World J Gastroenterol 2015 October 7; 21(37): 10493-10501 ISSN 1007-9327 (print) ISSN 2219-2840 (online)

Submit a Manuscript: http://www.wjgnet.com/esps/ Help Desk: http://www.wjgnet.com/esps/helpdesk.aspx DOI: 10.3748/wjg.v21.i37.10493

© 2015 Baishideng Publishing Group Inc. All rights reserved.

TOPIC HIGHLIGHT 2015 Advances in Gastric Cancer

Targeting mast cells in gastric cancer with special reference to bone metastases Christian Leporini, Michele Ammendola, Ilaria Marech, Giuseppe Sammarco, Rosario Sacco, Cosmo Damiano Gadaleta, Caroline Oakley, Emilio Russo, Giovambattista De Sarro, Girolamo Ranieri Christian Leporini, Emilio Russo, Giovambattista De Sarro, Department of Health Science, Clinical Pharmacology and Pharmacovigilance Unit and Pharmacovigilance’s Centre Calabria Region, University of Catanzaro “Magna Graecia” Medical School, Viale Europa - Germaneto, 88100 Catanzaro, Italy Michele Ammendola, Giuseppe Sammarco, Rosario Sacco, Department of Medical and Surgery Sciences, Clinical Surgery Unit, University of Catanzaro “Magna Graecia” Medical School, 88100 Catanzaro, Italy Ilaria Marech, Cosmo Damiano Gadaleta, Caroline Oakley, Girolamo Ranieri, Diagnostic and Interventional Radiology Unit with Integrated Section of Translational Medical Oncology, National Cancer Research Centre, “Giovanni Paolo II”, 70124 Bari, Italy Author contributions: Leporini C, Ammendola M, Marech I and Ranieri G conceived the review and performed the critical review of the literature; Sammarco G, Sacco R, Gadaleta CD, Russo E and De Sarro G contributed to the literature research and data analysis; all authors wrote the manuscript and Oakley c edited the manuscript. Conflict-of-interest statement: The authors declare no conflicts of interest. 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: Michele Ammendola, MD, Department of Medical and Surgery Sciences, Clinical Surgery Unit, University of Catanzaro “Magna Graecia” Medical School, Viale Europa Germaneto, 88100 Catanzaro, Italy. [email protected]

WJG|www.wjgnet.com

Telephone: +39-961-3694191 Fax: +39-961-3647556 Received: April 5, 2015 Peer-review started: April 7, 2015 First decision: April 23, 2015 Revised: June 15, 2015 Accepted: August 25, 2015 Article in press: August 25, 2015 Published online: October 7, 2015

Abstract Bone metastases from gastric cancer (GC) are considered a relatively uncommon finding; however, they are related to poorer prognosis. Both primary GC and its metastatic progression rely on angiogenesis. Several lines of evidence from GC patients strongly support the involvement of mast cells (MCs) positive to tryptase (MCPT) in primary gastric tumor angiogenesis. Recently, we analyzed infiltrating MCs and neovascularization in bone tissue metastases from primary GC patients, and observed a significant correlation between infiltrating MCPT and angiogenesis. Such a finding suggested the involvement of peritumoral MCPT by infiltrating surrounding tumor cells, and in bone metastasis angiogenesis from primary GC. Thus, an MCPT-stimulated angiogenic process could support the development of metastases in bone tissue. From this perspective, we aim to review the hypothetical involvement of tumorinfiltrating, peritumoral MCPT in angiogenesis-mediated GC cell growth in the bone microenvironment and in tumor-induced osteoclastic bone resorption. We also focus on the potential use of MCPT targeting agents, such as MCs tryptase inhibitors (gabexate mesylate, nafamostat mesylate) or c-KitR tyrosine kinase inhibitors (imatinib, masitinib), as possible new anti-angiogenic and anti-resorptive strategies for the treatment of GC patients

10493

October 7, 2015|Volume 21|Issue 37|

Leporini C et al . Mast cells in advanced gastric cancer

affected by bone metastases. Key words: Bone metastases; Gastric cancer; receptor activator of nuclear factor-kB; Angiogenesis; Osteoclastic bone resorption; Tryptase inhibitors; c-Kit receptor tyrosine kinase inhibitors; Anti-angiogenic therapy © The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.

Core tip: The activation of the stem cell factor/c-Kit receptor (c-KitR) pathway in mast cells (MCs), and tryptase release upon MCs degranulation have a pivotal role in tumor angiogenesis in several human malignancies. MCs positive to tryptase (MCPT) have been implicated in primary gastric cancer (GC) an­ giogenesis. Our preliminary findings indicated that bone metastasis angiogenesis from GC is also supported by infiltrating MCPTs. Overall, the evidence provides a rationale to evaluate c-KitR inhibitors that block MCs degranulation, or tryptase inhibitors that inhibit tryptase and/or the Proteinase-Activated Receptor-2 pathway, in clinical trials for bone metastasis GC patients. Leporini C, Ammendola M, Marech I, Sammarco G, Sacco R, Gadaleta CD, Oakley C, Russo E, De Sarro G, Ranieri G. Targeting mast cells in gastric cancer with special reference to bone metastases. World J Gastroenterol 2015; 21(37): 10493-10501 Available from: URL: http://www.wjgnet. com/1007-9327/full/v21/i37/10493.htm DOI: http://dx.doi. org/10.3748/wjg.v21.i37.10493

INTRODUCTION Gastric cancer (GC) is a major cause of cancer-related [1] mortality worldwide . According to GLOBOCAN estimates, there were 260000 cases of cardia GC and [1] 691000 cases of non-cardia GC in 2012 . Generally, at the time of diagnosis, most patients have unresectable [2] or metastatic disease . The most common site of distant metastases is the peritoneum, followed by [2,3] the liver, lung and bones . Bone metastases are a [4] relatively uncommon finding , occurring in between 1% and 20% of GCs. Notably, they represent a major discomfort because of the related pain, neurological [5,6] involvement and hypercalcemia syndrome . Recently, it was reported that bone metastases diagnosed by Fluorine-18- fluorodeoxyglucose positron emission tomography/computed tomography examinations [7] represented 10% of an evaluated series . In fact, many GC patients die from metastases in intraperitoneal organs before their bone metastatic sites are revealed. Both primary GC and its metastatic progression rely [8,9] on angiogenesis , which plays a crucial role in cancer development, inducing tumor growth, invasion and [10,11] metastasis . Moreover, cancer stem cells also promote GC metastasis via close physical cellular contact and paracrine signals released from the

WJG|www.wjgnet.com

tumor niche, both in vivo and in vitro. In fact, stromaassociated cancer stem cells promote the GC cell epithelial to mesenchymal transition (EMT), attracting circulating cancer cells to self-seed the primary tumor, [12] again though EMT . Interestingly, a large body of evidence supports the substantial involvement of mast cells (MCs) [9,13-17] in tumor angiogenesis . A preclinical pivotal study documented that the angiogenic response to subcutaneously growing B16-BL6 tumors was lower in genetically MC-deficient W/Wv mice compared +/+ with MC-sufficient littermate mice. Moreover, the latters showed a greater propensity to develop lung [18] metastases . Accordingly, a significant increase in MCs density (MCD) was observed in several animal [19-22] and human malignant tumors . MCs secrete several classical and non-classical pro-angiogenic factors, including vascular endothelial growth factor (VEGF), fibroblast growth factor-2 (FGF-2), platelet-derived growth factor-β (PDGF-β), interleukin-6 (IL-6), IL-8, thymidine phosphorylase [23-27] (TP), chymase and tryptase . Notably, tryptase is the most powerful non-classical pro-angiogenic factor released by MCs upon c-Kit [28] receptor (c-KitR) TK activation . Tryptase induced [29] endothelial cell (EC) proliferation in a matrigel assay and in vivo in a chick embryo chorioallantoic membrane [27] (CAM) assay : tryptase inhibitors suppressed both effects consistently. Tryptase activates the proteinaseactivated receptor-2 (PAR-2) expressed on ECs, directly [17,30] stimulating their proliferation . Interestingly, PAR-2 expression is greater in tumor tissues than in normal [31] tissues . Moreover, it also promotes the degradation of extracellular matrix (ECM) components via metal­ loproteinases activation, resulting in the release of ECMbound latent pro-angiogenic factors (e.g., VEGF and [32,33] FGF-2) . Most notably, many human studies have established a positive correlation between MCD positive to tryptase (MCDPT) and microvascular density (MVD) [9,13,14,34-39] in tumor tissue . Several findings from patients strongly support the crucial involvement of MCDPT in primary gastric [28,39-41] tumor angiogenesis . There is also a significant correlation between MCDPT and angiogenesis in bone [42] metastases from GC patients . Starting from these preliminary results, we aim to review: (1) the involvement of MCDPT in GC angio­ genesis; (2) the specific role that MCDPT might play in bone metastases angiogenesis; and (3) the potential targeting of MCDPT by tryptase or c-KitR tyrosine kinase (TK) inhibitors in metastatic gastric patients with special reference to bone metastases.

MCDPT AND TUMOR ANGIOGENESIS MCs are cells that originate in the bone marrow + [43] from pluripotent CD34 hematopoietic stem cells . Precursors of MCs migrate through the circulation to their target tissues, completing their maturation

10494

October 7, 2015|Volume 21|Issue 37|

Leporini C et al . Mast cells in advanced gastric cancer process into granulated cells, under the influence of [44] several microenvironment growth factors . The most important of the above factors is the ligand for the c-KitR TK, stem cell factor (SCF), secreted mainly by fibroblasts and ECs. SCF also regulates development, [33,45] survival and de novo proliferation of MCs . MCs express the high affinity IgE receptor, which, following IgE activation, triggers release of their stored proangiogenic factors. In this way a link between immunity and angiogenesis is established by MCs. Among the other IgE-independent MCs activation mechanisms, a wide variety of other surface receptors for cytokines, chemokines, immunoglobulins, complement and [46] bacterial products, are also described . MCs granules are key functional elements, cha­ racterized by two distinct secretory patterns: exocytosis [33] or piecemeal degranulation . Interestingly, this latter mechanism, representing a slow and selective pathway of cell secretion, has been more frequently observed in MCs infiltrating areas of chronic inflammation, such [47] as tumor tissues . Correspondingly, a link between MCs, chronic inflammation and cancer has been long [23,47] suggested . Thus, MCs are one of the earliest and major inflammatory cell types recruited into the tumor [9,13,14,39,48] microenvironment . In particular, several data from human studies have highlighted a strong linear correlation between MCDPT and pathological angiogenesis sustaining several solid tumors, such as human malignant melanoma, endometrial carcinoma, breast cancer, GC, colorectal cancer (CRC) [9,14,20,21,28,34-39,49-51] and pancreatic ductal adenocarcinoma . Remarkably, MCDPT correlated with angiogenesis, tumor aggressiveness and poor tumor outcome in most [9,16,23,33] clinical investigations , suggesting a prognostic [14,21,35,39,50-52] significance for MCDPT in several tumors . [28] Moreover, Ammendola et al demonstrated a positive correlation among MCDPT, c-KitR expressing cells and MVD in tumor tissue specimens from surgical GC patients, confirming that c-KitR activation on MCs surface, resulting in tryptase degranulation from activated [24,53] MCs, has a pivotal function in tumor angiogenesis . Thus, MCDPT may represent a novel and attra­ ctive therapeutic target of anti-angiogenic cancer [33,45] therapy . Indeed, MCDPT-targeted therapeutic approaches could limit angiogenesis in growing tumors, potentially decreasing tumor growth and metastasis. Therefore, tryptase inhibitors, such as gabexate [54] mesylate and nafamostat mesylate , or c-KitR [10,55,56] inhibitors, such as imatinib and masitinib , should be evaluated in clinical trials as new anti-angiogenic agents in combination with chemotherapy.

MCDPT, ANGIOGENESIS, AND IMMUNE SUPPRESSION IN HUMAN CANCERS: THE DUAL VALUE OF TARGETING MCDPT Different infiltrating cell populations form the tumor

WJG|www.wjgnet.com

microenvironment, enabling a pathological condition that directly promotes angiogenesis, tumor spread and [57] invasion and, concurrently, hamper the immune [58] response against tumors . However, a relationship between high MCD and improved overall survival has [59] also been reported in some human studies , signifying a diversified and not yet well-understood role for MCs in cancer. Note that, with reference to this relationship between high MCD and favorable prognosis, MCs are able to modulate both innate and adaptive immune [60] responses . In this context, MCs are involved in innate immunity by releasing tumor necrosis factor-α (TNF-α) and interleukins (IL-1, 4, 6) that, in turn, help to kill [24,46] tumors . In addition, MCs express both the major histocompatibility class (MCH) Ⅱ antigen and its costimulatory molecule, which activate adaptive T and B [61] cell responses against tumors . Finally, MCs cooperate [61] with natural killer cells to reject cancer cells . On [62] the other hand, Shin et al reported the critical involvement of MCs’ tryptase in eosinophils recruitment, suggesting a role in the induction of a pro-inflammatory microenvironment. In this way, MCDPT may also induce angiogenesis via several pro-inflammatory cytokines [23,33] that act as pro-angiogenic factors . Taken together, the data confirm the potential dual value of MCPT as a therapeutic target for tryptase inhibitors or c-KitR tyrosine kinase inhibitors, which could both prevent MCDPT-mediated immune suppression and promote/ unleash protective anti-tumor immune responses through the selective inhibition of MCDPT-mediated angiogenesis, resulting in tumor regression.

MCDPT AND ANGIOGENESIS IN HUMAN GC Several studies support the view that inflammation is a critical component of GC development and [24,53,63] progression . In turn, increasing evidence sug­ gests an intimate relationship between angiogenesis, [16] inflammation and GC progression . In the GC microen­ vironment, many types of inflammatory cells exist, and among them, MCs promote GC angiogenesis by secreting pro-angiogenic factors and cooperating with stromal and malignant cells. In turn, neovascularization helps to maintain inflammation by promoting the migration of [16,63,64] inflammatory cells to the site of inflammation . From a translational point of view, a significant positive correlation between MCDPT and MVD has been confirmed in cancer tissue specimens. Interestingly, specimens from patients with advanced histological stages of the disease showed a higher MCD than those [65,66] from patients with early stage disease . [9] Similarly, Ribatti et al observed that stage IV GC shows a higher degree of vascularization than the earlier stages, and that MCDPT increases in parallel with malignancy grade and is highly correlated with the extent of angiogenesis in primary tumor tissue. [67] Zhao et al reported a significant positive correlation

10495

October 7, 2015|Volume 21|Issue 37|

Leporini C et al . Mast cells in advanced gastric cancer Table 1 Principal studies that correlate mast cell density and mast cells density positive status to tryptase with angiogenesis or negative prognostic factors in gastric cancer patients Disease stage/ main stages

Chemotherapy

Patients (n )/site

Methods of MCs identification

Correlation

P value

Ribatti et al[9], 2010

All TNM stages (mainly Ⅱ-Ⅳ)

No

30

All TNM stages (mainly Ⅱ-Ⅳ) TNM stage Ⅲ TNM stage Ⅲ

No

60

Nr

25

Nr

19

MCD and Angiogenesis and advanced tumor stage MCD and advanced tumor stage MCDPT and MVD and c-KitR-EC MCDPT and no. of metastatic lymph nodes

< 0.05

Zhao et al[67], 2012

Immunohistochemistry primary anti-tryptase and anti-chymase Abs Immunohistochemistry primary anti-tryptase Ab Immunohistochemistry primary anti-tryptase Ab Immunohistochemistry primary anti-tryptase Ab

Ref.

Ammendola et al[28], 2013 Ammendola et al[32], 2013

NS 0.001 0.01

Ab/s: Antibody/antibodies; MCD: Mast cell density; NR: Not reported; MCDPT: Mast cells density positive to tryptase; c-KitR EC: c-Kit receptor expressing cell.

between the increased infiltration of MCDPT and the progression of disease in GC patients, documenting an association between a high level of tryptase expression and advanced tumor stage, which is a marker of poor prognosis. In agreement with the above findings, a more recent investigation evaluating biopsy specimens from 25 GC patients highlighted a positive correlation between MCDPT, c-KitR expressing cells and MVD, using [28] immunohistochemistry and image analysis methods . Furthermore, in a series of 41 gastrointestinal cancer patients, a positive correlation between MCDPT and the number of metastatic lymph nodes harvested, and between MCDPT in primary tumor tissue and [39] in metastatic lymph node tissue, were observed . These results suggested that MCs’ tryptase, like VEGF, could be involved in tumor lymphangiogenesis, which, in turn, is correlated with lymph node metastases, both in experimental cancer models and several [68] human cancers . Table 1 summarizes the above studies correlating MCD with angiogenesis or negative prognostic factors. Overall, the majority of studies in GC support the view that MCDPT is significantly correlated with the depth of invasion, lymph node metastases, lymphatic or blood invasion, the degree of histological differentiation, and the number of blood vessels surrounding GC cells. Most of the MCs located near the neovascularization areas were degranulated, suggesting the critical role of tryptase in angiogenesis [8] and tumor progression . Correspondingly, tumors injected into mice treated with inhibitors of MCsdegranulation presented decreased vascularization, [65,69] growth and metastasis . Data from an ultra-structural study in advanced GC patients highlighted that perivascular MCs containing only tryptase in their granules exhibited piecemeal degranulation that was significantly associated with microvascular basal lamina changes (irregular thickness, multiple layers and loose association with ECs and pericytes), which fitted with a remodeling of existing microvasculature. These changes are involved in the increased vascular permeability that characterizes

WJG|www.wjgnet.com

[70]

the tumor microvasculature . Interestingly, this suggested that MCs’ tryptase, through its direct and indirect proteolytic activity on the ECM, could induce fenestrations in the endothelium of tumor [71] microvasculature, like VEGF ; thus, specifically contributing to metastasis in GC. In light of these evidence-based considerations, serum tryptase released by MCs could be an indicator for future surgery, a valid predictive factor for hematogenous invasion or metastasis, and a promising prognosis marker both in early and advanced GC. Taken together, the evidence suggests that the accumulation of MCDPT at the periphery of GC tissue might lead to increased rates of tumor vascularization, thus promoting tumor growth and metastases to distant organs. Therefore, MCDPT may represent a valuable target of anti-angiogenic therapy, either by tryptase inhibitors or c-KitR inhibitors, which may prove useful therapeutic tools to control angiogenesismediated tumor growth, progression and metastasis in GC.

ROLE OF MCDPT IN BONE METASTASIS ANGIOGENESIS FROM PRIMARY GC Bone metastases from GC arise from a scattered metastatic spread of tumor cells in the bone marrow (most frequently located in the thoracic and lumbar [72] vertebrae) . These metastases are mainly osteolytic, [5,6,73-75] while osteoblastic implants are rarely reported . Notably, bone metastases in GC usually occur in the advanced stages of the disease, generally correlating [2,5,6,73,74] with poor prognosis . However, the dissemination of micrometastatic cells in the bone tissue may be evident in early GC, in which a poorly-differentiated carcinoma, the presence of signet-ring cell carcinoma, and/or the involvement of lymph node metastasis, seem [76] to be risk factors associated with bone metastases . In particular, in bone marrow micrometastases in early GC, subclinical seeding of tumor cells in the bone marrow (at the time of primary tumor resection) was detected by

10496

October 7, 2015|Volume 21|Issue 37|

Leporini C et al . Mast cells in advanced gastric cancer immunocytochemical evaluation of epithelial cytokeratin protein, a distinctive trait of epithelial cells, both normal and malignant, that would not normally be present [77] in the hematogenous marrow . According to Jauch [78] et al , cytokeratin-positive cells in the bone marrow represent a surrogate marker for general disseminative metastasis, rather than the beginning of metastatic growth in patients with GC. Interestingly, patients with cytokeratin positivity in their bone marrow had a higher [40,77] MVD compared with cytokeratin-negative patients . Correspondingly, VEGF-positive tumors associated with increased MVD show cytokeratin-positive cells in [8] the bone marrow . As a result, the above reported occurrence of such micrometastases (cytokeratinpositive cells) in the bone marrow is potentially closely [8,40] related to angiogenesis in the primary tumor . These observations suggested that angiogenesis plays an essential role in micrometastatic (subclinical) seeding of GC cells in the bone tissue, in the develop­ ment of bone marrow micrometastases into clinically manifest bone metastases (macroscopic disease) and in their potential to invade circulatory system for further distant dissemination from the bone microenvironment. Recently, we investigated infiltrating MCDPT and neovascularization in 15 bone tissue metastases selected from a series of 190 GC patients. We evaluated bone biopsies samples from these patients using immunohistochemistry and image analysis methods. The results showed a statistically significant correlation among MCDPT, MC-area-PT, MVD and endothelial [42] area . These correlations suggested the involvement of infiltrating MCDPT in bone metastasis angiogenesis from primary GC, as well as in primary tumor neovascularization. Therefore, an MCDPT-stimulated angiogenic process could support the development of metastases in the bone tissue. From a biological point of view, MCs may be both recruited and activated by [58,79] SCF , the ligand for c-KitR, and by other growth factors, such as VEGF, FGF-2 and TP, secreted by [8,27,29,80,81] metastasized bone marrow GC cells . Proangiogenic factors, released from activated MCs, may act in autocrine and paracrine manners, and [51,82] then stimulate MCs, ECs and GC cells, as [51] reported by Marech et al . Interestingly, the bone tumor microenvironment plays a crucial role in os­ teoclastogenesis by inducing the production of the receptor activator of nuclear factor-kB (RANK) ligand (RANKL) by marrow stromal cells and osteoblasts. Tumor cells secrete parathyroid hormone-related peptide (PTH-rP) and several other osteotropic factors, including IL-6, TNF, M-CSF and prostaglandin E2 (PGE2), which upregulate the expression of RANKL on the surface of marrow stromal cells and immature osteoblasts. RANKL then binds RANK on the surface of osteoclast precursors, ultimately resulting in osteoclast formation and bone resorption. Thus, tumor-induced osteoclastic bone resorption leads to the release of growth factors, such as TGF-β, FGFs, PDGFs, insulin-

WJG|www.wjgnet.com

like growth factors, and bone morphogenetic proteins by the bone matrix that promote tumor growth and further bone destruction by increasing the production of PTH-rP, as well as the above growth factors, resulting in [83] further RANKL upregulation . This reciprocal interplay between tumor cells and the bone tumor microenvironment results in a vicious circle that further increases tumor growth and bone [83] destruction . Crucially, MCDPT could foster this “symbiotic relationship” between bone destruction and tumor growth by releasing pivotal components of the [84,85] above-described vicious circle . The involvement of MCDPT both in metastatic bone resorption and in angiogenesis-mediated GC cell growth is also suggested by MCs ability to produce and [23] release TGF-β , which stimulates osteoclast activation [54,86] via the RANKL pathway . Furthermore, TGF-β, in the presence of M-CSF and in the absence of RANKL, could induce in vitro [87] osteoclast formation directly , suggesting that MCDPT, by releasing TGF-β, could also stimulate the RANKL-independent metastatic bone resorption in [81,88] the bone tissue metastases from GC patients . Based on the discussed data, MCDPT may potentially offer a novel and promising target of anti-angiogenic therapy to decrease both angiogenesis-mediated GC cell growth in the bone tissue and tumor-induced [2,8,42] osteoclastic bone resorption .

TARGETING MCDPT: A POSSIBLE ANTIANGIOGENIC AND ANTI-RESORPTIVE STRATEGY IN BONE METASTASIS GC PATIENTS The use of MCs’ tryptase inhibitors (gabexate mesylate, [17] nafamostat mesylate and tranilast) or c-KitR tyrosine [10,28,55] kinase inhibitors (imatinib and masitinib) could represent a potential anti-tumor strategy to inhibit both angiogenesis and RANKL-/non-RANKL-mediated [42] osteoclast activation . Unlike conventional chemotherapy, anti-angiogenic therapy does not induce a direct cytotoxic effect, but reduces neovascularization mainly by targeting small foci of proliferating cells of tumor-associated capillary endothelium or from metastatic sites. Thus, angiogenesis inhibitors generally do not cause suppression of the hematogenous bone marrow, hair loss or gastrointestinal symptoms like cytotoxic chemotherapeutics, which require pause periods to allow regeneration of normal cells. By virtue of these features, angiogenesis inhibitors need to be administered for a longer period compared with conventional cytotoxic chemotherapeutics and, consequently, anti-angiogenic [89] therapy is administered continuously . Therefore, anti-angiogenic agents may be effective for longterm administration to achieve prolonged dormancy of [40] primary GC and GC micrometastases .

10497

October 7, 2015|Volume 21|Issue 37|

Leporini C et al . Mast cells in advanced gastric cancer Furthermore, the characteristics of anti-angiogenic therapy have recently prompted the medical-scientific community to consider using anti-angiogenic agents in combination with traditional cytotoxic anti-cancer drugs to potentiate the clinical effectiveness of the latter by preventing their limitations. Moreover, MCs’ tryptase or c-KitR inhibitors could be added to avail­ able conventional cytotoxic drugs that target normal marrow and GC cells indiscriminately in the bone tissue from bone metastasis GC patients. In particular, these anti-angiogenic agents might temporarily inhibit the remodeling of tumor microvasculature and, consequently, the increase of tumor microvascular permeability in the bone metastases from primary GC patients. Starting from the consideration that tumorassociated vasculature represents the first barrier to the penetration of drugs into tumors, agents such as gabexate mesylate and nafamostat mesylate, or imatinib and masitinib, could act as anti-angiogenic modulators by impairing tumor vasculature functions in the bone tumor microenvironment, possibly resulting in the potentiation of the therapeutic response when administered in combination with cytotoxic chemo­ therapy, and potentially prolonging the survival time of GC patients with bone metastases. Notably, synergism between MCs’ tryptase inhibitors and classical chemotherapy drugs in cancer treatment is exemplified by the ability of nafamostat mesylate to inhibit both in vitro and in vivo chemotherapy[90,91] induced nuclear factor kappa-B (NF-kB) activation , which is well-known to contribute to the angiogenic [92] phenotype of several tumors , and chemoresistance and suboptimal therapeutic efficacy of cancer chemothe­ [93] rapy drugs . Correspondingly, it is interesting that nafamostat mesylate has been shown to enhance the anti-tumor effect of paclitaxel against GC with peritoneal dissemination by inhibiting paclitaxel-induced [94] NF-kB activation in mice . Thus, we speculate that a combination chemotherapy of nafamostat mesylate and classical cytotoxic drugs could potentially exert a synergistic anti-tumor effect in bone metastases from GC. Finally, MCDPT targeting agents, such as tryptase inhibitors or c-KitR receptor inhibitors, could represent a novel potential anti-angiogenic approach to inhibit tumor-induced osteoclastic bone resorption and bone destruction in bone metastasis GC patients. Intriguingly, the potential ability of MCDPT targeting to inhibit the RANKL-dependent vicious circle of cancer-induced bone destruction, supports further investigation of tryptase inhibitors or c-KitR inhibitors [95] in clinical trials comparing them with denosumab (a well-known inhibitor of the RANKL–RANK interaction), in terms of efficacy and safety, in the prevention of skeletal-related events in adult patients with bone metastases from GC. Importantly, unlike denosumab, MCDPT-targeting agents could decrease angiogenesismediated GC cell growth in the bone tissue and metastatic bone resorption, independently of their

WJG|www.wjgnet.com

inhibitory activity on the cycle of bone destruction and tumor growth mediated by RANKL. Therefore, it would be interesting to investigate whether combination therapy with these agents, which are potentially antiangiogenic and anti-resorptive, and denosumab may exert a clinically useful, synergistic effect in decrea­ sing tumor-induced osteoclastic bone resorption and angiogenesis-mediated bone metastasis progression in primary GC patients affected by bone metastases. Accordingly, clinical studies comparing the efficacy and safety profile of MCDPT targeting agents versus other [96] anti-resorptive drugs, such as bisphosphonates , in bone metastasis GC patients might be also worth performing.

CONCLUSION A survey of the literature data indicated that MCs are important players in tumor angiogenesis and [15,33,61] development , by releasing a panoply of angiogenic [23] factors (e.g., tryptase) . This is supported by human studies that highlighted a strong correlation between MCDPT and pathological angiogenesis in various solid [9,14,20,21,28,34-39,49-51] tumors . With special reference to GC, much evidence supports the involvement of MCDPT in primary gastric tumor [8,9,28,39-41] angiogenesis, thus promoting metastases . With regards to the subgroup of bone metastases, pilot published data demonstrated a significant correlation [42] between infiltrating MCDPT and angiogenesis . From a therapeutic point of view, tumors injected in mice treated with inhibitors of MCs-degranulation [65,69] presented decreased vascularization and metastasis . According to these data, targeting MCs is currently under [56,97] investigation, especially in GC patients . In particular, for the subgroup of patients with bone metastases from GC, the biological background suggests that MCDPT may stimulate the RANKL-dependent vicious circle of tumor-induced bone destruction and tumor growth, and the RANKL-independent metastatic bone resorption. This biological background further supports targeting of MCDPT as a therapeutic strategy in bone metastases. Finally, ad hoc clinical trials might be performed to compare, in terms of efficacy and safety, these potential anti-angiogenic and anti-resorptive agents with anti-resorptive drugs currently used clinically (i.e., denosumab and bisphosphonates) for the prevention of skeletal-related events in GC patients affected by bone metastases.

REFERENCES 1

2

10498

Colquhoun A, Arnold M, Ferlay J, Goodman KJ, Forman D, Soerjomataram I. Global patterns of cardia and non-cardia gastric cancer incidence in 2012. Gut 2015; Epub ahead of print [PMID: 25748648 DOI: 10.1136/gutjnl-2014-308915] Kim HS, Yi SY, Jun HJ, Lee J, Park JO, Park YS, Jang J, Kim HJ, Ko Y, Lim HY, Kang WK. Clinical outcome of gastric cancer patients with bone marrow metastases. Oncology 2007; 73: 192-197 [PMID: 18418012 DOI: 10.1159/000127386]

October 7, 2015|Volume 21|Issue 37|

Leporini C et al . Mast cells in advanced gastric cancer 3

4 5

6 7

8 9

10

11

12

13

14

15 16 17

18 19

Ekinci AŞ, Bal O, Ozatlı T, Türker I, Eşbah O, Demirci A, Budakoğlu B, Arslan UY, Eraslan E, Oksüzoğlu B. Gastric carcinoma with bone marrow metastasis: a case series. J Gastric Cancer 2014; 14: 54-57 [PMID: 24765538 DOI: 10.5230/ jgc.2014.14.1.54] Maccauro G, Spinelli MS, Mauro S, Perisano C, Graci C, Rosa MA. Physiopathology of spine metastasis. Int J Surg Oncol 2011; 2011: 107969 [PMID: 22312491 DOI: 10.1155/2011/107969] Amoroso V, Pittiani F, Grisanti S, Valcamonico F, Simoncini E, Ferrari VD, Marini G. Osteoblastic flare in a patient with advanced gastric cancer after treatment with pemetrexed and oxaliplatin: implications for response assessment with RECIST criteria. BMC Cancer 2007; 7: 94 [PMID: 17540044 DOI: 10.1186/1471-2407-7-94] Marti J, Sainz M. Unusual late metastasis from gastric carcinoma. Saudi J Gastroenterol 2011; 17: 423-424 [PMID: 22064345 DOI: 10.4103/1319-3767.87188] Ma DW, Kim JH, Jeon TJ, Lee YC, Yun M, Youn YH, Park H, Lee SI. 18F-fluorodeoxyglucose positron emission tomography-computed tomography for the evaluation of bone metastasis in patients with gastric cancer. Dig Liver Dis 2013; 45: 769-775 [PMID: 23831128 DOI: 10.1016/j.dld.2013.02.009] Lazăr D, Raica M, Sporea I, Tăban S, Goldiş A, Cornianu M. Tumor angiogenesis in gastric cancer. Rom J Morphol Embryol 2006; 47: 5-13 [PMID: 16838051] Ribatti D, Guidolin D, Marzullo A, Nico B, Annese T, Benagiano V, Crivellato E. Mast cells and angiogenesis in gastric carcinoma. Int J Exp Pathol 2010; 91: 350-356 [PMID: 20412338 DOI: 10.1111/ j.1365-2613.2010.00714.x] Ranieri G, Pantaleo M, Piccinno M, Roncetti M, Mutinati M, Marech I, Patruno R, Rizzo A, Sciorsci RL. Tyrosine kinase inhibitors (TKIs) in human and pet tumours with special reference to breast cancer: a comparative review. Crit Rev Oncol Hematol 2013; 88: 293-308 [PMID: 23768779 DOI: 10.1016/j.critrevonc.2013.05.0 09] Ranieri G, Gadaleta CD, Patruno R, Zizzo N, Daidone MG, Hansson MG, Paradiso A, Ribatti D. A model of study for human cancer: Spontaneous occurring tumors in dogs. Biological features and translation for new anticancer therapies. Crit Rev Oncol Hematol 2013; 88: 187-197 [PMID: 23561333 DOI: 10.1016/j.critrevonc.201 3.03.005] Xue Z, Wu X, Chen X, Liu Y, Wang X, Wu K, Nie Y, Fan D. Mesenchymal stem cells promote epithelial to mesenchymal transition and metastasis in gastric cancer though paracrine cues and close physical contact. J Cell Biochem 2015; 116: 618-627 [PMID: 25399738 DOI: 10.1002/jcb.25013] Ranieri G, Ammendola M, Patruno R, Celano G, Zito FA, Montemurro S, Rella A, Di Lecce V, Gadaleta CD, Battista De Sarro G, Ribatti D. Tryptase-positive mast cells correlate with angiogenesis in early breast cancer patients. Int J Oncol 2009; 35: 115-120 [PMID: 19513558] Gulubova M, Vlaykova T. Prognostic significance of mast cell num­ ber and microvascular density for the survival of patients with primary colorectal cancer. J Gastroenterol Hepatol 2009; 24: 1265-1275 [PMID: 17645466 DOI: 10.1111/j.1440-1746.2007.05009.x] Ribatti D, Crivellato E. Mast cells, angiogenesis and cancer. Adv Exp Med Biol 2011; 716: 270-288 [PMID: 21713661 DOI: 10.1007/ 978-1-4419-9533-9_14] Crivellato E, Nico B, Ribatti D. Mast cell contribution to tumor angiogenesis: a clinical approach. Eur Cytokine Netw 2009; 20: 197-206 [PMID: 20167559 DOI: 10.1684/ecn.2009.0167] Ammendola M, Leporini C, Marech I, Gadaleta CD, Scognamillo G, Sacco R, Sammarco G, De Sarro G, Russo E, Ranieri G. Targeting mast cells tryptase in tumor microenvironment: a potential antiangiogenetic strategy. Biomed Res Int 2014; 2014: 154702 [PMID: 25295247 DOI: 10.1155/2014/154702] Starkey JR, Crowle PK, Taubenberger S. Mast-cell-deficient W/Wv mice exhibit a decreased rate of tumor angiogenesis. Int J Cancer 1988; 42: 48-52 [PMID: 2455691] Ribatti D, Crivellato E. The controversial role of mast cells in tumor

WJG|www.wjgnet.com

20

21

22

23 24

25

26 27

28

29

30

31

32

33

34

10499

growth. Int Rev Cell Mol Biol 2009; 275: 89-131 [PMID: 19491054 DOI: 10.1016/S1937-6448(09)75004-X] Ammendola M, Sacco R, Sammarco G, Donato G, Zuccalà V, Luposella M, Patruno R, Marech I, Montemurro S, Zizzo N, Gadaleta CD, Ranieri G. Mast cells density positive to tryptase correlates with angiogenesis in pancreatic ductal adenocarcinoma patients having undergone surgery. Gastroenterol Res Pract 2014; 2014: 951957 [PMID: 24995015 DOI: 10.1155/2014/951957] Marech I, Ammendola M, Sacco R, Capriuolo GS, Patruno R, Rubini R, Luposella M, Zuccalà V, Savino E, Gadaleta CD, Ribatti D, Ranieri G. Serum tryptase, mast cells positive to tryptase and microvascular density evaluation in early breast cancer patients: possible translational significance. BMC Cancer 2014; 14: 534 [PMID: 25056597 DOI: 10.1186/1471-2407-14-534] Ammendola M, Sacco R, Sammarco G, Donato G, Montemurro S, Ruggieri E, Patruno R, Marech I, Cariello M, Vacca A, Gadaleta CD, Ranieri G. Correlation between serum tryptase, mast cells positive to tryptase and microvascular density in colo-rectal cancer patients: possible biological-clinical significance. PLoS One 2014; 9: e99512 [PMID: 24915568 DOI: 10.1371/journal.pone.0099512] Ribatti D, Crivellato E. Mast cells, angiogenesis, and tumour growth. Biochim Biophys Acta 2012; 1822: 2-8 [PMID: 21130163 DOI: 10.1016/j.bbadis.2010.11.010] Hassan S, Kinoshita Y, Kawanami C, Kishi K, Matsushima Y, Ohashi A, Funasaka Y, Okada A, Maekawa T, He-Yao W, Chiba T. Expression of protooncogene c-kit and its ligand stem cell factor (SCF) in gastric carcinoma cell lines. Dig Dis Sci 1998; 43: 8-14 [PMID: 9508539] Ranieri G, Labriola A, Achille G, Florio G, Zito AF, Grammatica L, Paradiso A. Microvessel density, mast cell density and thymidine phosphorylase expression in oral squamous carcinoma. Int J Oncol 2002; 21: 1317-1323 [PMID: 12429983] Ranieri G. Hot topic: targeting tumor angiogenesis: an update. Curr Med Chem 2012; 19: 937 [PMID: 22214460] Ribatti D, Ranieri G, Nico B, Benagiano V, Crivellato E. Tryptase and chymase are angiogenic in vivo in the chorioallantoic membrane assay. Int J Dev Biol 2011; 55: 99-102 [PMID: 21425085 DOI: 10.1387/ijdb.103138dr] Ammendola M, Sacco R, Sammarco G, Donato G, Zuccalà V, Romano R, Luposella M, Patruno R, Vallicelli C, Verdecchia GM, Cavaliere D, Montemurro S, Ranieri G. Mast Cells Positive to Tryptase and c-Kit Receptor Expressing Cells Correlates with Angiogenesis in Gastric Cancer Patients Surgically Treated. Gastroenterol Res Pract 2013; 2013: 703163 [PMID: 24348541 DOI: 10.1155/2013/703163] Blair RJ, Meng H, Marchese MJ, Ren S, Schwartz LB, Tonnesen MG, Gruber BL. Human mast cells stimulate vascular tube formation. Tryptase is a novel, potent angiogenic factor. J Clin Invest 1997; 99: 2691-2700 [PMID: 9169499 DOI: 10.1172/JCI119458] Morris DR, Ding Y, Ricks TK, Gullapalli A, Wolfe BL, Trejo J. Protease-activated receptor-2 is essential for factor VIIa and Xa-induced signaling, migration, and invasion of breast cancer cells. Cancer Res 2006; 66: 307-314 [PMID: 16397244 DOI: 10.1158/0008-5472.CAN-05-1735] Yoshii M, Jikuhara A, Mori S, Iwagaki H, Takahashi HK, Nishibori M, Tanaka N. Mast cell tryptase stimulates DLD-1 carcinoma through prostaglandin- and MAP kinase-dependent manners. J Pharmacol Sci 2005; 98: 450-458 [PMID: 16093613] Ammendola M, Zuccalà V, Patruno R, Russo E, Luposella M, Amorosi A, Vescio G, Sammarco G, Montemurro S, De Sarro G, Sacco R, Ranieri G. Tryptase-positive mast cells and angiogenesis in keloids: a new possible post-surgical target for prevention. Updates Surg 2013; 65: 53-57 [PMID: 23117746 DOI: 10.1007/s13304-0120183-y] Sperr WR, Czerwenka K, Mundigler G, Müller MR, Semper H, Klappacher G, Glogar HD, Lechner K, Valent P. Specific activation of human mast cells by the ligand for c-kit: comparison between lung, uterus and heart mast cells. Int Arch Allergy Immunol 1993; 102: 170-175 [PMID: 7691300] Ribatti D, Vacca A, Ria R, Marzullo A, Nico B, Filotico R, Roncali

October 7, 2015|Volume 21|Issue 37|

Leporini C et al . Mast cells in advanced gastric cancer

35

36

37

38

39

40 41

42

43

44

45 46 47 48

49

50

L, Dammacco F. Neovascularisation, expression of fibroblast growth factor-2, and mast cells with tryptase activity increase simultaneously with pathological progression in human malignant melanoma. Eur J Cancer 2003; 39: 666-674 [PMID: 12628847] Ribatti D, Ennas MG, Vacca A, Ferreli F, Nico B, Orru S, Sirigu P. Tumor vascularity and tryptase-positive mast cells correlate with a poor prognosis in melanoma. Eur J Clin Invest 2003; 33: 420-425 [PMID: 12760367] Ribatti D, Finato N, Crivellato E, Marzullo A, Mangieri D, Nico B, Vacca A, Beltrami CA. Neovascularization and mast cells with tryptase activity increase simultaneously with pathologic progression in human endometrial cancer. Am J Obstet Gynecol 2005; 193: 1961-1965 [PMID: 16325597 DOI: 10.1016/j.ajog.2005.04.055] Ribatti D, Finato N, Crivellato E, Guidolin D, Longo V, Mangieri D, Nico B, Vacca A, Beltrami CA. Angiogenesis and mast cells in human breast cancer sentinel lymph nodes with and without micrometastases. Histopathology 2007; 51: 837-842 [PMID: 17944928 DOI: 10.1111/j.1365-2559.2007.02869.x] Ribatti D, Belloni AS, Nico B, Salà G, Longo V, Mangieri D, Crivellato E, Nussdorfer GG. Tryptase- and leptin-positive mast cells correlate with vascular density in uterine leiomyomas. Am J Obstet Gynecol 2007; 196: 470.e1-470.e7 [PMID: 17466707 DOI: 10.1016/ j.ajog.2006.12.039] Ammendola M, Sacco R, Donato G, Zuccalà V, Russo E, Luposella M, Vescio G, Rizzuto A, Patruno R, De Sarro G, Montemurro S, Sammarco G, Ranieri G. Mast cell positivity to tryptase correlates with metastatic lymph nodes in gastrointestinal cancer patients treated surgically. Oncology 2013; 85: 111-116 [PMID: 23887206 DOI: 10.1159/000351145] Kakeji Y, Maehara Y, Sumiyoshi Y, Oda S, Emi Y. Angiogenesis as a target for gastric cancer. Surgery 2002; 131: S48-S54 [PMID: 11821787] Kakeji Y, Koga T, Sumiyoshi Y, Shibahara K, Oda S, Maehara Y, Sugimachi K. Clinical significance of vascular endothelial growth factor expression in gastric cancer. J Exp Clin Cancer Res 2002; 21: 125-129 [PMID: 12071518] Ammendola M, Marech I, Sammarco G, Zuccalà V, Luposella M, Zizzo N, Patruno R, Crovace A, Ruggieri E, Zito AF, Gadaleta CD, Sacco R, Ranieri G. Infiltrating mast cells correlate with angiogenesis in bone metastases from gastric cancer patients. Int J Mol Sci 2015; 16: 3237-3250 [PMID: 25648323 DOI: 10.3390/ ijms16023237] Gurish MF, Boyce JA. Mast cells: ontogeny, homing, and recruitment of a unique innate effector cell. J Allergy Clin Immunol 2006; 117: 1285-1291 [PMID: 16750988 DOI: 10.1016/ j.jaci.2006.04.017] Chen CC, Grimbaldeston MA, Tsai M, Weissman IL, Galli SJ. Identification of mast cell progenitors in adult mice. Proc Natl Acad Sci USA 2005; 102: 11408-11413 [PMID: 16006518 DOI: 10.1073/ pnas.0504197102] Iemura A, Tsai M, Ando A, Wershil BK, Galli SJ. The c-kit ligand, stem cell factor, promotes mast cell survival by suppressing apoptosis. Am J Pathol 1994; 144: 321-328 [PMID: 7508684] Marshall JS. Mast-cell responses to pathogens. Nat Rev Immunol 2004; 4: 787-799 [PMID: 15459670 DOI: 10.1038/nri1460] Crivellato E, Ribatti D. The mast cell: an evolutionary perspective. Biol Rev Camb Philos Soc 2010; 85: 347-360 [PMID: 19961471 DOI: 10.1111/j.1469-185X.2009.00105.x] Mangia A, Malfettone A, Rossi R, Paradiso A, Ranieri G, Simone G, Resta L. Tissue remodelling in breast cancer: human mast cell tryptase as an initiator of myofibroblast differentiation. Histopathology 2011; 58: 1096-1106 [PMID: 21707711 DOI: 10.1111/j.1365-2559.2011.03842.x] Benítez-Bribiesca L, Wong A, Utrera D, Castellanos E. The role of mast cell tryptase in neoangiogenesis of premalignant and malignant lesions of the uterine cervix. J Histochem Cytochem 2001; 49: 1061-1062 [PMID: 11457936] Acikalin MF, Oner U, Topçu I, Yaşar B, Kiper H, Colak E. Tumour angiogenesis and mast cell density in the prognostic assessment of colorectal carcinomas. Dig Liver Dis 2005; 37: 162-169 [PMID:

WJG|www.wjgnet.com

51

52

53

54 55

56

57

58

59

60

61

62

63

64 65

66

10500

15888280 DOI: 10.1016/j.dld.2004.09.028] Marech I, Gadaleta CD, Ranieri G. Possible prognostic and therapeutic significance of c-Kit expression, mast cell count and microvessel density in renal cell carcinoma. Int J Mol Sci 2014; 15: 13060-13076 [PMID: 25056544 DOI: 10.3390/ijms150713060] Marech I, Ammendola M, Gadaleta C, Zizzo N, Oakley C, Gadaleta CD, Ranieri G. Possible biological and translational significance of mast cells density in colorectal cancer. World J Gastroenterol 2014; 20: 8910-8920 [PMID: 25083063 DOI: 10.3748/wjg.v20.i27.8910] Ribatti D, Ranieri G, Basile A, Azzariti A, Paradiso A, Vacca A. Tumor endothelial markers as a target in cancer. Expert Opin Ther Targets 2012; 16: 1215-1225 [PMID: 22978444 DOI: 10.1517/1472 8222.2012.725047] Uchima Y, Sawada T, Hirakawa K. Action of antiproteases on pancreatic cancer cells. JOP 2007; 8: 479-487 [PMID: 17625304] Bai Y, Bandara G, Ching Chan E, Maric I, Simakova O, Bandara SN, Lu WP, Wise SC, Flynn DL, Metcalfe DD, Gilfillan AM, Wilson TM. Targeting the KIT activating switch control pocket: a novel mechanism to inhibit neoplastic mast cell proliferation and mast cell activation. Leukemia 2013; 27: 278-285 [PMID: 22907049 DOI: 10.1038/leu.2012.218] Marech I, Patruno R, Zizzo N, Gadaleta C, Introna M, Zito AF, Gadaleta CD, Ranieri G. Masitinib (AB1010), from canine tumor model to human clinical development: where we are? Crit Rev Oncol Hematol 2014; 91: 98-111 [PMID: 24405856 DOI: 10.1016/ j.critrevonc.2013.12.011] Mbeunkui F, Johann DJ. Cancer and the tumor microenvironment: a review of an essential relationship. Cancer Chemother Pharmacol 2009; 63: 571-582 [PMID: 19083000 DOI: 10.1007/ s00280-008-0881-9] Huang B, Lei Z, Zhang GM, Li D, Song C, Li B, Liu Y, Yuan Y, Unkeless J, Xiong H, Feng ZH. SCF-mediated mast cell infiltration and activation exacerbate the inflammation and immunosuppression in tumor microenvironment. Blood 2008; 112: 1269-1279 [PMID: 18524989 DOI: 10.1182/blood-2008-03-147033] Welsh TJ, Green RH, Richardson D, Waller DA, O’Byrne KJ, Bradding P. Macrophage and mast-cell invasion of tumor cell islets confers a marked survival advantage in non-small-cell lung cancer. J Clin Oncol 2005; 23: 8959-8967 [PMID: 16219934 DOI: 10.1200/ JCO.2005.01.4910] Galli SJ, Grimbaldeston M, Tsai M. Immunomodulatory mast cells: negative, as well as positive, regulators of immunity. Nat Rev Immunol 2008; 8: 478-486 [PMID: 18483499 DOI: 10.1038/ nri2327] Maltby S, Khazaie K, McNagny KM. Mast cells in tumor growth: angiogenesis, tissue remodelling and immune-modulation. Biochim Biophys Acta 2009; 1796: 19-26 [PMID: 19233249 DOI: 10.1016/ j.bbcan.2009.02.001] Shin K, Watts GF, Oettgen HC, Friend DS, Pemberton AD, Gurish MF, Lee DM. Mouse mast cell tryptase mMCP-6 is a critical link between adaptive and innate immunity in the chronic phase of Trichinella spiralis infection. J Immunol 2008; 180: 4885-4891 [PMID: 18354212] Mukherjee S, Bandyopadhyay G, Dutta C, Bhattacharya A, Karmakar R, Barui G. Evaluation of endoscopic biopsy in gastric lesions with a special reference to the significance of mast cell density. Indian J Pathol Microbiol 2009; 52: 20-24 [PMID: 19136773] Ribatti D, Crivellato E. Immune cells and angiogenesis. J Cell Mol Med 2009; 13: 2822-2833 [PMID: 19538473 DOI: 10.1111/ j.1582-4934.2009.00810.x] Yano H, Kinuta M, Tateishi H, Nakano Y, Matsui S, Monden T, Okamura J, Sakai M, Okamoto S. Mast cell infiltration around gastric cancer cells correlates with tumor angiogenesis and metastasis. Gastric Cancer 1999; 2: 26-32 [PMID: 11957067 DOI: 10.1007/s101209900023] Kondo K, Muramatsu M, Okamoto Y, Jin D, Takai S, Tanigawa N, Miyazaki M. Expression of chymase-positive cells in gastric cancer and its correlation with the angiogenesis. J Surg Oncol 2006; 93: 36-42; discussion 42-3 [PMID: 16353179 DOI: 10.1002/jso.20394]

October 7, 2015|Volume 21|Issue 37|

Leporini C et al . Mast cells in advanced gastric cancer 67

68 69

70

71 72 73 74 75

76

77

78

79 80

81

82

Zhao Y, Wu K, Cai K, Zhai R, Tao K, Wang G, Wang J. Increased numbers of gastric-infiltrating mast cells and regulatory T cells are associated with tumor stage in gastric adenocarcinoma patients. Oncol Lett 2012; 4: 755-758 [PMID: 23205096 DOI: 10.3892/ ol.2012.830] Wissmann C, Detmar M. Pathways targeting tumor lymphan­ giogenesis. Clin Cancer Res 2006; 12: 6865-6868 [PMID: 17145802 DOI: 10.1158/1078-0432.CCR-06-1800] Jiang YA, Zhang YY, Luo HS, Xing SF. Mast cell density and the context of clinicopathological parameters and expression of p185, estrogen receptor, and proliferating cell nuclear antigen in gastric carcinoma. World J Gastroenterol 2002; 8: 1005-1008 [PMID: 12439914] Caruso RA, Ieni A, Fabiano V, Basile G, Inferrera C. Perivascular mast cells in advanced gastric adenocarcinomas: an electron microscopic study. Anticancer Res 2004; 24: 2257-2263 [PMID: 15330170] Ferrara N. Role of vascular endothelial growth factor in the regulation of angiogenesis. Kidney Int 1999; 56: 794-814 [PMID: 10469350 DOI: 10.1046/j.1523-1755.1999.00610.x] Devkaran B, Jhobta R, Verma DK. Bony metastasis of gastric adenocarcinoma. Saudi J Gastroenterol 2009; 15: 137-138 [PMID: 19568584 DOI: 10.4103/1319-3767.49009] Ahn JB, Ha TK, Kwon SJ. Bone metastasis in gastric cancer patients. J Gastric Cancer 2011; 11: 38-45 [PMID: 22076200 DOI: 10.5230/jgc.2011.11.1.38] Chung YS, Choi TY, Ha CY, Kim HM, Lee KJ, Park CH, Fitzpatrick LA. An unusual case of osteoblastic metastasis from gastric carcinoma. Yonsei Med J 2002; 43: 377-380 [PMID: 12089748] Santos VM, Vieira TA, Marinho CS, Loures TP, Brandao BB, Botan RN. Infiltrating gastric adenocarcinoma with disseminated osteoblastic metastases. An Sist Sanit Navar 2013; 36: 153-157 [PMID: 23648509] Kobayashi M, Okabayashi T, Sano T, Araki K. Metastatic bone cancer as a recurrence of early gastric cancer -- characteristics and possible mechanisms. World J Gastroenterol 2005; 11: 5587-5591 [PMID: 16237749] Maehara Y, Hasuda S, Abe T, Oki E, Kakeji Y, Ohno S, Sugimachi K. Tumor angiogenesis and micrometastasis in bone marrow of patients with early gastric cancer. Clin Cancer Res 1998; 4: 2129-2134 [PMID: 9748130] Jauch KW, Heiss MM, Gruetzner U, Funke I, Pantel K, Babic R, Eissner HJ, Riethmueller G, Schildberg FW. Prognostic significance of bone marrow micrometastases in patients with gastric cancer. J Clin Oncol 1996; 14: 1810-1817 [PMID: 8656249] Zhang W, Stoica G, Tasca SI, Kelly KA, Meininger CJ. Modulation of tumor angiogenesis by stem cell factor. Cancer Res 2000; 60: 6757-6762 [PMID: 11118063] Nakae S, Suto H, Kakurai M, Sedgwick JD, Tsai M, Galli SJ. Mast cells enhance T cell activation: Importance of mast cell-derived TNF. Proc Natl Acad Sci USA 2005; 102: 6467-6472 [PMID: 15840716 DOI: 10.1073/pnas.0501912102] D’Amico L, Satolli MA, Mecca C, Castiglione A, Ceccarelli M, D’ Amelio P, Garino M, De Giuli M, Sandrucci S, Ferracini R, Roato I. Bone metastases in gastric cancer follow a RANKL-independent mechanism. Oncol Rep 2013; 29: 1453-1458 [PMID: 23404437 DOI: 10.3892/or.2013.2280] Ciurea R, Mărgăritescu C, Simionescu C, Stepan A, Ciurea M. VEGF and his R1 and R2 receptors expression in mast cells of oral squamous cells carcinomas and their involvement in tumoral angiogenesis. Rom J Morphol Embryol 2011; 52: 1227-1232 [PMID: 22203927]

83 84 85 86

87

88

89

90

91

92

93 94

95

96

97

Roodman GD. Mechanisms of bone metastasis. N Engl J Med 2004; 350: 1655-1664 [PMID: 15084698 DOI: 10.1056/NEJMra030831] Buijs JT, Juárez P, Guise TA. Therapeutic strategies to target TGF-β in the treatment of bone metastases. Curr Pharm Biotechnol 2011; 12: 2121-2137 [PMID: 21619539] Juárez P, Guise TA. TGF-β in cancer and bone: implications for treatment of bone metastases. Bone 2011; 48: 23-29 [PMID: 20699127 DOI: 10.1016/j.bone.2010.08.004] Teraoka H, Sawada T, Yamashita Y, Nakata B, Ohira M, Ishikawa T, Nishino H, Hirakawa K. TGF-beta1 promotes liver metastasis of pancreatic cancer by modulating the capacity of cellular invasion. Int J Oncol 2001; 19: 709-715 [PMID: 11562745] Itonaga I, Sabokbar A, Sun SG, Kudo O, Danks L, Ferguson D, Fujikawa Y, Athanasou NA. Transforming growth factor-beta induces osteoclast formation in the absence of RANKL. Bone 2004; 34: 57-64 [PMID: 14751563] Liu Y, Mueller BM. Protease-activated receptor-2 regulates vascular endothelial growth factor expression in MDA-MB-231 cells via MAPK pathways. Biochem Biophys Res Commun 2006; 344: 1263-1270 [PMID: 16650817 DOI: 10.1016/j.bbrc.2006.04.005] Gee MS, Procopio WN, Makonnen S, Feldman MD, Yeilding NM, Lee WM. Tumor vessel development and maturation impose limits on the effectiveness of anti-vascular therapy. Am J Pathol 2003; 162: 183-193 [PMID: 12507901 DOI: 10.1016/S0002-9440(10)63809-6] Uwagawa T, Misawa T, Sakamoto T, Ito R, Gocho T, Shiba H, Wakiyama S, Hirohara S, Sadaoka S, Yanaga K. A phase I study of full-dose gemcitabine and regional arterial infusion of nafamostat mesilate for advanced pancreatic cancer. Ann Oncol 2009; 20: 239-243 [PMID: 18836085 DOI: 10.1093/annonc/mdn640] Gocho T, Uwagawa T, Furukawa K, Haruki K, Fujiwara Y, Iwase R, Misawa T, Ohashi T, Yanaga K. Combination chemotherapy of serine protease inhibitor nafamostat mesilate with oxaliplatin targeting NF-κB activation for pancreatic cancer. Cancer Lett 2013; 333: 89-95 [PMID: 23348695 DOI: 10.1016/j.canlet.2013.01.019] Fujiwara Y, Furukawa K, Haruki K, Shimada Y, Iida T, Shiba H, Uwagawa T, Ohashi T, Yanaga K. Nafamostat mesilate can prevent adhesion, invasion and peritoneal dissemination of pancreatic cancer thorough nuclear factor kappa-B inhibition. J Hepatobiliary Pancreat Sci 2011; 18: 731-739 [PMID: 21484229 DOI: 10.1007/ s00534-011-0390-9] Arlt A, Schäfer H. NFkappaB-dependent chemoresistance in solid tumors. Int J Clin Pharmacol Ther 2002; 40: 336-347 [PMID: 12467302] Haruki K, Shiba H, Fujiwara Y, Furukawa K, Iwase R, Uwagawa T, Misawa T, Ohashi T, Yanaga K. Inhibition of nuclear factor-κB enhances the antitumor effect of paclitaxel against gastric cancer with peritoneal dissemination in mice. Dig Dis Sci 2013; 58: 123-131 [PMID: 22806547 DOI: 10.1007/s10620-012-2311-4] Dahiya N, Khadka A, Sharma AK, Gupta AK, Singh N, Brashier DB. Denosumab: A bone antiresorptive drug. Med J Armed Forces India 2015; 71: 71-75 [PMID: 25609868 DOI: 10.1016/ j.mjafi.2014.02.001] Roza T, Hakim L, van Poppel H, Joniau S. Bone-targeted therapies for elderly patients with renal cell carcinoma: current and future directions. Drugs Aging 2013; 30: 877-886 [PMID: 24072355 DOI: 10.1007/s40266-013-0117-5] Deplanque G, Demarchi M, Hebbar M, Flynn P, Melichar B, Atkins J, Nowara E, Moyé L, Piquemal D, Ritter D, Dubreuil P, Mansfield CD, Acin Y, Moussy A, Hermine O, Hammel P. A randomized, placebo-controlled phase III trial of masitinib plus gemcitabine in the treatment of advanced pancreatic cancer. Ann Oncol 2015; 26: 1194-1200 [PMID: 25858497 DOI: 10.1093/annonc/mdv133] P- Reviewer: Kim SS, Nowara E, Wei D S- Editor: Ma YJ L- Editor: Stewart G E- Editor: Wang CH

WJG|www.wjgnet.com

10501

October 7, 2015|Volume 21|Issue 37|

Published by Baishideng Publishing Group Inc 8226 Regency Drive, Pleasanton, CA 94588, USA Telephone: +1-925-223-8242 Fax: +1-925-223-8243 E-mail: [email protected] Help Desk: http://www.wjgnet.com/esps/helpdesk.aspx http://www.wjgnet.com

I S S N  1 0  0 7  -   9  3 2  7 3   7

9   7 7 1 0  0 7   9 3 2 0 45

© 2015 Baishideng Publishing Group Inc. All rights reserved.

Targeting mast cells in gastric cancer with special reference to bone metastases.

Bone metastases from gastric cancer (GC) are considered a relatively uncommon finding; however, they are related to poorer prognosis. Both primary GC ...
NAN Sizes 0 Downloads 8 Views