Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Immunol. Mar 27, 2016; 6(1): 9-18
Published online Mar 27, 2016. doi: 10.5411/wji.v6.i1.9
Role of tumor associated macrophages in regulating pancreatic cancer progression
Raul Caso, NYU School of Medicine, NYU Langone Medical Center, New York, NY 10016, United States
George Miller, Departments of Surgery and Cell Biology, NYU Langone Medical Center, New York, NY 10016, United States
Author contributions: Both authors contributed to this manuscript.
Conflict-of-interest statement: No potential 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: George Miller, MD, Departments of Surgery and Cell Biology, NYU Langone Medical Center, 430 E. 29th St., Suite 660, New York, NY 10016, United States. george.miller@nyumc.org
Telephone: +1-646-5012208 Fax: +1-646-5014564
Received: July 28, 2015
Peer-review started: July 29, 2015
First decision: October 13, 2015
Revised: December 8, 2015
Accepted: December 29, 2015
Article in press: January 4, 2016
Published online: March 27, 2016
Processing time: 244 Days and 7.1 Hours

Abstract

Pancreatic cancer has an overall 5-year survival rate of less than 5%. Unfortunately, patient survival has not substantially improved in the last couple of decades despite advances in treatment modalities that have been successful in other cancer types. The poor response of pancreatic cancer to therapy is a major obstacle faced by clinicians. Increasing attention is being paid to how tumor cells and non-tumor cells influence each other in the pancreatic tumor microenvironment. Tumor-associated macrophages (TAMs) are a highlight in this field because of their vast presence in the tumor microenvironment. TAMs promote angiogenesis, metastasis, and suppress the anti-tumor immune response. Here we review the current understanding of the role of TAMs in regulating the progression of pancreatic cancer.

Key Words: Pancreatic cancer, Tumor-associated macrophages, Tumor microenvironment, Macrophages

Core tip: Pancreatic cancer remains one of the most deadly cancers with dismal 5-year survival rates. Increasing importance is being given to the role of macrophages in pancreatic cancer. Tumor-associated promote angiogenesis, metastasis, and suppress the anti-tumor immune response. Targeting macrophages within the tumor microenvironment is an attractive novel therapeutic approach. Here we review the current understanding of the role of tumor-associated macrophages in the progression of pancreatic cancer.



PANCREATIC CANCER

Pancreatic cancer is the eighth leading cause of death from cancer in men and the ninth leading cause of death from cancer in women worldwide and is lethal in more than 95% of cases[1]. The incidence of pancreatic cancer ranges from 1 to 10 cases per 100000 people, is generally higher in developed countries, and has remained stable for the past 30 years relative to the incidence of other common solid tumors[2]. It is more common in the elderly and less than 20% of patients present with localized, potentially curable tumors[3]. The overall 5-year survival rate among patients with pancreatic cancer is less than 5%[4].

The causes of pancreatic cancer are not yet fully understood. Environmental factors have been implicated, yet evidence of a causative role only exists for tobacco use. The risk of pancreatic cancer in those that smoke is 2.5 to 3.6 times that in nonsmoking individuals[5]. Several medical conditions are associated with an increased risk of pancreatic cancer, including diabetes, chronic pancreatitis, chronic cirrhosis, a high-fat diet, and a high-cholesterol diet[3]. Additionally, it is estimated that 5% to 10% of cases are inherited, however currently there is no effective screening tool[6].

Pancreatic ductal adenocarcinoma (PDAC) is the most common form of pancreatic cancer. The cancer arises in the ductal epithelium and evolves from premalignant lesions to fully invasive cancer. The lesion called pancreatic intraepithelial neoplasia (PanIN) is the best characterized histologic precursor of pancreatic cancer[3]. The progression from dysplasia to invasive carcinoma is paralleled by the accumulation of mutations that include activation of the KRAS2 oncogene, inactivation of the tumor-suppressor gene CDKN2A, and inactivation of the tumor-suppressor genes TP53 and DPC4[7].

In the past two decades, almost no progress has been made in the clinical management or outcomes of patients with pancreatic cancer[1]. Approximately 70% of patients diagnosed with pancreatic cancer die from extensive metastatic disease while 30% present with limited metastasis, yet the majority of these patients have bulky primary tumors[8]. The poor response of pancreatic cancer to therapy is a major obstacle faced by clinicians. Patient survival has not substantially improved despite advances in treatment modalities that have proven successful in other cancers (i.e., breast, lung, colorectal, and melanoma)[9-11]. Novel biologic therapies such as cancer vaccines and methodologies using therapeutic gene transfer have also been either ineffective or shown limited promise[12]. The current standard chemotoxic drug, gemcitabine, has resulted in only a modest improvement in survival, and newer therapies show only marginal improvement[13]. Surgical resection is ultimately the most effective for pancreatic cancer. However, only 15% to 20% of patients are considered candidates for surgical resection[6] and outcomes of surgery alone are poor as less than 15% of resected patients are alive 5 years post-operatively[14]. Therefore, the development of new treatment strategies for the vast majority of patients afflicted with pancreatic cancer is vital.

PANCREATIC TUMOR MICROENVIRONMENT

The development of PDAC is marked by increasing desmoplasia, which results in a vast stroma that often exceeds the epithelial component of the tumor. Unlike most adenocarcinomas whose volume is comprised primarily of transformed cells, a pancreatic tumor is comprised of fibro-inflammatory stromal elements and islands of neoplastic epithelium[15,16]. Recent evidence suggests that far from being a passive observer, pancreatic tumor stroma affects both cancer progression and clinical outcome. In PDAC, activated pancreatic stellate cells have been shown to support tumor growth and immune dysfunction[17]. In particular, by releasing nutrient growth factors, such as insulin-like growth factor 2 and PDGF, into the tumor microenvironment, the stromal component of pancreatic cancer has been closely linked to both tumor growth and invasiveness[18-20]. In addition, chemotherapy resistance has been correlated with the extent of tumor desmoplasia as the stroma is thought to be a physical barrier to cytotoxic agents reaching the neoplastic epithelial cells[21,22]. Consequently, increasing attention is being paid to how tumor cells and non-tumor cells influence each other in the tumor microenvironment. Tumor-associated macrophages (TAMs) are a highlight in this field. Here we review the recent evidence for the role TAMs possess in promoting pancreatic cancer progression.

MACROPHAGE POLARIZATION

The immune system plays a critical role in the response to infection and elimination of foreign pathogens. In the context of cancer, the immune system recognizes antigens produced by tumor cells; therefore the cancer cell is deemed a foreign pathogen. Histopathological analyses of human tumors have provided evidence that variable numbers of infiltrating immune cells, such as macrophages, mast cells, granulocytes and myeloid-derived suppressor cells (MDSCs), are found in most cases infiltrating or surrounding tumor beds both in the core and at the invasive front of the tumor[23].

Macrophages play an important role in the innate immune system. They are essential in removing dead or dying cells and debris via phagocytosis, and activate the adaptive immune response[24]. After tissue damage, macrophages organize immune defenses and coordinate the tissue repair process. Normally, this process is self-limiting, yet if it doesn’t resolve it leads to chronic inflammation characterized by an alteration in the immune cell types involved, including an increase in infiltrating macrophages[25].

Macrophages are activated in response to environmental signals, including microbial products and cytokines. Analogous to the dichotomous differentiation of T helper cells giving rise to T helper type 1 (Th1) and T helper type 2 (Th2) T cells with distinct cytokine expression patterns and immunological functions, two fundamental macrophage phenotypes have been delineated. Activated macrophages are functionally divided into two subtypes: M1 (classical activated) and M2 (alternative activated)[26]. Th1-related cytokines such as Interferon gamma (IFN-γ), and lipopolysaccharide (LPS), polarize macrophages to M1[24]. M1 macrophages are characterized by IL-12hi, IL-23hi, tumor necrosis factor alpha (TNFα)hi, IL-10low, CXCL9hi, CXCL10hi, ROIhi, RNIhi, COX1low, COX2hi and iron uptake phenotype[24,27]. M1 macrophages preferentially induce anti-tumor Th1 cells, whereas Th2 cytokines such as IL-4 and IL-13 induce M2 macrophages[28]. These macrophages express IL-12low, TNFαlow, IL-10hi, IL-1decoyRhi, IL-1RAhi, Arginase1hi, CCL17hi, CCL18hi, CCL22hi, CCL24hi, COX1hi, COX2low, iron release, and increased phagocytic activity[24]. M2 macrophages initiate pro-tumorigenic actions by promoting angiogenesis, tissue repair, and inducing immune suppression via induction of Th2 cells[27]. It is worth noting that M1 and M2 macrophage designations are extreme ends of a fluid scale. In fact, there is ample literature describing the plasticity of macrophages. Macrophages receive signals from the microenvironment in which they reside[27] and it is the integration of these signals that ultimately determines the macrophage subtypes.

MACROPHAGE RECRUITMENT

The development and progression of pancreatic cancer has been linked to inflammation[29-31], with chronic inflammation being a risk factor for the development of PDAC[32-35]. The inflammatory microenvironment that is characteristic of PDAC supports tumorigenesis through paracrine crosstalk between tumor cells and immune cells[36]. Importantly, inflammatory conditions are not only prominent in the tumor microenvironment but also present in the peripheral blood of PDAC patients, thus highlighting a role for peripheral immune cells in disease progression[37]. Once established, PDAC is characterized by marked leukocyte infiltration.

TAMs are the main population of inflammatory cells found in the tumor microenvironment of solid tumors[38]. TAMs develop in response to tumor-induced cytokines, chemokines, and vascular endothelial growth factor (VEGF)[26,39,40]. Additionally, it is also well documented that granulocyte macrophage colony-stimulating factor (GM-CSF), released from the tumor microenvironment, regulates TAM recruitment, maturation, and differentiation. The matricellular glycoprotein secreted protein acidic and rich in cysteine (SPARC), an intracellular matrix protein, may influence macrophage infiltration and distribution in murine pancreatic tumors. Murine macrophages expressing F4/80 are more plentiful in tumors from wild-type SPARC mice and are distributed at the tumor margins[41]. Additionally, monocyte chemoattractants, such as the alarmins, may also play a role in the recruitment of monocytes and other macrophage precursors[42]. A factor released by dying tumor cells, the high mobility group box protein 1, is found in areas where TAMs reside[43].

TAMS CAN PROMOTE TUMOR PROGRESSION

The role that TAMs play in the tumor microenvironment remains controversial. For instance, in colorectal tumors, TAMs are pro-inflammatory, and are anti-tumor, which has been associated with a good prognosis[44]. However, in most tumors, the protumoral role of TAMs is supported by clinical studies that have reported a correlation between high macrophage content and poor prognosis[45-49]. Epidemiological data suggests that a tumor microenvironment that is rich in macrophages will lead to a more aggressive tumor with the potential for metastasis[50].

In pancreas cancer, there is evidence that TAMs infiltrate early lesions or PanINs and persist through invasive cancer[35]. It is well documented that early and advanced lesions have significant increases in macrophage infiltration, compared to normal pancreatic tissue. Moreover, evidence suggests that TAMs may regulate PanIN development. TAMs produce IL-6 in PanIN lesions of KrasG12D-expressing mice, therefore inducing STAT3 signaling and promoting cancer progression[51]. Additionally, pancreatic cancer cell proliferation via sonic hedgehog production has been linked to NFκB-activated monocytes[52]. Liou et al[33] identified the macrophage-secreted inflammatory cytokines RANTES and TNF as mediators of acinar cell dedifferentiation and tumor initiation. These cytokines act via the activation of NFκB and its target genes involved in regulating survival, proliferation, and degradation of extracellular matrix. Furthermore, the authors identified matrix metalloproteinases (MMPs) as targets that drive dedifferentiation and show that MMP inhibitors may be efficiently applied to block inflammation-induced dedifferentiation[33]. Helm et al[53] conducted studies on the functional impact of TAMs from human PDAC tissues on premalignant and malignant pancreatic ductal epithelial cells. Both cell types acquired an elongated cell shape along with an increased expression of vimentin and a reduced expression of epithelial E-cadherin when indirectly cocultured with TAMs from PDAC tissues[53]. When the cells were cocultured in the presence of polarized M1- and M2-macrophages, to elucidate whether pro- or anti-inflammatory properties account for these effects, similar to TAMs, both macrophage subsets induced epithelial-mesenchymal transition alterations and even greater invasiveness in the malignant epithelial cells[53]. Similar findings have been reported by others, further elucidating the impact of pancreatic cancer cell interaction with macrophages on the differentiation and function of macrophages and behaviors of pancreatic cancer cells[54]. Tumor growth inhibition following clodronate depletion of macrophages further clarifies the pivotal role macrophages have in tumor progression[55].

M2 macrophages

Macrophages often express an M2-phenotype at the tumor site[56]. During the early stages of pancreas tumor initiation and development, M1-macrophages are more abundant[57,58] and as the cancer progresses, macrophages switch to an M2-like phenotype[27,59-61]. In human patients, more M2-macrophages were detected in the context of pancreatic cancer than in chronic pancreatitis[62]. Furthermore, in pancreatic cancer patients a higher number of M2-macrophages was linked to larger tumor size, early liver recurrence, local recurrence, and reduced survival[62]. Similar results have been reported by others[63-65]. A higher percentage of M1-macrophages has been associated with longer survival[66].

Interestingly, Tjomsland et al[67] found evidence that high gene expression levels of CD68 (general macrophage marker) might be associated with poor prognosis and thus decreased survival following tumor resection, whereas a CD163-dominating M2-macrophage phenotype conferred a survival advantage[67]. Helm et al[68] have shown that human PDAC-associated TAMs concomitantly exhibit characteristics of pro-inflammatory M1-macrophages (e.g., HLA-DR, IL-1B, TNF-α) and anti-inflammatory M2-macrophages (e.g., CD163, IL-10). The authors also found HLA-DR and CD163 double positive cells by immunohistochemistry staining in pancreatic tissue of PDAC and chronic pancreatitis[68], thus confirming that macrophages with a mixed phenotype sharing pro- and anti-inflammatory properties are abundant in PDAC and are already present in the setting of chronic pancreatitis. These findings challenge the dichotomous concept of M1- and M2-macrophages. Importantly, their location in the tumor and the tumor stage might affect their ability to contribute to tumor promotion[27], which may explain conflicting findings.

Given the aforementioned findings, reprogramming TAMs towards a more anti-tumorigenic phenotype may prove promising. An agonist monoclonal antibody against CD40, a co-stimulatory protein found on professional antigen-presenting cells, has demonstrated efficacy in mouse models of PDAC[69]. By reprogramming M2 TAMs into M1 TAMs using a CD40 agonist, tumor immune surveillance was restored[69]. This increased the therapeutic efficacy of gemcitabine. These results show that tumor immunosurveillance can at times be governed strictly by innate immunity under the regulation of the CD40 pathway. A CD40 agonist has also demonstrated efficacy in human patients with PDAC[70] when delivered in combination with the chemotherapeutic agent gemcitabine, ostensibly via the anti-tumor activities of macrophages[71]. Low-dose tumor irradiation may enable recruitment of antitumor T cells[72]. Using a mouse model, Klug et al[72] have shown that irradiation triggers the polarization of M2-macrophages toward M1-macrophages that express iNOS. They further show that iNOS activity was responsible for vascular normalization and activation, T cell recruitment, and tumor rejection. The authors also obtained similar results when transferring irradiated macrophages and T cells, thus representing a promising adjuvant therapeutic strategy[72]. These results hold great promises in the treatment of pancreatic cancer.

Immune suppressive activities of TAMs

Tumor immunosuppression is a recognized mechanism for regulating tumor growth and when it succeeds, tumor progression continues mostly unchecked. TAMs have been described as key players of the tumor microenvironment contributing to immunosuppression by secreting TGF-β, IL-10, and arginase 1[73-75]. TGF-β promotes TAM polarization to an M2-phenotype, further promoting TGF-β release and immunosuppression[56]. Additionally, TGF-β promotes Th2 cell differentiation, resulting in an inefficient antitumor response[76]. Interestingly, Zhang et al[77] provide a novel model to explain the paradoxical role of TGF-β in the development of PDAC. Although TGF-β receptor signaling may inhibit cancer cell growth in the early stages of PDAC, its promoting role in angiogenesis may improve the long-term survival and progress of PDAC, thus it appears to be tumorigenesis-enhancing in the later stages of tumor progression[77]. TAM-derived IL-10 suppresses IL-12 expression[78], prevents in situ DC maturation[74,79], and suppresses IFN-γ release[80]. However, IL-10 may have a role in the antitumor immune response due to immunostimulating properties[81-83]. Arginase 1, a marker for M2-macrophages that is expressed in tumors, causes dysregulation of the T cell receptor (TCR) signal resulting in a deficient CD8+ T cell response[75,84]. Using a murine lung carcinoma model, Rodriguez et al[85] demonstrate that a subpopulation of mature tumor-associated myeloid cells express high levels of arginase 1. The authors also show that depletion of extracellular L-Arginine by tumor-associated myeloid cells blocked the expression of the TCR CD3ζ and inhibited antigen-specific T cell proliferation[85].

In human hepatocellular and ovarian carcinoma, CD14+ myeloid cells suppress autologous T cell proliferation and IFN-γ expression in vitro and nullify anti-tumor T cell activity during in vivo adoptive transfer experiments[86,87]. In pancreatic cancer, TAMs have a significant immunosuppressive role. CCR2 or CSF1R blockade with gemcitabine reduced TAM numbers, increased cytotoxic T cells, and decreased FOXP3+ Treg infiltration compared to gemcitabine alone[88]. These findings suggest that a reduction in macrophage infiltration at the tumor site resulted in an improved anti-tumor response in pancreatic cancer. More recently, using a retrospective cohort of patients with PDAC, Di Caro et al[89] report that the density of macrophages is a critical determinant of patient responsiveness to postsurgical conventional chemotherapy. In vitro, chemotherapy prevents the tumor-protective role of TAMs and reinstates their antitumor function, by a T-cell independent mechanism[89]. Targeting TAMs may be beneficial to tumor prognosis and in some cases be sufficient to ignite an effective antitumor action.

In addition to bona fide macrophages, there is extensive literature on abnormal accumulation of immature myeloid cells, known as MDSCs, which accumulate in the spleen and tumors as a consequence of tumor-associated changes in myelopoiesis and play a critical role in immunosuppression[90]. MDSCs represent a heterogeneous population of cells and are Gr-1+CD11b+[91]. Moreover, in vitro, Gr-1+CD11b+ MDSCs have the ability to impair T cell responses[92]. Although the relationship between MDSCs and TAMs is not yet entirely clear, in the context of tumor-derived factors, it has been suggested that MDSCs have the ability to differentiate in vitro into macrophages with immunosuppressive characteristics[93].

In pancreatic cancer, MDSCs may play a significant role in tumor progression. Using a murine model of PDAC, Bayne et al[94] demonstrate that GM-CSF, derived from the tumor, recruits Gr-1+CD11b+ myeloid cells that leads to the suppression of antitumor T cell response. In vivo abrogation of GM-CSF prevented the infiltration of MDSCs at the tumor site and blocked tumor progression[94]. Similar findings have been recently reported[95]. Interestingly, Pylayeva-Gupta et al[96] demonstrate that upregulation of GM-CSF in mouse pancreatic ductal epithelial cells is oncogenic KrasG12D-dependent. Mutational activation of Kras triggers the production of GM-CSF, promoting the expansion of immunosuppressive Gr-1+CD11b+ myeloid cells and leading to the evasion of CD8+ T cell-driven antitumor immunity. The suppression of GM-CSF production, in turn, inhibits the in vivo growth of KrasG12D tumors[96]. These findings provide insights into the challenges for designing effective therapeutic modalities targeting pancreatic cancer.

Strategies to reduce the effects of myeloid cell populations within the tumor microenvironment may offer great therapeutic potential. Using a mouse model of PDAC, Zhu et al[97] demonstrate that CSF1R blockade may enhance macrophage antigen presentation and T cell responses. CSF1R blockade upregulated the T cell checkpoint molecules PDL1 and CTLA4[97]. When PD1 and CTLA4 antagonists were combined with CSF1R blockade, this resulted in tumor regression. These findings provide a rationale to reprogram immunosuppressive myeloid cell populations in the tumor microenvironment under conditions that can significantly empower the therapeutic effects of checkpoint-based immunotherapeutics[97].

TAMs promote angiogenesis

Angiogenesis is a requirement for tumor growth and spread. TAMs have the capacity to regulate the vascular programming of tumors[27]. Upon activation, TAMs release multiple factors such as VEGF, PDGF, and TGF-β[98], all of which can promote angiogenesis. Vascular endothelial growth factor A (VEGFA) produced by TAMs is critical for angiogenesis and reverses the effects of macrophage depletion[99], whereas loss of VEGFA leads to vascular normalization[100].

However, while increased vascularization during tumor progression is recognized as an important step for the majority of solid tumors, in the context of pancreatic cancer this is unclear. Hypovascularized regions are characteristic of PDAC and has served as a diagnostic tool[101]. Moreover, this conflicts with the understanding that an angiogenic switch is required for tumor growth to occur[102]. Interestingly, in several animal models Sunitinib, an anti-angiogenic drug that targets VEGF and PDGF receptor signaling, showed progressive blockade of tumor growth but did not inhibit PDAC progression in a murine model[103]. In a murine model of pancreatic tumor of endocrine origin, which is highly vascularized and accounts for approximately 1% of pancreatic cancers in humans, Sunitinib reduced tumor formation[104]. Schmid et al[105,106] suggest that tumor vascularization may be promoted by circulating macrophages. The authors have shown that the chemoattractants SDF-1α and IL-1β collaborate with myeloid cell integrin-α4β1 to promote macrophage recruitment and adhesion to vascular endothelium resulting in tumor inflammation and growth. Inhibition of these molecules markedly decreased angiogenesis in pancreatic cancer models thus reducing tumor burden[105,106].

TAMs and tumor metastasis

A hallmark determining the severity of cancer is tumor metastasis, which is the result of tumor cells traveling through blood and lymphatic vessels to form ectopic tumors. Pancreatic cancer commonly metastasizes to the liver, peritoneum, lungs, and bones[104]. It has been suggested that TAMs have a role in tumor-cell migration, invasion, and metastasis[107]. There is an abundance of evidence showing how TAMs influence tumor cell migration. Precursors of TAMs, blood monocytes, may also contribute to the formation of an invasive microenvironment in a manner dependent on the secretion of soluble mediators such as TNF[108]. Conversely, pancreatic cancer cells induce differentiation of pro-tumor macrophages. Tumor-educated macrophages may promote pancreatic cancer cell invasion in vitro[109], thus underscoring the crosstalk that occurs between tumor cells and TAMs resulting in disease progression. It has been suggested that EGF released by TAMs interacts with CSF-1 produced by tumor cells to induce tumor cell migration[43]. TAMs also influence the tumor microenvironment by providing factors that promote tumor cell invasion, including proteases[110]. Macrophage-derived microRNA have also been suggested to regulate tumor invasion[111]. In pancreatic cancer of human origin, the macrophage inflammatory protein-3α (MIP-3α) has been linked to tumor cell invasion[112,113]. Interactions between MIP-3α and CCR6, the receptor expressed by PDAC cells, have been shown to induce PDAC cell proliferation, migration, and invasion in type IV collagen[112,113].

Pharmacologic inhibition of macrophage infiltration at the tumor site using the CSF1R inhibitors decreased liver metastasis in pancreatic tumor mouse models[88]. Focal adhesion kinase (FAK), which is a known regulator of cell migration, proliferation, apoptosis, and survival, has been linked to macrophage infiltration[114]. Using an inhibitor of FAK (PF-562,271), Stokes et al[114] found diminished migration of tumor cells, cancer-associated fibroblasts, and macrophages. Treatment of mice with this inhibitor reduced tumor growth, invasion, and metastases[114].

TAMs and cancer stem cells

Cancer stem cells (CSC) or cancer initiating cells are a specific subpopulation of cells with distinct stem cell properties, such as self-renewal and differentiation, with the ability to initiate tumorigenesis within tumors[115,116]. The interaction between CSCs and TAMs promotes tumor growth, maintains the CSC population, and reduces therapeutic efficacy. Furthermore, the histological grade of the malignancy has been found to positively correlate with the number of infiltrating macrophages because TAMs have been found distributed around CSC populations[117,118]. Wu et al[119] have shown that CSCs in glioma tissue recruit macrophages with an M2-phenotype that secrete IL-10 and TGF-β. The authors also found enhanced capacity of macrophages to inhibit T cell proliferation and therefore induce immunosuppression. In another study, blockade of macrophage infiltration lead to a reduction in the expression of ALDH, a CSC marker on pancreatic cancer cells[88].

Recently, Hou et al[120] found elevated expression levels of the CSC markers CD44/CD133 and TAM marker CD204 in human PDAC tissues. High coexpression of CD44/CD133 and CD204 was associated with shorter overall survival and disease-free survival. These findings suggest that for patients with PDAC the coexpression of CD44/CD133 and CD204 may be useful for predicting survival[120].

CONCLUSION

Pancreatic cancer remains one of the most common and deadly cancers. The limited success of chemotherapy in patients with pancreatic cancer is partly due to the complexity of the tumor microenvironment. Increasing attention is being given to the interactions of tumor cells and non-tumor cells found in the tumor microenvironment. In the pancreatic tumor microenvironment, TAMs are the most common cell population encountered. Accumulating evidence suggests that TAMs infiltrate early cancer lesions, subsequently promoting angiogenesis, tumor growth, spread, and immunosuppression. Additionally, the interaction between CSCs and TAMs promotes tumorigenicity, metastasis, maintains the CSC population, and impedes therapeutic efficacy. The success of studies inhibiting macrophage recruitments to the tumor microenvironment and re-activats their cancer cell killing activities provide evidence of novel therapies for future tumor management. Altogether, these findings suggest that targeting TAMs with anticancer therapies may represent a novel strategy to treat pancreatic cancer.

Footnotes

P- Reviewer: Corthay A, Ferrante A S- Editor: Ji FF L- Editor: A E- Editor: Wu HL

References
1.  Kuhn Y, Koscielny A, Glowka T, Hirner A, Kalff JC, Standop J. Postresection survival outcomes of pancreatic cancer according to demographic factors and socio-economic status. Eur J Surg Oncol. 2010;36:496-500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
2.  Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23762]  [Cited by in F6Publishing: 25229]  [Article Influence: 1940.7]  [Reference Citation Analysis (6)]
3.  Hidalgo M. Pancreatic cancer. N Engl J Med. 2010;362:1605-1617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2078]  [Cited by in F6Publishing: 2137]  [Article Influence: 152.6]  [Reference Citation Analysis (2)]
4.  Li D, Xie K, Wolff R, Abbruzzese JL. Pancreatic cancer. Lancet. 2004;363:1049-1057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1481]  [Cited by in F6Publishing: 1519]  [Article Influence: 76.0]  [Reference Citation Analysis (0)]
5.  Hassan MM, Bondy ML, Wolff RA, Abbruzzese JL, Vauthey JN, Pisters PW, Evans DB, Khan R, Chou TH, Lenzi R. Risk factors for pancreatic cancer: case-control study. Am J Gastroenterol. 2007;102:2696-2707.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 228]  [Cited by in F6Publishing: 223]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
6.  Ryan DP, Hong TS, Bardeesy N. Pancreatic adenocarcinoma. N Engl J Med. 2014;371:1039-1049.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1500]  [Cited by in F6Publishing: 1565]  [Article Influence: 156.5]  [Reference Citation Analysis (0)]
7.  Feldmann G, Beaty R, Hruban RH, Maitra A. Molecular genetics of pancreatic intraepithelial neoplasia. J Hepatobiliary Pancreat Surg. 2007;14:224-232.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 193]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
8.  Iacobuzio-Donahue CA, Fu B, Yachida S, Luo M, Abe H, Henderson CM, Vilardell F, Wang Z, Keller JW, Banerjee P. DPC4 gene status of the primary carcinoma correlates with patterns of failure in patients with pancreatic cancer. J Clin Oncol. 2009;27:1806-1813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 770]  [Cited by in F6Publishing: 821]  [Article Influence: 54.7]  [Reference Citation Analysis (0)]
9.  Katz MH, Fleming JB, Lee JE, Pisters PW. Current status of adjuvant therapy for pancreatic cancer. Oncologist. 2010;15:1205-1213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
10.  Nugent FW, Stuart K. Adjuvant and neoadjuvant therapy in curable pancreatic cancer. Surg Clin North Am. 2010;90:323-339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
11.  Winter JM, Brennan MF, Tang LH, D’Angelica MI, Dematteo RP, Fong Y, Klimstra DS, Jarnagin WR, Allen PJ. Survival after resection of pancreatic adenocarcinoma: results from a single institution over three decades. Ann Surg Oncol. 2012;19:169-175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 279]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
12.  He AR, Lindenberg AP, Marshall JL. Biologic therapies for advanced pancreatic cancer. Expert Rev Anticancer Ther. 2008;8:1331-1338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
13.  Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardière C. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817-1825.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4838]  [Cited by in F6Publishing: 5180]  [Article Influence: 398.5]  [Reference Citation Analysis (1)]
14.  Ferrone CR, Brennan MF, Gonen M, Coit DG, Fong Y, Chung S, Tang L, Klimstra D, Allen PJ. Pancreatic adenocarcinoma: the actual 5-year survivors. J Gastrointest Surg. 2008;12:701-706.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 184]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
15.  Miyamoto H, Murakami T, Tsuchida K, Sugino H, Miyake H, Tashiro S. Tumor-stroma interaction of human pancreatic cancer: acquired resistance to anticancer drugs and proliferation regulation is dependent on extracellular matrix proteins. Pancreas. 2004;28:38-44.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Korc M. Pancreatic cancer-associated stroma production. Am J Surg. 2007;194:S84-S86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 203]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
17.  Pandol S, Edderkaoui M, Gukovsky I, Lugea A, Gukovskaya A. Desmoplasia of pancreatic ductal adenocarcinoma. Clin Gastroenterol Hepatol. 2009;7:S44-S47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 165]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
18.  Tuveson DA, Shaw AT, Willis NA, Silver DP, Jackson EL, Chang S, Mercer KL, Grochow R, Hock H, Crowley D. Endogenous oncogenic K-ras(G12D) stimulates proliferation and widespread neoplastic and developmental defects. Cancer Cell. 2004;5:375-387.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  De Wever O, Mareel M. Role of tissue stroma in cancer cell invasion. J Pathol. 2003;200:429-447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 733]  [Cited by in F6Publishing: 707]  [Article Influence: 33.7]  [Reference Citation Analysis (0)]
20.  Abiatari I, Kleeff J, Li J, Felix K, Büchler MW, Friess H. Hsulf-1 regulates growth and invasion of pancreatic cancer cells. J Clin Pathol. 2006;59:1052-1058.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
21.  Hwang RF, Moore T, Arumugam T, Ramachandran V, Amos KD, Rivera A, Ji B, Evans DB, Logsdon CD. Cancer-associated stromal fibroblasts promote pancreatic tumor progression. Cancer Res. 2008;68:918-926.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 794]  [Cited by in F6Publishing: 890]  [Article Influence: 55.6]  [Reference Citation Analysis (0)]
22.  Kleeff J, Ishiwata T, Kumbasar A, Friess H, Büchler MW, Lander AD, Korc M. The cell-surface heparan sulfate proteoglycan glypican-1 regulates growth factor action in pancreatic carcinoma cells and is overexpressed in human pancreatic cancer. J Clin Invest. 1998;102:1662-1673.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 281]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
23.  Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer. 2012;12:298-306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3108]  [Cited by in F6Publishing: 3338]  [Article Influence: 278.2]  [Reference Citation Analysis (0)]
24.  Torroella-Kouri M, Rodríguez D, Caso R. Alterations in macrophages and monocytes from tumor-bearing mice: evidence of local and systemic immune impairment. Immunol Res. 2013;57:86-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
25.  Medzhitov R. Origin and physiological roles of inflammation. Nature. 2008;454:428-435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3548]  [Cited by in F6Publishing: 3954]  [Article Influence: 247.1]  [Reference Citation Analysis (0)]
26.  Sica A, Larghi P, Mancino A, Rubino L, Porta C, Totaro MG, Rimoldi M, Biswas SK, Allavena P, Mantovani A. Macrophage polarization in tumour progression. Semin Cancer Biol. 2008;18:349-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 812]  [Cited by in F6Publishing: 873]  [Article Influence: 54.6]  [Reference Citation Analysis (0)]
27.  Ruffell B, Affara NI, Coussens LM. Differential macrophage programming in the tumor microenvironment. Trends Immunol. 2012;33:119-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 649]  [Cited by in F6Publishing: 654]  [Article Influence: 54.5]  [Reference Citation Analysis (0)]
28.  Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32:593-604.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2742]  [Cited by in F6Publishing: 2894]  [Article Influence: 206.7]  [Reference Citation Analysis (0)]
29.  Lowenfels AB, Maisonneuve P, Cavallini G, Ammann RW, Lankisch PG, Andersen JR, Dimagno EP, Andrén-Sandberg A, Domellöf L. Pancreatitis and the risk of pancreatic cancer. International Pancreatitis Study Group. N Engl J Med. 1993;328:1433-1437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1255]  [Cited by in F6Publishing: 1110]  [Article Influence: 35.8]  [Reference Citation Analysis (0)]
30.  Evans A, Costello E. The role of inflammatory cells in fostering pancreatic cancer cell growth and invasion. Front Physiol. 2012;3:270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 47]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
31.  Yadav D, Lowenfels AB. The epidemiology of pancreatitis and pancreatic cancer. Gastroenterology. 2013;144:1252-1261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1151]  [Cited by in F6Publishing: 1239]  [Article Influence: 112.6]  [Reference Citation Analysis (0)]
32.  Raimondi S, Lowenfels AB, Morselli-Labate AM, Maisonneuve P, Pezzilli R. Pancreatic cancer in chronic pancreatitis; aetiology, incidence, and early detection. Best Pract Res Clin Gastroenterol. 2010;24:349-358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 402]  [Cited by in F6Publishing: 408]  [Article Influence: 29.1]  [Reference Citation Analysis (0)]
33.  Liou GY, Döppler H, Necela B, Krishna M, Crawford HC, Raimondo M, Storz P. Macrophage-secreted cytokines drive pancreatic acinar-to-ductal metaplasia through NF-κB and MMPs. J Cell Biol. 2013;202:563-577.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 211]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
34.  Fukunaga A, Miyamoto M, Cho Y, Murakami S, Kawarada Y, Oshikiri T, Kato K, Kurokawa T, Suzuoki M, Nakakubo Y. CD8+ tumor-infiltrating lymphocytes together with CD4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma. Pancreas. 2004;28:e26-e31.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Clark CE, Hingorani SR, Mick R, Combs C, Tuveson DA, Vonderheide RH. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res. 2007;67:9518-9527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 653]  [Cited by in F6Publishing: 717]  [Article Influence: 42.2]  [Reference Citation Analysis (0)]
36.  Vonderheide RH, Bayne LJ. Inflammatory networks and immune surveillance of pancreatic carcinoma. Curr Opin Immunol. 2013;25:200-205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 122]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
37.  Komura T, Sakai Y, Harada K, Kawaguchi K, Takabatake H, Kitagawa H, Wada T, Honda M, Ohta T, Nakanuma Y. Inflammatory features of pancreatic cancer highlighted by monocytes/macrophages and CD4+ T cells with clinical impact. Cancer Sci. 2015;106:672-686.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 52]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
38.  Lawrence T, Natoli G. Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nat Rev Immunol. 2011;11:750-761.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1363]  [Cited by in F6Publishing: 1560]  [Article Influence: 120.0]  [Reference Citation Analysis (0)]
39.  Lewis CE, Pollard JW. Distinct role of macrophages in different tumor microenvironments. Cancer Res. 2006;66:605-612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1523]  [Cited by in F6Publishing: 1608]  [Article Influence: 89.3]  [Reference Citation Analysis (1)]
40.  Mantovani A, Ming WJ, Balotta C, Abdeljalil B, Bottazzi B. Origin and regulation of tumor-associated macrophages: the role of tumor-derived chemotactic factor. Biochim Biophys Acta. 1986;865:59-67.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Puolakkainen PA, Brekken RA, Muneer S, Sage EH. Enhanced growth of pancreatic tumors in SPARC-null mice is associated with decreased deposition of extracellular matrix and reduced tumor cell apoptosis. Mol Cancer Res. 2004;2:215-224.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Coffelt SB, Scandurro AB. Tumors sound the alarmin(s). Cancer Res. 2008;68:6482-6485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 71]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
43.  Coffelt SB, Hughes R, Lewis CE. Tumor-associated macrophages: effectors of angiogenesis and tumor progression. Biochim Biophys Acta. 2009;1796:11-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 174]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
44.  Ong SM, Tan YC, Beretta O, Jiang D, Yeap WH, Tai JJ, Wong WC, Yang H, Schwarz H, Lim KH. Macrophages in human colorectal cancer are pro-inflammatory and prime T cells towards an anti-tumour type-1 inflammatory response. Eur J Immunol. 2012;42:89-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 102]  [Article Influence: 7.8]  [Reference Citation Analysis (1)]
45.  Balkwill F, Charles KA, Mantovani A. Smoldering and polarized inflammation in the initiation and promotion of malignant disease. Cancer Cell. 2005;7:211-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1284]  [Cited by in F6Publishing: 1278]  [Article Influence: 67.3]  [Reference Citation Analysis (0)]
46.  Tsutsui S, Yasuda K, Suzuki K, Tahara K, Higashi H, Era S. Macrophage infiltration and its prognostic implications in breast cancer: the relationship with VEGF expression and microvessel density. Oncol Rep. 2005;14:425-431.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  An T, Sood U, Pietruk T, Cummings G, Hashimoto K, Crissman JD. In situ quantitation of inflammatory mononuclear cells in ductal infiltrating breast carcinoma. Relation to prognostic parameters. Am J Pathol. 1987;128:52-60.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Bingle L, Brown NJ, Lewis CE. The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J Pathol. 2002;196:254-265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1459]  [Cited by in F6Publishing: 1434]  [Article Influence: 65.2]  [Reference Citation Analysis (0)]
49.  Balkwill F. Cancer and the chemokine network. Nat Rev Cancer. 2004;4:540-550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1747]  [Cited by in F6Publishing: 1749]  [Article Influence: 87.5]  [Reference Citation Analysis (0)]
50.  Nardin A, Abastado JP. Macrophages and cancer. Front Biosci. 2008;13:3494-3505.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Lesina M, Kurkowski MU, Ludes K, Rose-John S, Treiber M, Klöppel G, Yoshimura A, Reindl W, Sipos B, Akira S. Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer. Cancer Cell. 2011;19:456-469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 618]  [Cited by in F6Publishing: 650]  [Article Influence: 50.0]  [Reference Citation Analysis (0)]
52.  Yamasaki A, Kameda C, Xu R, Tanaka H, Tasaka T, Chikazawa N, Suzuki H, Morisaki T, Kubo M, Onishi H. Nuclear factor kappaB-activated monocytes contribute to pancreatic cancer progression through the production of Shh. Cancer Immunol Immunother. 2010;59:675-686.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 40]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
53.  Helm O, Held-Feindt J, Grage-Griebenow E, Reiling N, Ungefroren H, Vogel I, Krüger U, Becker T, Ebsen M, Röcken C. Tumor-associated macrophages exhibit pro- and anti-inflammatory properties by which they impact on pancreatic tumorigenesis. Int J Cancer. 2014;135:843-861.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 196]  [Article Influence: 19.6]  [Reference Citation Analysis (0)]
54.  Meng F, Li C, Li W, Gao Z, Guo K, Song S. Interaction between pancreatic cancer cells and tumor-associated macrophages promotes the invasion of pancreatic cancer cells and the differentiation and migration of macrophages. IUBMB Life. 2014;66:835-846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
55.  Partecke LI, Günther C, Hagemann S, Jacobi C, Merkel M, Sendler M, van Rooijen N, Käding A, Nguyen Trung D, Lorenz E. Induction of M2-macrophages by tumour cells and tumour growth promotion by M2-macrophages: a quid pro quo in pancreatic cancer. Pancreatology. 2013;13:508-516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
56.  Mantovani A, Sica A. Macrophages, innate immunity and cancer: balance, tolerance, and diversity. Curr Opin Immunol. 2010;22:231-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1025]  [Cited by in F6Publishing: 1077]  [Article Influence: 76.9]  [Reference Citation Analysis (0)]
57.  Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, Kagnoff MF, Karin M. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell. 2004;118:285-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1859]  [Cited by in F6Publishing: 1915]  [Article Influence: 95.8]  [Reference Citation Analysis (0)]
58.  Karin M, Greten FR. NF-kappaB: linking inflammation and immunity to cancer development and progression. Nat Rev Immunol. 2005;5:749-759.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2227]  [Cited by in F6Publishing: 2312]  [Article Influence: 121.7]  [Reference Citation Analysis (0)]
59.  Lin EY, Li JF, Gnatovskiy L, Deng Y, Zhu L, Grzesik DA, Qian H, Xue XN, Pollard JW. Macrophages regulate the angiogenic switch in a mouse model of breast cancer. Cancer Res. 2006;66:11238-11246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 765]  [Cited by in F6Publishing: 801]  [Article Influence: 44.5]  [Reference Citation Analysis (0)]
60.  Qian B, Deng Y, Im JH, Muschel RJ, Zou Y, Li J, Lang RA, Pollard JW. A distinct macrophage population mediates metastatic breast cancer cell extravasation, establishment and growth. PLoS One. 2009;4:e6562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 464]  [Cited by in F6Publishing: 509]  [Article Influence: 33.9]  [Reference Citation Analysis (0)]
61.  Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436-444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6912]  [Cited by in F6Publishing: 7811]  [Article Influence: 488.2]  [Reference Citation Analysis (0)]
62.  Yoshikawa K, Mitsunaga S, Kinoshita T, Konishi M, Takahashi S, Gotohda N, Kato Y, Aizawa M, Ochiai A. Impact of tumor-associated macrophages on invasive ductal carcinoma of the pancreas head. Cancer Sci. 2012;103:2012-2020.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 101]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
63.  Protti MP, De Monte L. Immune infiltrates as predictive markers of survival in pancreatic cancer patients. Front Physiol. 2013;4:210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 71]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
64.  Kurahara H, Shinchi H, Mataki Y, Maemura K, Noma H, Kubo F, Sakoda M, Ueno S, Natsugoe S, Takao S. Significance of M2-polarized tumor-associated macrophage in pancreatic cancer. J Surg Res. 2011;167:e211-e219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 380]  [Cited by in F6Publishing: 430]  [Article Influence: 28.7]  [Reference Citation Analysis (0)]
65.  Chen SJ, Zhang QB, Zeng LJ, Lian GD, Li JJ, Qian CC, Chen YZ, Chen YT, Huang KH. Distribution and clinical significance of tumour-associated macrophages in pancreatic ductal adenocarcinoma: a retrospective analysis in China. Curr Oncol. 2015;22:e11-e19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
66.  Ino Y, Yamazaki-Itoh R, Shimada K, Iwasaki M, Kosuge T, Kanai Y, Hiraoka N. Immune cell infiltration as an indicator of the immune microenvironment of pancreatic cancer. Br J Cancer. 2013;108:914-923.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 497]  [Cited by in F6Publishing: 625]  [Article Influence: 56.8]  [Reference Citation Analysis (0)]
67.  Tjomsland V, Niklasson L, Sandström P, Borch K, Druid H, Bratthäll C, Messmer D, Larsson M, Spångeus A. The desmoplastic stroma plays an essential role in the accumulation and modulation of infiltrated immune cells in pancreatic adenocarcinoma. Clin Dev Immunol. 2011;2011:212810.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 70]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
68.  Helm O, Mennrich R, Petrick D, Goebel L, Freitag-Wolf S, Röder C, Kalthoff H, Röcken C, Sipos B, Kabelitz D. Comparative characterization of stroma cells and ductal epithelium in chronic pancreatitis and pancreatic ductal adenocarcinoma. PLoS One. 2014;9:e94357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 56]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
69.  Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, Huhn RD, Song W, Li D, Sharp LL. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011;331:1612-1616.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1152]  [Cited by in F6Publishing: 1234]  [Article Influence: 94.9]  [Reference Citation Analysis (0)]
70.  Beatty GL, Torigian DA, Chiorean EG, Saboury B, Brothers A, Alavi A, Troxel AB, Sun W, Teitelbaum UR, Vonderheide RH. A phase I study of an agonist CD40 monoclonal antibody (CP-870,893) in combination with gemcitabine in patients with advanced pancreatic ductal adenocarcinoma. Clin Cancer Res. 2013;19:6286-6295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 306]  [Cited by in F6Publishing: 334]  [Article Influence: 30.4]  [Reference Citation Analysis (0)]
71.  Vonderheide RH, Bajor DL, Winograd R, Evans RA, Bayne LJ, Beatty GL. CD40 immunotherapy for pancreatic cancer. Cancer Immunol Immunother. 2013;62:949-954.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 69]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
72.  Klug F, Prakash H, Huber PE, Seibel T, Bender N, Halama N, Pfirschke C, Voss RH, Timke C, Umansky L. Low-dose irradiation programs macrophage differentiation to an iNOS⁺/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell. 2013;24:589-602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 661]  [Cited by in F6Publishing: 758]  [Article Influence: 68.9]  [Reference Citation Analysis (0)]
73.  Kurte M, López M, Aguirre A, Escobar A, Aguillón JC, Charo J, Larsen CG, Kiessling R, Salazar-Onfray F. A synthetic peptide homologous to functional domain of human IL-10 down-regulates expression of MHC class I and Transporter associated with Antigen Processing 1/2 in human melanoma cells. J Immunol. 2004;173:1731-1737.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Ben-Baruch A. Inflammation-associated immune suppression in cancer: the roles played by cytokines, chemokines and additional mediators. Semin Cancer Biol. 2006;16:38-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 247]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
75.  Bak SP, Alonso A, Turk MJ, Berwin B. Murine ovarian cancer vascular leukocytes require arginase-1 activity for T cell suppression. Mol Immunol. 2008;46:258-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 73]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
76.  Maeda H, Shiraishi A. TGF-beta contributes to the shift toward Th2-type responses through direct and IL-10-mediated pathways in tumor-bearing mice. J Immunol. 1996;156:73-78.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Zhang H, Liu C, Kong Y, Huang H, Wang C, Zhang H. TGFβ signaling in pancreatic ductal adenocarcinoma. Tumour Biol. 2015;36:1613-1618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
78.  Matsuda M, Salazar F, Petersson M, Masucci G, Hansson J, Pisa P, Zhang QJ, Masucci MG, Kiessling R. Interleukin 10 pretreatment protects target cells from tumor- and allo-specific cytotoxic T cells and downregulates HLA class I expression. J Exp Med. 1994;180:2371-2376.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Qin Z, Noffz G, Mohaupt M, Blankenstein T. Interleukin-10 prevents dendritic cell accumulation and vaccination with granulocyte-macrophage colony-stimulating factor gene-modified tumor cells. J Immunol. 1997;159:770-776.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Sica A, Saccani A, Bottazzi B, Polentarutti N, Vecchi A, van Damme J, Mantovani A. Autocrine production of IL-10 mediates defective IL-12 production and NF-kappa B activation in tumor-associated macrophages. J Immunol. 2000;164:762-767.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Lopez MV, Adris SK, Bravo AI, Chernajovsky Y, Podhajcer OL. IL-12 and IL-10 expression synergize to induce the immune-mediated eradication of established colon and mammary tumors and lung metastasis. J Immunol. 2005;175:5885-5894.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Miotto D, Lo Cascio N, Stendardo M, Querzoli P, Pedriali M, De Rosa E, Fabbri LM, Mapp CE, Boschetto P. CD8+ T cells expressing IL-10 are associated with a favourable prognosis in lung cancer. Lung Cancer. 2010;69:355-360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
83.  Mocellin S, Marincola FM, Young HA. Interleukin-10 and the immune response against cancer: a counterpoint. J Leukoc Biol. 2005;78:1043-1051.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 281]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
84.  Marigo I, Dolcetti L, Serafini P, Zanovello P, Bronte V. Tumor-induced tolerance and immune suppression by myeloid derived suppressor cells. Immunol Rev. 2008;222:162-179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 467]  [Cited by in F6Publishing: 489]  [Article Influence: 32.6]  [Reference Citation Analysis (0)]
85.  Rodriguez PC, Quiceno DG, Zabaleta J, Ortiz B, Zea AH, Piazuelo MB, Delgado A, Correa P, Brayer J, Sotomayor EM. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res. 2004;64:5839-5849.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 799]  [Cited by in F6Publishing: 882]  [Article Influence: 44.1]  [Reference Citation Analysis (0)]
86.  Kryczek I, Zou L, Rodriguez P, Zhu G, Wei S, Mottram P, Brumlik M, Cheng P, Curiel T, Myers L. B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma. J Exp Med. 2006;203:871-881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 563]  [Cited by in F6Publishing: 537]  [Article Influence: 29.8]  [Reference Citation Analysis (0)]
87.  Kuang DM, Zhao Q, Peng C, Xu J, Zhang JP, Wu C, Zheng L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J Exp Med. 2009;206:1327-1337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 591]  [Cited by in F6Publishing: 679]  [Article Influence: 45.3]  [Reference Citation Analysis (0)]
88.  Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, Belaygorod L, Carpenter D, Collins L, Piwnica-Worms D. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 2013;73:1128-1141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 639]  [Cited by in F6Publishing: 721]  [Article Influence: 60.1]  [Reference Citation Analysis (0)]
89.  Di Caro G, Cortese N, Castino GF, Grizzi F, Gavazzi F, Ridolfi C, Capretti G, Mineri R, Todoric J, Zerbi A. Dual prognostic significance of tumour-associated macrophages in human pancreatic adenocarcinoma treated or untreated with chemotherapy. Gut. 2015;Jul 8; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 179]  [Article Influence: 22.4]  [Reference Citation Analysis (0)]
90.  Gabrilovich DI, Velders MP, Sotomayor EM, Kast WM. Mechanism of immune dysfunction in cancer mediated by immature Gr-1+ myeloid cells. J Immunol. 2001;166:5398-5406.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9:162-174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5121]  [Cited by in F6Publishing: 5020]  [Article Influence: 334.7]  [Reference Citation Analysis (0)]
92.  Bronte V, Serafini P, De Santo C, Marigo I, Tosello V, Mazzoni A, Segal DM, Staib C, Lowel M, Sutter G. IL-4-induced arginase 1 suppresses alloreactive T cells in tumor-bearing mice. J Immunol. 2003;170:270-278.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Narita Y, Wakita D, Ohkur T, Chamoto K, Nishimura T. Potential differentiation of tumor bearing mouse CD11b+Gr-1+ immature myeloid cells into both suppressor macrophages and immunostimulatory dendritic cells. Biomed Res. 2009;30:7-15.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Bayne LJ, Beatty GL, Jhala N, Clark CE, Rhim AD, Stanger BZ, Vonderheide RH. Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell. 2012;21:822-835.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 686]  [Cited by in F6Publishing: 706]  [Article Influence: 58.8]  [Reference Citation Analysis (0)]
95.  Takeuchi S, Baghdadi M, Tsuchikawa T, Wada H, Nakamura T, Abe H, Nakanishi S, Usui Y, Higuchi K, Takahashi M. Chemotherapy-Derived Inflammatory Responses Accelerate the Formation of Immunosuppressive Myeloid Cells in the Tissue Microenvironment of Human Pancreatic Cancer. Cancer Res. 2015;75:2629-2640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 111]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
96.  Pylayeva-Gupta Y, Lee KE, Hajdu CH, Miller G, Bar-Sagi D. Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell. 2012;21:836-847.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 497]  [Cited by in F6Publishing: 509]  [Article Influence: 42.4]  [Reference Citation Analysis (0)]
97.  Zhu Y, Knolhoff BL, Meyer MA, Nywening TM, West BL, Luo J, Wang-Gillam A, Goedegebuure SP, Linehan DC, DeNardo DG. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014;74:5057-5069.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 971]  [Cited by in F6Publishing: 904]  [Article Influence: 90.4]  [Reference Citation Analysis (0)]
98.  Hao NB, Lü MH, Fan YH, Cao YL, Zhang ZR, Yang SM. Macrophages in tumor microenvironments and the progression of tumors. Clin Dev Immunol. 2012;2012:948098.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 499]  [Cited by in F6Publishing: 638]  [Article Influence: 53.2]  [Reference Citation Analysis (0)]
99.  Lin EY, Li JF, Bricard G, Wang W, Deng Y, Sellers R, Porcelli SA, Pollard JW. Vascular endothelial growth factor restores delayed tumor progression in tumors depleted of macrophages. Mol Oncol. 2007;1:288-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 133]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
100.  Stockmann C, Doedens A, Weidemann A, Zhang N, Takeda N, Greenberg JI, Cheresh DA, Johnson RS. Deletion of vascular endothelial growth factor in myeloid cells accelerates tumorigenesis. Nature. 2008;456:814-818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 349]  [Cited by in F6Publishing: 352]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
101.  Feig C, Gopinathan A, Neesse A, Chan DS, Cook N, Tuveson DA. The pancreas cancer microenvironment. Clin Cancer Res. 2012;18:4266-4276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 841]  [Cited by in F6Publishing: 951]  [Article Influence: 86.5]  [Reference Citation Analysis (0)]
102.  Mielgo A, Schmid MC. Impact of tumour associated macrophages in pancreatic cancer. BMB Rep. 2013;46:131-138.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Olson P, Chu GC, Perry SR, Nolan-Stevaux O, Hanahan D. Imaging guided trials of the angiogenesis inhibitor sunitinib in mouse models predict efficacy in pancreatic neuroendocrine but not ductal carcinoma. Proc Natl Acad Sci USA. 2011;108:E1275-E1284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 59]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
104.  Vincent A, Herman J, Schulick R, Hruban RH, Goggins M. Pancreatic cancer. Lancet. 2011;378:607-620.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1720]  [Cited by in F6Publishing: 1922]  [Article Influence: 147.8]  [Reference Citation Analysis (3)]
105.  Schmid MC, Avraamides CJ, Foubert P, Shaked Y, Kang SW, Kerbel RS, Varner JA. Combined blockade of integrin-α4β1 plus cytokines SDF-1α or IL-1β potently inhibits tumor inflammation and growth. Cancer Res. 2011;71:6965-6975.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 89]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
106.  Schmid MC, Avraamides CJ, Dippold HC, Franco I, Foubert P, Ellies LG, Acevedo LM, Manglicmot JR, Song X, Wrasidlo W. Receptor tyrosine kinases and TLR/IL1Rs unexpectedly activate myeloid cell PI3kγ, a single convergent point promoting tumor inflammation and progression. Cancer Cell. 2011;19:715-727.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 301]  [Cited by in F6Publishing: 313]  [Article Influence: 24.1]  [Reference Citation Analysis (0)]
107.  Condeelis J, Pollard JW. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell. 2006;124:263-266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1921]  [Cited by in F6Publishing: 1980]  [Article Influence: 110.0]  [Reference Citation Analysis (0)]
108.  Baran B, Bechyne I, Siedlar M, Szpak K, Mytar B, Sroka J, Laczna E, Madeja Z, Zembala M, Czyz J. Blood monocytes stimulate migration of human pancreatic carcinoma cells in vitro: the role of tumour necrosis factor - alpha. Eur J Cell Biol. 2009;88:743-752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 25]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
109.  Karnevi E, Andersson R, Rosendahl AH. Tumour-educated macrophages display a mixed polarisation and enhance pancreatic cancer cell invasion. Immunol Cell Biol. 2014;92:543-552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 67]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
110.  Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009;9:239-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2816]  [Cited by in F6Publishing: 2660]  [Article Influence: 177.3]  [Reference Citation Analysis (0)]
111.  Yang M, Chen J, Su F, Yu B, Su F, Lin L, Liu Y, Huang JD, Song E. Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells. Mol Cancer. 2011;10:117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 491]  [Cited by in F6Publishing: 531]  [Article Influence: 40.8]  [Reference Citation Analysis (0)]
112.  Kimsey TF, Campbell AS, Albo D, Wilson M, Wang TN. Co-localization of macrophage inflammatory protein-3alpha (Mip-3alpha) and its receptor, CCR6, promotes pancreatic cancer cell invasion. Cancer J. 2004;10:374-380.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Campbell AS, Albo D, Kimsey TF, White SL, Wang TN. Macrophage inflammatory protein-3alpha promotes pancreatic cancer cell invasion. J Surg Res. 2005;123:96-101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 55]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
114.  Stokes JB, Adair SJ, Slack-Davis JK, Walters DM, Tilghman RW, Hershey ED, Lowrey B, Thomas KS, Bouton AH, Hwang RF. Inhibition of focal adhesion kinase by PF-562,271 inhibits the growth and metastasis of pancreatic cancer concomitant with altering the tumor microenvironment. Mol Cancer Ther. 2011;10:2135-2145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 174]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
115.  Bjerkvig R, Johansson M, Miletic H, Niclou SP. Cancer stem cells and angiogenesis. Semin Cancer Biol. 2009;19:279-284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 39]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
116.  Tysnes BB, Bjerkvig R. Cancer initiation and progression: involvement of stem cells and the microenvironment. Biochim Biophys Acta. 2007;1775:283-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 53]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
117.  Pallini R, Ricci-Vitiani L, Banna GL, Signore M, Lombardi D, Todaro M, Stassi G, Martini M, Maira G, Larocca LM. Cancer stem cell analysis and clinical outcome in patients with glioblastoma multiforme. Clin Cancer Res. 2008;14:8205-8212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 266]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
118.  Zeppernick F, Ahmadi R, Campos B, Dictus C, Helmke BM, Becker N, Lichter P, Unterberg A, Radlwimmer B, Herold-Mende CC. Stem cell marker CD133 affects clinical outcome in glioma patients. Clin Cancer Res. 2008;14:123-129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 435]  [Cited by in F6Publishing: 461]  [Article Influence: 28.8]  [Reference Citation Analysis (0)]
119.  Wu A, Wei J, Kong LY, Wang Y, Priebe W, Qiao W, Sawaya R, Heimberger AB. Glioma cancer stem cells induce immunosuppressive macrophages/microglia. Neuro Oncol. 2010;12:1113-1125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 472]  [Cited by in F6Publishing: 472]  [Article Influence: 33.7]  [Reference Citation Analysis (0)]
120.  Hou YC, Chao YJ, Tung HL, Wang HC, Shan YS. Coexpression of CD44-positive/CD133-positive cancer stem cells and CD204-positive tumor-associated macrophages is a predictor of survival in pancreatic ductal adenocarcinoma. Cancer. 2014;120:2766-2777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 80]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]