Review Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Mar 7, 2015; 21(9): 2605-2613
Published online Mar 7, 2015. doi: 10.3748/wjg.v21.i9.2605
Role of periostin and its antagonist PNDA-3 in gastric cancer metastasis
Guo-Xiao Liu, Hong-Qing Xi, Xiao-Yan Sun, Bo Wei, Department of General Surgery, Chinese People’s Liberation Army General Hospital, Beijing 100853, China
Author contributions: All authors contributed equally to this work.
Supported by National Nature Science Foundation of China, No. 81101883.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Bo Wei, Professor, Department of General Surgery, Chinese People’s Liberation Army General Hospital, No. 28 Fuxing Road, Beijing 100853, China. 13381326128@163.com
Telephone: +86-10-66938271 Fax: +86-791-83849520-810
Received: August 23, 2014
Peer-review started: August 23, 2014
First decision: September 27, 2014
Revised: October 12, 2014
Accepted: December 14, 2014
Article in press: December 16, 2014
Published online: March 7, 2015

Abstract

The extracellular matrix component periostin is a secreted protein that functions as both a cell attachment protein and an autocrine or paracrine factor that signals through the cell adhesion molecule integrins αvβ3 and αvβ5. Periostin participates in normal physiological activities such as cardiac development, but is also involved in pathophysiological processes in vascular diseases, wound repair, bone formation, and tumor development. It is of increasing interest in tumor biology because it is frequently overexpressed in a variety of epithelial carcinomas and is functionally involved in multiple steps of metastasis progression. These include the maintenance of stemness, niche formation, EMT, the survival of tumor cells, and angiogenesis, all of which are indispensable for gastric cancer metastasis. Periostin has been reported to activate the PI-3K/AKT, Wnt, and FAK-mediated signaling pathways to promote metastasis. Therefore, periostin represents a potentially promising candidate for the inhibition of metastasis. In this review article, we summarize recent advances in knowledge concerning periostin, its antagonist PNDA-3, and their influence on such key processes in cancer metastasis as maintenance of stemness, niche formation, epithelial-to-mesenchymal transition, tumor cell survival, and angiogenesis. In particular, we focus our attention on the role of periostin in gastric cancer metastasis, speculate as to the usefulness of periostin as a therapeutic and diagnostic target for gastric cancer metastasis, and consider potential avenues for future research.

Key Words: Periostin, Cancer, Tumorigenesis, Extracellular matrix, Epithelial-to-mesenchymal transition, Metastasis, Aptamer, PNDA-3

Core tip: Periostin is involved in various signaling pathways, mediates the critical steps of a wide variety of human tumors, and is associated with tumor growth, invasiveness, and metastasis. Although some authors have written reviews about periostin, they are often fragmented and not very comprehensive. The purpose of this review is to summarize the most recent knowledge of periostin and its antagonist, as well as their structure and the role they play in cancer metastasis, including the maintenance of stemness, niche formation, epithelial-to-mesenchymal transition, the survival of tumor cells and angiogenesis, and avenues for future research.



INTRODUCTION

Gastric cancer is the fourth most commonly diagnosed cancer and the second leading cause of cancer death worldwide[1]. Metastases, rather than the primary tumor, are responsible for the majority of cancer deaths[2]. Gastric cancer metastasis is a very complex process involving epithelial-to-mesenchymal transition (EMT), extravasation, dissemination in the circulation, adhesion to and colonization of the distant site, angiogenesis, and survival. To complete this journey, gastric cancer cells use numerous strategies, all of which lead to a common ultimate goal: the dissemination of gastric cancer cells throughout the body and their survival to form secondary growths. These metastases are one of the greatest challenges to successful cancer therapy[3]. The prognosis for gastric cancer with concomitant liver metastasis is poor[4], with a 5-year survival rate as low as 0%-10% in unselected cases[5]. In 5.59% of gastric cancer patients, liver metastases are detected before or during surgery, while metastases occur in 21.75% following surgical treatment[6-11].

Gastric cancer metastasis is highly influenced by tumor microenvironment. For example, a change in nutrient availability or surrounding pH can influence tumor metastasis[12]. Extracellular matrix (ECM) proteins also play an important role in tumor cell metastasis by regulating stemness and the proliferation of cancer stem cells (CSCs)[13,14]. Therefore, an understanding of the roles played by ECM proteins present in the tumor microenvironment with regards to their influence on signaling pathways involved in cell-ECM interactions could aid the prevention and treatment of tumor metastasis.

The ECM protein periostin [formerly osteoblast specificity factor 2 (OSF2)] has been implicated in the process of gastric cancer metastasis[15]. Initially identified in the rat osteoblast cell line MC3T3-1, periostin contains 811 amino acid residues and is secreted by osteoblasts. It is also a member of the fasciclin family, and contains an NH2-end signal peptide sequence, a cysteine domain structure, four homologous repeats (120-160 amino acids), and a hydrophilic COOH-terminal structural domain[16]. Research from Coutu et al[17] has shown that periostin is a member of a novel vitamin K-dependent γ-carboxylated protein family characterized by the presence of fasciclin domains. Periostin participates in normal physiological processes, including cardiac development, though it also has pathophysiological roles in vascular diseases[18], wound repair[19], bone formation[20], and tumor development[21].

Overexpression of periostin has been identified in various malignancies, including those of the pancreas[22-25], bile duct[26], lung[27], ovary[28], colon[29], breast[30,31], and the head and neck[32]. In contrast, periostin is a suppressor of bladder cancer metastasis, with mutational analysis revealing that the C-terminal region is responsible for this effect[33,34]. To date, the role of periostin in gastric cancer development has not been studied sufficiently[15,35]. Gastric cancers can be classified into two main histological groups, diffuse or intestinal[36], and these have distinct genetic and molecular backgrounds, morphologies, and clinical features[37]. The expression and function of periostin appears to differ between these two gastric cancer subtypes and so further research is needed to understand this phenomenon.

Periostin is highly expressed in gastric epithelial tumors and its function is indispensable for successful angiogenesis and metastasis[35,38]. Clinical studies have demonstrated that high periostin expression or elevated serological levels of periostin correlate with tumor metastasis and poor prognosis[39,40]. Periostin participates in and promotes tumor metastasis through multiple signaling pathways, suggesting that the development of periostin inhibitors could aid the prevention of gastric cancer metastasis.

Gastric cancer cells bind to periostin via the integrins αvβ3 and αvβ5; however the epitope recognized by these integrins has not been formally identified. Orecchia et al[41] have explored whether particular periostin domains influence tumor growth and subsequent metastasis. They generated the monoclonal antibody OC-20 and used recombinant periostin fragments to characterize the epitope recognized by this antibody. Their study revealed that the epitope identified by monoclonal antibody OC-20 is also the binding site for the integrins αvβ3 and αvβ5, and is located in the second FAS1 domain of the protein. Additionally, the use of this antibody in vivo inhibits tumor growth and angiogenesis. The study also showed that the FAS1 domain of periostin plays an important role in tumor progression and subsequent metastasis. Further, the monoclonal antibody OC-20 could prove useful in further exploring the role of periostin in the metastasis of gastric cancer and may contribute to the design of novel anti-metastasis drugs.

EMT

EMT is critical in the development of tumor metastasis, facilitating the acquisition of invasive and metastatic potential by epithelial cells[42-45]. EMT was initially recognized as a key step of morphogenesis during embryonic development. Emerging evidence indicates that this important developmental program promotes tumor recurrence, drug resistance, and metastasis; features associated with a poor clinical outcome for patients with gastric cancer. Periostin has been demonstrated to be both a marker of metastasis and a potent EMT inducer[46]. Analysis of periostin expression by histology strongly implicates cancer-associated fibroblasts as the primary source of periostin. Here, periostin facilitates gastric cancer cell invasion by promoting EMT and establishing a pre-metastatic niche[47]. It has also been shown to promote the migration of cells that have undergone EMT, thereby facilitating their colonization of distant tissues[48]. Therefore, a better understanding of the signaling pathways that influence EMT is essential for the development of novel targeted therapeutics. However, the mechanism by which periostin induces EMT in gastric tumor cells remains unclear, and this should be a major focus of future research.

STEMNESS

Periostin can both induce stemness in tumor cells and facilitate maintenance of this stemness during the initiation of the colonization process[49]. CSCs exhibit a phenotype similar to that of adult stem cells isolated from the same tissue, and share the properties of self-renewal and differentiation with normal stem cells[50]. CSCs were first discovered in hematological malignancies[51] and have more recently been identified in solid tumors[52,53]. Dieter et al[54] have described how tumor invasion is initiated by a small group of CSCs, suggesting that they are extremely important for metastasis. Recently, Vermeulen et al[55] revealed that Wnt signaling is important for maintaining stemness in both normal colon stem cells and in colon cancer cells. Here, the presence of Wnt activity allows for the identification of colon CSCs and facilitates screening of colon tumors for mutations. Further, this study found that particular cells close to the CSC maintain this high Wnt activity, and are capable of activating the Wnt pathway in more differentiated tumor cells, which then regain clonogenicity or tumorigenicity. Recently, an interaction between periostin and the Wnt pathway ligands Wnt1 and Wnt3A was identified in a CSC population, and new emerging evidence indicates that EMT may enhance the stemness of CSCs, which in turn facilitates metastasis[56]. More clearly defining the relationship between periostin and CSC stemness in gastric cancer will aid in elucidating the mechanisms underlying gastric cancer metastasis.

NICHE FORMATION

The role of periostin in promoting adhesion within the CSC niche is a key component of the metastatic process. Tumor cells colonizing distant tissues must successfully interact with the target tissue microenvironment to establish metastasis. The expression of certain ECM components is upregulated in CSCs, which protects tumor cells from detection and destruction by the immune system following implantation. Periostin has been characterized as a niche component capable of promoting the survival of stem cells by enhancing Wnt signaling and promoting metastasis[57]. The niche environment can also promote mutation and evolution of the CSC, thereby increasing cell survival and metastatic potential[58]. The pro-survival effect of periostin on tumor cells and human microvascular endothelial cells was discovered by simulating the stress environment encountered in metastatic tumors. Stress conditions such as low oxygen, nutritional deficiencies, and reduced adhesion are very common in metastatic tumors and fast growing tumor masses[59]. Malanchi et al[57] also described how tumor cells infiltrating the lung can induce the expression of periostin in the niche, thereby initiating the colonization process.

Periostin can be secreted by cells in niches supporting both normal stem cells and CSCs. Here, periostin can act as an adhesion protein by facilitating interaction between the CSC and the niche. This protects stem cells from being influenced by external differentiation factors and so maintains them in an undifferentiated state. CSCs therefore evade differentiation and apoptosis, preserving their ability to colonize distant sites. Within the niche, tumor cells enter a resting state and store energy for later proliferation. As they adapt to their new environment within colonized tissues, the proliferation process is activated.

ANGIOGENESIS

Periostin promotes angiogenesis in tumor metastases, thereby facilitating survival and proliferation of tumor cells following their colonization of distant tissues. Angiogenesis encompasses endothelial cell proliferation, migration, and tube formation, all of which are necessary for tumor growth. Periostin is secreted and signals through the cell adhesion molecules integrins αvβ3 and αvβ5. In this pathway, periostin activates the PKB/AKT and FAK/Src signaling pathways, ultimately leading to increased angiogenesis, enhanced invasiveness and metastatic ability, and decreased apoptosis[39,60]. The induction of angiogenesis by periostin is partly achieved through upregulation of vascular endothelial growth factor (VEGF) receptor 2 (Flk-1/KDR) on endothelial cells by integrin signaling. VEGF and Flk-1/KDR have also been conclusively shown to be involved in the induction of angiogenesis during solid tumor development[61]. Periostin is differentially expressed in primary tumors and human colon cancer metastases, with studies showing that a high level of periostin expression is associated with metastatic colon tumor cells. Researchers have also identified elevated periostin expression in colon cancer cells inoculated into nude mice. Here, periostin promoted the growth of liver metastases, suggesting that periostin plays a significant role in the late stages of cancer progression. In support of this, periostin secreted by tumor cells has been shown to induce angiogenesis via paracrine signaling during the development of metastases[59]. Additional support for this is provided by the suggestion that periostin is a hypoxia-response gene that mediates cross-talk between gastric cancer cells and endothelial cells under hypoxic conditions, at least partially through the regulation of VEGF expression[62].

CELL SURVIVAL

Metastatic growth is controlled by the dynamic balance between cancer cell proliferation and apoptosis[63]. Therefore, factors that facilitate cell survival or inhibit programmed cell death contribute to the success of metastatic colonization. Studies have shown that fewer apoptotic cells were present in metastatic tumors that originated from periostin-producing cells when compared with control cells in vitro[59]. Consistently, periostin activated the Akt/PKB pathway in colon cancer cells through integrins αvβ3 and αvβ5, promoting cell survival[59]. To successfully establish at a metastatic site, tumor cells have to confront and overcome cellular stresses such as nutrient deprivation and hypoxia following their arrival. Interestingly, treatment of cells with an anti-periostin antibody potentiates the effects of 5-fluorouracil and augments apoptosis induced by chemotherapy in colon cancer cells[64]. Gastric and colon cells arise from the same progenitor cells during embryogenesis and share many characteristics. Therefore, periostin could be an effective target for the diagnosis and treatment of metastatic gastric cancer.

SOURCE OF PERIOSTIN

The source of periostin in tumors is a matter of controversy since periostin expression in most tumor cell lines is low. Interestingly, lower levels of periostin expression are detected in tumor cell lines when compared with similar tumor tissues, and periostin production by non-epithelial cells present in tumors of gastric origin has been demonstrated via analysis of publicly available microarray data sets[15]. Several independent studies have reported production of periostin by stromal cells[40,65], whereas others have detected periostin mRNA in the cytoplasm of cancer cells. In gastric cancer tissue, fibroblastic stromal cells present in a dense collagenous matrix show strong immunoreactivity for periostin[47]. Here, double-staining for α-SMA+ and periostin in advanced invasive cancer showed that α-SMA+ fibroblasts were embedded in the cancer stroma, which also contained abundant periostin. Furthermore, periostin mRNA was detected in fibroblastic stromal cells, but not in carcinoma cells, suggesting that periostin is produced by stromal myofibroblasts rather than by neoplastic cells in gastric cancer. An analysis of publicly available microarray data sets has subsequently revealed that no gastric cancer cell lines express periostin mRNA except YCC11[15]. Cells of this type harbor a unique, non-benign single-nucleotide variant in RPS6KA6, which could influence periostin expression via the activation of CREB, a known inducer of periostin expression[66]. These findings suggest that the stroma is an active participant in gastric cancer metastasis, and that focusing on the stroma in future research may help achieve a better understanding of the mechanisms underlying tumor progression and facilitate therapeutics based on stromal targets.

PERIOSTIN REGULATION

Periostin has been found to be potently upregulated by both bone morphogenetic protein-2 and transforming growth factor (TGF)-β[67,68]. In this context, it appears that periostin serves as an effector of the pro-metastatic activity of TGF-β during gastric cancer metastasis. Periostin has been revealed to be regulated by twist as well, with twist capable of binding the periostin promoter to upregulate periostin expression in cancer cells[69]. The hypoxia-responsive growth factors FGF-1 and angiotensin II also increase periostin expression in pulmonary arterial smooth muscle cells by activating the PI3-K/Akt/p70S6K, Ras/MEK1/2/ERK1/2, and Ras/p38MAPK signaling pathways[70]. Further, periostin expression is regulated by IL-4 and IL-13 in lung fibroblasts, and by Wnt-3 in mouse mammary epithelial cells[71,72]. Pancreatic stellate cells are also stimulated to secrete periostin by FGF-A, FGF-B, PDGF-aa, and PDGF-bb. However, much data concerning periostin regulators is derived from studies of embryonic or adult processes. Further research is required to characterize the regulation of periostin under metastatic conditions. An understanding of the distinct mechanisms that regulate periostin in this context will enhance patient evaluation and facilitate the design of innovative therapeutic approaches to gastric cancer metastasis.

TRANSFORMING GROWTH FACTOR, BETA-INDUCED

Transforming growth factor, beta-induced (TGFBI) and periostin are both TGF-β-induced ECM proteins possessing FAS1 domains[73]. TGFBI mRNA expression shows a pattern complementary to that of its highly homologous relative, periostin[74]. TGFBI contains an N-terminal secretory signal peptide and a C-terminal RGD motif followed by a cysteine-rich domain and four internal homologous repeats that could serve as a bridge between cells expressing appropriate ligands[75,76]. TGFBI has been shown to play an important role in the invasion of colon and pancreatic cancers. TGFBI is also expressed in mesothelial cells, especially in those from patients with advanced gastric cancer. It has been suggested that expression of TGFBI in peritoneal mesothelial cells during gastric cancer is both a novel marker of the metastatic behavior of gastric cancer and an important component of the process of peritoneal carcinomatosis[77]. These effects have been shown to result from interactions between the integrin receptors and the FAS1 domain of TGFBI[78]. Interestingly, although periostin and TGFBI share remarkable sequence and structural homology, the slight differences in their C-terminal domains may lead to differences in their effects at different stages of metastasis. TGFBI appears to promote colon cancer metastasis primarily during extravasation, which is an early stage of metastasis, by inducing dissociation of VE-cadherin junctions between endothelial cells through the activation of the integrin αvβ5-Src signaling pathway. Therefore, cancers over-expressing TGFBI may have an increased metastatic potential, leading to poor prognosis[75]. Periostin facilitates colon cancer metastasis by increasing cell survival at a later stage of cancer metastasis. Therefore, close examination of the C-terminal domains of periostin may enhance our understanding of gastric cancer metastasis.

NUCLEIC ACID APTAMERS

The potential use of aptamers in cancer therapy was recognized upon their development almost a quarter of a century ago[79]. PNDA-3 is a recently described modified nucleic acid that can specifically bind to the third or fourth FAS1 domain structure of periostin to inhibit its function. The development of targeted therapy has become a main focus of cancer treatment in recent years[80,81]. Aptamers have many potential advantages over other therapeutic tools, including enhanced stability, easy generation and modification, low immunogenicity, and low toxicity[82-85]. Aptamers often inhibit protein function following specific binding, and the lack of immunogenicity or toxicity, high penetrability, and easy clearance, make them attractive candidates to develop as inhibitors of tumor metastasis[86-88]. The production cost of RNA is lower than DNA and is difficult to be degraded, so the selected aptamer is more suitable for treatment in vivo and diagnosis in vitro. Scientists also combine aptamer with nanotechnology to develop a smartly-designed fluorescent probe (aptamer beacon) according to the character of high affinity and high specificity of the aptamer, to detect the content of target molecules through the intensity of the fluorescent signal. Aptamer is therefore a very promising antagonist molecule. Aptamers targeting cell-surface proteins have recently been developed as promising delivery vehicles, diagnostic tools, and treatment tools for targeting cancer[89,90].

Aptamers are used as aptamer-nanoparticle conjugates for smart drug delivery and in combination with nanoparticles for biomedical sensing and detection because of their outstanding properties. Aptamer-nanoparticle conjugates enable active controlled delivery of drugs that are incorporated in the nanoparticles when they are bind to a disease site because of the aptamer affinity to this site. Aptamers combined with nanoparticles are nanosystems well qualified for the development of new biomedical devices for analytical, imaging, drug delivery, and many other medical applications. In this way, nanoparticle-based bioimaging and smart drug delivery are enabled, especially by use of systematically developed aptamers for cancer-associated biomarkers.

Most anticancer pharmaceuticals have destructive effects on both gastric cancer cells and normal cells. Aptamers can facilitate cell-specific drug delivery in a selective way to sick gastric cancer cells because of their specific binding, which can not only enhance therapeutic effects, but also diminish adverse effects. Moreover, simultaneous in vivo detection and therapy for gastric cancer cells lowers the burden for gastric cancer patients. However, the widespread phenomenon of possible nonspecific accumulation of nanoparticles in the liver must be taken into account.

Problems and solutions

Although aptamers have a large number of advantages, it seems strange that they account for only a small part of modern therapeutic drugs. There main problems impeding the widespread application of aptamers in disease and approaches that could significantly expand the range of aptamer. The average time of aptamer decay in blood depends on conformational structure and oligonucleotide concentration. Since such a time is inappropriate for most therapeutic applications, some methods for protecting aptamers from degradation by nucleases have been developed. One of the methods used to generate nuclease-resistant aptamers is to perform SELEX with oligonucleotides containing modified nucleotides[79]. Furthermore, the removal of aptamers in vivo via renal filtration complicates their wide application. Most aptamers can be easily excreted by kidneys. Conjugation of aptamers with polyethylene glycol is the best solution to this problem. Additionally, aptamer generation usually requires purified target molecules. Some proteins are difficult to purify because of their chemical properties. Sometimes, aptamers generated against target proteins expressed in one kind of cells do not bind to the same proteins expressed in other kinds of cells as a result of post-translational modifications[91]. The modified SELEX protocol can be used to select aptamers recognizing cell-surface proteins[92,93]. Aptamers that recognize particular targets can also bind to molecules with a similar structure regardless of their high specificity. Aptamer cross-reactivity can be a barrier to their therapeutic application, since the possible side effects may be caused by aptamers interacting with other proteins; however, this problem can be settled by performing a SELEX negative selection step with similar molecules in structure.

PNDA-3

PNDA-3 blocks the interaction between periostin and its cell surface receptor. Its use could therefore facilitate the effective diagnosis and treatment of tumor metastasis. PNDA-3 impairs the activation of signaling pathways that rely on ligand binding to integrins αvβ3 and αvβ5. This produces a strong inhibitory effect on tumor cell adhesion, migration, and implantation in a mouse model. Importantly, the effect of PNDA-3 on the inhibition of tumor growth in this model is much higher in vivo than in vitro. This phenomenon could be explained by the role of periostin in the niche prior to colonization. Therefore, the presence of PNDA-3 in the tumor microenvironment affects the function of not only cancer cells, but also the surrounding endothelial and other tumor-related cells.

The effectiveness of periostin antagonism by PNDA-3 has been assessed in vivo using a tumor growth and metastasis model that employs metastatic 4T1 cells implanted into female BALB/c mice mammary fat pads. After 20 d, post-injection tumors in all mice showed a similar distribution, including the lymph nodes, liver, spleen, and lungs. However, large metastases (> 2 mm) were commonly found in the lung tissue of the control group, with few such metastases identified in the PNDA-3 treatment group. Use of a similar approach whereby gastric cancer cells are implanted into the stomach could enable the evaluation of PNDA’s efficiency in reducing gastric cancer metastasis to the liver. Further investigations revealed an anti-angiogenic effect of PNDA-3, with microvascular density significantly reduced in the PNDA-3 treatment group. Therefore, the decreased activation of signaling pathways that rely on integrins αvβ3 and αvβ5, such as the FAK/Src pathway, not only affects tumor metastasis itself, but also inhibits angiogenesis within metastases[94]. Employing PNDA-3 in a mouse model of gastric cancer could therefore aid in understanding how to control liver metastases of stomach cancer from the source.

To ascertain whether the specificity of PNDA-3 for periostin is constant in vivo, Cy3-labeled PNDA-3 was injected intravenously into tumor-bearing mice. Cy3-labeled PNDA-3 was detected in areas of metastatic tumor formation for a period of time, indicating that it could be used as a novel effective method for the diagnosis of gastric cancer metastasis. PNDA-3 could also impair metastasis by inhibiting the function of periostin, allowing it to function in both the treatment and prevention of gastric cancer metastasis.

CONCLUSION

Targeted therapies have become the key strategic focus in the development of cancer treatment in recent years. Nucleic acid aptamers are single-stranded DNA or RNA molecules designed to bind to proteins and regulate their activity and function. These aptamers represent an emerging class of targeted therapeutic molecules[95,96]. While the mutations that cause cancer change gene expression patterns first, the cell and tissue morphology changes occur later. Conventional methods of gastric cancer diagnosis are primarily focused on morphological abnormalities, and therefore cannot identify the early stages of gastric cancer. We have described how a modified DNA aptamer, PNDA-3, could be modified for in vivo use. PNDA-3 blocks the interaction between periostin and its cell surface receptors, reducing the activation of signaling pathways that rely on integrins αvβ3 and αvβ5. This results in weaker FAK/Src signal pathway activity, potently inhibiting the maintenance of stemness, niche formation, EMT, and angiogenesis in cancer metastasis. These results suggest that molecules targeting periostin may be promising tools for the inhibition of cancer metastasis. The study of periostin may well be a promising direction for future research into the inhibition of gastric cancer metastasis.

Footnotes

P- Reviewer: Zhang MX S- Editor: Yu J L- Editor: Rutherford A E- Editor: Wang CH

References
1.  Orditura M, Galizia G, Sforza V, Gambardella V, Fabozzi A, Laterza MM, Andreozzi F, Ventriglia J, Savastano B, Mabilia A. Treatment of gastric cancer. World J Gastroenterol. 2014;20:1635-1649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 404]  [Cited by in F6Publishing: 451]  [Article Influence: 45.1]  [Reference Citation Analysis (5)]
2.  Chambers AF, Groom AC, MacDonald IC. Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2:563-572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2801]  [Cited by in F6Publishing: 2694]  [Article Influence: 122.5]  [Reference Citation Analysis (0)]
3.  Zhang Z, Niu B, Chen J, He X, Bao X, Zhu J, Yu H, Li Y. The use of lipid-coated nanodiamond to improve bioavailability and efficacy of sorafenib in resisting metastasis of gastric cancer. Biomaterials. 2014;35:4565-4572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 79]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
4.  Kwok CM, Wu CW, Lo SS, Shen KH, Hsieh MC, Lui WY. Survival of gastric cancer with concomitant liver metastases. Hepatogastroenterology. 2004;51:1527-1530.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Liu J, Chen L. Current status and progress in gastric cancer with liver metastasis. Chin Med J (Engl). 2011;124:445-456.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Marrelli D, Roviello F, De Stefano A, Fotia G, Giliberto C, Garosi L, Pinto E. Risk factors for liver metastases after curative surgical procedures for gastric cancer: a prospective study of 208 patients treated with surgical resection. J Am Coll Surg. 2004;198:51-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 47]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
7.  Koga R, Yamamoto J, Ohyama S, Saiura A, Seki M, Seto Y, Yamaguchi T. Liver resection for metastatic gastric cancer: experience with 42 patients including eight long-term survivors. Jpn J Clin Oncol. 2007;37:836-842.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 82]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
8.  Sakamoto Y, Sano T, Shimada K, Esaki M, Saka M, Fukagawa T, Katai H, Kosuge T, Sasako M. Favorable indications for hepatectomy in patients with liver metastasis from gastric cancer. J Surg Oncol. 2007;95:534-539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 123]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
9.  Cheon SH, Rha SY, Jeung HC, Im CK, Kim SH, Kim HR, Ahn JB, Roh JK, Noh SH, Chung HC. Survival benefit of combined curative resection of the stomach (D2 resection) and liver in gastric cancer patients with liver metastases. Ann Oncol. 2008;19:1146-1153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 140]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
10.  Tiberio GA, Coniglio A, Marchet A, Marrelli D, Giacopuzzi S, Baiocchi L, Roviello F, de Manzoni G, Nitti D, Giulini SM. Metachronous hepatic metastases from gastric carcinoma: a multicentric survey. Eur J Surg Oncol. 2009;35:486-491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 47]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
11.  Ueda K, Iwahashi M, Nakamori M, Nakamura M, Naka T, Ishida K, Ojima T, Yamaue H. Analysis of the prognostic factors and evaluation of surgical treatment for synchronous liver metastases from gastric cancer. Langenbeck’s archives of surgery /Deutsche Gesellschaft fur Chirurgie. 2009;394:647-653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 55]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
12.  Bloom AB, Zaman MH. Influence of the microenvironment on cell fate determination and migration. Physiol Genomics. 2014;46:309-314.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 46]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
13.  Friedl P, Wolf K. Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer. 2003;3:362-374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2304]  [Cited by in F6Publishing: 2230]  [Article Influence: 106.2]  [Reference Citation Analysis (0)]
14.  Jacks T, Weinberg RA. Taking the study of cancer cell survival to a new dimension. Cell. 2002;111:923-925.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 231]  [Cited by in F6Publishing: 239]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
15.  Li JS, Sun GW, Wei XY, Tang WH. Expression of periostin and its clinicopathological relevance in gastric cancer. World J Gastroenterol. 2007;13:5261-5266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
16.  Takeshita S, Kikuno R, Tezuka K, Amann E. Osteoblast-specific factor 2: cloning of a putative bone adhesion protein with homology with the insect protein fasciclin I. Biochem J. 1993;294:271-278.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Coutu DL, Wu JH, Monette A, Rivard GE, Blostein MD, Galipeau J. Periostin, a member of a novel family of vitamin K-dependent proteins, is expressed by mesenchymal stromal cells. J Biol Chem. 2008;283:17991-18001.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 137]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
18.  Stanton LW, Garrard LJ, Damm D, Garrick BL, Lam A, Kapoun AM, Zheng Q, Protter AA, Schreiner GF, White RT. Altered patterns of gene expression in response to myocardial infarction. Circ Res. 2000;86:939-945.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 240]  [Cited by in F6Publishing: 257]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
19.  Roy S, Patel D, Khanna S, Gordillo GM, Biswas S, Friedman A, Sen CK. Transcriptome-wide analysis of blood vessels laser captured from human skin and chronic wound-edge tissue. Proc Natl Acad Sci USA. 2007;104:14472-14477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 96]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
20.  Kruzynska-Frejtag A, Wang J, Maeda M, Rogers R, Krug E, Hoffman S, Markwald RR, Conway SJ. Periostin is expressed within the developing teeth at the sites of epithelial-mesenchymal interaction. Dev Dyn. 2004;229:857-868.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 120]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
21.  Kudo Y, Siriwardena BS, Hatano H, Ogawa I, Takata T. Periostin: novel diagnostic and therapeutic target for cancer. Histol Histopathol. 2007;22:1167-1174.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Baril P, Gangeswaran R, Mahon PC, Caulee K, Kocher HM, Harada T, Zhu M, Kalthoff H, Crnogorac-Jurcevic T, Lemoine NR. Periostin promotes invasiveness and resistance of pancreatic cancer cells to hypoxia-induced cell death: role of the beta4 integrin and the PI3k pathway. Oncogene. 2007;26:2082-2094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 221]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
23.  Erkan M, Kleeff J, Gorbachevski A, Reiser C, Mitkus T, Esposito I, Giese T, Büchler MW, Giese NA, Friess H. Periostin creates a tumor-supportive microenvironment in the pancreas by sustaining fibrogenic stellate cell activity. Gastroenterology. 2007;132:1447-1464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 235]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
24.  Fukushima N, Kikuchi Y, Nishiyama T, Kudo A, Fukayama M. Periostin deposition in the stroma of invasive and intraductal neoplasms of the pancreas. Mod Pathol. 2008;21:1044-1053.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 80]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
25.  Kanno A, Satoh K, Masamune A, Hirota M, Kimura K, Umino J, Hamada S, Satoh A, Egawa S, Motoi F. Periostin, secreted from stromal cells, has biphasic effect on cell migration and correlates with the epithelial to mesenchymal transition of human pancreatic cancer cells. Int J Cancer. 2008;122:2707-2718.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 102]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
26.  Utispan K, Thuwajit P, Abiko Y, Charngkaew K, Paupairoj A, Chau-in S, Thuwajit C. Gene expression profiling of cholangiocarcinoma-derived fibroblast reveals alterations related to tumor progression and indicates periostin as a poor prognostic marker. Mol Cancer. 2010;9:13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 107]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
27.  Sasaki H, Dai M, Auclair D, Fukai I, Kiriyama M, Yamakawa Y, Fujii Y, Chen LB. Serum level of the periostin, a homologue of an insect cell adhesion molecule, as a prognostic marker in nonsmall cell lung carcinomas. Cancer. 2001;92:843-848.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Gillan L, Matei D, Fishman DA, Gerbin CS, Karlan BY, Chang DD. Periostin secreted by epithelial ovarian carcinoma is a ligand for alpha(V)beta(3) and alpha(V)beta(5) integrins and promotes cell motility. Cancer Res. 2002;62:5358-5364.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Kikuchi Y, Kashima TG, Nishiyama T, Shimazu K, Morishita Y, Shimazaki M, Kii I, Horie H, Nagai H, Kudo A. Periostin is expressed in pericryptal fibroblasts and cancer-associated fibroblasts in the colon. J Histochem Cytochem. 2008;56:753-764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 102]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
30.  Grigoriadis A, Mackay A, Reis-Filho JS, Steele D, Iseli C, Stevenson BJ, Jongeneel CV, Valgeirsson H, Fenwick K, Iravani M. Establishment of the epithelial-specific transcriptome of normal and malignant human breast cells based on MPSS and array expression data. Breast Cancer Res. 2006;8:R56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 102]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
31.  Shao R, Bao S, Bai X, Blanchette C, Anderson RM, Dang T, Gishizky ML, Marks JR, Wang XF. Acquired expression of periostin by human breast cancers promotes tumor angiogenesis through up-regulation of vascular endothelial growth factor receptor 2 expression. Mol Cell Biol. 2004;24:3992-4003.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 239]  [Cited by in F6Publishing: 254]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
32.  Kudo Y, Ogawa I, Kitajima S, Kitagawa M, Kawai H, Gaffney PM, Miyauchi M, Takata T. Periostin promotes invasion and anchorage-independent growth in the metastatic process of head and neck cancer. Cancer Res. 2006;66:6928-6935.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 156]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
33.  Kim CJ, Yoshioka N, Tambe Y, Kushima R, Okada Y, Inoue H. Periostin is down-regulated in high grade human bladder cancers and suppresses in vitro cell invasiveness and in vivo metastasis of cancer cells. Int J Cancer. 2005;117:51-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 72]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
34.  Yoshioka N, Fuji S, Shimakage M, Kodama K, Hakura A, Yutsudo M, Inoue H, Nojima H. Suppression of anchorage-independent growth of human cancer cell lines by the TRIF52/periostin/OSF-2 gene. Exp Cell Res. 2002;279:91-99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 56]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
35.  Liu Y, Liu BA. Enhanced proliferation, invasion, and epithelial-mesenchymal transition of nicotine-promoted gastric cancer by periostin. World J Gastroenterol. 2011;17:2674-2680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
36.  Lauren P. The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. An attempt at a histo-clinical classification. Acta Pathol Microbiol Scand. 1965;64:31-49.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Hu B, El Hajj N, Sittler S, Lammert N, Barnes R, Meloni-Ehrig A. Gastric cancer: Classification, histology and application of molecular pathology. J Gastrointest Oncol. 2012;3:251-261.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Morra L, Moch H. Periostin expression and epithelial-mesenchymal transition in cancer: a review and an update. Virchows Arch. 2011;459:465-475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 189]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
39.  Contié S, Voorzanger-Rousselot N, Litvin J, Clézardin P, Garnero P. Increased expression and serum levels of the stromal cell-secreted protein periostin in breast cancer bone metastases. Int J Cancer. 2011;128:352-360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 70]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
40.  Sasaki H, Yu CY, Dai M, Tam C, Loda M, Auclair D, Chen LB, Elias A. Elevated serum periostin levels in patients with bone metastases from breast but not lung cancer. Breast Cancer Res Treat. 2003;77:245-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 114]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
41.  Orecchia P, Conte R, Balza E, Castellani P, Borsi L, Zardi L, Mingari MC, Carnemolla B. Identification of a novel cell binding site of periostin involved in tumour growth. Eur J Cancer. 2011;47:2221-2229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 40]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
42.  Peinado H, Olmeda D, Cano A. Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer. 2007;7:415-428.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2365]  [Cited by in F6Publishing: 2426]  [Article Influence: 142.7]  [Reference Citation Analysis (0)]
43.  Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol. 2006;7:131-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2893]  [Cited by in F6Publishing: 3008]  [Article Influence: 167.1]  [Reference Citation Analysis (0)]
44.  Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2:442-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4877]  [Cited by in F6Publishing: 4961]  [Article Influence: 225.5]  [Reference Citation Analysis (0)]
45.  Huber MA, Kraut N, Beug H. Molecular requirements for epithelial-mesenchymal transition during tumor progression. Curr Opin Cell Biol. 2005;17:548-558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1362]  [Cited by in F6Publishing: 1408]  [Article Influence: 74.1]  [Reference Citation Analysis (0)]
46.  Yan W, Shao R. Transduction of a mesenchyme-specific gene periostin into 293T cells induces cell invasive activity through epithelial-mesenchymal transformation. J Biol Chem. 2006;281:19700-19708.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 129]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
47.  Kikuchi Y, Kunita A, Iwata C, Komura D, Nishiyama T, Shimazu K, Takeshita K, Shibahara J, Kii I, Morishita Y. The niche component periostin is produced by cancer-associated fibroblasts, supporting growth of gastric cancer through ERK activation. Am J Pathol. 2014;184:859-870.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 63]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
48.  Lindsley A, Snider P, Zhou H, Rogers R, Wang J, Olaopa M, Kruzynska-Frejtag A, Koushik SV, Lilly B, Burch JB. Identification and characterization of a novel Schwann and outflow tract endocardial cushion lineage-restricted periostin enhancer. Dev Biol. 2007;307:340-355.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Sampieri K, Fodde R. Cancer stem cells and metastasis. Semin Cancer Biol. 2012;22:187-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 158]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
50.  Wei B, Chen L, Li R, Tian J. Stem cells in gastrointestinal cancers: a matter of choice in cell fate determination. Expert Rev Anticancer Ther. 2010;10:1621-1633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
51.  Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367:645-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3316]  [Cited by in F6Publishing: 3222]  [Article Influence: 107.4]  [Reference Citation Analysis (0)]
52.  Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB. Identification of human brain tumour initiating cells. Nature. 2004;432:396-401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5422]  [Cited by in F6Publishing: 5376]  [Article Influence: 268.8]  [Reference Citation Analysis (0)]
53.  Prince ME, Sivanandan R, Kaczorowski A, Wolf GT, Kaplan MJ, Dalerba P, Weissman IL, Clarke MF, Ailles LE. Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci USA. 2007;104:973-978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1548]  [Cited by in F6Publishing: 1574]  [Article Influence: 92.6]  [Reference Citation Analysis (0)]
54.  Dieter SM, Ball CR, Hoffmann CM, Nowrouzi A, Herbst F, Zavidij O, Abel U, Arens A, Weichert W, Brand K. Distinct types of tumor-initiating cells form human colon cancer tumors and metastases. Cell Stem Cell. 2011;9:357-365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 243]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
55.  Vermeulen L, De Sousa E Melo F, van der Heijden M, Cameron K, de Jong JH, Borovski T, Tuynman JB, Todaro M, Merz C, Rodermond H. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol. 2010;12:468-476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1301]  [Cited by in F6Publishing: 1412]  [Article Influence: 100.9]  [Reference Citation Analysis (1)]
56.  Li P, Yang R, Gao WQ. Contributions of epithelial-mesenchymal transition and cancer stem cells to the development of castration resistance of prostate cancer. Mol Cancer. 2014;13:55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 109]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
57.  Malanchi I, Santamaria-Martínez A, Susanto E, Peng H, Lehr HA, Delaloye JF, Huelsken J. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 2012;481:85-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 943]  [Cited by in F6Publishing: 1018]  [Article Influence: 78.3]  [Reference Citation Analysis (0)]
58.  Mitra SK, Schlaepfer DD. Integrin-regulated FAK-Src signaling in normal and cancer cells. Curr Opin Cell Biol. 2006;18:516-523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1084]  [Cited by in F6Publishing: 1170]  [Article Influence: 65.0]  [Reference Citation Analysis (0)]
59.  Bao S, Ouyang G, Bai X, Huang Z, Ma C, Liu M, Shao R, Anderson RM, Rich JN, Wang XF. Periostin potently promotes metastatic growth of colon cancer by augmenting cell survival via the Akt/PKB pathway. Cancer Cell. 2004;5:329-339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 416]  [Cited by in F6Publishing: 424]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
60.  Ouyang G, Liu M, Ruan K, Song G, Mao Y, Bao S. Upregulated expression of periostin by hypoxia in non-small-cell lung cancer cells promotes cell survival via the Akt/PKB pathway. Cancer Lett. 2009;281:213-219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 80]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
61.  Stacker SA, Caesar C, Baldwin ME, Thornton GE, Williams RA, Prevo R, Jackson DG, Nishikawa S, Kubo H, Achen MG. VEGF-D promotes the metastatic spread of tumor cells via the lymphatics. Nat Med. 2001;7:186-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 905]  [Cited by in F6Publishing: 887]  [Article Influence: 38.6]  [Reference Citation Analysis (0)]
62.  Qiu F, Shi CH, Zheng J, Liu YB. Periostin mediates the increased pro-angiogenic activity of gastric cancer cells under hypoxic conditions. J Biochem Mol Toxicol. 2013;27:364-369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
63.  Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19834]  [Cited by in F6Publishing: 18704]  [Article Influence: 779.3]  [Reference Citation Analysis (0)]
64.  Tai IT, Dai M, Chen LB. Periostin induction in tumor cell line explants and inhibition of in vitro cell growth by anti-periostin antibodies. Carcinogenesis. 2005;26:908-915.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 80]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
65.  Sasaki H, Dai M, Auclair D, Kaji M, Fukai I, Kiriyama M, Yamakawa Y, Fujii Y, Chen LB. Serum level of the periostin, a homologue of an insect cell adhesion molecule, in thymoma patients. Cancer Lett. 2001;172:37-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
66.  Zang ZJ, Ong CK, Cutcutache I, Yu W, Zhang SL, Huang D, Ler LD, Dykema K, Gan A, Tao J. Genetic and structural variation in the gastric cancer kinome revealed through targeted deep sequencing. Cancer Res. 2011;71:29-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 68]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
67.  Horiuchi K, Amizuka N, Takeshita S, Takamatsu H, Katsuura M, Ozawa H, Toyama Y, Bonewald LF, Kudo A. Identification and characterization of a novel protein, periostin, with restricted expression to periosteum and periodontal ligament and increased expression by transforming growth factor beta. J Bone Miner Res. 1999;14:1239-1249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 730]  [Cited by in F6Publishing: 749]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
68.  Ji X, Chen D, Xu C, Harris SE, Mundy GR, Yoneda T. Patterns of gene expression associated with BMP-2-induced osteoblast and adipocyte differentiation of mesenchymal progenitor cell 3T3-F442A. J Bone Miner Metab. 2000;18:132-139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 106]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
69.  Oshima A, Tanabe H, Yan T, Lowe GN, Glackin CA, Kudo A. A novel mechanism for the regulation of osteoblast differentiation: transcription of periostin, a member of the fasciclin I family, is regulated by the bHLH transcription factor, twist. J Cell Biochem. 2002;86:792-804.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 104]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
70.  Li P, Oparil S, Feng W, Chen YF. Hypoxia-responsive growth factors upregulate periostin and osteopontin expression via distinct signaling pathways in rat pulmonary arterial smooth muscle cells. J Appl Physiol (1985). 2004;97:1550-1558; discussion 1549.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Takayama G, Arima K, Kanaji T, Toda S, Tanaka H, Shoji S, McKenzie AN, Nagai H, Hotokebuchi T, Izuhara K. Periostin: a novel component of subepithelial fibrosis of bronchial asthma downstream of IL-4 and IL-13 signals. J Allergy Clin Immunol. 2006;118:98-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 474]  [Cited by in F6Publishing: 497]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
72.  Haertel-Wiesmann M, Liang Y, Fantl WJ, Williams LT. Regulation of cyclooxygenase-2 and periostin by Wnt-3 in mouse mammary epithelial cells. J Biol Chem. 2000;275:32046-32051.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 79]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
73.  Ruan K, Bao S, Ouyang G. The multifaceted role of periostin in tumorigenesis. Cell Mol Life Sci. 2009;66:2219-2230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 241]  [Cited by in F6Publishing: 257]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
74.  Norris RA, Kern CB, Wessels A, Wirrig EE, Markwald RR, Mjaatvedt CH. Detection of betaig-H3, a TGFbeta induced gene, during cardiac development and its complementary pattern with periostin. Anat Embryol (Berl). 2005;210:13-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 24]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
75.  Ma C, Rong Y, Radiloff DR, Datto MB, Centeno B, Bao S, Cheng AW, Lin F, Jiang S, Yeatman TJ. Extracellular matrix protein betaig-h3/TGFBI promotes metastasis of colon cancer by enhancing cell extravasation. Genes Dev. 2008;22:308-321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 180]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
76.  Skonier J, Neubauer M, Madisen L, Bennett K, Plowman GD, Purchio AF. cDNA cloning and sequence analysis of beta ig-h3, a novel gene induced in a human adenocarcinoma cell line after treatment with transforming growth factor-beta. DNA Cell Biol. 1992;11:511-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 365]  [Cited by in F6Publishing: 389]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
77.  Li Z, Miao Z, Jin G, Li X, Li H, Lv Z, Xu HM. βig-h3 supports gastric cancer cell adhesion, migration and proliferation in peritoneal carcinomatosis. Mol Med Rep. 2012;6:558-564.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Litvin J, Zhu S, Norris R, Markwald R. Periostin family of proteins: therapeutic targets for heart disease. Anat Rec A Discov Mol Cell Evol Biol. 2005;287:1205-1212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 69]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
79.  Lakhin AV, Tarantul VZ, Gening LV. Aptamers: problems, solutions and prospects. Acta Naturae. 2013;5:34-43.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Sawyers C. Targeted cancer therapy. Nature. 2004;432:294-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 793]  [Cited by in F6Publishing: 770]  [Article Influence: 38.5]  [Reference Citation Analysis (0)]
81.  Zangemeister-Wittke U. Antibodies for targeted cancer therapy -- technical aspects and clinical perspectives. Pathobiology. 2005;72:279-286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
82.  Germer K, Leonard M, Zhang X. RNA aptamers and their therapeutic and diagnostic applications. Int J Biochem Mol Biol. 2013;4:27-40.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Li J, Tan S, Chen X, Zhang CY, Zhang Y. Peptide aptamers with biological and therapeutic applications. Curr Med Chem. 2011;18:4215-4222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
84.  Kaur G, Roy I. Therapeutic applications of aptamers. Expert Opin Investig Drugs. 2008;17:43-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 63]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
85.  Ulrich H, Trujillo CA, Nery AA, Alves JM, Majumder P, Resende RR, Martins AH. DNA and RNA aptamers: from tools for basic research towards therapeutic applications. Comb Chem High Throughput Screen. 2006;9:619-632.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 70]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
86.  Wong TY, Liew G, Mitchell P. Clinical update: new treatments for age-related macular degeneration. Lancet. 2007;370:204-206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 95]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
87.  Daniels DA, Chen H, Hicke BJ, Swiderek KM, Gold L. A tenascin-C aptamer identified by tumor cell SELEX: systematic evolution of ligands by exponential enrichment. Proc Natl Acad Sci USA. 2003;100:15416-15421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 425]  [Cited by in F6Publishing: 409]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
88.  Mi Z, Guo H, Russell MB, Liu Y, Sullenger BA, Kuo PC. RNA aptamer blockade of osteopontin inhibits growth and metastasis of MDA-MB231 breast cancer cells. Mol Ther. 2009;17:153-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 114]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
89.  Cerchia L, de Franciscis V. Nucleic acid-based aptamers as promising therapeutics in neoplastic diseases. Methods Mol Biol. 2007;361:187-200.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Cerchia L, de Franciscis V. Targeting cancer cells with nucleic acid aptamers. Trends Biotechnol. 2010;28:517-525.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 157]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
91.  Liu Y, Kuan CT, Mi J, Zhang X, Clary BM, Bigner DD, Sullenger BA. Aptamers selected against the unglycosylated EGFRvIII ectodomain and delivered intracellularly reduce membrane-bound EGFRvIII and induce apoptosis. Biol Chem. 2009;390:137-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 68]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
92.  Sun W, Du L, Li M. Advances and perspectives in cell-specific aptamers. Curr Pharm Des. 2011;17:80-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
93.  Ye M, Hu J, Peng M, Liu J, Liu J, Liu H, Zhao X, Tan W. Generating Aptamers by Cell-SELEX for Applications in Molecular Medicine. Int J Mol Sci. 2012;13:3341-3353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 88]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
94.  Lee YJ, Kim IS, Park SA, Kim Y, Lee JE, Noh DY, Kim KT, Ryu SH, Suh PG. Periostin-binding DNA aptamer inhibits breast cancer growth and metastasis. Mol Ther. 2013;21:1004-1013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 78]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
95.  Ireson CR, Kelland LR. Discovery and development of anticancer aptamers. Mol Cancer Ther. 2006;5:2957-2962.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 346]  [Cited by in F6Publishing: 334]  [Article Influence: 19.6]  [Reference Citation Analysis (0)]
96.  Que-Gewirth NS, Sullenger BA. Gene therapy progress and prospects: RNA aptamers. Gene Ther. 2007;14:283-291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 160]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]