info@biomedres.us   +1 (502) 904-2126   One Westbrook Corporate Center, Suite 300, Westchester, IL 60154, USA   Site Map
ISSN: 2574 -1241

Impact Factor : 0.548

  Submit Manuscript

Review ArticleOpen Access

Cancer-Immune Microenvironment: A Review Volume 49- Issue 5

Girum Tefera Belachew*

  • Department of Biotechnology, College of Natural and Computational Sciences, Debre Birhan University, Ethiopia

Received: April 02, 2023;   Published: April 24, 2023

*Corresponding author: Girum Tefera Belachew, Department of Biotechnology, College of Natural and Computational Sciences, Debre Birhan University, P.O. Box 445, Debre Birhan, Ethiopia

DOI: 10.26717/BJSTR.2023.49.007879

Abstract PDF

ABSTRACT

Malignant growth cells, stromal tissue, and extracellular network make up the tumor microenvironment. The tumor microenvironment is significantly influenced by the immune system. Malignant tumors are not a collection of altered cells, but rather an additional organ made up of non-cancerous cells that make up a large portion of the tumor mass and have turned bad and lost the ability to maintain the communication necessary for maintaining the tissue’s homeostasis. The tumor microenvironment is made up of a variety of important components, such as tumor parenchyma cells, fibroblasts, mesenchymal cells, blood, and lymphatic arteries, as well as tumor-invading immune cells, chemokines, and cytokines. Another fundamental reason for linking the tumor genotype to the participating immune cells is the release of tumor-inferred chemokines, which are controlled by certain oncogenes. According to ongoing research using a BRAFV600E and Pten-deficient mouse model of melanoma, constitutive tumor-inborn WNT/- catenin signaling is associated with poor immunological penetration and insufficient anticancer T cells, largely because CD103+ DC recruitment and recurrence are decreased. Even before dispersed malignant tumor cells reach a secondary organ, immunological alterations brought on by the tumor have an impact on the development of metastatic infection. Systemic immune tolerance and alterations in the characteristics of surrounding myeloid cells can favorably influence a tumor’s capacity to develop a metastatic location. Currently, it is widely believed that the immunological circumstances in the tumor microenvironment play a fundamentally important role in the anticipation, development, and progression of tumors. Studies on a variety of malignant tumors have provided compelling evidence that the state of the tumor microenvironment is closely related to the course of the disease. The review’s objective is to provide an overview of the impact of host immunological factors on the development of the tumor microenvironment and subsequent illness.

Keywords: Cancer; Microenvironment; Prognosis; Therapy; Tumor

Abbreviations: TME: Tumor Microenvironment; VEGF: Vascular Endothelial Growth Factor; PDGF: Platelet-Derived Growth Factor; MMPs; Matrix Metalloproteinase; MDSC: Myeloid-Derived Suppressor Cells; TLRs: Toll-Like Receptors; BCG: Bacillus Calmette-Guérin

Introduction

Malignant growth cells, stromal tissue, and extracellular network make up the tumor microenvironment. The tumor microenvironment is significantly influenced by the immune system. Certainly, extensive research has been done in the preceding few decades on the puzzling relationship between malignant development cells and the host immune response. Several immune deficits have been linked to accelerated tumor growth in both animal and human models [1,2]. It has been extensively documented [3-5] that transplant patients who receive long-term immunosuppressive medication have a greater tumor incidence.

Moreover, mice with weakened immune systems because of genetic modifications get cancer more frequently [6-9]. It is currently widely accepted that strong immune system tumor surveillance is necessary to maintain the host’s homeostasis. The immune system’s cancer reconnaissance may ultimately fail, although playing a crucial role in host defense. Prior to becoming clinically noticeable, the immune system first eliminates cancer cells. After there comes an equilibrium phase, during which less immunogenic tumor changes are determined until the malignancy eventually «escapes» immune monitoring [10,11]. Yet, the persistent inflammation brought on by chronic conditions may also promote the emergence of new cancers [12].

Gastric, colorectal, hepatic, and cervical cancers are all closely linked to basic, ongoing provoking reactions [13,14]. This type of expression of several immunological gene products during ongoing inflammation appears to create an excellent milieu for the emergence and spread of cancer [10,14]. Interestingly, extensive genomes studies being conducted on cancer patients have revealed a link between the tumor microenvironment’s characteristics—specifically, the level of host tissue inflammation—and a better prognosis for the patient [15- 17]. An immunosuppressed microenvironment with a predominance of natural immune components routinely benefits the tumor. On the other hand, patients who maintain active, pro-inflammatory immune responses inside the tumor microenvironment have superior results [18,19]. The review’s objective is to provide an overview of the impact of host immunological factors on the development of the tumor microenvironment and subsequent illness.

Figure 1:

biomedres-openaccess-journal-bjstr

The Tumor Microenvironment

Malignant tumors are not a collection of altered cells but rather an additional organ made up of non-cancerous cells that make up a large portion of the tumor mass but have turned bad and lost the ability to maintain a communication that would have enabled tissue homeostasis [20]. These cells include immunological cells as well as fibroblasts, adipocytes, pericytes, vascular endothelial cells, and others [21]. Comparatively to what occurs during organogenesis during development, tumor and stromal cells co-proliferate, and communication between the various segments results in a constant phenotypic and practical adaptability. Via junctions, receptors, and a variety of indicators produced by the many cell types enclosed in a three- dimensional extracellular network, dynamic equal correspondence between cells and the surroundings is guided (ECM). This combines ECM-rebuilding enzymes with glycoproteins, proteoglycans, cytokines, and growth factors, providing both fundamental assistance and accurate information [22]. The ability of stromal and immune cells to digest cells and function alters dynamically when tissue homeostasis is disrupted [23]. The tumor microenvironment (TME) is made up of this intricate network (Figure 1) [24], and tumor research must make a substantial effort to create a multidimensional map that will make clear the highways and byways of the front line of malignant tumours.

Characteristics of the Tumor Microenvironment

The tumor microenvironment contains a variety of important components, such as tumor parenchyma cells, fibroblasts, mesenchymal cells, blood, and lymphatic arteries, as well as tumorinvading immune cells, chemokines, and cytokines [25]. These numerous and varied components meet the definition of a complex system, wherein the interdependence between the pieces is multilevel, multiscale, and nonlinear in nature [26]. Each of these components has the potential to significantly contribute to the development and spread of tumors. The construction and renovation of the extracellular network is the responsibility of these non-immune segments, and tumor associated fibroblasts are a major source of growth factors that promote the development of carcinoma cells [27]. While existing blood and lymphatic arteries may serve as routes for local assault and distant metastasis, the formation of new blood vessels is essential for tumor progression as the mass increases [28,29]. Many studies have demonstrated that the development of factors like vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), and matrix metalloproteinase (MMPs), which stimulate vein development, contribute to the spread of tumor cells and predict poor patient endurance [29].

Other host cell lineages, like mesenchymal stem cells, can separate into the many cell types required to fuel angiogenesis as the disease progresses in addition to producing new cancer cells [30]. Yet, due to their fundamental role in the development of tumors and the management of cancer, the immunological components of the tumor microenvironment have gained attention in recent years. Malignant tumor outcomes are mostly determined by immune cells that penetrate tumors, including myeloid-derived suppressor cells (MDSC), tumor-related macrophages, and cytotoxic lymphocytes. Several studies have shown that increased MDSC and tumorassociated macrophage concentrations promote tumor growth via a variety of suppressive mechanisms [31,32]. On the other hand, different aggressive tumor cells have a good prognosis when cytotoxic lymphocytes are present in the tumor microenvironment [33-35].Chemokines and cytokines are two additional immunological components of the tumor microenvironment that may alter the local balance of proregulatory and anticancer immune responses [36,37].Innate immunity components, such as the toll-like receptors (TLRs), can identify risk signals in the microenvironment, such as heat shock proteins, nucleic acids, and HMGB1 transformed, dying, or dead tumor cells, and these signals can trigger anticancer immune responses [38,39].

Figure 2:

biomedres-openaccess-journal-bjstr

Interconnectivity of Tumor Genotypes and Phenotypes and the Tumor Immune Microenvironment

It remains to be established how the composition of the tumor immune microenvironment is influenced by cytokines and chemokines released by cancer, cancer oncogenes, and mutation landscapes (TIME). Although there are many models that can demonstrate relationships between immunological configuration and cancer genotype/phenotype, these models are not sufficiently robust to allow this agreement to be immediately used toward therapeutic intervention [40,41] (Figure 2).

Tumor-Derived Chemokines

Another fundamental reason for linking the tumor genotype to the participating immune cells is the release of tumor-inferred chemokines, which are controlled by certain oncogenes. Current data from a BRAFV600E and Pten-deficient mouse model of melanoma suggest that constitutive tumor-inborn WNT/-catenin signaling is associated with poor immunological penetration and insufficient anticancer T cells, mostly because CD103+ DC recruitment and recurrence are reduced [42]. In vitro DC migration assays and transcriptional analysis of tumor cells have revealed that constitutive WNT/-catenin signaling causes reduced production of Ccl4, a potent chemo attractant for a variety of myeloid cells, including CD103+ DCs. This finding may help to explain why CD103+ DC recruitment is reduced and CD8+ T cell penetration into the tumor microenvironment is similarly poor. Several studies in mice have shown that tumor discharged CCL2 causes the enrollment of CCR2+ old-style monocytes in the tumor, where they split into TAMs, a protumoral myeloid population [43]. This is true even if the precise oncogenic determinant of expression is unclear.

The Immune Environment in Metastasis

Even before dispersed malignant tumor cells reach a secondary organ, immunological alterations brought on by the tumor have an impact on the development of metastatic infection. Systemic immune tolerance and alterations in the characteristics of surrounding myeloid cells can favorably influence a tumor’s capacity to develop a metastatic location. Certain combinations of immune populaces that have the ability to both promote and suppress metastasis development quickly gather tumor cells as they spread to distant tissue locations [43,44]. An astonishing amount of evidence supports the pro-metastatic ability of both macrophages and classically inflammatory monocytes [45-47]. Mice lacking Csj-1, which is required for the development of CSF-1-subordinate cells, as well as monocytes and macrophages, demonstrate delayed progression of mammary cancer to metastasis, according to a novel study using the MMTV-PyMT breast cancer mouse model [44]. According to recent research, macrophages and their ancestors populations observed in pre-metastatic tumor locations greatly advance metastasis in addition to TAMs in the primary malignancy [47,48]. According to studies conducted on mice, CD4+ T cell-derived IL-4 indirectly promotes breast malignant tumor spread by regulating macrophage phenotype, so demonstrating a role for both the innate and adaptive immune systems in stifling beneficial anticancer effects [45].

Unusual waves of myeloid cells absorb tumor material as leading metastatic tumor cells arrive and die, delivering antigen to both proand antitumor immune compartments, according to a recent study using multiphoton intravital imaging of the lung pre-metastatic location in mice [48]. In any event, the majority of the tumor material is overpowered by monocytes, which may sequester significant tumor antigen from stimulatory DC populations. Moreover, a decline in monocytes results in increased antigen loads in those DCs.Nonclassical or «watching» monocytes have been shown to exhibit antimetastatic capabilities, despite the fact that classical inflammatory monocytes have a proven capability for metastatic advancing [49]. Neutrophils play fundamental roles in tumor improvement, just like monocytes and macrophages. Neutrophil levels are elevated in the blood and accumulate in peripheral organs during tumor growth, contrary to what several preclinical mouse cancer models have demonstrated [50-54]. Neutrophils’ roles in metastasis, however, are still debatable. While some studies have focused on neutrophils’ anti-metastatic activity [54,55], others have found that they have prometastatic characteristics [50,52,56-59]. By direct cytotoxicity against spreading malignant tumor cells, tumor-entrapped neutrophils have been shown to prevent lung metastasis in the 4T1 mouse breast cancer model [54]. Furthermore, according to a recent study, a subpopulation of neutrophils that communicate with the MET protooncogene protects against the growth of metastasis [56]. On the other hand, it has been discovered that neutrophils, in the MMTV-PyMT breast cancer model, promote metastasis to the lung by increasing the number of metastasis-starting malignant tumor cells through the release of leukotrienes [50]. Neutrophils have also been discovered to promote metastasis by stymieing antitumor immunity in a model of lobular breast carcinoma [52]. Cancer-incited IL17-delivering T cells are responsible for the systemic growth and polarization of pro-metastatic neutrophils [52], demonstrating the close interaction between the innate and adaptive immune systems during metastasis. A growing body of evidence suggests that immunostimulatory myeloid cells can also enhance anticancer T cell responses, even though much of what is known about immunological organization at the metastatic location focuses on cells with immunosuppressive capabilities. Despite consuming the majority of tumor antigen, macrophages usually fail to successfully activate T cells in vitro [60], which is consistent with their recently reported pro-tumorigenic activity. In any case, CD103+ DCs are far superior T cell activators [60,61], and their absence leads to a crucial expansion in pneumonic metastasis [48], suggesting that even in the metastatic site, CD103+ DCs are important for inciting potent antitumor CD8+ T cell responses. This is despite the fact that their presence in cancer and metastatic sores is sparse. Collectively, these data support the idea that treating myeloid cells to reduce immunosuppression and stimulate T cell responses may be a useful immunotherapeutic approach to treating patients with metastatic malignant tumor cells.

Cancer Immunotherapy

The host immune system is being strengthened by medications. The holy grail of tumor immunotherapy remains the ability of the host immune system to recognize and eradicate cancer cells with minimal systemic damage [62]. William B. (Coley’s poison) pioneered the use of immunotherapy as the primary therapy for the management of lethal malignancies in 1891.In patients with delicate tissue sarcomas, Dr. Coley directly injected streptococcus bacteria into tumors, muscle tissue, or intravenously «to provoke erysipelas and the immune system» to attack the tumor [63]. Despite emerging clinical use of chemotherapy and radiation therapy, its use was eventually discontinued 40 years later [64] due to its extreme toxicity and lack of consistent results. Coley’s original theories regarding tumor immunotherapy, which still hold true today, suggest that activating immunity can undoubtedly result in tumor dismissal. In the 1970s, Morales et al. established the viability of the bacterium Bacillus Calmette-Guérin (BCG) in the treatment of superficial bladder cancer, which was the first of the advanced applications of Coley’s standard [65]. This clinical indication is supported by recent research by Old et al. that demonstrates the antitumor effects of BCG in a mouse model [66]. In addition to his work on BCG, Old also conducted extensive research and worked on the interpretation of tumor necrosis factor in 1975 [67]. However, the idea that the immune system might be crucial in the treatment of many malignant tumors remained outside the purview of conventional oncology [68]. The disclosure and depiction of dendritic cells by Ralph Steinman in 1973, the portrayal of MHC constraint in 1974 by Zinkernagel and Doherty’s, the documentation of NK cell activity in 1975 by Eva Klein’s, the concentrate in gigantic size of cytokines in breast malignant cells, renal cell cancer, glioblastoma, lymphoma, and melanoma during the 1980s, began the state-of-theart safe based cancer treatment in clinical medication [68].

Conclusion

Currently, it is widely believed that the immunological circumstances in the tumor microenvironment play a fundamentally important role in the anticipation, development, and progression of tumors. Studies on a variety of malignant tumors have provided compelling evidence that the state of the tumor microenvironment is closely related to the course of the disease. Whether a pro-tumor or anti-tumor immune response predominates in the microenvironment depends on the presence of immune cell types or chemicals. For malignant tumor immunotherapies to be successful in the future, it is now widely accepted that the immune response must be altered from one that elevates tumors to one that damages them. Controlling the immunological borders that define the tumor microenvironment may work to affect the balance of host responses leading to effective immunization. Improved knowledge of the functions of immune cells and chemicals in the tumor microenvironment will be crucial for the development of more effective novel treatments.

Data Availability

All of the required data will be available upon request to the corresponding author

Authors’ Contributions

The author wrote the review article alone.

Acknowledgment

The author is grateful to thank those individuals who gave help directly or indirectly.

Financial Support and Sponsorship

There is no financial support and sponsorship.

Conflicts of Interest

There are no conflicts of interest.

References

  1. Chow Melvyn T, Andreas Möller, Mark J Smyth (2012) Inflammation and immune surveillance in cancer. In Seminars in cancer biology 2(1): 23-32.
  2. Smyth Mark J, Gavin P Dunn, Robert D Schreiber (2006) Cancer immunosurveillance and immunoediting: the roles of immunity in suppressing tumor development and shaping tumor immunogenicity. Advances in immunology 90: 1-50.
  3. Euvrard, Sylvie, Jean Kanitakis, Alain Claudy (2003) Skin cancers after organ transplantation. New England Journal of Medicine 348(17): 1681-1691.
  4. EQ Sanchez, S Marubashi, G Jung, Marlon F Levy, Robert M Goldstein, et al. (2002) De novo tumors after liver transplantation: a single- institution experience. Liver Transplantation 8(3): 285-291.
  5. FO Zwald, LJ Christenson, EM Billingsley, N C Zeitouni, D Ratner, J Bordeaux, et al. (2010) Melanoma in solid organ transplant recipients. American Journal of Transplantation 10(5): 1297-1304.
  6. M Girardi, DE Oppenheim, CR Steele, JM Lewis, E Glusac, et al. (2001) Regulation of cutaneous malignancy by γδ T cells. Science 294(5542): 605-609.
  7. MJ Smyth, KY T Thia, S E A Street, E Cretney, J A Trapani, et al. (2000) Differential tumor surveillance by natural killer (NK) and NKT cells. Journal of Experimental Medicine 191(4): 661-668.
  8. SE A Street, E Cretney, M J Smyth (2001) Perforin and interferon-γ activities independently control tumor initiation, growth, and metastasis Blood 97(1):192-197.
  9. MF Van Den Broek, D K¨agi, F Ossendorp, R Toes, S Vamvakas, et al. (1996) Decreased tumor surveillance in perforin- deficient mice. Journal of Experimental Medicine 184(5): 1781-1790.
  10. GP Dunn, AT Bruce, H Ikeda, L J Old, R D Schreiber (2002) Cancer immunoediting: from immunosurveillance to tumor escape. Nature Immunology 3(11): 991-998.
  11. AM Engel, I M Svane, J Rygaard, O Werdelin (1997) MCA sarcomas induced in scid mice are more immunogenic than MCA sarcomas induced in congenic, immunocompetent mice. Scandinavian Journal of Immunology 45(5): 463-470.
  12. BB Aggarwal (2009) Inflammation, a silent killer in cancer is not so silent! Current Opinion in Pharmacology 9(4): 347-350.
  13. BB Aggarwal, S Shishodia, S K Sandur, M K Pandey, G Sethi (2006) Inflammation and cancer: how hot is the link? Biochemical Pharmacology 72(11): 1605-1621.
  14. F Balkwill, A Mantovani (2001) Inflammation and cancer: back to Virchow? The Lancet 357(9255): 539-545.
  15. V Chew, J Chen, D Lee, Evelyn Loh, Joyce Lee, et al. (2012) Chemokine-driven lymphocyte infiltration: an early intratumoural event determining long-term survival in resectable hepatocellular carcinoma. Gut 61(3): 427-438.
  16. D S Hsu, M K Kim, B S Balakumaran, Chaitanya R Acharya, Carey K Anders, et al. (2010) Immune signatures predict prognosis in localized cancer. Cancer Investigation 28(7): 765-773.
  17. K Suzuki, S S Kachala, K Kadota, Ronglai Shen, Qianxing Mo, et al. (2011) Prognostic immune markers in non-small cell lung cancer. Clinical Cancer Research 17(16): 5247-5256.
  18. F Pages, J Galon, MC Dieu Nosjean, E Tartour, C Sautes Fridman, et al. (2010) Immune infiltration in human tumors: a prognostic factor that should not be ignored. Oncogene 29(8): 1093-1102.
  19. V Chew, C Tow, M Teo, Hing Lok Wong, Jasmine Chan, et al. (2010) Inflammatory tumour microenvironment is associated with superior survival in hepatocellular carcinoma patients. Journal of Hepatology 52(3): 370-379.
  20. Radisky, Derek (2002) Putting tumours in context. Nature Reviews (Cancer) 1(1): 46- 54.
  21. Hanahan Douglas, Lisa M Coussens (2012) Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer cell 21(3): 309-322.
  22. Pickup Michael W, Janna K Mouw, Valerie M Weaver (2014) The extracellular matrix modulates the hallmarks of cancer. EMBO reports 15(12): 1243-1253.
  23. Buck Michael D, Ryan T Sowell, Susan M Kaech, Erika L Pearce (2017) Metabolic instruction of immunity. Cell 169(4): 570-586.
  24. Galli Filippo, Jesus Vera Aguilera, Belinda Palermo, Svetomir N Markovic, Paola Nisticò, et al. (2020) Relevance of immune cell and tumor microenvironment imaging in the new era of immunotherapy. Journal of Experimental & Clinical Cancer Research 39: 1-21.
  25. Weber Cynthia E, Paul C Kuo (2012) The tumor microenvironment. Surgical oncology 21(3): 172-177.
  26. ZN Oltvai, A L Barabasi (2002) Systems biology: life’s complexity pyramid. Science 298(5594): 763-764.
  27. NA Bhowmick, EG Neilson, HL Moses (2004) Stromal fibroblasts in cancer initiation and progression. Nature 432(7015): 332-337.
  28. P Carmeliet, RK Jain (2000) Angiogenesis in cancer and other diseases. Nature 407(6801): 249-257.
  29. Weis Sara M, David A Cheresh (2011) Tumor angiogenesis: molecular pathways and therapeutic targets. Nature medicine 17(11): 1359-1370.
  30. AB Mohseny, PCW Hogendoorn (2011) Concise review: mesenchymal tumors: when stem cells go mad. Stem Cells 29(3): 397-403.
  31. Ostrand Rosenberg Suzanne, Pratima Sinha (2009) Myeloid-derived suppressor cells: linking inflammation and cancer. The Journal of Immunology 182(8): 4499-4506.
  32. A Mantovani, T Schioppa, C Porta, P Allavena, A Sica (2006) Role of tumor-associated macrophages in tumor progression and invasion. Cancer and Metastasis Reviews 25(3): 315-322.
  33. M Tosolini, A Kirilovsky, B Mlecnik, Tessa Fredriksen, Stéphanie Mauger, et al. (2011) Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, Th2, Treg, Th17) in patients with colorectal cancer. Cancer Research 71(4): 1263-1271.
  34. J Wilson, F Balkwill (2002) The role of cytokines in the epithelial cancer microenvironment. Seminars in Cancer Biology 12(2): 113-120.
  35. F Balkwill (2004) Cancer and the chemokine network. Nature Reviews Cancer 4(7): 540-550.
  36. P Matzinger (2002) The danger model: a renewed sense of self. Science 296(5566): 301-305.
  37. GP Sims, DC Rowe, ST Rietdijk, R Herbst, AJ Coyle (2010) HMGB1 and RAGE in inflammation and cancer. Annual Review of Immunology 28: 367-388.
  38. Spranger Stefani, Daisy Dai, Brendan Horton, Thomas F Gajewski (2017) Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer cell 31(5): 711-723.
  39. Binnewies Mikhail, Edward W Roberts, Kelly Kersten, Vincent Chan, Douglas F Fearon, et al. (2018) Understanding the tumor immune microenvironment (TIME) for effective therapy. Nature medicine 24(5): 541-550.
  40. Spranger Stefani, Riyue Bao, Thomas F Gajewski (2015) Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 523(7559): 231-235.
  41. Qian Bin Zhi, Jiufeng Li, Hui Zhang, Takanori Kitamura, Jinghang Zhang, et al. (2011) CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 475(7355): 222-225.
  42. Lin Elaine Y, Andrew V Nguyen, Robert G Russell, Jeffrey W Pollard (2001) Colony- stimulating factor 1 promotes progression of mammary tumors to malignancy. Journal of Experimental Medicine 193(6): 727-740.
  43. DeNardo David G, Jairo B Barreto, Pauline Andreu, Lesley Vasquez, David Tawfik, et al. (2009) CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer cell 16(2): 91-102.
  44. Qian Bin Zhi, Hui Zhang, Jiufeng Li, Tianfang He, Eun Jin Yeo, et al. (2015) FLT1 signaling in metastasis-associated macrophages activates an inflammatory signature that promotes breast cancer metastasis. Journal of Experimental Medicine 212(9): 1433-1448.
  45. Kitamura Takanori, Bin Zhi Qian, Daniel Soong, Luca Cassetta, Roy Noy, et al. (2015) CCL2-induced chemokine cascade promotes breast cancer metastasis by enhancing retention of metastasis-associated macrophages. Journal of Experimental Medicine 212(7): 1043-1059.
  46. Headley Mark B, Adriaan Bins, Alyssa Nip, Edward W Roberts, Mark R Looney, et al. (2016) Visualization of immediate immune responses to pioneer metastatic cells in the lung. Nature 531(7595): 513-517.
  47. Hanna Richard N, Caglar Cekic, Duygu Sag, Robert Tacke, Graham D Thomas, et al. (2015) Patrolling monocytes control tumor metastasis to the lung. Science 350(6263): 985-990.
  48. Wculek Stefanie K, Ilaria Malanchi (2015) Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature 528(7582): 413-417.
  49. Nywening Timothy M, Andrea Wang Gillam, Dominic E Sanford, Brian A Belt, Roheena Z Panni, et al. (2016) Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single-centre, open-label, dose-finding, non- randomised, phase 1b trial. The lancet oncology 17(5): 651-662.
  50. Coffelt Seth B, Kelly Kersten, Chris W Doornebal, Jorieke Weiden, Kim Vrijland, et al. (2015) IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature 522(7556): 345-348.
  51. Casbon, Amy Jo, Damien Reynaud, Chanhyuk Park, Emily Khuc, et al. (2015) Invasive breast cancer reprograms early myeloid differentiation in   the   bone   marrow   to   generate   immunosuppressive neutrophils.  Proceedings of the National Academy of Sciences 112(6): 566-575.
  52. Granot Zvi, Erik Henke, Elizabeth A Comen, Tari A King, Larry Norton, et al. (2011) Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer cell 20(3): 300-314.
  53. Finisguerra Veronica, Giusy Di Conza, Mario Di Matteo, Jens Serneels, Sandra Costa, et al. (2015) MET is required for the recruitment of anti-tumoural neutrophils. Nature 522(7556): 349-353.
  54. Steele Colin W, Saadia A Karim, Joshua DG Leach, Peter Bailey, Rosanna Upstill Goddard, et al. (2016) CXCR2 inhibition profoundly suppresses metastases and augments immunotherapy in pancreatic ductal adenocarcinoma. Cancer cell 29(6): 832-845.
  55. Kowanetz Marcin, Xiumin Wu, John Lee, Martha Tan, Thijs Hagenbeek, et al. (2010) Granulocyte-colony stimulating factor promotes lung metastasis through mobilization of Ly6G+ Ly6C+ granulocytes. Proceedings of the National Academy of Sciences 107(50): 21248-21255.
  56. Jamieson Thomas, Mairi Clarke, Colin W Steele, Michael S Samuel, Jens Neumann, et al. (2012) Inhibition of CXCR2 profoundly suppresses inflammation-driven and spontaneous tumorigenesis. The Journal of clinical investigation 122(9): 3127-3144.
  57. Bald Tobias, Thomas Quast, Jennifer Landsberg, Meri Rogava, Nicole Glodde, et al. (2014) Ultraviolet-radiation-induced inflammation promotes angiotropism and metastasis in melanoma. Nature 507(7490): 109-113.
  58. Broz Miranda L, Mikhail Binnewies, Bijan Boldajipour, Amanda E Nelson, Joshua L Pollack, et al. (2014) Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer cell 26(5): 638-652.
  59. Roberts Edward W, Miranda L Broz, Mikhail Binnewies, Mark B Headley, Amanda E Nelson, et al. (2016) Critical role for CD103(+)/CD141(+) dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer cell 30(2): 324-336.
  60. Marin Acevedo Julian A, Aixa E Soyano, Bhagirathbhai Dholaria, Keith L Knutson, Yanyan Lou (2018) Cancer immunotherapy beyond immune checkpoint inhibitors. Journal of hematology & oncology 11(1): 1-25.
  61. McCarthy, Edward F (2006) The toxins of William B. Coley and the treatment of bone and soft- tissue sarcomas. The Iowa orthopaedic journal 26: 154.
  62. Parish Christopher R (2003) Cancer immunotherapy: The past, the present   and   the future. Immunology and cell biology 81(2): 106-113.
  63. Morales A, D Eidinger, A W Bruce (2002) Intracavitary Bacillus Calmette-Guerin in the treatment of superficial bladder tumors. The Journal of urology 167(2): 891-894.
  64. Old Lloyd J, Donald A Clarke, Baruj Benacerraf (1959) Effect of Bacillus Calmette-Guerin infection on transplanted tumours in the mouse. Nature 184(4682): 291-292.
  65. Sarraf CE (1994) Tumor-necrosis-factor and cell-death in tumors. International journal of oncology 5(6): 1333-1339.
  66. Decker William K, Rodrigo F da Silva, Mayra H Sanabria, Laura S Angelo, Fernando Guimarães, et al. (2017) Cancer immunotherapy: historical perspective of a clinical revolution and emerging preclinical animal models. Frontiers in Immunology 8: 829.
  67. Egen Jackson G, Wenjun Ouyang, Lawren C Wu (2020) Human anti-tumor immunity: insights from immunotherapy clinical trials. Immunity 52(1): 36-54.
  68. JP Sleeman, W Thiele (2009) Tumor metastasis and the lymphatic vasculature. International Journal of Cancer 125(12): 2747-2756.