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 + 2452 word(s) 2452 2021-10-13 11:58:27 |
2 The format is correct + 217 word(s) 2669 2021-10-27 04:55:28 |

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.
Hussain, K. Tumor-Associated Macrophages. Encyclopedia. Available online: https://encyclopedia.pub/entry/15423 (accessed on 29 March 2024).
Hussain K. Tumor-Associated Macrophages. Encyclopedia. Available at: https://encyclopedia.pub/entry/15423. Accessed March 29, 2024.
Hussain, Khiyam. "Tumor-Associated Macrophages" Encyclopedia, https://encyclopedia.pub/entry/15423 (accessed March 29, 2024).
Hussain, K. (2021, October 26). Tumor-Associated Macrophages. In Encyclopedia. https://encyclopedia.pub/entry/15423
Hussain, Khiyam. "Tumor-Associated Macrophages." Encyclopedia. Web. 26 October, 2021.
Tumor-Associated Macrophages
Edit

Resident macrophage populations within tumors are termed tumor-associated macrophages (TAMs) and can comprise up to half of the tumor mass. In established solid malignancies, the anti-tumor functions of TAMs such as phagocytosis and cytotoxic activity are suppressed, and TAMs are subverted to facilitate tumor growth.

tumor-associated macrophages resistance antibody-dependent cellular phagocytosis antibody immunotherapy phagocytosis checkpoints

1. Tumor-Associated Macrophages

Macrophages are highly plastic cells that respond and adapt to the TME in which they are resident [1][2]. Macrophage functions range from organogenesis, the capture and elimination of pathogens, tissue homeostasis, wound healing and tumorigenesis [3][4]. In solid malignancies, TAM populations can impact tumors through a multitude of complex and often opposing mechanisms, including those impacting; cell death, immunoregulation, and angiogenesis, with the net result being either pro- or antitumor. However, recently a consensus has emerged whereby most TAMs in large tumors are thought to contribute to tumor progression by increasing cancer cell invasiveness, angiogenesis and immunosuppression [5].
TAMs originate from both bone-marrow-derived hematopoietic and non-hematopoietic lineages [6][7]. In early tumorigenesis, tissue resident macrophages accumulate within tumors and account for the majority of TAMs [8]. In the brain, tissue resident macrophages (known as microglia) arise from the yolk sac, and are distinct from hematopoietic precursors in the yolk sac or fetal liver, and proliferate within tissues throughout adulthood [9]. Furthermore, it has been reported that in murine gliomas, typically only 25% of TAMs originate from circulating monocytes, with the majority derived from tissue resident microglia [10][11]. In the liver, macrophages called Kupffer cells arise from both the yolk sac and embryonic hematopoietic stem cells [12]. In adulthood, the tissue microenvironment determines to what extent circulating blood monocytes replace these tissue resident macrophages [8]. As tumors increase in size and intratumoral vascular networks form, monocytes become the dominant source of TAMs [3][13][14]. The recruitment of TAMs to tumor sites is mediated by previously resident TAMs, cancer cells, and fibroblasts, secreting a range of chemokines including: chemokines (C-C motif) ligand (CCL)2, CCL5, CCL7, and chemokine (C-X3-C motif) ligand 1 (CX3CL1), as well as cytokines such as macrophage colony-stimulating factor (M-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), and vascular endothelial growth factor (VEGF) [15][16][17][18]. Furthermore, classical monocytes (CD14hiFcγRIIIalo in humans and CD11bhiLy6C+ in mice) are recruited as a tumor progresses and differentiate into TAMs, often in a CCL2-CCR2-dependent manner. Indeed, inhibition of CCR2 signaling blocks TAM recruitment and thus reduces TAM frequency, improving the survival of tumor-burdened mice in certain murine tumor models [3].
TAMs acquire immunosuppressive or immunostimulatory gene expression patterns in response to the dynamic and varied TME in large tumors. The expression patterns of these genes can be loosely categorized as pro- or antitumor in the context of disease prognosis and their potential impact on anti-cancer therapies (Figure 1). TAMs typically express myeloid surface markers such as CD68, CD163 (class A macrophage scavenger receptor), CD206 (mannose receptor, C type 1), macrophage galactose-type lectin (MGL), macrophage receptor with collagenous structure (MARCO), programmed cell death ligand 1 (PD-L1) and V-domain Ig suppressor of T cell activation (VISTA) [19][20][21][22]. In particular, CD68, CD163 and CD206 are extensively used to identify and quantify TAMs, in addition to their being used as prognostic markers for several tumor types [23].
Figure 1. TAM-associated markers. Expression of genes in TAMs that phenotypically and functionally associate with protumor (blue) and antitumor (red) outcomes in the context of tumor progression and/or efficacy of direct targeting mAb therapy (adapted from [24]).

2. TAM Activation States

The intrinsic heterogeneity of macrophages was historically stratified into two broad activation states: M1 (for proinflammatory or classically activated macrophages) and M2 (for anti-inflammatory or alternatively activated macrophages) [25][26][27]. Although it is often reported that TAMs more closely resemble M2 macrophages, the M1/M2 dichotomy is now thought to be too reductionist and these states are likely to be examples within a spectrum of activation states [28][29]. M1 macrophages are generated following stimulation with the interferon-γ (IFN-γ) alone or in concert with bacterial components, e.g., lipopolysaccharide (LPS) or pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) [30]. Phenotypic regulation of M1-like macrophages is regulated via multiple transcription factors, such as IRF-1, STAT-1 and NF-κB [31][32][33]. These induce a pro-inflammatory phenotype in M1 macrophages, which is additionally regulated by the transcription factors: IRF-4, STAT6, PPAR-γ, the protein degradation adaptor protein, Tribbles homolog 1 (TRIB1), and chromatin modifiers including, histone demethylases and Jumonji domain-containing protein D3 (JMJD3) [32][34]. In contrast, M2 macrophages are polarized by several factors, and can be further subdivided into M2a, M2b, and M2c [30]. M2a macrophages are generated following exposure to IL-4 and/or IL-13. M2b macrophages are induced by immune complexes (ICs), LPS, certain Toll-like receptor (TLRs) agonists, or the IL-1 receptor antagonist (IL-1ra) [35][36][37]. M2c macrophages can be induced in response to exposure to IL-10, transforming growth factor-β (TGF-β), or glucocorticoids (GCs) [38][39]. TAMs with enhanced expression of CD163, CD204, CD206, stabilin-1, arginase-1, and matrix metallopeptidase 9 (MMP9), and elevated production of IL-10, VEGF, and prostaglandin E2 (PGE2), generally show M2-like characteristics [40][30][2][41]. In addition to implications for tumor neogenesis as well as “wound healing”, the status of ‘M2-like’ macrophages has ramifications for multiple treatment modalities.

3. Protumor Functions of TAMs

TAMs possess an ‘M2-like’ phenotype and function that promotes immunosuppression, metastases, and angiogenesis (Figure 2). Tissue resident macrophages and TAMs can phagocytose, and lyse cancer cells, activate NK cells and induce T helper 1 (Th1) immune responses [42][43][44]. However, TAMs are broadly associated with poor prognosis in several tumor types, including cholangiocarcinoma, glioma, Hodgkin lymphoma and ovarian and breast cancers [41]. Increased frequencies of CD163+, CD204+ and CD206+ TAMs correlate with tumor progression and worse clinical prognosis [31]. Furthermore, in some malignant tumors, the density and quantity of TAM infiltration is associated with higher Ki-67 expression, indicating elevated cancer cell proliferation [45].
Figure 2. Major protumor functions of tumor-associated macrophages. TAMs mediate suppression of effector T cells via the secretion of soluble proteins and through the expression of inhibitory cell surface molecules. TAMs also produce several factors that promote extracellular matrix (ECM) degradation, which facilitates tumor metastasis. Furthermore, TAMs secrete cytokines that promote angiogenesis, consequently accelerating tumor growth.
TAMs also produce high levels of cytokines and chemokines, which recruit or induce immunosuppressive cell types at tumor sites. Thymically derived natural Treg traffic and infiltrate to tumor sites via several chemokine receptors, in particular CCR4 [46].
In addition to inducing Treg cells and MDSCs at the tumor site, TAMs actively participate in the immunosuppression of effector T cells. TAM-derived arginine and tryptophan suppresses CD3 ζ-chain expression in T cells, resulting in the inhibition of effector T cell activation [47][48].
TAMs express several enzymes, cytokines and chemokines that promote tumor metastases, such that TAM frequencies positively correlate with cancer cell invasiveness and metastasis [49].
TAMs are also important promoters of angiogenesis in the TME. They function to degrade the tumor basement membrane, via the production of MMPs and cathepsins, and secrete proangiogenic growth factors such as VEGF, PDGF, bFGF and TGF-β that induce new vasculature in growing tumors [50][51].
In summary, TAMs promote tumor growth through multiple mechanisms that are attributed to ‘M2-like’ phenotypes induced within the TME, highlighting a need to develop strategies that either delete these cells or repolarize them to proinflammatory antitumor states. In the context of direct targeting mAb immunotherapies, TAMs can function to phagocytose mAb-opsonized cells, and novel strategies to target the so-called ‘phagocytosis checkpoints’ to enhance the phagocytic functions of these cells are also currently under investigation.

4. TAM-Mediated Depletion of Cancer Cells

Tumor-targeting mAbs such as Rituximab, Herceptin and Cetuximab, recruit ADCP-mediating macrophages to directly eliminate cancer cells [52][53][54][55][56][57]. Checkpoint inhibitor mAbs such as Ipilimumab were previously thought to function solely via receptor blockade and expansion of effector T cells [58]. However, additionally, Ipilimumab has been reported to work optimally through the depletion of tumor-infiltrating immunosuppressive Treg cells, also indicating a role for ADCP-mediating myeloid cells [59][60][61]. Although several cell types are functionally capable of phagocytosing and destroying host cells, including epithelial cells, mesenchymal cells and fibroblasts, neutrophils, and monocytes [62][63][64], macrophages are ‘professional phagocytes’ and the principal effector cells in efferocytosis (clearance of apoptotic cells) and ADCP [65].
IgG antibodies can trigger ADCP indirectly via activation of the classical complement pathway, where iC3b-opsonized target cells can bind to complement receptor 3 (CR3, integrin αMβ2) to elicit engulfment by ‘sinking phagocytosis’ [66]. Importantly, the macrophage cell surface receptors required for ADCP are less varied than for efferocytosis, with ADCP in the context of mAbs like Rituximab almost entirely dependent on FcγRs that bind the Fc portion of IgG antibodies. Human macrophages express the activating high affinity FcγRI and low affinity FcγRIIa and FcγRIIIa [67][68], as well as the inhibitory FcγRIIb. Antibody-bound target cells interact with FcγRI, FcγRIIa and FcγRIIIa for optimal ADCP (FcγRI, FcγRIII and FcγRIV in the mouse), whereas engagement with the sole inhibitory FcγR, FcγRIIb (FcγRII in mice), attenuates phagocytic function [67]. The expression levels and cellular distribution of FcγR on effector cells are of crucial importance in mAb therapy outcomes. Furthermore, human IgG1 and murine IgG2a, and IgG2c isotypes preferentially engage, activating above inhibitory FcγR, eliciting stronger ADCP (relative to human IgG2 or murine IgG1), and therefore are the preferred IgG isotypes for direct tumor-targeting mAbs [69][70][71].
After engagement, activating FcγRs cluster and phosphorylate ITAM in their cytoplasmic domains or associated gamma chains [67]. This induces the formation of the phagocytic synapse and thence, actin polymerization leads to the formation of the phagocytic cup [72]. The macrophage then extends pseudopodia around the opsonized target cell, engulfing it in a process termed zippering phagocytosis [73]. Actin filaments subsequently rearrange within the macrophage, causing its cell membrane to encompass the target cell, which leads to its inclusion into a phagosome. The phagosome fuses with endosomes and then lysosomes [74], followed by a marked reduction in pH (∼4.5) and generation of ROS [75], leading to the destruction of the phagocytosed cell [76]. The inhibitory FcγRIIb possesses an ITIM in its cytoplasmic domain, and the interaction of IgG Fc regions or immune complexes results in the recruitment of src homology 2 (SH2) domain containing inositol polyphosphate 5-phosphatase (SHIP), curtailing signaling from activating FcγR and consequently ADCP [77].
A seminal study by Clynes et al. [78] observed that nude mice deficient in the common gamma chain (FcRγ−/−/nu/nu mice), which consequently lack expression and signaling from the activating FcγRs, were unable to control human breast carcinoma BT474M1 growth in response to trastuzumab treatment. This implicated a role for activating-FcγR-bearing myeloid cells and NK cells in therapeutic outcomes. Importantly, mice deficient in the inhibitory FcγRIIb showed potent antibody-mediated target cell killing. The latter result not only demonstrated that FcγR-dependent mechanisms contribute substantially to the action of direct targeting mAbs, but implicated macrophages as key effectors cells in direct targeting mAb immunotherapy, given that NK cells do not express FcγRIIb in mice or humans [78].
Subsequent studies using intravital microscopy have reported that following anti-CD20 mAb therapy in murine models, Kupffer cells in the liver sinusoids, phagocytose circulating mAb-opsonized malignant B lymphoma cells [75][79][80], including in human CD20 transgenic mice [80]. Anti-CD20 mediated depletion of lymphoma cells in adoptive transfer models or the Eμ-Myc B cell lymphoma model has been shown to be dependent on activating FcγRs. Furthermore, the clodronate-mediated elimination of macrophages abrogated anti-CD20 therapy in this mouse model, further highlighting the indispensable role of macrophages in malignant B cell depletion [81].

5. Antibody-Mediated Modulation of TAM Recruitment, Survival, and Effector Functions

Strategies to diminish the protumor functions of TAMs include the suppression of TAM generation, monocyte recruitment, and the repolarization of TAMs to proinflammatory phenotypes. Additionally, a compelling TAM targeting strategy has emerged that aims to target ‘phagocytosis checkpoints’ to enhance ‘eat me’ and block ‘don’t eat me’ signaling in tumors (Figure 3). Table 1 summarizes TAM-targeting mAbs in early phase trials that have been developed to reduce protumor TAM frequencies or augment antitumor immune responses in cancer patients.

Figure 3. TAM cell surface molecule candidates for mAb targeting. TAM cell surface molecules that can potentially be targeted by mAbs to modify TAM frequencies or repolarization to a proinflammatory phenotype in the TME. These mAb targets are grouped according to the predominant effect resulting from stimulation of their natural ligand. However, mAb-mediated targeting of these molecules may exert further functional changes in TAMs and healthy mononuclear phagocytes.
Table 1. TAM-targeting mAbs in completed or active trials. These mAbs have been investigated in or are in active clinical trials, either as single agents, or in combination with chemotherapeutic agents, checkpoint inhibitors, Fc fusion proteins or TLR agonists.
Target Compound Sponsor Phase Indication Status ClinicalTrials.gov identifier
CCR2 Plozalizumab Southwest Oncology Group II Metastatic cancer, unspecified adult solid tumor Completed NCT01015560
CCL2 Carlumab Centocor Research & Development, Inc. II Prostate cancer Completed NCT00992186
CSF-1R AMG820 Amgen I Solid tumors Completed NCT0144404
Emactuzumab (RG7155) Roche I Solid tumors Completed NCT01494688
IMC-SC4 Eli Lilly I Breast and prostate cancer Active NCT02265536
CD40 SEA-CD40 Seagen Inc. I Non-small-cell lung carcinoma, squamous solid tumors Active NCT02376699
LVGN7409 Lyvgen Biopharma Holdings Limited I Solid tumors Active NCT04635995
CDX-1140 Celldex Therapeutics I/II Melanoma Active NCT04364230
APX005M Apexigen, Inc. II Soft tissue sarcoma Active NCT03719430
ADC-1013 Janssen Research & Development, LLC I Advanced solid neoplasms Active NCT02829099
ChiLob 7/4 Cancer Research UK I B-cell lymphoma Completed NCT01561911
Selicrelumab Hoffmann-La Roche I/II Pancreatic adenocarcinoma Active NCT03193190
FcγRIIb BI-1206 BioInvent International AB I/II Indolent B-cell non-Hodgkin lymphoma Active NCT03571568
SIRPα BI 765063 OSE Immunotherapeutics I Solid tumor Active NCT03990233
CC-95251 Celgene I Neoplasms Active NCT03783403
GS-0189 Gilead Sciences I Non-Hodgkin lymphoma Active NCT04502706
VISTA CI-8993 Curis, Inc. I Solid tumor Active NCT04475523

6. TAM Recruitment and Survival

6.1. CSF-1R

CSF-1R is a tyrosine kinase receptor expressed on all myeloid cells. Its ligands are M-CSF (CSF-1), GM-CSF (CSF-2) and IL-34, and their binding to CSF-1R induces differentiation, recruitment to tumor sites, and the survival of monocytes and macrophages [82]. The ‘M2-like’ TAM phenotype has been reported to be mediated by the growth factor M-CSF in addition to the Th2 cytokines: IL-4/IL-13, and Treg-cell-derived IL-10, in the TME [30]. Since the presence of CSF-1R+ TAMs correlates with poor survival in several tumor types [83], targeting CSF-1R represents an attractive strategy to eliminate or potentially repolarize these cells. Mononuclear phagocytes are almost completely absent in CSFR1−/− mice [84]. Accordingly, mAbs targeting either CSF-1R or its ligand M-CSF have been developed. The antitumor and antimetastatic activities of anti-CSF-1R mAb have been demonstrated in subcutaneous EL4 lymphoma and MMTV-PyMT breast tumor models [85].

6.2. CCR2/CCL2

CCL2 is a key chemokine which mediates macrophage recruitment to tumor sites. The anti-CCR2 mAb MLN1202 has been successfully used in patients at risk for atherosclerotic cardiovascular disease to reduce markers of inflammation [86]. Targeting CCR2 or its ligand, CCL2, with mAbs to block TAM recruitment has also been investigated in mice with orthotopic MDA-MB-231 human breast cancer tumors. Here, treatment with anti-CCL2 mAb reduced TAM accumulation, consequently reducing angiogenesis and tumor growth [87]. Furthermore, Carlumab, a human IgG1 anti-CCL2 mAb, has been investigated in clinical trials for patients with various solid tumors. However, this strategy was not sufficiently efficacious, even when combined with chemotherapy [88]. Likely explanations include the broad redundancy in the chemokine system, which contains dozens of different ligands and receptors. Indeed, tissue resident macrophages in particular, which differentiate into the most protumor fraction of the myeloid compartment, may be independent of regulation by any single chemokine receptor or ligand [89].

7. Conclusions

Although TAMs are indispensable effector cells in direct targeting mAb immunotherapy, the immunosuppressive TME markedly reduces their ability to elicit ADCP and deplete mAb-opsonized targets. Currently, several early phase clinical trials are investigating different TAM-targeting mAbs, and in particular, anti-CD40 agonistic mAbs hold great potential to repolarize TAM activation states in the TME. Furthermore, mAb-mediated FcγRIIb blockade is also a promising candidate for the enhancement of ADCP in the context of anti-CD20 mAb therapies. Recently, high-dimensional single-cell RNA sequencing has shed new light on the variety of myeloid cells in the TME. Furthermore, this has also revealed novel TAM-associated cell surface markers and signaling pathways, with the potential for targeted intervention to reshape the tumor myeloid cell landscape to in turn enhance clinical outcomes.

References

  1. Sica, A.; Mantovani, A. Macrophage plasticity and polarization: In vivo veritas. J. Clin. Investig. 2012, 122, 787–795.
  2. Pollard, J.W. Tumour-educated macrophages promote tumour progression and metastasis. Nat. Rev. Cancer 2004, 4, 71–78.
  3. Qian, B.Z.; Pollard, J.W. Macrophage diversity enhances tumor progression and metastasis. Cell 2010, 141, 39–51.
  4. Laoui, D.; Van Overmeire, E.; Movahedi, K.; Van den Bossche, J.; Schouppe, E.; Mommer, C.; Nikolaou, A.; Morias, Y.; De Baetselier, P.; Van Ginderachter, J.A. Mononuclear phagocyte heterogeneity in cancer: Different subsets and activation states reaching out at the tumor site. Immunobiology 2011, 216, 1192–1202.
  5. Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416.
  6. Zhu, Y.; Herndon, J.M.; Sojka, D.K.; Kim, K.W.; Knolhoff, B.L.; Zuo, C.; Cullinan, D.R.; Luo, J.; Bearden, A.R.; Lavine, K.J.; et al. Tissue-Resident Macrophages in Pancreatic Ductal Adenocarcinoma Originate from Embryonic Hematopoiesis and Promote Tumor Progression. Immunity 2017, 47, 323–338.e6.
  7. Bowman, R.L.; Klemm, F.; Akkari, L.; Pyonteck, S.M.; Sevenich, L.; Quail, D.F.; Dhara, S.; Simpson, K.; Gardner, E.E.; Iacobuzio-Donahue, C.A.; et al. Macrophage Ontogeny Underlies Differences in Tumor-Specific Education in Brain Malignancies. Cell Rep. 2016, 17, 2445–2459.
  8. Epelman, S.; Lavine, K.J.; Randolph, G.J. Origin and functions of tissue macrophages. Immunity 2014, 41, 21–35.
  9. Ginhoux, F.; Guilliams, M. Tissue-Resident Macrophage Ontogeny and Homeostasis. Immunity 2016, 44, 439–449.
  10. Hambardzumyan, D.; Gutmann, D.H.; Kettenmann, H. The role of microglia and macrophages in glioma maintenance and progression. Nat. Neurosci. 2016, 19, 20–27.
  11. Muller, A.; Brandenburg, S.; Turkowski, K.; Muller, S.; Vajkoczy, P. Resident microglia, and not peripheral macrophages, are the main source of brain tumor mononuclear cells. Int. J. Cancer 2015, 137, 278–288.
  12. Schulz, C.; Gomez Perdiguero, E.; Chorro, L.; Szabo-Rogers, H.; Cagnard, N.; Kierdorf, K.; Prinz, M.; Wu, B.; Jacobsen, S.E.; Pollard, J.W.; et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 2012, 336, 86–90.
  13. Arwert, E.N.; Harney, A.S.; Entenberg, D.; Wang, Y.; Sahai, E.; Pollard, J.W.; Condeelis, J.S. A Unidirectional Transition from Migratory to Perivascular Macrophage Is Required for Tumor Cell Intravasation. Cell Rep. 2018, 23, 1239–1248.
  14. Franklin, R.A.; Liao, W.; Sarkar, A.; Kim, M.V.; Bivona, M.R.; Liu, K.; Pamer, E.G.; Li, M.O. The cellular and molecular origin of tumor-associated macrophages. Science 2014, 344, 921–925.
  15. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444.
  16. Brown, C.E.; Vishwanath, R.P.; Aguilar, B.; Starr, R.; Najbauer, J.; Aboody, K.S.; Jensen, M.C. Tumor-derived chemokine MCP-1/CCL2 is sufficient for mediating tumor tropism of adoptively transferred T cells. J. Immunol. 2007, 179, 3332–3341.
  17. Pollard, J.W. Macrophages define the invasive microenvironment in breast cancer. J. Leukoc. Biol. 2008, 84, 623–630.
  18. Zhang, J.; Lu, Y.; Pienta, K.J. Multiple roles of chemokine (C-C motif) ligand 2 in promoting prostate cancer growth. J. Natl. Cancer Inst. 2010, 102, 522–528.
  19. Mattiola, I.; Tomay, F.; De Pizzol, M.; Silva-Gomes, R.; Savino, B.; Gulic, T.; Doni, A.; Lonardi, S.; Astrid Boutet, M.; Nerviani, A.; et al. The macrophage tetraspan MS4A4A enhances dectin-1-dependent NK cell-mediated resistance to metastasis. Nat. Immunol. 2019, 20, 1012–1022.
  20. Gordon, S.R.; Maute, R.L.; Dulken, B.W.; Hutter, G.; George, B.M.; McCracken, M.N.; Gupta, R.; Tsai, J.M.; Sinha, R.; Corey, D.; et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 2017, 545, 495–499.
  21. Blando, J.; Sharma, A.; Higa, M.G.; Zhao, H.; Vence, L.; Yadav, S.S.; Kim, J.; Sepulveda, A.M.; Sharp, M.; Maitra, A.; et al. Comparison of immune infiltrates in melanoma and pancreatic cancer highlights VISTA as a potential target in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2019, 116, 1692–1697.
  22. Kato, S.; Okamura, R.; Kumaki, Y.; Ikeda, S.; Nikanjam, M.; Eskander, R.; Goodman, A.; Lee, S.; Glenn, S.T.; Dressman, D.; et al. Expression of TIM3/VISTA checkpoints and the CD68 macrophage-associated marker correlates with anti-PD1/PDL1 resistance: Implications of immunogram heterogeneity. Oncoimmunology 2020, 9, 1708065.
  23. Dander, E.; Fallati, A.; Gulic, T.; Pagni, F.; Gaspari, S.; Silvestri, D.; Cricri, G.; Bedini, G.; Portale, F.; Buracchi, C.; et al. Monocyte-macrophage polarization and recruitment pathways in the tumour microenvironment of B-cell acute lymphoblastic leukaemia. Br. J. Haematol. 2021, 193, 1157–1171.
  24. Molgora, M.; Colonna, M. Turning enemies into allies-reprogramming tumor-associated macrophages for cancer therapy. Med 2021, 2, 666–681.
  25. Pace, J.L.; Russell, S.W.; Schreiber, R.D.; Altman, A.; Katz, D.H. Macrophage activation: Priming activity from a T-cell hybridoma is attributable to interferon-gamma. Proc. Natl. Acad. Sci. USA 1983, 80, 3782–3786.
  26. Celada, A.; Gray, P.W.; Rinderknecht, E.; Schreiber, R.D. Evidence for a gamma-interferon receptor that regulates macrophage tumoricidal activity. J. Exp. Med. 1984, 160, 55–74.
  27. Mills, C.D.; Kincaid, K.; Alt, J.M.; Heilman, M.J.; Hill, A.M. M-1/M-2 macrophages and the Th1/Th2 paradigm. J. Immunol. 2000, 164, 6166–6173.
  28. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20.
  29. Xue, J.; Schmidt, S.V.; Sander, J.; Draffehn, A.; Krebs, W.; Quester, I.; De Nardo, D.; Gohel, T.D.; Emde, M.; Schmidleithner, L.; et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity 2014, 40, 274–288.
  30. Mantovani, A.; Sozzani, S.; Locati, M.; Allavena, P.; Sica, A. Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002, 23, 549–555.
  31. Komohara, Y.; Jinushi, M.; Takeya, M. Clinical significance of macrophage heterogeneity in human malignant tumors. Cancer Sci. 2014, 105, 1–8.
  32. Mantovani, A.; Sica, A.; Locati, M. Macrophage polarization comes of age. Immunity 2005, 23, 344–346.
  33. Taub, D.D.; Cox, G.W. Murine Th1 and Th2 cell clones differentially regulate macrophage nitric oxide production. J. Leukoc. Biol. 1995, 58, 80–89.
  34. Wynn, T.A.; Chawla, A.; Pollard, J.W. Macrophage biology in development, homeostasis and disease. Nature 2013, 496, 445–455.
  35. Bianchini, R.; Roth-Walter, F.; Ohradanova-Repic, A.; Flicker, S.; Hufnagl, K.; Fischer, M.B.; Stockinger, H.; Jensen-Jarolim, E. IgG4 drives M2a macrophages to a regulatory M2b-like phenotype: Potential implication in immune tolerance. Allergy 2019, 74, 483–494.
  36. Bumgardner, S.A.; Zhang, L.; LaVoy, A.S.; Andre, B.; Frank, C.B.; Kajikawa, A.; Klaenhammer, T.R.; Dean, G.A. Nod2 is required for antigen-specific humoral responses against antigens orally delivered using a recombinant Lactobacillus vaccine platform. PLoS ONE 2018, 13, e0196950.
  37. Yang, C.; Zhang, D.M.; Song, Z.B.; Hou, Y.Q.; Bao, Y.L.; Sun, L.G.; Yu, C.L.; Li, Y.X. Protumoral TSP50 Regulates Macrophage Activities and Polarization via Production of TNF-alpha and IL-1beta, and Activation of the NF-kappaB Signaling Pathway. PLoS ONE 2015, 10, e0145095.
  38. Martinez, F.O.; Helming, L.; Gordon, S. Alternative activation of macrophages: An immunologic functional perspective. Annu. Rev. Immunol. 2009, 27, 451–483.
  39. Zizzo, G.; Hilliard, B.A.; Monestier, M.; Cohen, P.L. Efficient clearance of early apoptotic cells by human macrophages requires M2c polarization and MerTK induction. J. Immunol. 2012, 189, 3508–3520.
  40. Bingle, L.; Brown, N.J.; Lewis, C.E. The role of tumour-associated macrophages in tumour progression: Implications for new anticancer therapies. J. Pathol. 2002, 196, 254–265.
  41. Heusinkveld, M.; van der Burg, S.H. Identification and manipulation of tumor associated macrophages in human cancers. J. Transl. Med. 2011, 9, 216.
  42. Biswas, S.K.; Sica, A.; Lewis, C.E. Plasticity of macrophage function during tumor progression: Regulation by distinct molecular mechanisms. J. Immunol. 2008, 180, 2011–2017.
  43. Molgora, M.; Supino, D.; Mavilio, D.; Santoni, A.; Moretta, L.; Mantovani, A.; Garlanda, C. The yin-yang of the interaction between myelomonocytic cells and NK cells. Scand. J. Immunol. 2018, 88, e12705.
  44. Garrido-Martin, E.M.; Mellows, T.W.P.; Clarke, J.; Ganesan, A.P.; Wood, O.; Cazaly, A.; Seumois, G.; Chee, S.J.; Alzetani, A.; King, E.V.; et al. M1(hot) tumor-associated macrophages boost tissue-resident memory T cells infiltration and survival in human lung cancer. J. Immunother. Cancer 2020, 8, e000778.
  45. Leuverink, E.M.; Brennan, B.A.; Crook, M.L.; Doherty, D.A.; Hammond, I.G.; Ruba, S.; Stewart, C.J. Prognostic value of mitotic counts and Ki-67 immunoreactivity in adult-type granulosa cell tumour of the ovary. J. Clin. Pathol. 2008, 61, 914–919.
  46. Watanabe, M.; Kanao, K.; Suzuki, S.; Muramatsu, H.; Morinaga, S.; Kajikawa, K.; Kobayashi, I.; Nishikawa, G.; Kato, Y.; Zennami, K.; et al. Increased infiltration of CCR4-positive regulatory T cells in prostate cancer tissue is associated with a poor prognosis. Prostate 2019, 79, 1658–1665.
  47. Munn, D.H.; Sharma, M.D.; Baban, B.; Harding, H.P.; Zhang, Y.; Ron, D.; Mellor, A.L. GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity 2005, 22, 633–642.
  48. Rodriguez, P.C.; Quiceno, D.G.; Ochoa, A.C. L-arginine availability regulates T-lymphocyte cell-cycle progression. Blood 2007, 109, 1568–1573.
  49. Condeelis, J.; Pollard, J.W. Macrophages: Obligate partners for tumor cell migration, invasion, and metastasis. Cell 2006, 124, 263–266.
  50. Kessenbrock, K.; Plaks, V.; Werb, Z. Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell 2010, 141, 52–67.
  51. Murdoch, C.; Muthana, M.; Coffelt, S.B.; Lewis, C.E. The role of myeloid cells in the promotion of tumour angiogenesis. Nat. Rev. Cancer 2008, 8, 618–631.
  52. Yakes, F.M.; Chinratanalab, W.; Ritter, C.A.; King, W.; Seelig, S.; Arteaga, C.L. Herceptin-induced inhibition of phosphatidylinositol-3 kinase and Akt Is required for antibody-mediated effects on p27, cyclin D1, and antitumor action. Cancer Res. 2002, 62, 4132–4141.
  53. Gong, Q.; Ou, Q.; Ye, S.; Lee, W.P.; Cornelius, J.; Diehl, L.; Lin, W.Y.; Hu, Z.; Lu, Y.; Chen, Y.; et al. Importance of cellular microenvironment and circulatory dynamics in B cell immunotherapy. J. Immunol. 2005, 174, 817–826.
  54. Uchida, J.; Hamaguchi, Y.; Oliver, J.A.; Ravetch, J.V.; Poe, J.C.; Haas, K.M.; Tedder, T.F. The innate mononuclear phagocyte network depletes B lymphocytes through Fc receptor-dependent mechanisms during anti-CD20 antibody immunotherapy. J. Exp. Med. 2004, 199, 1659–1669.
  55. Beers, S.A.; French, R.R.; Chan, H.T.; Lim, S.H.; Jarrett, T.C.; Vidal, R.M.; Wijayaweera, S.S.; Dixon, S.V.; Kim, H.; Cox, K.L.; et al. Antigenic modulation limits the efficacy of anti-CD20 antibodies: Implications for antibody selection. Blood 2010, 115, 5191–5201.
  56. Biburger, M.; Aschermann, S.; Schwab, I.; Lux, A.; Albert, H.; Danzer, H.; Woigk, M.; Dudziak, D.; Nimmerjahn, F. Monocyte subsets responsible for immunoglobulin G-dependent effector functions in vivo. Immunity 2011, 35, 932–944.
  57. Lehmann, B.; Biburger, M.; Bruckner, C.; Ipsen-Escobedo, A.; Gordan, S.; Lehmann, C.; Voehringer, D.; Winkler, T.; Schaft, N.; Dudziak, D.; et al. Tumor location determines tissue-specific recruitment of tumor-associated macrophages and antibody-dependent immunotherapy response. Sci. Immunol. 2017, 2, eaah6413.
  58. Wei, S.C.; Levine, J.H.; Cogdill, A.P.; Zhao, Y.; Anang, N.A.S.; Andrews, M.C.; Sharma, P.; Wang, J.; Wargo, J.A.; Pe’er, D.; et al. Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade. Cell 2017, 170, 1120–1133.e17.
  59. Arce Vargas, F.; Furness, A.J.S.; Litchfield, K.; Joshi, K.; Rosenthal, R.; Ghorani, E.; Solomon, I.; Lesko, M.H.; Ruef, N.; Roddie, C.; et al. Fc Effector Function Contributes to the Activity of Human Anti-CTLA-4 Antibodies. Cancer Cell 2018, 33, 649–663.e4.
  60. Simpson, T.R.; Li, F.; Montalvo-Ortiz, W.; Sepulveda, M.A.; Bergerhoff, K.; Arce, F.; Roddie, C.; Henry, J.Y.; Yagita, H.; Wolchok, J.D.; et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J. Exp. Med. 2013, 210, 1695–1710.
  61. Romano, E.; Kusio-Kobialka, M.; Foukas, P.G.; Baumgaertner, P.; Meyer, C.; Ballabeni, P.; Michielin, O.; Weide, B.; Romero, P.; Speiser, D.E. Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc. Natl. Acad. Sci. USA 2015, 112, 6140–6145.
  62. Monks, J.; Rosner, D.; Geske, F.J.; Lehman, L.; Hanson, L.; Neville, M.C.; Fadok, V.A. Epithelial cells as phagocytes: Apoptotic epithelial cells are engulfed by mammary alveolar epithelial cells and repress inflammatory mediator release. Cell Death Differ. 2005, 12, 107–114.
  63. Tso, G.H.; Law, H.K.; Tu, W.; Chan, G.C.; Lau, Y.L. Phagocytosis of apoptotic cells modulates mesenchymal stem cells osteogenic differentiation to enhance IL-17 and RANKL expression on CD4+ T cells. Stem Cells 2010, 28, 939–954.
  64. Seeberg, J.C.; Loibl, M.; Moser, F.; Schwegler, M.; Buttner-Herold, M.; Daniel, C.; Engel, F.B.; Hartmann, A.; Schlotzer-Schrehardt, U.; Goppelt-Struebe, M.; et al. Non-professional phagocytosis: A general feature of normal tissue cells. Sci. Rep. 2019, 9, 11875.
  65. Weiskopf, K.; Weissman, I.L. Macrophages are critical effectors of antibody therapies for cancer. mAbs 2015, 7, 303–310.
  66. Golay, J.; Taylor, R.P. The Role of Complement in the Mechanism of Action of Therapeutic Anti-Cancer mAbs. Antibodies 2020, 9, 58.
  67. Nimmerjahn, F.; Ravetch, J.V. Fcgamma receptors as regulators of immune responses. Nat. Rev. Immunol. 2008, 8, 34–47.
  68. Nagelkerke, S.Q.; Tacke, C.E.; Breunis, W.B.; Tanck, M.W.T.; Geissler, J.; Png, E.; Hoang, L.T.; van der Heijden, J.; Naim, A.N.M.; Yeung, R.S.M.; et al. Extensive Ethnic Variation and Linkage Disequilibrium at the FCGR2/3 Locus: Different Genetic Associations Revealed in Kawasaki Disease. Front. Immunol. 2019, 10, 185.
  69. Nimmerjahn, F.; Ravetch, J.V. Translating basic mechanisms of IgG effector activity into next generation cancer therapies. Cancer Immun. 2012, 12, 13.
  70. White, A.L.; Dou, L.; Chan, H.T.; Field, V.L.; Mockridge, C.I.; Moss, K.; Williams, E.L.; Booth, S.G.; French, R.R.; Potter, E.A.; et al. Fcgamma receptor dependency of agonistic CD40 antibody in lymphoma therapy can be overcome through antibody multimerization. J. Immunol. 2014, 193, 1828–1835.
  71. Hamaguchi, Y.; Xiu, Y.; Komura, K.; Nimmerjahn, F.; Tedder, T.F. Antibody isotype-specific engagement of Fcgamma receptors regulates B lymphocyte depletion during CD20 immunotherapy. J. Exp. Med. 2006, 203, 743–753.
  72. Lee, W.L.; Mason, D.; Schreiber, A.D.; Grinstein, S. Quantitative analysis of membrane remodeling at the phagocytic cup. Mol. Biol. Cell 2007, 18, 2883–2892.
  73. Griffin, F.M., Jr.; Griffin, J.A.; Leider, J.E.; Silverstein, S.C. Studies on the mechanism of phagocytosis. I. Requirements for circumferential attachment of particle-bound ligands to specific receptors on the macrophage plasma membrane. J. Exp. Med. 1975, 142, 1263–1282.
  74. Pitt, A.; Mayorga, L.S.; Stahl, P.D.; Schwartz, A.L. Alterations in the protein composition of maturing phagosomes. J. Clin. Investig. 1992, 90, 1978–1983.
  75. Gul, N.; Babes, L.; Siegmund, K.; Korthouwer, R.; Bogels, M.; Braster, R.; Vidarsson, G.; ten Hagen, T.L.; Kubes, P.; van Egmond, M. Macrophages eliminate circulating tumor cells after monoclonal antibody therapy. J. Clin. Investig. 2014, 124, 812–823.
  76. Flannagan, R.S.; Jaumouille, V.; Grinstein, S. The cell biology of phagocytosis. Annu. Rev. Pathol. 2012, 7, 61–98.
  77. Li, X.; Wu, J.; Carter, R.H.; Edberg, J.C.; Su, K.; Cooper, G.S.; Kimberly, R.P. A novel polymorphism in the Fcgamma receptor IIB (CD32B) transmembrane region alters receptor signaling. Arthritis Rheum. 2003, 48, 3242–3252.
  78. Clynes, R.A.; Towers, T.L.; Presta, L.G.; Ravetch, J.V. Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets. Nat. Med. 2000, 6, 443–446.
  79. Montalvao, F.; Garcia, Z.; Celli, S.; Breart, B.; Deguine, J.; Van Rooijen, N.; Bousso, P. The mechanism of anti-CD20-mediated B cell depletion revealed by intravital imaging. J. Clin. Investig. 2013, 123, 5098–5103.
  80. Grandjean, C.L.; Montalvao, F.; Celli, S.; Michonneau, D.; Breart, B.; Garcia, Z.; Perro, M.; Freytag, O.; Gerdes, C.A.; Bousso, P. Intravital imaging reveals improved Kupffer cell-mediated phagocytosis as a mode of action of glycoengineered anti-CD20 antibodies. Sci. Rep. 2016, 6, 34382.
  81. Minard-Colin, V.; Xiu, Y.; Poe, J.C.; Horikawa, M.; Magro, C.M.; Hamaguchi, Y.; Haas, K.M.; Tedder, T.F. Lymphoma depletion during CD20 immunotherapy in mice is mediated by macrophage FcgammaRI, FcgammaRIII, and FcgammaRIV. Blood 2008, 112, 1205–1213.
  82. Lenzo, J.C.; Turner, A.L.; Cook, A.D.; Vlahos, R.; Anderson, G.P.; Reynolds, E.C.; Hamilton, J.A. Control of macrophage lineage populations by CSF-1 receptor and GM-CSF in homeostasis and inflammation. Immunol. Cell Biol. 2012, 90, 429–440.
  83. Zhang, Q.W.; Liu, L.; Gong, C.Y.; Shi, H.S.; Zeng, Y.H.; Wang, X.Z.; Zhao, Y.W.; Wei, Y.Q. Prognostic significance of tumor-associated macrophages in solid tumor: A meta-analysis of the literature. PLoS ONE 2012, 7, e50946.
  84. Erblich, B.; Zhu, L.; Etgen, A.M.; Dobrenis, K.; Pollard, J.W. Absence of colony stimulation factor-1 receptor results in loss of microglia, disrupted brain development and olfactory deficits. PLoS ONE 2011, 6, e26317.
  85. Strachan, D.C.; Ruffell, B.; Oei, Y.; Bissell, M.J.; Coussens, L.M.; Pryer, N.; Daniel, D. CSF1R inhibition delays cervical and mammary tumor growth in murine models by attenuating the turnover of tumor-associated macrophages and enhancing infiltration by CD8(+) T cells. Oncoimmunology 2013, 2, e26968.
  86. Gilbert, J.; Lekstrom-Himes, J.; Donaldson, D.; Lee, Y.; Hu, M.; Xu, J.; Wyant, T.; Davidson, M.; Group, M.L.N.S. Effect of CC chemokine receptor 2 CCR2 blockade on serum C-reactive protein in individuals at atherosclerotic risk and with a single nucleotide polymorphism of the monocyte chemoattractant protein-1 promoter region. Am. J. Cardiol. 2011, 107, 906–911.
  87. Fujimoto, H.; Sangai, T.; Ishii, G.; Ikehara, A.; Nagashima, T.; Miyazaki, M.; Ochiai, A. Stromal MCP-1 in mammary tumors induces tumor-associated macrophage infiltration and contributes to tumor progression. Int. J. Cancer 2009, 125, 1276–1284.
  88. Brana, I.; Calles, A.; LoRusso, P.M.; Yee, L.K.; Puchalski, T.A.; Seetharam, S.; Zhong, B.; de Boer, C.J.; Tabernero, J.; Calvo, E. Carlumab, an anti-C-C chemokine ligand 2 monoclonal antibody, in combination with four chemotherapy regimens for the treatment of patients with solid tumors: An open-label, multicenter phase 1b study. Target. Oncol. 2015, 10, 111–123.
  89. Argyle, D.; Kitamura, T. Targeting Macrophage-Recruiting Chemokines as a Novel Therapeutic Strategy to Prevent the Progression of Solid Tumors. Front. Immunol. 2018, 9, 2629.
More
Information
Subjects: Oncology
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: 794
Revisions: 2 times (View History)
Update Date: 29 Mar 2022
1000/1000