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 + 1771 word(s) 1771 2021-04-20 08:37:26 |
2 format correct Meta information modification 1771 2021-04-28 10:50:56 |

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.
Marcucci, F. Immune Checkpoint Molecules. Encyclopedia. Available online: https://encyclopedia.pub/entry/9137 (accessed on 08 July 2024).
Marcucci F. Immune Checkpoint Molecules. Encyclopedia. Available at: https://encyclopedia.pub/entry/9137. Accessed July 08, 2024.
Marcucci, Fabrizio. "Immune Checkpoint Molecules" Encyclopedia, https://encyclopedia.pub/entry/9137 (accessed July 08, 2024).
Marcucci, F. (2021, April 28). Immune Checkpoint Molecules. In Encyclopedia. https://encyclopedia.pub/entry/9137
Marcucci, Fabrizio. "Immune Checkpoint Molecules." Encyclopedia. Web. 28 April, 2021.
Immune Checkpoint Molecules
Edit

Antibodies against inhibitory immune checkpoint molecules (ICPMs), referred to as immune checkpoint inhibitors (ICIs), have gained a prominent place in cancer therapy. Several ICIs in clinical use have been engineered to be devoid of effector functions because of the fear that ICIs with preserved effector functions could deplete immune cells, thereby curtailing antitumor immune responses. ICPM ligands (ICPMLs), however, are often overexpressed on a sizeable fraction of tumor cells of many tumor types and these tumor cells display an aggressive phenotype with changes typical of tumor cells undergoing an epithelial-mesenchymal transition. Moreover, immune cells expressing ICPMLs are often endowed with immunosuppressive or immune-deviated functionalities. Taken together, these observations suggest that compounds with the potential of depleting cells expressing ICPMLs may become useful tools for tumor therapy.

immune checkpoint epithelial-mesenchymal transition overexpression ADC bispecific CAR T cells effector functions oncolytic virus combination therapy

1. Mechanisms Underlying the Overexpression of ICPMLs on Tumor Cells

Overexpression of ICPMLs on tumor cells can be the result of different stimuli, either cell-autonomous stimuli or stimuli from the tumor microenvironment (TME). The mechanisms underlying the overexpression of ICPMLs on tumor cells have been most thoroughly investigated for PD-L1 and have been reviewed recently [1]. As regards tumor cell-autonomous stimuli, overexpression of PD-L1 can be the result of intrachromosomal or extrachromosomal events. Copy number alterations in chromosomal region 9p24.1 that encompasses the loci for PD-L1 and PD-L2, inversions, deletions, translocations, generation of chimeric fusion transcripts, and disruption or mutation of the 3′-untranslated region of the PD-L1 gene are intrachromosomal events that can lead to PD-L1 overexpression [2][3][4]. Tumor cell-autonomous, extrachromosomal events are receptor-activating mutations or receptor overexpression [5], gain-of-function or loss-of-function mutations affecting intracellular signaling molecules [6][7], activation or overexpression of transcription factors (e.g., hypoxia-inducible factor-α, signal transducer and activator of transcription (STAT) 3, MYC) [8][9][10]. More recently, also epigenetic mechanisms have been reported to induce or contribute to the overexpression of tumor cell-associated PD-L1 [11][12]. Tumor cell-exogenous stimuli that can lead to the overexpression of PD-L1 are cytokines (e.g., interferon (IFN)-γ, tumor necrosis factor (TNF)-α) [13][14] and various other stimuli from the TME like hypoxia or pseudohypoxia [10][15], antitumor drugs (chemotherapeutics, targeted therapeutics) [16] or metabolites (e.g., lactate) [17]. While the mechanisms leading to the overexpression of other tumor cell-associated ICPMLs have been much less investigated, they appear to be similar to those for PD-L1. Thus, hypoxia or pseudohypoxia lead to the overexpression of B7-H4 [18], CD70 [19], CD47 [20]. Antitumor drugs lead to the overexpression of CD70 and B7-H3 [21][22]. Activation of the Ras-Raf-MEK-extracellular signal-regulated kinase pathway leads to overexpression of CD155 and CD137 [23][24], Hedgehog signaling to overexpression of CD155 [24], the Janus kinase 2-STAT3 pathway to overexpression of fibrinogen-like protein 1 (FGL1) [25]. While the stimuli that induce overexpression of ICPMLs on tumor cells appear to be similar, in some instances subtle differences in the intracellular signaling pathways regulating the expression of two different ICPMLs have been observed [26], suggesting that these differences may explain the different patterns of expression that have been observed between different tumor cell-associated ICPMLs (see Section 3).

2. The Consequences of the Expression of ICPMLs on the Biology of Tumor Cells

In addition to transmitting signals to other cells (mostly immune cells) upon engagement of their cognate receptors [27][28][29][30] tumor cell-associated ICPMLs also exert cell-autonomous functions. Thus, their expression is associated with changes whereby tumor cells acquire enhanced capabilities to migrate, invade and metastasize to distant organs, undergo faster growth and metabolic alterations [31][32], acquire tumor-initiating potential [20][21][33] as well as resistance to antitumor drugs and apoptosis [1]. Collectively, these changes, when they are accompanied by the expression of specific transcription factors and molecular modifications [34] are referred to as tumor cell epithelial-mesenchymal transition (EMT) [35][36]. Indeed, the causal relationship between ICPML expression on tumor cells and EMT has been shown in several instances with a variety of technical approaches (e.g., siRNA, CRISPR/Cas) [37][38][39][40][41]. Expression of ICPMLs on tumor cells can be both a consequence [42], as well as a cause of tumor cell EMT [37][38][43][44][45], suggesting the existence of a positive feedback loop between the expression of ICPMLs and EMT [1]. Interestingly, tumor cell EMT can also have immunosuppressive effects [46] and it has recently been shown that loss of the epithelial marker E-cadherin, a hallmark of EMT, reduces responsiveness to ICIs in a mouse melanoma model [47].

As regards individual ICPMLs, the following have been reported to be associated with tumor cell EMT: PD-1 [48][49] PD-L1 [48][49][50][51], PD-L2 [48][49], B7-H3 [49], B7-H4 [37][52], CTLA-4 [48][49], OX40 [48], CD47 [53][54][55], CD137 ligand [56], CD155 [38], FGL1 [40], T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) [49][57] and B- and T-lymphocyte attenuator (BTLA, CD272) [49]. Other ICPMLs, while not having been formally associated with EMT (e.g., CD70, galectin-9), are expressed by tumor cells displaying EMT-related functionalities [58][59][60][61].

The data discussed so far suggest the existence of a close association between expression of ICPMLs on tumor cells and EMT and raise the question as to whether this association is absolute. In fact, data show that the association of an ICPML (PD-L1) and EMT on tumor cells is not coincident [33]. Moreover, as already mentioned, tumor cell-associated PD-L1 expression can be induced by intrachromosomal events. In these cases, PD-L1 overexpression is independent of tumor cell EMT [2][4][62], but it cannot be excluded that it may contribute to the induction of tumor cell EMT. The observation that genomic amplification targeting PD-L1 and PD-L2 is enriched in triple-negative breast cancer (TNBC), a cancer type with a predominantly mesenchymal phenotype suggests that this may, indeed, be the case [63].

Moreover, the lack of coincidence between ICPML expression and tumor cell EMT may also be the consequence of EMT plasticity, whereby tumor cells undergoing EMT cover a whole spectrum of phenotypes spanning from a fully epithelial to a fully mesenchymal one [64]. This suggests the possibility, for example, that an individual ICPML on tumor cells may be expressed at EMT initiation, when epithelial markers still predominate over mesenchymal markers. Moreover, heterogeneity of EMT marker expression is paralleled by the heterogeneity of ICPML expression [65]. Such heterogeneity applies both to individual ICPMLs, with ICPML-negative and -positive tumor cells coexisting within the same tumor [66], as well as to different ICPMLs showing non-overlapping or partially overlapping expression within the same tumor cell population. As regards the heterogeneous expression of different ICPMLs, it has been reported, for example, that a fraction of PD-L1-negative melanomas expressed high levels of CD155 and this was associated with a poor response to anti-PD-1/anti-CTLA4 therapy [67]. Moreover, expression of B7-H4 was prevalent among immune-cold TNBCs, and correlated inversely with that of PD-L1 [68][69]. In hepatocellular carcinoma tissues, FGL1 and PD-L1 had distinct distribution and relationships with each other [70]. Expression of Herpes virus entry mediator (HVEM) was found to be broader than that of PD-L1 on cells of melanoma metastases from 116 patients [71]. Moreover, in some situations, administration of an anti-ICPM antibody (anti-PD-1) has been shown to lead to the upregulation of an ICPML (TIM-3) [72].

3. Why Non-Depleting Antibodies Have Been Used against Inhibitory ICPMLs

Given the points discussed so far and, in particular, the close association between tumor cell expression of ICPML and an aggressive phenotype, it is somehow surprising to note that most of the ICIs against ICPMLs that are in current clinical use, have been selected so to be devoid of cell-depleting activity.

In fact, ICI antibodies of IgG1 isotype are able, in addition to inhibit the interaction with the cognate ICPM, to induce cytotoxic or phagocytic effects (antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), complement-dependent cytotoxicity (CDC)) on cells expressing the targeted antigen. As to currently used antibodies, the anti-PD-L1 mAb atezolizumab has an aglycosylated Fc region devoid of effector functions, and the anti-PD-L1 mAb durvalumab is of IgG1 isotype with three mutations in the Fc domain resulting in greatly reduced ADCC and CDC [73]. A notable exception to this picture is the anti-PD-L1 mAb avelumab, which will be discussed later. The reason as to why several clinically approved ICIs have been selected to be devoid of effector functions is due to the fact that ICPMLs can be expressed not only on tumor cells, but also on cells of the innate and adaptive immune system and that their depletion through ADCC, CDC or ADCP might lead to undesired immunosuppressive effects. In fact, taking a closer look to ICPML-expressing immune cells, one may reach the conclusion that their depletion may not be necessarily of harm, because in many instances such cells have immunosuppressive effects. In the following, we will briefly discuss this knowledge which has been obtained mainly for PD-L1.

Tumor-associated dendritic cells (DCs) upregulate PD-L1 in response to T-cell derived inflammatory cytokines like IFN-γ [74], while M1 macrophages do so in response to another inflammatory cytokine, interleukin (IL)-1β [75]. PD-L1+ DCs can lead to functional inactivation of T cells upon interaction with PD-1 [76]. Similarly, other PD-L1+ antigen-presenting cells like macrophages can induce anergy in T cells upon interaction with PD-1 [77], explaining why expression of PD-L1 on immune cells, rather than tumor cells, has been found in some studies to correlate with a favorable response to anti-PD-1 therapy [78]. Additionally, B7-H4 is expressed on immunosuppressive tumor-associated macrophages (TAM) [79]. Moreover, upon PD-1/PD-L1 interaction, macrophages can produce increased levels of immunosuppressive cytokines like IL-10, but reduced levels of inflammatory cytokines like IL-6 [77][80]. Additionally, tumor-infiltrating T cells can express PD-L1 upon activation and this PD-L1 is important for T-cell survival [81]. Ligation of T cell-associated PD-L1 can have immunosuppressive effects by promoting M2 polarization of macrophages, reducing the production of inflammatory cytokines and inducing an anergic phenotype or apoptosis in T-cells [82][83]. PD-L1 expression has also been documented on non-tumor cells of the TME that may play tumor-promoting and immunosuppressive roles like cancer-associated fibroblasts (CAF) [84]. Eventually, mice lacking CD155 on both tumor-infiltrating myeloid cells as well as tumor cells showed greater reduction of tumor growth and metastasis compared to mice lacking CD155 only on tumor cells [85]. Importantly, the immunosuppressive effects of ICPMLs may be context-dependent as has been shown for PD-L1, with tumor-associated PD-L1 playing a predominantly immunosuppressive role in some tumor types, and PD-L1 expressed on tumor-associated immune cells playing a predominantly immunosuppressive role in other tumor types [86][87]).

So far, we have listed several downsides related to ICPML expression on immune cells. There are, however, some observations suggesting that the expression of PD-L1 on immune cells may contribute to antitumor effects of the immune response. Thus, some tumors were shown to induce expression of PD-L1 on natural killer (NK) cells and this led to enhanced NK-cell function. These PD-L1-positive NK cells could be activated with an anti-PD-L1 antibody to perform increased antitumor activity [88]. Depletion of PD-L1-expressing NK cells led to the suppression of this antitumor mechanism. Still another possibility to be considered is that depletion of ICPML+ immunosuppressive cells triggers direct tumor-promoting effects of the immune system like those that may occur during hyperprogressive disease observed during ICI therapy [89][90][91].

References

  1. Marcucci, F.; Rumio, C.; Corti, A. Tumor cell-associated immune checkpoint molecules—Drivers of malignancy and stemness. Biochim. Biophys. Acta 2017, 1868, 571–583.
  2. George, J.; Saito, M.; Tsuta, K.; Iwakawa, R.; Shiraishi, K.; Scheel, A.H.; Uchida, S.; Watanabe, S.I.; Nishikawa, R.; Noguchi, M.; et al. Genomic amplification of CD274 (PD-L1) in small-cell lung cancer. Clin. Cancer Res. 2017, 23, 1220–1226.
  3. Ota, K.; Azuma, K.; Kawahara, A.; Hattori, S.; Iwama, E.; Tanizaki, J.; Harada, T.; Matsumoto, K.; Takayama, K.; Takamori, S.; et al. Induction of PD-L1 expression by the EML4-ALK oncoprotein and downstream signaling pathways in non-small cell lung cancer. Clin. Cancer Res. 2015, 21, 4014–4021.
  4. Kataoka, K.; Shiraishi, Y.; Takeda, Y.; Sakata, S.; Matsumoto, M.; Nagano, S.; Maeda, T.; Nagata, Y.; Kitanaka, A.; Mizuno, S.; et al. Aberrant PD-L1 expression through 3’-UTR disruption in multiple cancers. Nature 2016, 534, 402–406.
  5. Balan, M.; Mier y Teran, E.; Waaga-Gasser, A.M.; Gasser, M.; Choueiri, T.K.; Freeman, G.; Pal, S. Novel roles of c-Met in the survival of renal cancer cells through the regulation of HO-1 and PD-L1 expression. J. Biol. Chem. 2015, 290, 8110–8120.
  6. Parsa, A.T.; Waldron, J.S.; Panner, A.; Crane, C.A.; Parney, I.F.; Barry, J.J.; Cachola, K.E.; Murray, J.C.; Tihan, T.; Jensen, M.C.; et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat. Med. 2007, 13, 84–88.
  7. Lastwika, K.J.; Wilson, W., 3rd; Li, Q.K.; Norris, J.; Xu, H.; Ghazarian, S.R.; Kitagawa, H.; Kawabata, S.; Taube, J.M.; Yao, S.; et al. Control of PD-L1 expression by oncogenic activation of the AKT–mTOR pathway in non–small cell lung cancer. Cancer Res. 2016, 76, 227–238.
  8. Marzec, M.; Zhang, Q.; Goradia, A.; Raghunath, P.N.; Liu, X.; Paessler, M.; Wang, H.Y.; Wysocka, M.; Cheng, M.; Ruggeri, B.A.; et al. Oncogenic kinase NPM/ALK induces through STAT3 expression of immunosuppressive protein CD274 (PD-L1, B7-H1). Proc. Natl. Acad. Sci. USA 2008, 105, 20852–20857.
  9. Sun, Y.; Yu, M.; Qu, M.; Ma, Y.; Zheng, D.; Yue, Y.; Guo, S.; Tang, L.; Li, G.; Zheng, W.; et al. Hepatitis B virus-triggered PTEN/β-catenin/c-Myc signaling enhances PD-L1 expression to promote immune evasion. Am. J. Physiol. Gastrointest. Liver Physiol. 2020, 318, G162–G173.
  10. Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014, 211, 781–790.
  11. Xiong, W.; Deng, H.; Huang, C.; Zen, C.; Jian, C.; Ye, K.; Zhong, Z.; Zhao, X.; Zhu, L. MLL3 enhances the transcription of PD-L1 and regulates anti-tumor immunity. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 454–463.
  12. Huang, K.C.; Chiang, S.F.; Chen, W.T.; Chen, T.W.; Hu, C.H.; Yang, P.C.; Ke, T.W.; Chao, K.S.C. Decitabine augments chemotherapy-induced PD-L1 upregulation for PD-L1 blockade in colorectal cancer. Cancers 2020, 12, 462.
  13. 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.
  14. Lee, Y.; Shin, J.H.; Longmire, M.; Wang, H.; Kohrt, H.E.; Chang, H.Y.; Sunwoo, J.B. CD44+ cells in head and neck squamous cell carcinoma suppress T-cell-mediated immunity by selective constitutive and inducible expression of PD-L1. Clin. Cancer Res. 2016, 22, 3571–3581.
  15. Messai, Y.; Gad, S.; Noman, M.Z.; Le Teuff, G.; Couve, S.; Janji, B.; Kammerer, S.F.; Rioux-Leclerc, N.; Hasmim, M.; Ferlicot, S.; et al. Renal cell carcinoma programmed death-ligand 1, a new direct target of hypoxia-inducible factor-2α, is regulated by von Hippel-Lindau gene mutation status. Eur. Urol. 2016, 70, 623–632.
  16. Peng, J.; Hamanishi, J.; Matsumura, N.; Abiko, K.; Murat, K.; Baba, T.; Yamaguchi, K.; Horikawa, N.; Hosoe, Y.; Murphy, S.K.; et al. Chemotherapy induces programmed cell death-ligand 1 overexpression via the nuclear factor-κB to foster an immunosuppressive tumor microenvironment in ovarian cancer. Cancer Res. 2015, 75, 5034–5045.
  17. Feng, J.; Yang, H.; Zhang, Y.; Wei, H.; Zhu, Z.; Zhu, B.; Yang, M.; Cao, W.; Wang, L.; Wu, Z. Tumor cell-derived lactate induces TAZ-dependent upregulation of PD-L1 through GPR81 in human lung cancer cells. Oncogene 2017, 36, 5829–5839.
  18. Jeon, Y.K.; Park, S.G.; Choi, I.W.; Lee, S.W.; Lee, S.M.; Choi, I. Cancer cell-associated cytoplasmic B7-H4 is induced by hypoxia through hypoxia-inducible factor-1α and promotes cancer cell proliferation. Biochem. Biophys. Res. Commun. 2015, 459, 277–283.
  19. Ruf, M.; Mittmann, C.; Nowicka, A.M.; Hartmann, A.; Hermanns, T.; Poyet, C.; van den Broek, M.; Sulser, T.; Moch, H.; Schraml, P. pVHL/HIF-regulated CD70 expression is associated with infiltration of CD27+ lymphocytes and increased serum levels of soluble CD27 in clear cell renal cell carcinoma. Clin. Cancer Res. 2015, 21, 889–898.
  20. Zhang, H.; Lu, H.; Xiang, L.; Bullen, J.W.; Zhang, C.; Samanta, D.; Gilkes, D.M.; He, J.; Semenza, G.L. HIF-1 regulates CD47 expression in breast cancer cells to promote evasion of phagocytosis and maintenance of cancer stem cells. Proc. Natl. Acad. Sci. USA 2015, 112, E6215–E6223.
  21. Riether, C.; Schürch, C.M.; Flury, C.; Hinterbrandner, M.; Drück, L.; Huguenin, A.L.; Baerlocher, G.M.; Radpour, R.; Ochsenbein, A.F. Tyrosine kinase inhibitor-induced CD70 expression mediates drug resistance in leukemia stem cells by activating Wnt signaling. Sci. Transl. Med. 2015, 7, 298ra119.
  22. Li, H.; Huang, C.; Zhang, Z.; Feng, Y.; Wang, Z.; Tang, X.; Zhong, K.; Hu, Y.; Guo, G.; Zhou, L.; et al. MEK inhibitor augments antitumor activity of B7-H3-redirected bispecific antibody. Front. Oncol. 2020, 10, 1527.
  23. Glorieux, C.; Huang, P. Regulation of CD137 expression through K-Ras signaling in pancreatic cancer cells. Cancer Commun. 2019, 39, 41.
  24. Kučan Brlić, P.; Lenac Roviš, T.; Cinamon, G.; Tsukerman, P.; Mandelboim, O.; Jonjić, S. Targeting PVR (CD155) and its receptors in anti-tumor therapy. Cell. Mol. Immunol. 2019, 16, 40–52.
  25. Wang, J.; Wei, W.; Tang, Q.; Lu, L.; Luo, Z.; Li, W.; Lu, Y.; Pu, J. Oxysophocarpine suppresses hepatocellular carcinoma growth and sensitizes the therapeutic blockade of anti-Lag-3 via reducing FGL1 expression. Cancer Med. 2020, 9, 7125–7136.
  26. Li, R.; Zatloukalova, P.; Muller, P.; Gil-Mir, M.; Kote, S.; Wilkinson, S.; Kemp, A.J.; Hernychova, L.; Wang, Y.; Ball, K.L.; et al. The MDM2 ligand Nutlin-3 differentially alters expression of the immune blockade receptors PD-L1 and CD276. Cell. Mol. Biol. Lett. 2020, 25, 41.
  27. Janakiram, M.; Shah, U.A.; Liu, W.; Zhao, A.; Schoenberg, M.P.; Zang, X. The third group of the B7-CD28 immune checkpoint family: HHLA2, TMIGD2, B7x, and B7-H3. Immunol. Rev. 2017, 276, 26–39.
  28. Andrews, L.P.; Yano, H.; Vignali, D.A.A. Inhibitory receptors and ligands beyond PD-1, PD-L1 and CTLA-4: Breakthroughs or backups. Nat. Immunol. 2019, 20, 1425–1434.
  29. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008, 26, 677–704.
  30. Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–64.
  31. Ni, L.; Dong, C. New B7 family checkpoints in human cancers. Mol. Cancer Ther. 2017, 16, 1203–1211.
  32. Marcucci, F.; Rumio, C. Glycolysis-induced drug resistance in tumors-A response to danger signals? Neoplasia 2021, 23, 234–245.
  33. Zhi, Y.; Mou, Z.; Chen, J.; He, Y.; Dong, H.; Fu, X.; Wu, Y. B7H1 expression and epithelial-to-mesenchymal transition phenotypes on colorectal cancer stem-like cells. PLoS ONE 2015, 10, e0135528.
  34. Sanchez-Tillo, E.; Liu, Y.; De Barrios, O.; Siles, L.; Fanlo, L.; Cuatrecasas, M.; Darling, D.S.; Dean, D.C.; Castells, A.; Postigo, A. EMT-activating transcription factors in cancer: Beyond EMT and tumor invasiveness. Cell. Mol. Life Sci. 2012, 69, 3429–3456.
  35. Dongre, A.; Weinberg, R.A. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat. Rev. Mol. Cell Biol. 2019, 20, 69–84.
  36. Marcucci, F.; Stassi, G.; De Maria, R. Epithelial–mesenchymal transition: A new target in anticancer drug discovery. Nat. Rev. Drug Discov. 2016, 15, 311–325.
  37. Feng, Y.; Yang, Z.; Zhang, C.; Che, N.; Liu, X.; Xuan, Y. B7-H4 induces epithelial-mesenchymal transition and promotes colorectal cancer stemness. Pathol. Res. Pract. 2021, 218, 153323.
  38. Zheng, Q.; Gao, J.; Yin, P.; Wang, W.; Wang, B.; Li, Y.; Zhao, C. CD155 contributes to the mesenchymal phenotype of triple-negative breast cancer. Cancer Sci. 2020, 111, 383–394.
  39. Ge, Y.; Chen, W.; Zhang, X.; Wang, H.; Cui, J.; Liu, Y.; Ju, S.; Tian, X.; Ju, S. Nuclear-localized costimulatory molecule 4-1BBL promotes colon cancer cell proliferation and migration by regulating nuclear Gsk3β, and is linked to the poor outcomes associated with colon cancer. Cell Cycle 2020, 19, 577–591.
  40. Zhang, Y.; Qiao, H.X.; Zhou, Y.T.; Hong, L.; Chen, J.H. Fibrinogen-like-protein 1 promotes the invasion and metastasis of gastric cancer and is associated with poor prognosis. Mol. Med. Rep. 2018, 18, 1465–1472.
  41. Li, J.; Chen, L.; Xiong, Y.; Zheng, X.; Xie, Q.; Zhou, Q.; Shi, L.; Wu, C.; Jiang, J.; Wang, H. Knockdown of PD-L1 in human gastric cancer cells inhibits tumor progression and improves the cytotoxic sensitivity to CIK therapy. Cell. Physiol. Biochem. 2017, 41, 907–920.
  42. Noman, M.Z.; Janji, B.; Abdou, A.; Hasmim, M.; Terry, S.; Tan, T.Z.; Mami-Chouaib, F.; Thiery, J.P.; Chouaib, S. The immune checkpoint ligand PD-L1 is upregulated in EMT-activated human breast cancer cells by a mechanism involving ZEB-1 and miR-200. Oncoimmunology 2017, 6, e1263412.
  43. Jiang, B.; Zhang, T.; Liu, F.; Sun, Z.; Shi, H.; Hua, D.; Yang, C. The co-stimulatory molecule B7-H3 promotes the epithelial-mesenchymal transition in colorectal cancer. Oncotarget 2016, 7, 31755–31771.
  44. Shan, B.; Man, H.; Liu, J.; Wang, L.; Zhu, T.; Ma, M.; Xv, Z.; Chen, X.; Yang, X.; Li, P. TIM-3 promotes the metastasis of esophageal squamous cell carcinoma by targeting epithelial-mesenchymal transition via the Akt/GSK-3β/Snail signaling pathway. Oncol. Rep. 2016, 36, 1551–1561.
  45. Wang, Y.; Wang, H.; Zhao, Q.; Xia, Y.; Hu, X.; Guo, J. PD-L1 induces epithelial-to-mesenchymal transition via activating SREBP-1c in renal cell carcinoma. Med. Oncol. 2015, 32, 212.
  46. Romeo, E.; Caserta, C.A.; Rumio, C.; Marcucci, F. The vicious cross-talk between tumor cells with an EMT phenotype and cells of the immune system. Cells 2019, 8, 460.
  47. Shields, B.D.; Koss, B.; Taylor, E.M.; Storey, A.J.; West, K.L.; Byrum, S.D.; Mackintosh, S.G.; Edmondson, R.; Mahmoud, F.; Shalin, S.C.; et al. Loss of E-cadherin inhibits CD103 antitumor activity and reduces checkpoint blockade responsiveness in melanoma. Cancer Res. 2019, 79, 1113–1123.
  48. Mak, M.P.; Tong, P.; Diao, L.; Cardnell, R.J.; Gibbons, D.L.; William, W.N.; Skoulidis, F.; Parra, E.R.; Rodriguez-Canales, J.; Wistuba, I.I.; et al. A patient-derived, pan-cancer EMT signature identifies global molecular alterations and immune target enrichment following epithelial-to-mesenchymal transition. Clin. Cancer Res. 2016, 22, 609–620.
  49. Lou, Y.; Diao, L.; Cuentas, E.R.P.; Denning, W.L.; Chen, L.; Fan, Y.H.; Byers, L.A.; Wang, J.; Papadimitrakopoulou, V.A.; Behrens, C.; et al. Epithelial–mesenchymal transition is associated with a distinct tumor microenvironment including elevation of inflammatory signals and multiple immune checkpoints in lung adenocarcinoma. Clin. Cancer Res. 2016, 22, 3630–3642.
  50. Emran, A.A.; Chatterjee, A.; Rodger, E.J.; Tiffen, J.C.; Gallagher, S.J.; Eccles, M.R.; Hersey, P. Reactive oxygen species modulate macrophage immunosuppressive phenotype through the up-regulation of PD-L1. Proc. Natl. Acad. Sci. USA 2019, 116, 4326–4335.
  51. Ock, C.Y.; Kim, S.; Keam, B.; Kim, M.; Kim, T.M.; Kim, J.H.; Jeon, Y.K.; Lee, J.S.; Kwon, S.K.; Hah, J.H.; et al. PD-L1 expression is associated with epithelial-mesenchymal transition in head and neck squamous cell carcinoma. Oncotarget 2016, 7, 15901–15914.
  52. Xie, N.; Cai, J.B.; Zhang, L.; Zhang, P.F.; Shen, Y.H.; Yang, X.; Lu, J.C.; Gao, D.M.; Kang, Q.; Liu, L.X.; et al. Upregulation of B7-H4 promotes tumor progression of intrahepatic cholangiocarcinoma. Cell Death Dis. 2017, 8, 3205.
  53. Pai, S.; Bamodu, O.A.; Lin, Y.K.; Lin, C.S.; Chu, P.Y.; Chien, M.H.; Wang, L.S.; Hsiao, M.; Yeh, C.T.; Tsai, J.T. CD47-SIRPα signaling induces epithelial-mesenchymal transition and cancer stemness and links to a poor prognosis in patients with oral squamous cell carcinoma. Cells 2019, 8, 1658.
  54. Noman, M.Z.; Van Moer, K.; Marani, V.; Gemmill, R.M.; Tranchevent, L.C.; Azuaje, F.; Muller, A.; Chouaib, S.; Thiery, J.P.; Berchem, G.; et al. CD47 is a direct target of SNAI1 and ZEB1 and its blockade activates the phagocytosis of breast cancer cells undergoing EMT. Oncoimmunology 2018, 7, e1345415.
  55. Zhao, H.; Wang, J.; Kong, X.; Li, E.; Liu, Y.; Du, X.; Kang, Z.; Tang, Y.; Kuang, Y.; Yang, Z.; et al. CD47 promotes tumor invasion and metastasis in non-small cell lung cancer. Sci. Rep. 2016, 6, 29719.
  56. Grimmig, T.; Gasser, M.; Moench, R.; Zhu, L.J.; Nawalaniec, K.; Callies, S.; Wagner, M.; Polat, B.; Mothi, S.S.; Luo, Y.; et al. Expression of tumor-mediated CD137 ligand in human colon cancer indicates dual signaling effects. Oncoimmunology 2019, 8, e1651622.
  57. Xiao, Y.; Qing, J.; Li, B.; Chen, L.; Nong, S.; Yang, W.; Tang, X.; Chen, Z. TIM-3 participates in the invasion and metastasis of nasopharyngeal carcinoma via SMAD7/SMAD2/SNAIL1 axis-mediated epithelial-mesenchymal transition. OncoTargets Ther. 2020, 13, 1993–2006.
  58. Bertino, P.; Premeaux, T.A.; Fujita, T.; Haun, B.K.; Marciel, M.P.; Hoffmann, F.W.; Garcia, A.; Yiang, H.; Pastorino, S.; Carbone, M.; et al. Targeting the C-terminus of galectin-9 induces mesothelioma apoptosis and M2 macrophage depletion. Oncoimmunology 2019, 8, 1601482.
  59. Ge, H.; Mu, L.; Jin, L.; Yang, C.; Chang, Y.E.; Long, Y.; DeLeon, G.; Deleyrolle, L.; Mitchell, D.A.; Kubilis, P.S.; et al. Tumor associated CD70 expression is involved in promoting tumor migration and macrophage infiltration in GBM. Int. J. Cancer 2017, 141, 1434–1444.
  60. Pich, C.; Sarrabayrouse, G.; Teiti, I.; Mariamé, B.; Rochaix, P.; Lamant, L.; Favre, G.; Maisongrosse, V.; Tilkin-Mariamé, A.F. Melanoma-expressed CD70 is involved in invasion and metastasis. Br. J. Cancer 2016, 114, 63–70.
  61. Aggarwal, S.; He, T.; Fitzhugh, W.; Rosenthal, K.; Field, B.; Heidbrink, J.; Mesmer, D.; Ruben, S.M.; Moore, P.A. Immune modulator CD70 as a potential cisplatin resistance predictive marker in ovarian cancer. Gynecol. Oncol. 2009, 115, 430–437.
  62. Budczies, J.; Bockmayr, M.; Denkert, C.; Klauschen, F.; Gröschel, S.; Darb-Esfahani, S.; Pfarr, N.; Leichsenring, J.; Onozato, M.L.; Lennerz, J.K.; et al. Pan-cancer analysis of copy number changes in programmed death-ligand 1 (PD-L1, CD274)—Associations with gene expression, mutational load, and survival. Genes Chromosomes Cancer 2016, 55, 626–639.
  63. Barrett, M.T.; Anderson, K.S.; Lenkiewicz, E.; Andreozzi, M.; Cunliffe, H.E.; Klassen, C.L.; Dueck, A.C.; McCullough, A.E.; Reddy, S.K.; Ramanathan, R.K.; et al. Genomic amplification of 9p24.1 targeting JAK2, PD-L1, and PD-L2 is enriched in high-risk triple negative breast cancer. Oncotarget 2015, 6, 26483–26493.
  64. Pastushenko, I.; Brisebarre, A.; Sifrim, A.; Fioramonti, M.; Revenco, T.; Boumahdi, S.; Van Keymeulen, A.; Brown, D.; Moers, V.; Lemaire, S.; et al. Identification of the tumour transition states occurring during EMT. Nature 2018, 556, 463–468.
  65. Calderaro, J.; Rousseau, B.; Amaddeo, G.; Mercey, M.; Charpy, C.; Costentin, C.; Luciani, A.; Zafrani, E.S.; Laurent, A.; Azoulay, D.; et al. Programmed death ligand 1 expression in hepatocellular carcinoma: Relationship with clinical and pathological features. Hepatology 2016, 64, 2038–2046.
  66. Madore, J.; Vilain, R.E.; Menzies, A.M.; Kakavand, H.; Wilmott, J.S.; Hyman, J.; Yearley, J.H.; Kefford, R.F.; Thompson, J.F.; Long, G.V.; et al. PD-L1 expression in melanoma shows marked heterogeneity within and between patients: Implications for anti-PD-1/PD-L1 clinical trials. Pigment. Cell Melanoma Res. 2015, 28, 245–253.
  67. Lepletier, A.; Madore, J.; O’Donnell, J.S.; Johnston, R.L.; Li, X.Y.; McDonald, E.; Ahern, E.; Kuchel, A.; Eastgate, M.; Pearson, S.A.; et al. Tumor CD155 expression is associated with resistance to anti-PD1 immunotherapy in metastatic melanoma. Clin. Cancer Res. 2020, 26, 3671–3681.
  68. Song, X.; Zhou, Z.; Li, H.; Xue, Y.; Lu, X.; Bahar, I.; Kepp, O.; Hung, M.C.; Kroemer, G.; Wan, Y. Pharmacologic suppression of B7-H4 glycosylation restores antitumor immunity in immune-cold breast cancers. Cancer Discov. 2020, 10, 1872–1893.
  69. Cheng, H.; Borczuk, A.; Janakiram, M.; Ren, X.; Lin, J.; Assal, A.; Halmos, B.; Perez-Soler, R.; Zang, X. Wide expression and significance of alternative immune checkpoint molecules, B7x and HHLA2, in PD-L1–negative human lung cancers. Clin. Cancer Res. 2018, 24, 1954–1964.
  70. Guo, M.; Yuan, F.; Qi, F.; Sun, J.; Rao, Q.; Zhao, Z.; Huang, P.; Fang, T.; Yang, B.; Xia, J. Expression and clinical significance of LAG-3, FGL1, PD-L1 and CD8 + T cells in hepatocellular carcinoma using multiplex quantitative analysis. J. Transl. Med. 2020, 18, 306.
  71. Malissen, N.; Macagno, N.; Granjeaud, S.; Granier, C.; Moutardier, V.; Gaudy-Marqueste, C.; Habel, N.; Mandavit, M.; Guillot, B.; Pasero, C.; et al. HVEM has a broader expression than PD-L1 and constitutes a negative prognostic marker and potential treatment target for melanoma. Oncoimmunology 2019, 8, e1665976.
  72. Koyama, S.; Akbay, E.A.; Li, Y.Y.; Herter-Sprie, G.S.; Buczkowski, K.A.; Richards, W.G.; Gandhi, L.; Redig, A.J.; Rodig, S.J.; Asahina, H.; et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat. Commun. 2016, 7, 10501.
  73. Stewart, R.; Morrow, M.; Hammond, S.A.; Mulgrew, K.; Marcus, D.; Poon, E.; Watkins, A.; Mullins, S.; Chodorge, M.; Andrews, J.; et al. Identification and characterization of MEDI4736, an antagonistic anti-PD-L1 monoclonal antibody. Cancer Immunol. Res. 2015, 3, 1052–1062.
  74. Casella, G.; Rasouli, J.; Thome, R.; Descamps, H.C.; Vattikonda, A.; Ishikawa, L.; Boehm, A.; Hwang, D.; Zhang, W.; Xiao, D.; et al. Interferon-γ/Interleukin-27 axis induces programmed death ligand 1 expression in monocyte-derived dendritic cells and restores immune tolerance in central nervous system autoimmunity. Front. Immunol. 2020, 11, 576752.
  75. Zong, Z.; Zou, J.; Mao, R.; Ma, C.; Li, N.; Wang, J.; Wang, X.; Zhou, H.; Zhang, L.; Shi, Y. M1 macrophages induce PD-L1 expression in hepatocellular carcinoma cells through IL-1β signaling. Front. Immunol. 2019, 10, 1643.
  76. Shklovskaya, E.; Rizos, H. spatial and temporal changes in PD-L1 expression in cancer: The role of genetic drivers, tumor microenvironment and resistance to therapy. Int. J. Mol. Sci. 2020, 21, 7139.
  77. Lee, Y.; Moon, Y.; Hyung, K.; Yoo, J. Macrophage PD-L1 strikes back: PD-1/PDL1 interaction drives macrophages toward regulatory subsets. Adv. Biosci. 2013, 4, 19–29.
  78. Herbst, R.S.; Soria, J.C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.; Gettinger, S.N.; et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014, 515, 563–567.
  79. Kryczek, I.; Zou, L.; Rodriguez, P.; Zhu, G.; Wei, S.; Mottram, P.; Brumlik, M.; Cheng, P.; Curiel, T.; Myers, L.; et al. B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma. J. Exp. Med. 2006, 203, 871–881.
  80. Hao, N.B.; Lü, M.H.; Fan, Y.H.; Cao, Y.L.; Zhang, Z.R.; Yang, S.M. Macrophages in tumor microenvironments and the progression of tumors. Clin. Dev. Immunol. 2012, 2012, 948098.
  81. Pulko, V.; Harris, K.J.; Liu, X.; Gibbons, R.M.; Harrington, S.M.; Krco, C.J.; Kwon, E.D.; Dong, H. B7-h1 expressed by activated CD8 T cells is essential for their survival. J. Immunol. 2011, 187, 5606–5614.
  82. Liu, X.; Wu, X.; Cao, S.; Harrington, S.M.; Yin, P.; Mansfield, A.S.; Dong, H. B7-H1 antibodies lose antitumor activity due to activation of p38 MAPK that leads to apoptosis of tumor-reactive CD8(+) T cells. Sci. Rep. 2016, 6, 36722.
  83. Diskin, B.; Adam, S.; Cassini, M.F.; Sanchez, G.; Liria, M.; Aykut, B.; Buttar, C.; Li, E.; Sundberg, B.; Salas, R.D.; et al. PD-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer. Nat. Immunol. 2020, 21, 442–454.
  84. Teramoto, K.; Igarashi, T.; Kataoka, Y.; Ishida, M.; Hanaoka, J.; Sumimoto, H.; Daigo, Y. Clinical significance of PD-L1-positive cancer-associated fibroblasts in pN0M0 non-small cell lung cancer. Lung Cancer 2019, 137, 56–63.
  85. Li, X.Y.; Das, I.; Lepletier, A.; Addala, V.; Bald, T.; Stannard, K.; Barkauskas, D.; Liu, J.; Aguilera, A.R.; Takeda, K.; et al. CD155 loss enhances tumor suppression via combined host and tumor-intrinsic mechanisms. J. Clin. Investig. 2018, 128, 2613–2625.
  86. Juneja, V.R.; McGuire, K.A.; Manguso, R.T.; LaFleur, M.W.; Collins, N.; Haining, W.N.; Freeman, G.J.; Sharpe, A.H. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J. Exp. Med. 2017, 214, 895–904.
  87. Kleinovink, J.W.; van Hall, T.; Ossendorp, F.; Fransen, M.F. PD-L1 immune suppression in cancer: Tumor cells or host cells? Oncoimmunology 2017, 6, e1294299.
  88. Dong, W.; Wu, X.; Ma, S.; Wang, Y.; Nalin, A.P.; Zhu, Z.; Zhang, J.; Benson, D.M.; He, K.; Caligiuri, M.A.; et al. The mechanism of anti-PD-L1 antibody efficacy against PD-L1-negative tumors identifies NK cells expressing PD-L1 as a cytolytic effector. Cancer Discov. 2019, 9, 1422–1437.
  89. Champiat, S.; Dercle, L.; Ammari, S.; Massard, C.; Hollebecque, A.; Postel-Vinay, S.; Chaput, N.; Eggermont, A.; Marabelle, A.; Soria, J.C.; et al. Hyperprogressive disease is a new pattern of progression in cancer patients treated by anti-PD-1/PDL1. Clin. Cancer Res. 2017, 23, 1920–1928.
  90. Ferrara, R.; Mezquita, L.; Texier, M.; Lahmar, J.; Audigier-Valette, C.; Tessonnier, L.; Mazieres, J.; Zalcman, G.; Brosseau, S.; Le Moulec, S.; et al. Hyperprogressive disease in patients with advanced non-small cell lung cancer treated with PD-1/PD-L1 inhibitors or with single-agent chemotherapy. JAMA Oncol. 2018, 4, 1543–1552.
  91. Marcucci, F.; Rumio, C. The tumor-promoting effects of the adaptive immune system: A cause of hyperprogressive disease in cancer? Cell. Mol. Life Sci. 2021, 78, s00018–s00020.
More
Information
Subjects: Cell Biology
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: 390
Revisions: 2 times (View History)
Update Date: 28 Apr 2021
1000/1000
Video Production Service