 plays an important role in the communication of tumor

PT

cells with their surrounding microenvironment. The interaction of CXCL12 and its

RI

receptors subsequently excite the downstream signaling pathways to affect tumor

SC

angiogenesis, tumor cell proliferation and chemoresistance, and thus represents a

NU

potential target for cancer therapy. Outpouring molecules targeting CXCL12/CXCR4 axis in tumor microenvironment combined with traditional chemotherapy have drawn

MA

more and more attentions, which will be a promising method in anti-cancer therapies. Our review focuses on these roles of CXCL12 and summarizes strategies for treating

PT E

D

cancer by disrupting this interaction with special emphasis on the CXCR4/CXCL12

AC

CE

axis.

ACCEPTED MANUSCRIPT Meng W et al. 2017

Introduction Tumor microenvironment (TME) is the local environment of tumorigenesis and tumor growth, composed of tumor cells and surroundings such as blood vessels, the extracellular matrix (ECM), other non-malignant cells such as bone marrow-derived

PT

dendritic cells, mesenchymal stem/stromal cells, fibroblasts, pericytes, immune cells,

RI

and the expression of their products, metabolic material, and also signaling molecules

SC

(Fig.1) (Junttila and de Sauvage, 2013; Quail and Joyce, 2013). TME affect whole

NU

process of tumors from happening to transfer, and tumor cells in turn influence the physical and chemical properties of the TME to make TME beneficial even stimulative

MA

to the growth of tumor(Mahadevan and Von Hoff, 2007).

D

Chemokines are a very important part of the cross talk between tumor cells and the

PT E

tumor microenvironment(Mantovani et al., 2008). Chemokine receptors and their specific chemokines control and regulate the tumor development, including leukocyte

CE

infiltration, tumor related angiogenesis, activation of the specific immune response to the tumor of host, stimulating tumor cell proliferation by autocrine or paracrine way, as

AC

well as control of tumor cell movements(Mbeunkui and Johann, 2009). The chemokine ligand C-X-C motif chemokine ligand 12 (CXCL12) and its receptor chemokine receptor 4 (CXCR4) are two key factors in the cross-talking, what makes them promising targets for cancer therapy. In this review, we summarize the role of CXCL12 in tumor growth, angiogenesis, metastasis, and TME-mediated chemoresistance.

ACCEPTED MANUSCRIPT Meng W et al. 2017

The CXCL12/CXCR4 axis Chemokines are small proteins possessing a common structural feature of conserved cysteine residues at the N-terminus with a molecular mass of between 8 and 10 kDa(Baggiolini, 1998). Chemokines have a common structural composition.

PT

According to the number of and relative spacing of the N-terminal cysteine residues,

RI

chemokines are divided into four families (CXC, CX3C, CC, and C)(Le et al., 2004;

SC

Vindrieux et al., 2009). CXCL12, also known as stromal-derived factor 1 (SDF-1), is

NU

widely secreted in different tissues by stromal cells, fibroblasts and epithelial cells in six different isoforms, encoded on chromosome 10q11. CXCR4 is an evolutionarily

MA

highly conserved G protein coupled receptor (GPCR) expressed on a great diversity of cell types, including lymphocytes, hematopoietic stem cells, endothelial cells,

D

epithelial cells, stromal fibroblasts and cancer cells. This allows them to migrate along

in many

PT E

CXCL12 gradients. It has been observed that the level of CXCL12/CXCR4 is increased types of cancer, such as breast cancer, pancreatic cancer, ovarian carcinoma,

CE

cervical carcinoma, leukemia diseases and the rest(Mahadevan and Von Hoff, 2007;

AC

Shen et al., 2009; Zeng et al., 2009; Ling et al., 2013; Kim and Park, 2014; Li et al., 2016). Furthermore, CXCR4 expression has been defined as a prognostic factor in several human tumor types. As shown in the Fig.1, in the tumor microenvironment, some conditions like hypoxia or toxins increase the levels of CXCL12 and thus generate the migration of tumor cells expressing CXCR4, leading them to safer residence constantly. And the heterotrimeric G protein dissociates into subunits (the GTP-bound α,β,and γ) when CXCR4 binding to CXCL12(Chatterjee et al., 2014),

ACCEPTED MANUSCRIPT Meng W et al. 2017

thus contributing to the tumor angiogenesis, tumor cell survival, proliferation and chemoresistance through the excitation of a variety of downstream signaling pathways(Burger and Kipps, 2006; Wojcechowskyj et al., 2011).

PT

CXCL12 enhances adhesion ability of tumor cells CXCL12 regulates adhesion of tumor cell with laminin, fibrinogen, stromal cells

RI

and endothelial cell by activating various cell surface adhesion molecules (e.g.

SC

integrins). It has been shown that CXCL12 could increase the adhesion rate of

NU

pancreatic cancer cells to laminin (the main component of basement membrane) and enhance the capacity of cells to penetrate the matrigel (artificial reconstruction

MA

basement membrane)(Mori et al., 2004). Compared with normal cells, the prostate

D

cancer cells pretreated with CXCL12 significantly represented higher adhesion of

PT E

osteosarcomas and endothelial cell lines in vitro in a dose-dependent manner(Taichman et al., 2002). In another similar assay, CXCL12 was found to increase adhesion of PC-3

CE

cell to the human umbilical vein endothelial cell monolayer(Kukreja et al., 2005). Moreover, CXCL12 increased the level of integrin a5β3 expression in the tumor cell

AC

membrane, and thus enhanced the adhesion of prostate tumor cells to human endothelium or extracellular matrix proteins laminin, collagen, and fibronectin(Engl et al., 2006). The effects of CXCL12 on the expression and activity of integrins on cell surfaces may be crucial in adhesion to fibronectin and collagen I in prostate cancer cell(Dehghani et al., 2014). The adhesion of HeLa cells to fibronectin and laminin was increased when CXCL12 was added in the medium. In ovarian cancer cell lines, CXCL12 induced integrin β1 expression which leads to the increased adhesion of

ACCEPTED MANUSCRIPT Meng W et al. 2017

tumor cells to laminin(Shen et al., 2009). In small cell lung cancer cell lines, CXCL12/CXCR4 interaction could induce the expression of integrin and promote adhesion of cancer cells to vascular cell adhesion molecule-1 (VCAM-1), fibronectin and collagen and also vascular endothelial cells(Hartmann et al., 2005).

PT

CXCL12 promotes tumor angiogenesis and metastasis

RI

CXCL12/CXCR4 axis is closely related to angiogenesis which supplying nutrient

SC

to tumor cells and giving rise to excretion of tumor cells metabolites efficiently.

NU

CXCL12 can stimulate angiogenesis directly or indirectly. Vascular endothelial growth factor (VEGF) is an important cytokine that induces tumor angiogenesis and promotes

MA

tumor metastasis. VEGF can induce endothelial cells to express MMP-2, MMP-9, to

D

stimulate chemotaxis of endothelial cell and the formation of capillary channels, and

PT E

thus indirectly regulate angiogenesis. Salvucci and other studies have shown that CXCL12 could induce endothelial cells to express VEGF, and VEGF in turn could

CE

promote the expression of CXCL12 in vascular endothelial cell(Salvucci et al., 2002). Moreover, VEGF also induces expression of CXCR4 in cancer cells in autocrine way,

AC

thereby promotes cancer cells’ migration along CXCL12 gradients.

Metastasis is an important biological characteristic of malignant tumors. CXCL12 can not only induce expression of matrix metalloproteinases (MMP) in cancer cells, but also up-regulate the activity of MMP in tumor microenvironment, which lead to the promoted tumor invasion and metastasis. CXCL12 could increase MMP-9 and MT1-MMP expression and activities, which regulated myeloma cells’ movement in the

ACCEPTED MANUSCRIPT Meng W et al. 2017

bone marrow(Parmo-Cabanas et al., 2006). By stimulating different types of cells to secrete MMP-2 CXCL12/CXCR4 axis could improve the migration of nerve cells along the corpus callosum(Mao et al., 2016). CXCR4 was highly expressed in tumor cell of patients with liver metastases of colorectal cancer, while high expression of its

PT

ligand CXCL12 was found in the most common metastatic sites of colorectal cancer,

RI

such as lymph nodes, liver, lung(Kim et al., 2006). Sun et al also confirmed that

SC

down-regulation of CXCR4 expression could also inhibit distant metastasis of prostate cancer(Sun et al., 2005). In addition, the use of neutralizing antibodies, specific

NU

peptides, and siRNAs to block CXCR4 expression significantly inhibited the metastasis

MA

of breast cancer to lymph nodes and lungs(Skobe et al., 2001). Though the molecular mechanism of tumor metastasis has not yet been fully elucidated, various studies have

D

indicated that the role of CXCL12/CXCR4 axis is significant during the progress.

PT E

CXCL12 contributes to tumor cell growth and survival

CE

Studies have shown that CXCL12/CXCR4 axis can regulate a variety of tumor cell proliferation(Kryczek et al., 2007; Wu et al., 2008). The binding of CXCL12 to its

AC

receptor CXCR4 can induce proliferation of various type of tumor cells by activating the extracellular signal-regulated kinase (ERK) and AKT signaling pathway. Activated ERK phosphorylates and regulates other cellular proteins and transcription factors, bringing about changes in gene expression and cell cycle process. Activated AKT plays a key role in tumor cell survival through inactivation of BCL-2 antagonist resulting in cell survival. CXCL12/CXCR4 signaling via AKT also resulted in inactivation of GSK3β and stabilization of β-catenin. Stabilized β-catenin moved to the nucleus

ACCEPTED MANUSCRIPT Meng W et al. 2017

activating gene transcription and promoting proliferation(Barbero et al., 2003). CXCL12-dependent tumor proliferation contributed to tumor growth by a sustained inhibition of cyclic AMP (cAMP) production(Yang et al., 2007). CXCL12 also protected CXCR4-positive cancer cells from apoptosis induced by both serum-free

PT

culture and chemo administration. In addition, tumor cells could express CXCL12 in

RI

paracrine form, stimulating tumor stromal cells to produce TNF, which in turn

SC

promoted tumor cell growth. These studies suggest that the CXCL12 / CXCR4 biological axis not only directly promotes cell proliferation but also indirectly through

NU

anti-apoptotic effects(Marchesi et al., 2004).

MA

Targeting the CXCR4/CXCL12 axis in tumor microenvironment

D

Up to now, several molecules have been developed to target CXCL12 or CXCR4

PT E

for the purpose of interfering with the tumor growth, and have stronger lethal effect with combination of other pharmaceuticals(Table.1). CTCE-9908 is a CXCL12

CE

analogue with inhibited and agonist activity. In two murine models of osteosarcoma, CTCE-9908 reduced osteosarcoma cells’ growth and adhesion, and the metastatic

AC

dissemination of cancer cells(Kim et al., 2008). Compared with controls receiving scrambled protein, the treatment with CTCE-9908 experimental group showed lower primary tumor growth rate in transgenic mouse model of breast cancer. In addition, CTCE-9908 gave rise to reduced protein expression of vascular endothelial growth factor (VEGF) and p-AKT/AKT(Hassan et al., 2011). Olaptesed pegol(ola-PEG), a high-affinity anti-CXCL12 spiegelmer represented as a successful agent targeting the interaction between bone marrow niches and tumor cells, thereby blocking or

ACCEPTED MANUSCRIPT Meng W et al. 2017

disrupting bone marrow colonization by multiple myeloma cells in a cancer xenograft model. It also diminished tumor mobilization, homing, and growth within the bone marrow niches. Combinations of ola-PEG with other chemotherapeutic agents were also likely to lead to synergistic effects(Roccaro et al., 2014). Plerixafor(AMD3100), a

PT

CXCR4 inhibitor and a hematopoetic stem cell mobilizer, competitively inhibited

RI

CXCL12 binding to CXCR4, which resulted in inhibition of initial establishment of

SC

prostate tumors in the bone(Conley-LaComb et al., 2016). AMD3100 blocked ligand-receptor binding of CXCL12/CXCR4 and reduced growth of ovarian cancer

NU

cells, in addition, modestly promoted overall survival of mice with metastatic ovarian

MA

cancer(Ray et al., 2011). AMD3100 undermined the interaction between tumor and stroma, inhibited the adhesion of leukemic cells and weakened the cell migration ability

D

to bone marrow and niche. It also brought about mobilization of the leukemia cells to

PT E

enhance cytotoxicity of cytrarabine(Ara-C) on account of the absence of the protection of the tumor microenvironment. Treatment of AMD3100 combined with G-CSF had a

CE

stronger impact on therapy induced by Ara-C(Shen et al., 2016). In mouse model of

AC

breast cancer, AMD3465 decreased the cancer growth and metastases of breast cancer cells, by acting on the interaction of tumor cells and the tumor microenvironment(Ling et al., 2013). Compared with the conventional method of drug delivery, the targeted delivery of CXCR4-A-mFc antagonists by oncolytic Vaccinia viruses (OVV) showed an enhanced antitumor effect in destroying tumor vasculature and inhibiting breast cancer metastasis(Gil et al., 2013). In ovarian cancer, by inhibiting the expression of CXCL12, miR-448 inhibited cell proliferation, migration and invasion, serving as

ACCEPTED MANUSCRIPT Meng W et al. 2017

tumor suppressor(Lv et al., 2015). As a component of Epimedium koreanum, Baohuoside I could suppress cervical and breast cancer metastasis by downregulating CXCR4 expression(Kim and Park, 2014).

PT

CXCR7 impact on CXCL12 biology in tumor study For many years it was believed that CXCR4 was the only receptor for CXCL12.

RI

When chemotactic response to T lymphocytes of CXCL12 were studied, Balabanian et

SC

al., found that CXCL12 bound to RDC1 (previously used as an orphan receptor) and

NU

anti-RDC1 monoclonal antibodies probably inhibited the chemotactic response to T lymphocytes of CXCL12(Balabanian et al., 2005). Burns et al. discovered that on the

MA

13th day of the CXCR4 knockout mice embryonic development, the liver cells can still

D

bind CXCL12 in the study by chance. RDC1 had similarity with CXCR4 in amino acid

PT E

conserved sequences, so renamed CXCR7(Burns et al., 2006).

Membrane-associated CXCR7 is expressed on many tumor cell lines, on activated

CE

endothelial cells, on fetal liver cells, and on T lymphocytes(Balabanian et al., 2005;

AC

Burns et al., 2006). The expression of CXCR7 was found to be strongly positive in 97% (106/109)

human

breast

cancer

vascular

endothelial

cells

detected

by

immunohistochemistry, but almost undetectable in normal mammary vascular endothelial cells(Miao et al., 2007). Studies have shown that CXCR7 could reduce tumor cell apoptosis and promote its proliferation. Compared with wild-type cells, after 5 days’ treatment, the number of viable cells in CXCR7-transfected MDA MB 435s cells was significantly higher, although no difference was found in the total cells(Burns

ACCEPTED MANUSCRIPT Meng W et al. 2017

et al., 2006). CXCR7 also regulated expression of pro-angiogenic factors interleukin-8 and vascular endothelial growth factor, which may be involved in the regulation of angiogenesis(Wang et al., 2008). CXCL12/CXCR7 axis is also involved in the tumor invasion and metastasis process. Compared with wild-type cells, the number of cells

PT

adhered to human dermal microvascular endothelial cells (HDMEC) was significantly

RI

higher, and the invasive ability of CXCR7-over-expressed prostate cancer or C4-2B

SC

cells was higher in the presence of CXCL12. CXCR7 enhanced the invasiveness and metastasis of tumor cells by regulating the levels of cell adhesion molecules (FN1,

NU

CDH11 and CD44) and matrix metalloproteinase (MMP), such as MMP3, MMP10,

MA

MMP11 and HPSE(Wang et al., 2008). In vitro experiments it was also confirmed that CXCR7-transfected MDA MB 435s cells were more adherent to human umbilical vein

D

endothelial cells (HUVECs) than untransfected wild-type cells(Burns et al., 2006).

PT E

Similarly, in in vivo experiments inducing lung metastasis model in mice injected with tumor cells in the tail vein, tumors caused by injecting cells with 4T1-CXCR7-RNAi

CE

into the lungs were smaller than injecting the wild-type 4T1 WT cells, while the tumors

AC

caused by CXCR7-over-expressed cells transferred to the lungs were larger than the wild-type 4T1 WT cells’ group, indicating that CXCR7 expression in breast cancer cells enhanced the ability to metastasize to the lungs and proliferate in the lungs(Miao et al., 2007).

Conclusion It is well recognized that the interaction between tumor cells and TME is a rising target to improve anti-cancer treatment caused by protection of TME (Fig.1). Extensive

ACCEPTED MANUSCRIPT Meng W et al. 2017

bench studies indicate that targeting the CXCL12/CXCR4 axis may have beneficial actions for sensitizing tumor cells to chemical therapy; however, clinical researches to date are still lagged behind due to multiple reasons such as the limit of pharmacokinetic properties. Additional studies with new agents are required to prove whether

PT

interference of the CXCR4/CXCL12/CXCR7 axis in TME may increase the

RI

effectiveness of cancer therapy, and the process of drug approval needs to be

SC

accelerated.

NU

Acknowledgements

The work was supported by the project from the Natural Science Foundation of

MA

Zhejiang province (R15H080001), and the "Double First-rate" project initiatives of

PT E

Conflict of interest

D

Zhejiang University.

AC

CE

The authors declare no competing financial interests.

ACCEPTED MANUSCRIPT Meng W et al. 2017

Reference Baggiolini, M., 1998. Chemokines and leukocyte traffic. Nature 392, 565-8. Balabanian, K., Lagane, B., Infantino, S., Chow, K.Y., Harriague, J., Moepps, B., Arenzana-Seisdedos,

the orphan receptor RDC1 in T lymphocytes. J Biol Chem 280, 35760-6.

PT

F., Thelen, M. and Bachelerie, F., 2005. The chemokine SDF-1/CXCL12 binds to and signals through

RI

Barbero, S., Bonavia, R., Bajetto, A., Porcile, C., Pirani, P., Ravetti, J.L., Zona, G.L., Spaziante, R.,

SC

Florio, T. and Schettini, G., 2003. Stromal cell-derived factor 1 alpha stimulates human glioblastoma cell

NU

growth through the activation of both extracellular signal-regulated kinases 1/2 and Akt. Cancer Research 63, 1969-1974.

MA

Burger, J.A. and Kipps, T.J., 2006. CXCR4: a key receptor in the crosstalk between tumor cells and their microenvironment. Blood 107, 1761-1767.

PT E

D

Burns, J.M., Summers, B.C., Wang, Y., Melikian, A., Berahovich, R., Miao, Z., Penfold, M.E., Sunshine, M.J., Littman, D.R., Kuo, C.J., Wei, K., McMaster, B.E., Wright, K., Howard, M.C. and Schall, T.J., 2006. A novel chemokine receptor for SDF-1 and I-TAC involved in cell survival, cell

CE

adhesion, and tumor development. J Exp Med 203, 2201-13.

AC

Chatterjee, S., Behnam Azad, B. and Nimmagadda, S., 2014. The intricate role of CXCR4 in cancer. Adv Cancer Res 124, 31-82. Conley-LaComb, M.K., Semaan, L., Singareddy, R., Li, Y., Heath, E.I., Kim, S., Cher, M.L. and Chinni, S.R., 2016. Pharmacological targeting of CXCL12/CXCR4 signaling in prostate cancer bone metastasis. Mol Cancer 15, 68.

ACCEPTED MANUSCRIPT Meng W et al. 2017

Dehghani, M., Kianpour, S., Zangeneh, A. and Mostafavi-Pour, Z., 2014. CXCL12 Modulates Prostate Cancer Cell Adhesion by Altering the Levels or Activities of beta1-Containing Integrins. Int J Cell Biol 2014, 981750. Engl, T., Relja, B., Marian, D., Blumenberg, C., Muller, I., Beecken, W.D., Jones, J., Ringel, E.M.,

PT

Bereiter-Hahn, J., Jonas, D. and Blaheta, R.A., 2006. CXCR4 chemokine receptor mediates prostate

RI

tumor cell adhesion through alpha5 and beta3 integrins. Neoplasia 8, 290-301.

SC

Gil, M., Seshadri, M., Komorowski, M.P., Abrams, S.I. and Kozbor, D., 2013. Targeting CXCL12/CXCR4 signaling with oncolytic virotherapy disrupts tumor vasculature and inhibits breast

NU

cancer metastases. Proc Natl Acad Sci U S A 110, E1291-300.

MA

Hartmann, T.N., Burger, J.A., Glodek, A., Fujii, N. and Burger, M., 2005. CXCR4 chemokine receptor and integrin signaling co-operate in mediating adhesion and chemoresistance in small cell lung cancer

D

(SCLC) cells. Oncogene 24, 4462-71.

PT E

Hassan, S., Buchanan, M., Jahan, K., Aguilar-Mahecha, A., Gaboury, L., Muller, W.J., Alsawafi, Y., Mourskaia, A.A., Siegel, P.M., Salvucci, O. and Basik, M., 2011. CXCR4 peptide antagonist inhibits

CE

primary breast tumor growth, metastasis and enhances the efficacy of anti-VEGF treatment or docetaxel

AC

in a transgenic mouse model. Int J Cancer 129, 225-32. Junttila, M.R. and de Sauvage, F.J., 2013. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature 501, 346-54. Kim, B. and Park, B., 2014. Baohuoside I suppresses invasion of cervical and breast cancer cells through the downregulation of CXCR4 chemokine receptor expression. Biochemistry 53, 7562-9. Kim, J., Mori, T., Chen, S.L., Amersi, F.F., Martinez, S.R., Kuo, C., Turner, R.R., Ye, X., Bilchik, A.J., Morton, D.L. and Hoon, D.S.B., 2006. Chemokine receptor CXCR4 expression in patients with

ACCEPTED MANUSCRIPT Meng W et al. 2017

melanoma and colorectal cancer liver metastases and the association with disease outcome. Annals of Surgery 244, 113-120. Kim, S.Y., Lee, C.H., Midura, B.V., Yeung, C., Mendoza, A., Hong, S.H., Ren, L., Wong, D., Korz, W., Merzouk, A., Salari, H., Zhang, H., Hwang, S.T., Khanna, C. and Helman, L.J., 2008. Inhibition of the

PT

CXCR4/CXCL12 chemokine pathway reduces the development of murine pulmonary metastases. Clin

RI

Exp Metastasis 25, 201-11.

SC

Kryczek, I., Wei, S., Keller, E., Liu, R. and Zou, W., 2007. Stroma-derived factor (SDF-1/CXCL12) and human tumor pathogenesis. Am J Physiol Cell Physiol 292, C987-95.

NU

Kukreja, P., Abdel-Mageed, A.B., Mondal, D., Liu, K. and Agrawal, K.C., 2005. Up-regulation of

MA

CXCR4 expression in PC-3 cells by stromal-derived factor-1 alpha (CXCL12) increases endothelial adhesion and transendothelial migration: Role of MEK-ERK signaling pathway-dependent NF-kappa B

D

activation. Cancer Research 65, 9891-9898.

PT E

Le, Y., Zhou, Y., Iribarren, P. and Wang, J., 2004. Chemokines and chemokine receptors: their manifold roles in homeostasis and disease. Cell Mol Immunol 1, 95-104.

CE

Li, S.H., Dong, W.C., Fan, L. and Wang, G.S., 2016. Suppression of chronic lymphocytic leukemia

AC

progression by CXCR4 inhibitor WZ811. American Journal of Translational Research 8, 3812-3821. Ling, X., Spaeth, E., Chen, Y., Shi, Y., Zhang, W., Schober, W., Hail, N., Jr., Konopleva, M. and Andreeff, M., 2013. The CXCR4 antagonist AMD3465 regulates oncogenic signaling and invasiveness in vitro and prevents breast cancer growth and metastasis in vivo. PLoS One 8, e58426. Lv, Y., Lei, Y., Hu, Y., Ding, W., Zhang, C. and Fang, C., 2015. miR-448 negatively regulates ovarian cancer cell growth and metastasis by targeting CXCL12. Clin Transl Oncol 17, 903-9.

ACCEPTED MANUSCRIPT Meng W et al. 2017

Mahadevan, D. and Von Hoff, D.D., 2007. Tumor-stroma interactions in pancreatic ductal adenocarcinoma. Mol Cancer Ther 6, 1186-97. Mantovani, A., Allavena, P., Sica, A. and Balkwill, F., 2008. Cancer-related inflammation. Nature 454, 436-44.

PT

Mao, W.F., Yi, X., Qin, J.B., Tian, M.L. and Jin, G.H., 2016. CXCL12/CXCR4 Axis Improves

SC

Brain Injury in Rats. Neurochemical Research 41, 1315-1322.

RI

Migration of Neuroblasts Along Corpus Callosum by Stimulating MMP-2 Secretion After Traumatic

Marchesi, F., Monti, P., Leone, B.E., Zerbi, A., Vecchi, A., Piemonti, L., Mantovani, A. and Allavena,

NU

P., 2004. Increased survival, proliferation, and migration in metastatic human pancreatic tumor cells

MA

expressing functional CXCR4. Cancer Res 64, 8420-7.

Mbeunkui, F. and Johann, D.J., Jr., 2009. Cancer and the tumor microenvironment: a review of an

D

essential relationship. Cancer Chemother Pharmacol 63, 571-82.

PT E

Miao, Z., Luker, K.E., Summers, B.C., Berahovich, R., Bhojani, M.S., Rehemtulla, A., Kleer, C.G., Essner, J.J., Nasevicius, A., Luker, G.D., Howard, M.C. and Schall, T.J., 2007. CXCR7 (RDC1)

CE

promotes breast and lung tumor growth in vivo and is expressed on tumor-associated vasculature. Proc

AC

Natl Acad Sci U S A 104, 15735-40. Mori, T., Doi, R., Koizumi, M., Toyoda, E., Ito, D., Kami, K., Masui, T., Fujimoto, K., Tamamura, H., Hiramatsu, K., Fujii, N. and Imamura, M., 2004. CXCR4 antagonist inhibits stromal cell-derived factor 1-induced migration and invasion of human pancreatic cancer. Mol Cancer Ther 3, 29-37. Parmo-Cabanas, M., Molina-Ortiz, I., Matias-Roman, S., Garcia-Bernal, D., Carvajal-Vergara, X., Valle, I., Pandiella, A., Arroyo, A.G. and Teixido, J., 2006. Role of metalloproteinases MMP-9 and

ACCEPTED MANUSCRIPT Meng W et al. 2017

MT1-MMP in CXCL12-promoted myeloma cell invasion across basement membranes. J Pathol 208, 108-18. Quail, D.F. and Joyce, J.A., 2013. Microenvironmental regulation of tumor progression and metastasis. Nat Med 19, 1423-37.

PT

Ray, P., Lewin, S.A., Mihalko, L.A., Schmidt, B.T., Luker, K.E. and Luker, G.D., 2011. Noninvasive

RI

imaging reveals inhibition of ovarian cancer by targeting CXCL12-CXCR4. Neoplasia 13, 1152-61.

SC

Roccaro, A.M., Sacco, A., Purschke, W.G., Moschetta, M., Buchner, K., Maasch, C., Zboralski, D., Zollner, S., Vonhoff, S., Mishima, Y., Maiso, P., Reagan, M.R., Lonardi, S., Ungari, M., Facchetti, F.,

NU

Eulberg, D., Kruschinski, A., Vater, A., Rossi, G., Klussmann, S. and Ghobrial, I.M., 2014. SDF-1

MA

inhibition targets the bone marrow niche for cancer therapy. Cell Rep 9, 118-28. Salvucci, O., Yao, L., Villalba, S., Sajewicz, A., Pittaluga, S. and Tosato, G., 2002. Regulation of

D

endothelial cell branching morphogenesis by endogenous chemokine stromal-derived factor-1. Blood

PT E

99, 2703-11.

Shen, X., Wang, S., Wang, H., Liang, M., Xiao, L. and Wang, Z., 2009. The role of SDF-1/CXCR4 axis

CE

in ovarian cancer metastasis. J Huazhong Univ Sci Technolog Med Sci 29, 363-7.

AC

Shen, Z.H., Zeng, D.F., Kong, P.Y., Ma, Y.Y. and Zhang, X., 2016. AMD3100 and G-CSF disrupt the cross-talk between leukemia cells and the endosteal niche and enhance their sensitivity to chemotherapeutic drugs in biomimetic polystyrene scaffolds. Blood Cells Mol Dis 59, 16-24. Skobe, M., Hawighorst, T., Jackson, D.G., Prevo, R., Janes, L., Velasco, P., Riccardi, L., Alitalo, K., Claffey, K. and Detmar, M., 2001. Induction of tumor lymphangiogenesis by VEGF-C promotes breast cancer metastasis. Nat Med 7, 192-8.

ACCEPTED MANUSCRIPT Meng W et al. 2017

Sun, Y.X., Schneider, A., Jung, Y., Wang, J., Dai, J.L., Wang, J.C., Cook, K., Osman, N.I., Koh-Paige, A.J., Shim, H., Pienta, K.J., Keller, E.T., McCauley, L.K. and Taichman, R.S., 2005. Skeletal localization and neutralization of the SDF-1(CXCL12)/CXCR4 axis blocks prostate cancer metastasis and growth in osseous sites in vivo. Journal of Bone and Mineral Research 20, 318-329.

PT

Taichman, R.S., Cooper, C., Keller, E.T., Pienta, K.J., Taichman, N.S. and McCauley, L.K., 2002. Use of

RI

the stromal cell-derived factor-1/CXCR4 pathway in prostate cancer metastasis to bone. Cancer

SC

Research 62, 1832-1837.

Vindrieux, D., Escobar, P. and Lazennec, G., 2009. Emerging roles of chemokines in prostate cancer.

NU

Endocr Relat Cancer 16, 663-73.

MA

Wang, J.H., Shiozawa, Y.S., Wang, J.C., Wang, Y., Jung, Y.H., Pienta, K.J., Mehra, R., Loberg, R. and Taichman, R.S., 2008. The role of CXCR7/RDC1 as a chemokine receptor for CXCL12/SDF-1 in

D

prostate cancer. Journal of Biological Chemistry 283, 4283-4294.

PT E

Wojcechowskyj, J.A., Lee, J.Y., Seeholzer, S.H. and Doms, R.W., 2011. Quantitative phosphoproteomics of CXCL12 (SDF-1) signaling. PLoS One 6, e24918.

CE

Wu, M., Chen, Q., Li, D., Li, X., Li, X., Huang, C., Tang, Y., Zhou, Y., Wang, D., Tang, K., Cao, L.,

AC

Shen, S. and Li, G., 2008. LRRC4 inhibits human glioblastoma cells proliferation, invasion, and proMMP-2 activation by reducing SDF-1 alpha/CXCR4-mediated ERK1/2 and Akt signaling pathways. J Cell Biochem 103, 245-55. Yang, L., Jackson, E., Woerner, B.M., Perry, A., Piwnica-Worms, D. and Rubin, J.B., 2007. Blocking CXCR4-mediated cyclic AMP suppression inhibits brain tumor growth in vivo. Cancer Res 67, 651-8. Zeng, Z.H., Shi, Y.X., Samudio, I.J., Wang, R.Y., Ling, X.Y., Frolova, O., Levis, M., Rubin, J.B., Negrin, R.R., Estey, E.H., Konoplev, S., Andreeff, M. and Konopleva, M., 2009. Targeting the leukemia

ACCEPTED MANUSCRIPT Meng W et al. 2017

microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in

AC

CE

PT E

D

MA

NU

SC

RI

PT

AML. Blood 113, 6215-6224.

ACCEPTED MANUSCRIPT Meng W et al. 2017

Figure legends Figure 1. CXCL12/CXCR4 axis plays a critical role in regulation of tumor growth, metastasis, and development of chemoresistance. In the tumor microenvironment, cancer-associated fibroblast (CAF) and mesenchymal stem/stromal

PT

cells (MSC) constitutively express CXCL12, this paracrine signaling enhances the

RI

proliferation and survival of CXCR4-positive tumor cells. Moreover, low oxygen level

SC

in the microenvironment significantly improve expression of CXCL12, as well as the

NU

surface CXCR4 levels in tumor cells. CXCL12 binding promotes a three-dimensional CXCR4 conformation favoring Gαi protein dissociation into α and βγ subunits, and

MA

triggers multiple signal transduction pathways that are able to regulate intracellular calcium flux, transcription, proliferation and cell survival. Furthermore, these

D

CXCR4-positive tumour cells can migrate along the CXCL12 gradient to distant

PT E

organs, such as the CXCL12-rich bone marrow microenvironment, leading to the metastases. Evidence shows that CXCL12 agonist and CXCR4 antagonists can inhibit

CE

the cross-talk between tumour and stromal cells and mobilize tumor cells from this

AC

protective microenvironment, making them more sensitive to conventional drugs. Combination chemotherapies have been developed.

ACCEPTED MANUSCRIPT

AC

CE

PT E

D

MA

NU

SC

RI

PT

Meng W et al. 2017

ACCEPTED MANUSCRIPT Meng W et al. 2017

Table 1. Effective targeting of CXCL12/CXCR4 in the microenvironment Descriptio

Indication/Conditio

n

n

Function

R4

NU

antibody

Small

BLM-induced

1070)

molecule CXCR4

Chow LN,

pulmonary and

leukocyte

Schreiner P, et

CCl4-induced

mobilization and

al. 2016

hepatic fibrosis in

decreasing

(PMID:

mice

extracellular

26998906)

D

PT E

HIV infections

AC

CE

NCT01374503

Increasing

MA

AMD070(AMD1

antagonist

RI

Healthy volunteers

ence

SC

Anti-CXC

ALX-0651

Trial number/refer

PT

Name

matrix deposition.

HIV-1 entry

NCT00089466

inhibitor

HIV infections,X4

HIV-1 entry

tropic virus

inhibitor

HIV infections

Inhibiting membrane fusion

NCT00361101

NCT00063804

ACCEPTED MANUSCRIPT Meng W et al. 2017

and viral entry

Small

Glioma,Anaplastic

HSC mobilization NCT01339039

bil, plerixafor)

molecule

Astrocytoma,Anapla

in patients with

CXCR4

stic

NHL and multiple

antagonist

Oligodendroglioma,

myeloma

PT

AMD3100(Mozo

RI

Mixed Anaplastic

NU

Acute Myeloid

SC

Oligoastrocytoma

NCT01141543

Leukemic Cells

MA

Leukemia(AML)

Mobilization of

Chronic lymphocytic

NCT01373229

PT E

D

leukemia

Mobilization and

Neoplasms

Transplantation

CE

Hematologic

NCT00914849

of HLA-Matched

AC

Sibling Donor Hematopoietic Stem Cells

Chronic

Make CLL/SLL

Lymphocytic

cells more

Leukemia

sensitive to being

NCT00694590

ACCEPTED MANUSCRIPT Meng W et al. 2017

(CLL);Small

killed by

Lymphocytic

rituximab

Lymphoma (SLL)

Disrupting the

Leukemia(AML)

interaction

NCT00512252

PT

Acute Myeloid

RI

between AML

SC

blasts and the

MA

NU

marrow

D

Myelokathexis

AC

CE

PT E

(WHIM syndrome)

microenvironmen t

Mobilizing

NCT00967785

hematopoietic stem cells

Multiple

Stem cell

Myeloma;Lymphom

mobilization

NCT02098109

a, Non-Hodgkin

Lymphoma

Mobilization and Collection of Peripheral Hematopoietic

NCT02221492

ACCEPTED MANUSCRIPT Meng W et al. 2017

Stem Cells

Mobilization of

,Neuroblastoma,

Haematopoietic

Brain Tumors

Stem Cells Into

NCT01288573

PT

Ewing sarcoma

RI

Peripheral Blood,

Mobilize

Multiple Myeloma

NCT00396383

SC

Peripheral Blood

NU

Progenitor Cells

AC

CE

PT E

D

MA

Healthy volunteers

Peripheral Blood

NCT00082329

Hematopoietic Progenitor Cell Mobilization with G-CSF

Chronic

Interrupting

Lymphocytic

communication

Leukemia,

between stromal

Lymphoma,

cells and cancer

NCT01610999

Multiple Myeloma

AMD3465

CXCR4

Primary Chronic

Enhancing

Zeng Z,

Lymphocytic

chemotherapy-in

Samudio IJ, et

ACCEPTED MANUSCRIPT Meng W et al. 2017

Leukemia,Acute

duced apoptosis

al. 2006

Myelogenous

(PMID:

Leukemia

17172414)

Acute Myeloid

Mobilized of

Leukemia

AML cells and

PT

antagonist

Zeng, Z. H., et al. 2009

RI

progenitor cells to (PMID: 18955566)

SC

the blood

NU

circulation

Allosteric

ATI-2341

AC

CE

PT E

D

MA

agonist

Baohuoside I

Mobilizer of bone

Tchernychev,

marrow

B., et al. 2010

polymorphonucle

(PMID:

ar neutrophils and

21139054)

hematopoietic stem and progenitor cells

CXCR4

cervical cancer and

Suppression of

Kim, B. and B.

expression

breast cancer

cancer metastasis

Park, et al. 2014 (PMID:

regulator

25407882)

BL-8040

Peptide

Acute Myeloid

NCT01838395

ACCEPTED MANUSCRIPT Meng W et al. 2017

(T140

(BKT-140)

Leukemia

analogue) Mobilization of

Leukemia

leukemic blasts

Chronic Myeloid

Mobilization of

Leukemia

CML leukemia

NCT02502968

NCT02115672

RI

PT

Acute Myeloid

SC

stem cells

Peptide (T140

PT E

(BKT-140)

Multiple Myeloma

D

BL-8040

MA

NU

Healthy volunteers

AC

BMS-936564

CE

analogue)

(MDX-1338)

Mobilization of

NCT02073019

Hematopoietic Stem Cells

Induction of the

NCT01010880

exit of blood cells from the bone marrow to the peripheral blood

Anti-CXC

Acute Myeloid

R4

Leukemia,Diffuse

antibody

large B-cell leukemia,Follicular lymphoma

NCT01120457

ACCEPTED MANUSCRIPT Meng W et al. 2017

Acute Myeloid

NCT02305563

Leukemia

Jurkat human

Mobilizer of

agonist

lymphoblastoid cell

polymorphonucle

al. 2005

peptide

line

ar neutrophil and

(PMID:

hematopoietic

15730853)

PT

CXCR4

SC

CTCE-0021

NCT01359657

RI

Multiple myeloma

Pelus, L. M., et

CXCL12

CTCE-9908

MA

NU

stem and

prostate cancer

Decreasing the

Wong, D., et

invasion of tumor

al. 2014

D

mimetic

progenitor cells

esophageal cancer

AC

CE

PT E

(PMID: 24472670)

Reducing

Drenckhan, A.,

metastases and

et al. 2013

growth of tumor

(PMID: 23117118)

prostate tumor

Reduction of

Porvasnik, S.,

tumor cell

et al. 2009

proliferation

(PMID:

ACCEPTED MANUSCRIPT Meng W et al. 2017

19588526)

breast cancer

Reduction of

Huang, E. H.,

metastases

et al. 2009

Faber A,

mimetic

bone marrow

Roderburg C,

recovery

et al. 2007

SC

Mobilization

MA

PT E

organic

D

Small

KRH-1636

AC

CE

molecule

LY2510924

19482312)

CXCL12

NU

CTCE-0214

RI

PT

(PMID:

CXCR4

Metastatic Clear Cell

antagonist

Renal Cell

(PMID: 17541466)

Anti–HIV-1

Ichiyama K, et

activity

al. 2003 (PMID: 12642669)

NCT01391130

Carcinoma

Extensive Stage Small Cell Lung Carcinoma

NCT01439568

ACCEPTED MANUSCRIPT Meng W et al. 2017

Small

MSX122

anti-tumor

Solid Tumors

organic

NCT00591682

activity

molecule

Healthy Volunteers

Effect in

NCT00976378

PT

Anti-CXC L12

autologous stem

antibody

cell

RI

NOX-A12

SC

transplantation

MA

NU

HSC transplantation

PT E

D

Multiple myeloma

Mobilization of Hematopoietic Stem Cells

Mobilization of

CE

AC

POL6326

Small

Metastatic Breast

molecule

Cancer

NCT01521533

plasma cells

Chronic lymphocytic Mobilization of leukemia

NCT01194934

NCT01486797

CLL cells

NCT01837095

Hematological

Mobilization and

Malignancies

Transplantation of HLA-Matched Sibling Donor

NCT01413568

ACCEPTED MANUSCRIPT Meng W et al. 2017

Hematopoietic Stem Cells

Hematopoietic

Multiple Myeloma

NCT01105403

PT

stem cell

Healthy volunteers

RI

mobilization

Hematopoietic

NCT01841476

SC

stem cell

Multiple

antagonist

Myeloma, Non-Hod

stem cell

gkin

mobilization

MA

CXCR4

Hematopoietic

NCT01018979

D

TG-0054

NU

mobilization

PT E

Lymphoma, Hodgki

AC

CE

n Disease

Multiple

Hematopoietic

Myeloma, Non-Hod

stem cell

gkin

mobilization

NCT01458288

Lymphoma, Hodgki n Disease

Multiple

Mobilizing

Myeloma, Non-Hod

hematopoietic

NCT02104427

ACCEPTED MANUSCRIPT Meng W et al. 2017

stem cells

Lymphoma, Hodgki

combined with

n Disease

G-CSF

Healthy volunteers

Mobilization of

NCT00822341

PT

gkin

CXCR4

Chronic

Decreasing cell

Li, S. H., et al.

antagonist

Lymphocytic

viability,

2016 (PMID:

migration;inhibit

27725861)

AC

CE

PT E

D

MA

NU

Leukemia

SC

WZ811

RI

stem cell

tumor growth;make higher sensitivity to docetaxel

ACCEPTED MANUSCRIPT Meng W et al. 2017

Abbreviations

CXCL12: C-X-C motif chemokine ligand 12

Tumor microenvironment

GPCR:

G protein coupled receptor

CXCR4:

chemokine receptor 4

CXCR7:

chemokine receptor 7

NU

VCAM-1: vascular cell adhesion molecule-1

SC

RI

PT

TME:

Vascular endothelial growth factor

MMP:

matrix metalloproteinases

ERK:

extracellular signal-regulated kinase

Ara-C:

cytrarabine

ECM:

extracellular matrix

AC

CE

PT E

D

MA

VEGF:

HDMEC:

human dermal microvascular endothelial cell

MSC:

mesenchymal stem/stromal cell

HUVEC:

CAF:

human umbilical vein endothelial cell

cancer-associated fibroblast

The role of CXCL12 in tumor microenvironment.

The chemokine ligand C-X-C motif chemokine ligand 12 (CXCL12) is a kind of small molecules of cytokines that widely expressed in diversified tissues. ...
723KB Sizes 6 Downloads 10 Views