Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1888 word(s) 1888 2021-08-10 11:35:47 |
2 h + 265 word(s) 2153 2021-08-18 05:38:58 | |
3 h + 265 word(s) 2153 2021-08-18 05:39:34 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Zhang, J. Tumor-Associated Macrophages (TAMs). Encyclopedia. Available online: https://encyclopedia.pub/entry/13273 (accessed on 20 April 2024).
Zhang J. Tumor-Associated Macrophages (TAMs). Encyclopedia. Available at: https://encyclopedia.pub/entry/13273. Accessed April 20, 2024.
Zhang, Jinhua. "Tumor-Associated Macrophages (TAMs)" Encyclopedia, https://encyclopedia.pub/entry/13273 (accessed April 20, 2024).
Zhang, J. (2021, August 17). Tumor-Associated Macrophages (TAMs). In Encyclopedia. https://encyclopedia.pub/entry/13273
Zhang, Jinhua. "Tumor-Associated Macrophages (TAMs)." Encyclopedia. Web. 17 August, 2021.
Tumor-Associated Macrophages (TAMs)
Edit

Tumor-associated macrophages (TAMs) are a major component of the immune cells of the TME. They play a prominent role by secreting cytokines and chemokines and coordinating with inflammatory mechanisms to promote tumor development, invasion, metastasis, immunosuppression, angiogenesis, and drug tolerance. Different subtypes of TAMs have different functions, which can be dynamically changed in response to various signals from cancer cells or the TME.

colorectal cancer tumor microenvironment (TME) macrophage polarization therapy prognosis

1. Introduction

Colorectal cancer, whose abnormal cells grow in the colon or the rectum of the large intestine, is the third most common malignant tumor worldwide. The global incidence of colorectal cancer (CRC) has increased in recent years. According to the global cancer statistics for 2020 released by the International Agency for Research on Cancer (IARC), there were approximately 1.9 million new cases of CRC and 940,000 cancer deaths worldwide in 2020; its global mortality rate ranks second [1]. Some Asian countries, such as Japan and Malaysia, also have high CRC incidence. In China, the incidence of CRC has risen to become the fourth most common malignancy with the fifth highest mortality rate [2], making CRC a major public health issue.

The main risk factors of CRC are increasing age and genetic, lifestyle, and environmental factors. Genetic and epigenetic alterations inside the cell—such as the activation of oncogenes and proliferative signals from the abnormal microenvironment surrounding the cell—play the intrinsic role, while lifestyle or environmental factors such as obesity, inadequate exercise, tobacco or alcohol use, and processed meat consumption constitute the exogenous causes of CRC, interacting synergistically with the endogenous factors to promote CRC occurrence and development [3][4].

Over the past two decades, increasing evidence has shown that the tumor microenvironment (TME) plays an equally significant role in tumor initiation, progression, and metastasis as the genetic and epigenetic changes in cancer cells. The components of the tumor microenvironment include all of the nonmalignant stroma cells inside the tumor other than the tumor cells, including fibroblasts, endothelial cells, immune cells, and platelets [5][6]. Paget proposed the “seed and soil” theory in 1889 and conducted an in-depth analysis of the molecular characteristics of “seeds” (cancer cells) [7][8][9][10][11]. By studying the “soil” formed by cancer cells and host immune cells, scientists found that immune cells usually bind to cancer cells and obtain specific biological phenotypes via interactions with them. As a result, the TME is a unique environment which develops alongside tumor progression. It is now widely recognized that neglection of the complex changes in tumor microenvironment during tumor development is one important reason for the failure of current targeted therapies against tumor cells. Therefore, new therapeutic strategies targeting the component cells of the TME can be combined with traditional treatments to benefit CRC patients in individual medicine.

Tumor-associated macrophages (TAMs) are a major component of the immune cells of the TME. They play a prominent role by secreting cytokines and chemokines and coordinating with inflammatory mechanisms to promote tumor development, invasion, metastasis, immunosuppression, angiogenesis, and drug tolerance [12][13][14][15][16][17][18][19]. Different subtypes of TAMs have different functions, which can be dynamically changed in response to various signals from cancer cells or the TME. Studies have shown that TAMs are associated with poor prognosis in most solid tumors; however, their role is slightly more complicated in CRC, in which reeducating the polarization of TAMs may facilitate tumor immunotherapy [17][20][21][22][23][24]. The current review focuses on the phenotypic polarization of TAMs in CRC, the underlying functional mechanisms, and how these mechanisms can be used as potential targets for the treatment and prognosis of CRC.

2. The Functional Mechanism of TAMs in CRC

In the TME, TAMs and tumor cells promote tumor cell proliferation through the secretion of cytokines. In vitro studies confirmed that colon cancer cells upregulated the expression of RGC-32 in macrophages by secreting TGF-β1, and RGC-32 promoted the migration of macrophages and further accelerated the proliferation of colon cancer cells [25]. Yu, X. et al. reported that overexpression of C-X-C-motif receptor 4 (CXCR4) in the intestinal epithelial mucosa can promote epithelial–mesenchymal transition (EMT) and macrophage infiltration in colonic tissue, leading to colitis-associated tumorigenesis and progression [26]. Tacconi, C. et al. found that TAMs expressed VEGFR3, inhibited antitumor immunity, and promoted primary colorectal cancer growth through the VEGFC/VEGFR3 axis [27]. Furthermore, by coculturing TAMs and CT26 colon cancer cells in vitro, it was found that the oxidative stress regulated by TAMs can affect the proliferation of colon cancer cells. TAMs can maintain the level of reactive oxygen species (ROS) by regulating the activity of NADPH oxidase, thereby maintaining the redox state of the TME and promoting tumor cell proliferation [28].

The TME is mainly composed of stromal cells and immune cells such as macrophages, T lymphocytes, natural killer (NK) cells, dendritic cells (DC), neutrophils, and myeloid-derived suppressor cells (MDSCs) [5][29][30]. As the main component of the TME, the initial mechanism of TAMs is to recruit and activate T cells and NK cells by presenting tumor antigens, producing chemokines and cytokines, inhibiting the immune microenvironment of colon cancer, and exerting an immunosuppressive effect [31]. The release of chemokines mediated by TAMs, such as CCL2, CCL3, CCL4, CCL5, and CCL20, further contributes to Treg cell recruitment into the TME, and TAMs suppress the antitumor effects of T cells and NK cells [12][32][33].

TAMs highly express the ligands PD-1 and CTLA-4 (along with PDL1, B7-H1, and other ligands); suppress the cytotoxic function of T cells, NKT cells, and NK cells; and further reduce the body’s ability to kill colon cancer cells [34]. Peritoneal macrophages secrete IL-17, which enhances G-MDSCs accumulation, increases the proportion of Th17 cells, and ultimately promotes CAC development [35]. Furthermore, mutant p53 regulates macrophages through exosomes miR-1246 to increase the activity of TGF-β and promote the anti-inflammatory immunosuppression of macrophages [36].

Metabolism regulates the differentiation, mobilization, phenotypic polarization, and function of macrophages. The metabolic pathways are significantly different among heterogeneous macrophages. In cancer, the macrophage metabolism reprogramming induced by cytokines and other mediators from tumor cells and the TME involves changes in metabolism-related enzymes, metabolites, and metabolic pathways [37][38][39].

3. Potential Applications of TAMs in Therapy of CRC

Given the significance of TAMs in CRC—as discussed in the previous section: TAMs facilitate tumor proliferation, invasion, migration, and angiogenesis; suppress antitumor immunity; regulate metabolism; and interact with the microbiota—there has been growing interest in new strategies that target TAMs in CRC treatments. Although preclinical studies have obtained some promising evidence that supports a combination of these approaches with traditional methods such as chemotherapy or radiation therapy, new therapeutic approaches targeting TAMs should be carefully evaluated for efficacy and safety in clinical trials.

Blocking the infiltration of mononuclear cells in tumor-related inflammatory tissues is a promising strategy for the treatment of primary tumors. Chanmee, T. et al. confirmed that colon cancer TAMs induce an enhanced expression of the transcription factors HIF-1, CXCL-12, and CXCR4 in the hypoxic TME environment. Targeting the HIF-1/CXCR4 pathway blocks the accumulation of TAMs [12]. Furthermore, NT157 represents a new class of anticancer drugs that targets both the IGF-1 receptor (IGF-1R) and the STAT3 oncogenic signaling pathway, exerting an inhibitory effect on tumor cells. Studies have shown that NT157 inhibits the expression of tumorigenic cytokines, chemokines, and growth factors, such as IL-6, IL-11 and IL-23, CCL2, CCL5, CXCL7, CXCL5, intercellular adhesion molecule-1 (ICAM1), and TGF-β, thereby inhibiting TAMs in the TME [40]. Mantovani et al. found that TAMs derived from monocytes in colon cancer have the ability to differentiate. Thus, a combination therapy that blocks differentiation is urgently needed to effectively target these cells. TNF-γ can induce monocyte or macrophage recruitment to the TME of colon cancer and inhibit their differentiation into TAMs in vivo [41].

The plasticity of macrophages allows researchers to re-educate TAMs. Since TAMs mainly exhibit the M2 phenotype and promote angiogenesis and immunosuppression [42][43], TAMs can be re-educated by inducing polarization from the M2 to the M1 phenotype. For instance, Georgoudaki, A.M. et al. investigated the effect of immune checkpoint therapy by inhibiting the expression of macrophage receptor with collagenous structure (MARCO) by TAMs, which repolarized TAMs to the M1 type in a mouse MC38 colon cancer model and induced antitumor activity [44]. As a small-molecule immunotherapy, tasquinimod reduces the immunosuppressive potential of the TME by altering the number and frequency of tumor-infiltrating myeloid cells [45]. Olsson, A. et al. found that tasquinimod targets early-stage tumor-infiltrating myeloid cells and induces phenotype switching from the proangiogenic and immunosuppressive M2-like phenotype to the proinflammatory M1-like phenotype, which alters the TME to promote immunomodulation, prevent angiogenesis, and inhibit metastasis [46].

As evolutionarily conserved tumor suppressors, T2 RNases can inhibit tumor growth in vivo by balancing the M1/M2 macrophage ratio in tumors and recruiting adaptive antitumor CD8 + T cells [47]. Furthermore, Halama, N. and his colleagues also confirmed that inhibiting CCR5 can repolarize the phenotype of TAMs from M2 to M1 by regulating the STAT3/SOCS3 signaling pathway in TAMs, thereby exerting antitumor effects in a phase I clinical trial of patients with CRC liver metastases [48].

4. TAMs and Prognosis in CRC

The role of TAMs seems to be complicated in regard to colon cancer progression, as they are reported to perform both tumor-suppressive and tumor-promoting activities [49]. Some studies have shown that TAMs are associated with better CRC patient prognosis, while others have associated TAMs with poor prognosis. A summary of the related literature is shown in Table 1.

Table 1. Literature reports on the associations between TAMs and the prognosis of CRC patients.

Study Result Expression in TAMs Sample Size (Case) Reference
Benign prognosis      
High-density CD68+ TAM subtypes in CRC tissues were significantly associated with good 5-year overall survival (OS) rates High-density CD68+ 6115 [50]
The ratio of CD68+ macrophages to colon cancer cells is associated with improved survival in colon cancer patients High CD68+/colon cancer cells ratio 205 [51]
Adjuvant chemotherapy significantly improved recurrence-free survival (RFS) and OS for patients with high CD206+/CD68+ ratio of TAMs High CD206+/CD68+ ratio 835 [52]
Both CD68+- and VEGF-expressing TAMs were predictive of improved survival rates in stage II and stage III colon cancer patients CD68+ and VEGF 131 [53]
High infiltration of M1 macrophages is correlated with better prognosis in CRC in a stage-dependent manner High M1+ 485 [54]
Poor prognosis      
Infiltration of TAMs CD68+/iNOS in the tumor stroma is a negative prognostic factor CD68+/iNOS 89 [55]
An increased ratio of CD163+/CD68+ in the tumor invasive front (TF) was positively correlated with shorter CRC RFS and OS times High CD163+/CD68+ 81 [56]
An increase in the proportion of M2/M1 type TAMs was positively correlated with an increase in liver metastases in patients with colorectal cancer High M2/M1 ratio 120 [57]
A decrease in the number of infiltrating CD68+ TAMs in the tumor stroma was associated with longer RFS and OS times in advanced CRC patients receiving bevacizumab combined with chemotherapy High CD68+ 123 [58]
The combination of FSP-1+ CAFs and CD163+ M2 TAMs was associated with poor survival rates more significantly than when these markers were studied alone FSP-1+ CAFs and CD163+ M2 TAMs 289 [59]

Some studies have shown that CD68 + TAMs are mostly distributed in CRC tumor stroma, mainly along the front edge of the invasion, and CD68 + TAMs infiltrated into this site can improve the prognosis of CRC patients [50][51][60][61]. Feng, Q. et al. recruited two independent cohorts of consecutively enrolled patients at one medical center with pathological stage II colon cancer after radical resection. In both cohorts, adjuvant chemotherapy significantly prolonged the recurrence-free survival (RFS) and overall survival (OS) rate of patients with a high CD206/CD68 ratio. This suggests that the CD206/CD68 ratio is probably a better biomarker for prognosis and prediction of stage II colon cancer after adjuvant chemotherapy [52]. However, TAM infiltration alone was not highly significant in prognostic analysis, while the presence of both CD68- and VEGF-expressing TAMs was predictive of better survival rates in stage II and stage III colon cancer patients [53]. In addition, Najbauer, J. et al. found that high M1 macrophage infiltration is correlated with a better prognostic situation in CRC patients in a stage-dependent manner [54].

Nevertheless, different types and locations of TAMs have different prognostic significance for CRC patients. Infiltration of CD68 + TAMs and M2 TAMs is associated with poor CRC prognosis [62][63][64]. Infiltration of CD68 + /iNOS − TAMs in the tumor stroma is a negative prognostic factor [55]. An increased CD163 + /CD68 + ratio in the tumor invasive front (TF) was positively correlated with shorter RFS and OS rates in CRC [56]. In addition, an increase in the proportion of M2/M1 type TAMs was positively correlated with liver metastases in patients with colorectal cancer [57]. In a retrospective study of 123 patients with advanced CRC who were treated with bevacizumab combined with chemotherapy, the RFS and OS rates of CRC patients with low tumor interstitial CD68 + TAMs were significantly higher. This suggests that an increase in the number of CD68 + TAMs infiltrating the tumor stroma may reduce the efficacy of bevacizumab combined with chemotherapy in patients with advanced CRC [58]. In addition, Herrera, M. et al. demonstrated that the combination of FSP-1 + CAFs (cancer-associated fibroblasts) and CD163 + M2 TAMs was associated with poor survival rates more significantly than when these markers were studied alone [59].

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249.
  2. Wei, W.; Zeng, H.; Zheng, R.; Zhang, S.; An, L.; Chen, R.; Wang, S.; Sun, K.; Matsuda, T.; Bray, F.; et al. Cancer registration in China and its role in cancer prevention and control. Lancet Oncol. 2020, 21, e342–e349.
  3. David, A.R.; Zimmerman, M.R. Cancer: An old disease, a new disease or something in between? Nat. Rev. Cancer 2010, 10, 728–733.
  4. Thanikachalam, K.; Khan, G. Colorectal Cancer and Nutrition. Nutrients 2019, 11, 164.
  5. Huang, H.W.; Chang, C.C.; Wang, C.S.; Lin, K.H. Association between Inflammation and Function of Cell Adhesion Molecules Influence on Gastrointestinal Cancer Development. Cells 2021, 10, 67.
  6. Kim, J.; Bae, J.S. Tumor-Associated Macrophages and Neutrophils in Tumor Microenvironment. Mediat. Inflamm 2016, 2016, 6058147.
  7. Mikula-Pietrasik, J.; Uruski, P.; Tykarski, A.; Ksiazek, K. The peritoneal “soil” for a cancerous “seed”: A comprehensive review of the pathogenesis of intraperitoneal cancer metastases. Cell Mol. Life Sci. 2018, 75, 509–525.
  8. Mogi, K.; Yoshihara, M.; Iyoshi, S.; Kitami, K.; Uno, K.; Tano, S.; Koya, Y.; Sugiyama, M.; Yamakita, Y.; Nawa, A.; et al. Ovarian Cancer-Associated Mesothelial Cells: Transdifferentiation to Minions of Cancer and Orchestrate Developing Peritoneal Dissemination. Cancers 2021, 13, 1352.
  9. Zhuyan, J.; Chen, M.; Zhu, T.; Bao, X.; Zhen, T.; Xing, K.; Wang, Q.; Zhu, S. Critical steps to tumor metastasis: Alterations of tumor microenvironment and extracellular matrix in the formation of pre-metastatic and metastatic niche. Cell Biosci. 2020, 10, 89.
  10. Sellner, F.; Thalhammer, S.; Klimpfinger, M. Tumour Evolution and Seed and Soil Mechanism in Pancreatic Metastases of Renal Cell Carcinoma. Cancers 2021, 13, 1342.
  11. Srinivasan, E.S.; Tan, A.C.; Anders, C.K.; Pendergast, A.M.; Sipkins, D.A.; Ashley, D.M.; Fecci, P.E.; Khasraw, M. Salting the Soil: Targeting the Microenvironment of Brain Metastases. Mol. Cancer Ther. 2021, 20, 455–466.
  12. Chanmee, T.; Ontong, P.; Konno, K.; Itano, N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers 2014, 6, 1670–1690.
  13. Ge, Z.; Ding, S. The Crosstalk Between Tumor-Associated Macrophages (TAMs) and Tumor Cells and the Corresponding Targeted Therapy. Front. Oncol. 2020, 10, 590941.
  14. Yang, Q.; Guo, N.; Zhou, Y.; Chen, J.; Wei, Q.; Han, M. The role of tumor-associated macrophages (TAMs) in tumor progression and relevant advance in targeted therapy. Acta Pharm. Sin. B 2020, 10, 2156–2170.
  15. Goswami, K.K.; Ghosh, T.; Ghosh, S.; Sarkar, M.; Bose, A.; Baral, R. Tumor promoting role of anti-tumor macrophages in tumor microenvironment. Cell Immunol. 2017, 316, 1–10.
  16. Malekghasemi, S.; Majidi, J.; Baghbanzadeh, A.; Abdolalizadeh, J.; Baradaran, B.; Aghebati-Maleki, L. Tumor-Associated Macrophages: Protumoral Macrophages in Inflammatory Tumor Microenvironment. Adv. Pharm. Bull. 2020, 10, 556–565.
  17. DeNardo, D.G.; Ruffell, B. Macrophages as regulators of tumour immunity and immunotherapy. Nat. Rev. Immunol. 2019, 19, 369–382.
  18. Ngambenjawong, C.; Gustafson, H.H.; Pun, S.H. Progress in tumor-associated macrophage (TAM)-targeted therapeutics. Adv. Drug Deliv. Rev. 2017, 114, 206–221.
  19. Van Overmeire, E.; Laoui, D.; Keirsse, J.; Van Ginderachter, J.A.; Sarukhan, A. Mechanisms driving macrophage diversity and specialization in distinct tumor microenvironments and parallelisms with other tissues. Front. Immunol. 2014, 5, 127.
  20. Zhu, Y.; Zhou, J.; Feng, Y.; Chen, L.; Zhang, L.; Yang, F.; Zha, H.; Wang, X.; Han, X.; Shu, C.; et al. Control of Intestinal Inflammation, Colitis-Associated Tumorigenesis, and Macrophage Polarization by Fibrinogen-Like Protein 2. Front. Immunol. 2018, 9, 87.
  21. Zhao, G.; Liu, L.; Peek, R.M., Jr.; Hao, X.; Polk, D.B.; Li, H.; Yan, F. Activation of Epidermal Growth Factor Receptor in Macrophages Mediates Feedback Inhibition of M2 Polarization and Gastrointestinal Tumor Cell Growth. J. Biol. Chem. 2016, 291, 20462–20472.
  22. Yang, J.M.; Lee, H.S.; Seo, J.H.; Park, J.H.; Gelman, I.H.; Lo, E.H.; Kim, K.W. Structural environment built by AKAP12+ colon mesenchymal cells drives M2 macrophages during inflammation recovery. Sci. Rep. 2017, 7, 42723.
  23. Sui, H.; Tan, H.; Fu, J.; Song, Q.; Jia, R.; Han, L.; Lv, Y.; Zhang, H.; Zheng, D.; Dong, L.; et al. The active fraction of Garcinia yunnanensis suppresses the progression of colorectal carcinoma by interfering with tumorassociated macrophage-associated M2 macrophage polarization in vivo and in vitro. FASEB J. 2020, 34, 7387–7403.
  24. Lian, G.; Chen, S.; Ouyang, M.; Li, F.; Chen, L.; Yang, J. Colon Cancer Cell Secretes EGF to Promote M2 Polarization of TAM Through EGFR/PI3K/AKT/mTOR Pathway. Technol. Cancer Res. Treat. 2019, 18, 1533033819849068.
  25. Zhao, P.; Wang, B.; Zhang, Z.; Zhang, W.; Liu, Y. Response gene to complement 32 expression in macrophages augments paracrine stimulation-mediated colon cancer progression. Cell Death Dis. 2019, 10, 776.
  26. Yu, X.; Wang, D.; Wang, X.; Sun, S.; Zhang, Y.; Wang, S.; Miao, R.; Xu, X.; Qu, X. CXCL12/CXCR4 promotes inflammation-driven colorectal cancer progression through activation of RhoA signaling by sponging miR-133a-3p. J. Exp. Clin. Cancer Res. 2019, 38, 32.
  27. Tacconi, C.; Ungaro, F.; Correale, C.; Arena, V.; Massimino, L.; Detmar, M.; Spinelli, A.; Carvello, M.; Mazzone, M.; Oliveira, A.I.; et al. Activation of the VEGFC/VEGFR3 Pathway Induces Tumor Immune Escape in Colorectal Cancer. Cancer Res. 2019, 79, 4196–4210.
  28. Luput, L.; Licarete, E.; Sesarman, A.; Patras, L.; Alupei, M.C.; Banciu, M. Tumor-associated macrophages favor C26 murine colon carcinoma cell proliferation in an oxidative stress-dependent manner. Oncol. Rep. 2017, 37, 2472–2480.
  29. Nagai, N.; Kudo, Y.; Aki, D.; Nakagawa, H.; Taniguchi, K. Immunomodulation by Inflammation during Liver and Gastrointestinal Tumorigenesis and Aging. Int. J. Mol. Sci. 2021, 22, 2238.
  30. Zhongbo, Z.; Xiangdong, Z.; Shuo, Y. Yin-yang effect of tumour cells in breast cancer: From mechanism of crosstalk between tumour-associated macrophages and cancer-associated adipocytes. Am. J. Cancer Res. 2020, 10, 383–392.
  31. Chen, L.; Wang, S.; Wang, Y. IL-6 influences the polarization of macrophages and the formation and growth of colorectal tumor. Oncotarget 2018, 9, 17443–17454.
  32. Han, Q.; Shi, H.; Liu, F. CD163(+) M2-type tumor-associated macrophage support the suppression of tumor-infiltrating T cells in osteosarcoma. Int. Immunopharmacol. 2016, 34, 101–106.
  33. Liu, J.; Zhang, N.; Li, Q.; Zhang, W.; Ke, F.; Leng, Q.; Wang, H.; Chen, J.; Wang, H. Tumor-associated macrophages recruit CCR6+ regulatory T cells and promote the development of colorectal cancer via enhancing CCL20 production in mice. PLoS ONE 2011, 6, e19495.
  34. Kondo, A.; Yamashita, T.; Tamura, H.; Zhao, W.; Tsuji, T.; Shimizu, M.; Shinya, E.; Takahashi, H.; Tamada, K.; Chen, L.; et al. Interferon-gamma and tumor necrosis factor-alpha induce an immunoinhibitory molecule, B7-H1, via nuclear factor-kappaB activation in blasts in myelodysplastic syndromes. Blood 2010, 116, 1124–1131.
  35. Zhang, Y.; Wang, J.; Wang, W.; Tian, J.; Yin, K.; Tang, X.; Ma, J.; Xu, H.; Wang, S. IL-17A produced by peritoneal macrophages promote the accumulation and function of granulocytic myeloid-derived suppressor cells in the development of colitis-associated cancer. Tumour. Biol. 2016, 37, 15883–15891.
  36. Cooks, T.; Pateras, I.S.; Jenkins, L.M.; Patel, K.M.; Robles, A.I.; Morris, J.; Forshew, T.; Appella, E.; Gorgoulis, V.G.; Harris, C.C. Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR-1246. Nat. Commun. 2018, 9, 771.
  37. Netea-Maier, R.T.; Smit, J.W.A.; Netea, M.G. Metabolic changes in tumor cells and tumor-associated macrophages: A mutual relationship. Cancer Lett. 2018, 413, 102–109.
  38. Mehla, K.; Singh, P.K. Metabolic Regulation of Macrophage Polarization in Cancer. Trends Cancer 2019, 5, 822–834.
  39. Vitale, I.; Manic, G.; Coussens, L.M.; Kroemer, G.; Galluzzi, L. Macrophages and Metabolism in the Tumor Microenvironment. Cell Metab. 2019, 30, 36–50.
  40. Sanchez-Lopez, E.; Flashner-Abramson, E.; Shalapour, S.; Zhong, Z.; Taniguchi, K.; Levitzki, A.; Karin, M. Targeting colorectal cancer via its microenvironment by inhibiting IGF-1 receptor-insulin receptor substrate and STAT3 signaling. Oncogene 2016, 35, 2634–2644.
  41. Mantovani, A.; Sozzani, S.; Locati, M. Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2005, 23, 549–555.
  42. Noy, R.; Pollard, J.W. Tumor-associated macrophages: From mechanisms to therapy. Immunity 2014, 41, 49–61.
  43. Salmaninejad, A.; Valilou, S.F.; Soltani, A.; Ahmadi, S.; Abarghan, Y.J.; Rosengren, R.J.; Sahebkar, A. Tumor-associated macrophages: Role in cancer development and therapeutic implications. Cell. Oncol. 2019, 42, 591–608.
  44. Georgoudaki, A.M.; Prokopec, K.E.; Boura, V.F.; Hellqvist, E.; Sohn, S.; Ostling, J.; Dahan, R.; Harris, R.A.; Rantalainen, M.; Klevebring, D.; et al. Reprogramming Tumor-Associated Macrophages by Antibody Targeting Inhibits Cancer Progression and Metastasis. Cell Rep. 2016, 15, 2000–2011.
  45. Shen, L.; Sundstedt, A.; Ciesielski, M.; Miles, K.M.; Celander, M.; Adelaiye, R.; Orillion, A.; Ciamporcero, E.; Ramakrishnan, S.; Ellis, L.; et al. Tasquinimod modulates suppressive myeloid cells and enhances cancer immunotherapies in murine models. Cancer Immunol. Res. 2015, 3, 136–148.
  46. Olsson, A.; Nakhlé, J.; Sundstedt, A.; Plas, P.; Bauchet, A.-L.; Pierron, V.; Bruetschy, L.; Deronic, A.; Törngren, M.; Liberg, D.; et al. Tasquinimod triggers an early change in the polarization of tumor associated macrophages in the tumor microenvironment. J. ImmunoTherapy Cancer 2015, 3, 53.
  47. De Vito, A.; Orecchia, P.; Balza, E.; Reverberi, D.; Scaldaferri, D.; Taramelli, R.; Noonan, D.M.; Acquati, F.; Mortara, L. Overexpression of Murine Rnaset2 in a Colon Syngeneic Mouse Carcinoma Model Leads to Rebalance of Intra-Tumor M1/M2 Macrophage Ratio, Activation of T Cells, Delayed Tumor Growth, and Rejection. Cancers 2020, 12, 717.
  48. Halama, N.; Zoernig, I.; Berthel, A.; Kahlert, C.; Klupp, F.; Suarez-Carmona, M.; Suetterlin, T.; Brand, K.; Krauss, J.; Lasitschka, F.; et al. Tumoral Immune Cell Exploitation in Colorectal Cancer Metastases Can Be Targeted Effectively by Anti-CCR5 Therapy in Cancer Patients. Cancer Cell 2016, 29, 587–601.
  49. Nielsen, S.R.; Schmid, M.C. Macrophages as Key Drivers of Cancer Progression and Metastasis. Mediat. Inflamm 2017, 2017, 9624760.
  50. Li, J.; Li, L.; Li, Y.; Long, Y.; Zhao, Q.; Ouyang, Y.; Bao, W.; Gong, K. Tumor-associated macrophage infiltration and prognosis in colorectal cancer: Systematic review and meta-analysis. Int. J. Colorectal. Dis. 2020, 35, 1203–1210.
  51. Koelzer, V.H.; Canonica, K.; Dawson, H.; Sokol, L.; Karamitopoulou-Diamantis, E.; Lugli, A.; Zlobec, I. Phenotyping of tumor-associated macrophages in colorectal cancer: Impact on single cell invasion (tumor budding) and clinicopathological outcome. Oncoimmunology 2016, 5, e1106677.
  52. Feng, Q.; Chang, W.; Mao, Y.; He, G.; Zheng, P.; Tang, W.; Wei, Y.; Ren, L.; Zhu, D.; Ji, M.; et al. Tumor-associated Macrophages as Prognostic and Predictive Biomarkers for Postoperative Adjuvant Chemotherapy in Patients with Stage II Colon Cancer. Clin. Cancer Res. 2019, 25, 3896–3907.
  53. Khorana, A.A.; Ryan, C.K.; Cox, C.; Eberly, S.; Sahasrabudhe, D.M. Vascular endothelial growth factor, CD68, and epidermal growth factor receptor expression and survival in patients with Stage II and Stage III colon carcinoma: A role for the host response in prognosis. Cancer 2003, 97, 960–968.
  54. Edin, S.; Wikberg, M.L.; Dahlin, A.M.; Rutegard, J.; Oberg, A.; Oldenborg, P.A.; Palmqvist, R. The distribution of macrophages with a M1 or M2 phenotype in relation to prognosis and the molecular characteristics of colorectal cancer. PLoS ONE 2012, 7, e47045.
  55. Waniczek, D.; Lorenc, Z.; Snietura, M.; Wesecki, M.; Kopec, A.; Muc-Wierzgon, M. Tumor-Associated Macrophages and Regulatory T Cells Infiltration and the Clinical Outcome in Colorectal Cancer. Arch. Immunol. Ther. Exp. 2017, 65, 445–454.
  56. Yang, C.; Wei, C.; Wang, S.; Shi, D.; Zhang, C.; Lin, X.; Dou, R.; Xiong, B. Elevated CD163(+)/CD68(+) Ratio at Tumor Invasive Front is Closely Associated with Aggressive Phenotype and Poor Prognosis in Colorectal Cancer. Int. J. Biol. Sci. 2019, 15, 984–998.
  57. Cui, Y.L.; Li, H.K.; Zhou, H.Y.; Zhang, T.; Li, Q. Correlations of tumor-associated macrophage subtypes with liver metastases of colorectal cancer. Asian Pac. J. Cancer Prev. 2013, 14, 1003–1007.
  58. Dost Gunay, F.S.; Kirmizi, B.A.; Ensari, A.; Icli, F.; Akbulut, H. Tumor-associated Macrophages and Neuroendocrine Differentiation Decrease the Efficacy of Bevacizumab Plus Chemotherapy in Patients with Advanced Colorectal Cancer. Clin. Colorectal Cancer 2019, 18, e244–e250.
  59. Herrera, M.; Herrera, A.; Dominguez, G.; Silva, J.; Garcia, V.; Garcia, J.M.; Gomez, I.; Soldevilla, B.; Munoz, C.; Provencio, M.; et al. Cancer-associated fibroblast and M2 macrophage markers together predict outcome in colorectal cancer patients. Cancer Sci. 2013, 104, 437–444.
  60. Forssell, J.; Oberg, A.; Henriksson, M.L.; Stenling, R.; Jung, A.; Palmqvist, R. High macrophage infiltration along the tumor front correlates with improved survival in colon cancer. Clin. Cancer Res. 2007, 13, 1472–1479.
  61. Zhou, Q.; Peng, R.Q.; Wu, X.J.; Xia, Q.; Hou, J.H.; Ding, Y.; Zhou, Q.M.; Zhang, X.; Pang, Z.Z.; Wan, D.S.; et al. The density of macrophages in the invasive front is inversely correlated to liver metastasis in colon cancer. J. Transl. Med. 2010, 8, 13.
  62. Wei, C.; Yang, C.; Wang, S.; Shi, D.; Zhang, C.; Lin, X.; Liu, Q.; Dou, R.; Xiong, B. Crosstalk between cancer cells and tumor associated macrophages is required for mesenchymal circulating tumor cell-mediated colorectal cancer metastasis. Mol. Cancer 2019, 18, 64.
  63. Lan, J.; Sun, L.; Xu, F.; Liu, L.; Hu, F.; Song, D.; Hou, Z.; Wu, W.; Luo, X.; Wang, J.; et al. M2 Macrophage-Derived Exosomes Promote Cell Migration and Invasion in Colon Cancer. Cancer Res. 2019, 79, 146–158.
  64. Yin, Y.; Yao, S.; Hu, Y.; Feng, Y.; Li, M.; Bian, Z.; Zhang, J.; Qin, Y.; Qi, X.; Zhou, L.; et al. The Immune-microenvironment Confers Chemoresistance of Colorectal Cancer through Macrophage-Derived IL6. Clin. Cancer Res. 2017, 23, 7375–7387.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 511
Revisions: 3 times (View History)
Update Date: 18 Aug 2021
1000/1000