You're using an outdated browser. Please upgrade to a modern browser for the best experience.
Submitted Successfully!
Thank you for your contribution! You can also upload a video entry or images related to this topic. For video creation, please contact our Academic Video Service.
Version Summary Created by Modification Content Size Created at Operation
1 Nathan Karin + 1680 word(s) 1680 2021-12-20 10:43:54 |
2 format corrected. Beatrix Zheng Meta information modification 1680 2022-01-06 04:24:23 |

Video Upload Options

We provide professional Academic Video Service to translate complex research into visually appealing presentations. Would you like to try it?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Karin, N. Chemokines in the Landscape of Cancer Immunotherapy. Encyclopedia. Available online: https://encyclopedia.pub/entry/17759 (accessed on 24 December 2025).
Karin N. Chemokines in the Landscape of Cancer Immunotherapy. Encyclopedia. Available at: https://encyclopedia.pub/entry/17759. Accessed December 24, 2025.
Karin, Nathan. "Chemokines in the Landscape of Cancer Immunotherapy" Encyclopedia, https://encyclopedia.pub/entry/17759 (accessed December 24, 2025).
Karin, N. (2022, January 05). Chemokines in the Landscape of Cancer Immunotherapy. In Encyclopedia. https://encyclopedia.pub/entry/17759
Karin, Nathan. "Chemokines in the Landscape of Cancer Immunotherapy." Encyclopedia. Web. 05 January, 2022.
Chemokines in the Landscape of Cancer Immunotherapy
Edit

“Hot” tumors are those that show signs of inflammation, meaning they have been invaded by effector T cells rushing to fight the cancerous cells. Evidence suggests that the limited success of ICI-based immunotherapies is related to attempts to treat patients with “cold tumors” that either do not contain effector T cells or in which these cells are markedly suppressed by regulatory T cells (Tregs). Chemokines are a well-defined group of proteins with chemotactic properties. We focus on key chemokines that not only attract leukocytes to tumor sites but also shape their biological properties. We propose using stabilized forms of two of them: CXL9 and CXCL10, to enhance anti-tumor immunity and possibly transform cold tumors into hot ones. Additionally, we discuss the possibility of targeting or deleting a key subset of Tregs that are CCR8+ Tregs and are highly dominant at the tumor site of several cold tumors. This may convert these cold tumors into hot tumors, and thus extend the success of immunotherapy beyond its current limits. 

chemokines chemokine receptors cancer immunotherapy CXCL9 CXCL10 CCR8 CCL1 regulatory T cells immune checkpoint inhibitors

1. Introduction

Chemokines are small proteins that have mostly been associated with directing leukocyte migration, and in affecting the dynamics of cancer, inflammation, and immune regulation [1][2][3]. As for cancer, many chemokines are produced by cancer cells that also possess their receptors [4][5]. So far, sixteen out of nineteen human chemokine receptors have been detected in cancer cells [6]. Key examples are CXCR4, CXCR1/2, CCR2, CXCR3, CCR5, and their ligands [1]. All became targets for cancer therapy [1][4][5][7][8][9]. The traditional view has been that chemokines mostly support tumor growth and survival either by a direct effect on tumor cells that possess their receptors [5] or by indirect mechanisms [5][10][11][12][13][14]. These indirect mechanisms mostly include interactions with their receptors on endothelial cells within the tumor microenvironment (TME), to induce growth factors production, and also in attracting bone marrow (BM)-derived cells to the tumor site. These cells then assist tumor growth and suppress the activities of anti-tumor effector T cells that limit tumor growth [5][10][11][12][13][14]. The major BM-derived cells that are known to support tumor growth and suppress anti-tumor immune reactivity are tumor-associated macrophages (TAMS), myeloid-derived suppressor cells (MDSC), neutrophilic cells, and regulatory T cells (Tregs). All of them suppress anti-tumor immune reactivity, and some of them directly support tumor growth [5][10][11][12][13][14]. Altogether, it implies that chemokines and their receptors are valid targets for cancer therapy [15]. Yet, thus far attempts to block many of these chemokines or their receptors showed limited success in human cancers. A possible mechanism of tumor escape may involve the rapid selection of resistant tumor cells [4]. The other possible explanation could be redundancy between chemokines [16][17].
The breakthrough of using monoclonal antibodies to immune checkpoint inhibitors (ICI) (also referred to as immune checkpoint blockers, ICB) opened new therapeutic opportunities [18][19][20][21][22][23][24][25][26]. The first successful approach of ICI has been the use of anti-cytotoxic T lymphocyte antigen 4 (CTLA-4) inhibitors in metastatic melanoma [25][27][28][29][30], and continuing with blocking the interactions between program cell death 1 (PD-1) and its ligands: program cell death ligand 1 (PDL-1) and program cell death ligand 2 (PDL-2) [25][27][28][29][30]. These blockers have been approved for about 20 different indications [23][26][31][32][33][34][35][36]. As a part of their mechanism of action, these ICIs enhanced the activity of tumor-specific effector CD4+ and CD8+ T cells [31][32][34][37]. Yet, immune checkpoint therapies (ICT) for many cancer diseases still show limited success [21][31][38][39][40][41]. Moreover, even in diseases with a significant positive response to ICI a relatively high number of patients are poor responders, and/or develop severe immune-related toxicities. This led to intensive research in two complementary avenues. The first focuses on developing tools for personalized-based medicine enabling to predict success on a personalized basis and excludes patients that following therapy have a high risk of developing immune-related toxicities [42][43][44][45][46][47][48]. The other avenue is spending efforts on developing new immunotherapeutic tools that would be used, either alone, or in combination with “conventional” ICI, and extend their therapeutic landscape.
It is believed that one of the major reasons for which the success of ICI is limited is that therapy is applied on diseases that either lack infiltration of effector CD8+ T cells or include massive accumulation of Tregs that suppress their activities [26][31][32][33][34][35][36]. These tumors are known as “cold” tumors [49][50][51]. Turning “cold tumors” into “hot tumors” by enhancing the activity of tumor-specific infiltrating effector T cells may extend the relative number of responders to ICI [49][50][51][52]. Likewise, in tumors enriched with Tregs, it is likely that blocking their activity or depleting these cells from the TME would turn cold tumors into hot.

2. Regulatory T Cells in Cancer Diseases, and Chemokine Receptor-Based Selective Depletion of These Cells for Cancer Immunotherapy

Maintenance of immunological self-tolerance by suppressing self-reacting T cells, as well as restraining the activities of effector T cells in response to infectious stimuli, thus, limiting chronic inflammatory conditions, is largely regulated by CD4+ regulatory T cells; [53][54]. These cells fall into two major subsets: those that express the transcription factor forkhead box P3 (FOXp3), also known as regulatory T cells (Tregs), and those that are FOXp3-negative but produce high levels of IL-10, also known as T regulatory -1 cells (Tr1) [53][54][55]. Those that are FOXp3+ commonly do not express the IL-7 alpha chain CD127, which is essential for IL-7 signaling required for converting T cells into memory cells [56][57][58]. These cells are of major interest for their key role in regulating cancer disease, mostly in suppressing the anti-cancer reactivity of effector T cells [59]. There are three major approaches for inhibiting Tregs and their ability to limit anticancer effector T cells: 1. Blocking the migration and accumulation of Tregs at the tumor site. 2. Inhibiting their suppressive activities within the tumor site and 3. Depletion of Tregs within the tumor site. Of these approaches, depleting Tregs is likely to be the most dramatic and possibly effective way. Yet systemic depletion of Tregs may result in major impairment of immune regulation. For example, a loss-of-function mutation in the gene encoding FOXp3 leads to a very severe autoimmune syndrome in humans named immune deficiency poly-endocrinopathy enteropathy X-linked (IPEX) syndrome [60].
Chemokines and chemokine receptors are thought to be involved in the selective migration of Tregs to the tumor site, and also in their potentiation within this site. Tregs express several chemokine receptors among them: CCR8, CCR4, CXCR3, CCR2, CCR6, and CCR5 [61]. Among these receptors, the CCR4-CCL22/CCL17 and the CCR8-CCL1 axis have been of major interest for both selective migrations of Tregs to tumor sites and their potentiation there. Moreover, their selective accumulation within the tumor site may suggest that selective depletion of CCR4+ or CCR8+ Tregs may enhance anti-cancer immunity while having a very limited effect on Tregs in the periphery. This subject is further discussed below.

2.1. CCR4+ Tregs

CCR4 is a chemokine receptor with two ligands CCL22 and CCL17. Both ligands but mostly CCL22 are largely involved in directing the recruitment and induction of suppressive function of Tregs at the tumor site [62][63][64][65][66][67][68][69][70][71]. This includes breast cancer, cervical cancer, glioblastoma, squamous cell carcinoma (SCC) colorectal cancer (CRC), and Pancreatic ductal adenocarcinoma (PDAC) [62][63][64][65][66][67][68][69][70][71]. Aside from Tregs, CCR4 is present in other leukocytes, among them CD4+ Th2 cells, NK cells, and macrophages [70][72][73][74]. It is also abundant on cancer cells, among them breast cancer [67]. Olkhanud et al. used a highly metastatic breast cancer (4T1) model in which CCR4 is largely expressed on cancer cells and Tregs, and demonstrated the pivotal role of CCR4 in recruiting and inducing NK cells and Tregs to limit tumor development and metastatic spread [71]. This does not exclude the possibility that targeting CCR4 would be more effective in several cancer diseases in which cancer cells are also CCR4+, among them breast cancer. In human cancers, major target diseases are several solid tumors, B-cell lymphomas, T-cells lymphomas, and leukemia in which not only CCR4 is highly expressed within the tumor microenvironment by Tregs, NK cells, and tumor cells, but mostly in those that poor prognosis has been associated with high expression of CCR4 on these cells [66][67][75][76][77][78]. Currently, there are two small chemical class II antagonists produced by Astra-Zeneca that block Tregs recruitment (AZD-2098, Marketed, and AZD-1678 in preclinical studies), a small chemical class II antagonist that blocks the interaction between CCL22 and CCR4 (FLX-475 produced by FLx-Bio) in phase 1/2 clinical trials as monotherapy or in combination with anti-PD-1 (Merck), a humanized mAb (KW-0761) capable of inducing ADCC to CCR4+ cells in phase 1a monotherapy for solid tumors, in combination with anti-PD-1 (Merck) for B cell lymphoma (phase 1/2), in combination with anti-PD-L1 or anti-CTLA-4 (Astra-Zeneca) in Phase 1b for solid tumors, and combination with anti-PD-1 (BMS) for solid tumors (very recently reviewed in [61]).

2.2. CCR8+ Tregs

CCR8 is a chemokine receptor mostly, but not exclusively, expressed by FOXp3+ Tregs [72][79][80][81][82][83]. Human CCR8 has four known ligands: CCL1, CCL8, CCL16, and CCL18 [84], whereas in murine only 3 of them are expressed: CCL1, CCL8, and CCL16 [85][86][87]. In both humans and mice, CCR8 is the only known receptor for CCL1 [80], whereas the other CCR8 ligands bind several chemokine receptors, as well as decoy receptors [85][86][87]. Four years ago, we identified CCR8+ Tregs as master drivers of the immune regulation [88]. In this study, we observed that the relative number of CCR8+ Tregs that is very low in the periphery increases along with the development of experimental autoimmune encephalomyelitis (EAE), a T cell-mediated autoimmune disease of the central nervous system (CNS). This study also observed that within the CNS CCR8+ Tregs are potentiated by CCL1, possibly in an autocrine manner, which makes them “driver” regulatory cells that restrain the progression of the disease [88]. Independently, Plitas et al. showed that in several human tumors, particularly “cold tumors” such as breast cancer, that these cells are highly dominant [89]. Along with this, recently it has been reported that anti-CCR8 mAb could be used to limit cancer growth in several cancer models [90][91][92]. One of the major reasons for which the success of ICI is limited is that therapy is applied on diseases that are designated as “cold tumors” that either lack infiltration of effector CD8+ T cells, or include massive accumulation of Tregs that suppress their activities [49][50][93][94][95][96]. Anti CCR8 mAb, mostly depleting antibodies, are currently under preclinical development by several lead companies.

References

  1. Chow, M.T.; Luster, A.D. Chemokines in Cancer. Cancer Immunol. Res. 2014, 2, 1125–1131.
  2. Luster, A.D. Chemokines—Chemotactic Cytokines That Mediate Inflammation. N. Engl. J. Med. 1998, 338, 436–445.
  3. Sokol, C.L.; Luster, A.D. The Chemokine System in Innate Immunity. Cold Spring Harb. Perspect. Biol. 2015, 7, a016303.
  4. Murphy, P.M. Chemokines and the Molecular Basis of Cancer Metastasis. N. Engl. J. Med. 2001, 345, 833–835.
  5. Zlotnik, A.; Burkhardt, A.M.; Homey, B. Homeostatic chemokine receptors and organ-specific metastasis. Nat. Rev. Immunol. 2011, 11, 597–606.
  6. Balkwill, F. Cancer and the chemokine network. Nat. Rev. Cancer. 2004, 4, 540–550.
  7. Poeta, V.M.; Massara, M.; Capucetti, A.; Bonecchi, R. Chemokines and Chemokine Receptors: New Targets for Cancer Immunotherapy. Front. Immunol. 2019, 10, 379.
  8. Bule, P.; Aguiar, S.I.; Aires-Da-Silva, F.; Dias, J.N.R. Chemokine-Directed Tumor Microenvironment Modulation in Cancer Immunotherapy. Int. J. Mol. Sci. 2021, 22, 9804.
  9. Zhou, W.; Guo, S.; Liu, M.; Burow, M.E.; Wang, G. Targeting CXCL12/CXCR4 Axis in Tumor Immunotherapy. Curr. Med. Chem. 2019, 26, 3026–3041.
  10. Giuliano, S.; Guyot, M.; Grépin, R.; Pagès, G. The ELR+CXCL chemokines and their receptors CXCR1/CXCR2: A signaling axis and new target for the treatment of renal cell carcinoma. OncoImmunology 2014, 3, e28399.
  11. Biasci, D.; Smoragiewicz, M.; Connell, C.M.; Wang, Z.; Gao, Y.; Thaventhiran, J.E.D.; Basu, B.; Magiera, L.; Johnson, T.I.; Bax, L.; et al. CXCR4 inhibition in human pancreatic and colorectal cancers induces an integrated immune response. Proc. Natl. Acad. Sci. USA 2020, 117, 28960–28970.
  12. Epstein, R.J. The CXCL12–CXCR4 chemotactic pathway as a target of adjuvant breast cancer therapies. Nat. Rev. Cancer 2004, 4, 901–909.
  13. Staller, P.; Sulitkova, J.; Lisztwan, J.; Moch, H.; Oakeley, E.J.; Krek, W. Chemokine receptor CXCR4 downregulated by von Hippel–Lindau tumour suppressor pVHL. Nature 2003, 425, 307–311.
  14. Schall, T.J.; Proudfoot, A.E.I. Overcoming hurdles in developing successful drugs targeting chemokine receptors. Nat. Rev. Immunol. 2011, 11, 355–363.
  15. Nagarsheth, N.; Wicha, M.S.; Zou, W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol. 2017, 17, 559–572.
  16. Dyer, D.P.; Medina-Ruiz, L.; Bartolini, R.; Schuette, F.; Hughes, C.E.; Pallas, K.; Vidler, F.; Macleod, M.K.L.; Kelly, C.J.; Lee, K.M.; et al. Chemokine Receptor Redundancy and Specificity Are Context Dependent. Immunity 2019, 50, 378–389.e5.
  17. Mantovani, A. The chemokine system: Redundancy for robust outputs. Immunol. Today 1999, 20, 254–257.
  18. Noguchi, E.; Shien, T.; Iwata, H. Current status of PD-1/PD-L1 blockade immunotherapy in breast cancer. Jpn. J. Clin. Oncol. 2021, 51, 321–332.
  19. Majidpoor, J.; Mortezaee, K. The efficacy of PD-1/PD-L1 blockade in cold cancers and future perspectives. Clin. Immunol. 2021, 226, 108707.
  20. Versluis, J.M.; Long, G.; Blank, C.U. Learning from clinical trials of neoadjuvant checkpoint blockade. Nat. Med. 2020, 26, 475–484.
  21. de Miguel, M.; Calvo, E. Clinical Challenges of Immune Checkpoint Inhibitors. Cancer Cell 2020, 38, 326–333.
  22. Robert, C.; Lanoy, E.; Besse, B. One or Two Immune Checkpoint Inhibitors? Cancer Cell 2019, 36, 579–581.
  23. Sun, C.; Mezzadra, R.; Schumacher, T.N. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018, 48, 434–452.
  24. Jerby-Arnon, L.; Shah, P.; Cuoco, M.; Rodman, C.; Su, M.-J.; Melms, J.; Leeson, R.; Kanodia, A.; Mei, S.; Lin, J.-R.; et al. A Cancer Cell Program Promotes T Cell Exclusion and Resistance to Checkpoint Blockade. Cell 2018, 175, 984–997.e24.
  25. Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61.
  26. Borghaei, H.; Paz-Ares, L.; Horn, L.; Spigel, D.R.; Steins, M.; Ready, N.E.; Chow, L.Q.; Vokes, E.E.; Felip, E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 1627–1639.
  27. Pfister, D.; Núñez, N.G.; Pinyol, R.; Govaere, O.; Pinter, M.; Szydlowska, M.; Gupta, R.; Qiu, M.; Deczkowska, A.; Weiner, A.; et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nat. Cell Biol. 2021, 592, 450–456.
  28. Sharma, P.; Allison, J.P. Dissecting the mechanisms of immune checkpoint therapy. Nat. Rev. Immunol. 2020, 20, 75–76.
  29. Sharma, P.; Allison, J.P. Immune Checkpoint Targeting in Cancer Therapy: Toward Combination Strategies with Curative Potential. Cell 2015, 161, 205–214.
  30. Quezada, S.; Simpson, T.R.; Peggs, K.S.; Merghoub, T.; Vider, J.; Fan, X.; Blasberg, R.; Yagita, H.; Muranski, P.; Antony, P.A.; et al. Tumor-reactive CD4+ T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts. J. Exp. Med. 2010, 207, 637–650.
  31. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355.
  32. 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.
  33. Nishino, M.; Sholl, L.M.; Hatabu, H.; Ramaiya, N.H.; Hodi, F.S. Anti–PD-1–Related Pneumonitis during Cancer Immunotherapy. N. Engl. J. Med. 2015, 373, 288–290.
  34. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 2015, 373, 23–34.
  35. Kleffel, S.; Posch, C.; Barthel, S.R.; Mueller, H.; Schlapbach, C.; Guenova, E.; Elco, C.P.; Lee, N.; Juneja, V.R.; Zhan, Q.; et al. Melanoma Cell-Intrinsic PD-1 Receptor Functions Promote Tumor Growth. Cell 2015, 162, 1242–1256.
  36. Callahan, M.K.; Postow, M.A.; Wolchok, J.D. CTLA-4 and PD-1 Pathway Blockade: Combinations in the Clinic. Front. Oncol. 2015, 4, 385.
  37. Topalian, S.L.; Drake, C.G.; Pardoll, D.M. Immune Checkpoint Blockade: A Common Denominator Approach to Cancer Therapy. Cancer Cell 2015, 27, 450–461.
  38. Janjigian, Y.Y.; Wolchok, J.D.; Ariyan, C.E. Eradicating micrometastases with immune checkpoint blockade: Strike while the iron is hot. Cancer Cell 2021, 39, 738–742.
  39. Koh, S.-B.; Ellisen, L.W. Immune activation and evolution through chemotherapy plus checkpoint blockade in triple-negative breast cancer. Cancer Cell 2021, 39, 1562–1564.
  40. Morad, G.; Helmink, B.A.; Sharma, P.; Wargo, J.A. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell 2021, 184, 5309–5337.
  41. Pitt, J.M.; Vétizou, M.; Daillère, R.; Roberti, M.P.; Yamazaki, T.; Routy, B.; Lepage, P.; Boneca, I.G.; Chamaillard, M.; Kroemer, G.; et al. Resistance Mechanisms to Immune-Checkpoint Blockade in Cancer: Tumor-Intrinsic and -Extrinsic Factors. Immunity 2016, 44, 1255–1269.
  42. Toribio-Vázquez, C.; Rivas, J.G.; Yebes, A.; Carrión, D.M.; Quesada-Olarte, J.; Trelles, C.R.; Álvarez-Maestro, M.; Van Der Poel, H.; Martínez-Piñeiro, L. Immunotherapy toxicity. Diagnosis and treatment. Diagn. Treat. Arch. Esp Urol. 2020, 73, 906–917.
  43. Pirozzi, F.; Poto, R.; Aran, L.; Cuomo, A.; Galdiero, M.R.; Spadaro, G.; Abete, P.; Bonaduce, D.; Marone, G.; Tocchetti, C.G.; et al. Cardiovascular Toxicity of Immune Checkpoint Inhibitors: Clinical Risk Factors. Curr. Oncol. Rep. 2021, 23, 13.
  44. Fan, Y.; Xie, W.; Huang, H.; Wang, Y.; Li, G.; Geng, Y.; Hao, Y.; Zhang, Z. Association of Immune Related Adverse Events with Efficacy of Immune Checkpoint Inhibitors and Overall Survival in Cancers: A Systemic Review and Meta-analysis. Front. Oncol. 2021, 11, 633032.
  45. Das, R.; Bar, N.; Ferreira, M.; Newman, A.; Zhang, L.; Bailur, J.K.; Bacchiocchi, A.; Kluger, H.; Wei, W.; Halaban, R.; et al. Early B cell changes predict autoimmunity following combination immune checkpoint blockade. J. Clin. Investig. 2018, 128, 715–720.
  46. Dougan, M.; Pietropaolo, M. Time to dissect the autoimmune etiology of cancer antibody immunotherapy. J. Clin. Investig. 2020, 130, 51–61.
  47. Hwang, W.L.; Pike, L.R.G.; Royce, T.J.; Mahal, B.; Loeffler, J.S. Safety of combining radiotherapy with immune-checkpoint inhibition. Nat. Rev. Clin. Oncol. 2018, 15, 477–494.
  48. Siwicki, M.; Gort-Freitas, N.A.; Messemaker, M.; Bill, R.; Gungabeesoon, J.; Engblom, C.; Zilionis, R.; Garris, C.; Gerhard, G.M.; Kohl, A.; et al. Resident Kupffer cells and neutrophils drive liver toxicity in cancer immunotherapy. Sci. Immunol. 2021, 6, eabi7083.
  49. Haanen, J.B. Converting Cold into Hot Tumors by Combining Immunotherapies. Cell 2017, 170, 1055–1056.
  50. Sevenich, L. Turning “Cold” Into “Hot” Tumors—Opportunities and Challenges for Radio-Immunotherapy Against Primary and Metastatic Brain Cancers. Front. Oncol. 2019, 9, 163.
  51. Mueller, K.L. Blocking PI3K makes cold tumors hot. Science 2016, 354, 1246.
  52. Baruch, E.N.; Youngster, I.; Ben-Betzalel, G.; Ortenberg, R.; Lahat, A.; Katz, L.; Adler, K.; Dick-Necula, D.; Raskin, S.; Bloch, N.; et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 2021, 371, 602–609.
  53. Sakaguchi, S.; Ono, M.; Setoguchi, R.; Yagi, H.; Hori, S.; Fehervari, Z.; Shimizu, J.; Takahashi, T.; Nomura, T. Foxp3+CD25+CD4+ natural regulatory T cells in dominant self-tolerance and autoimmune disease. Immunol. Rev. 2006, 212, 8–27.
  54. Shevach, E.M. Biological Functions of Regulatory T Cells. Adv. Immunol. 2011, 112, 137–176.
  55. Roncarolo, M.G.; Gregori, S.; Battaglia, M.; Bacchetta, R.; Fleischhauer, K.; Levings, M.K. Interleukin-10-secreting type 1 regulatory T cells in rodents and humans. Immunol. Rev. 2006, 212, 28–50.
  56. Cieri, N.; Camisa, B.; Cocchiarella, F.; Forcato, M.; Oliveira, G.; Provasi, E.; Bondanza, A.; Bordignon, C.; Peccatori, J.; Ciceri, F.; et al. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood 2013, 121, 573–584.
  57. Li, J.; Huston, G.; Swain, S.L. IL-7 Promotes the Transition of CD4 Effectors to Persistent Memory Cells. J. Exp. Med. 2003, 198, 1807–1815.
  58. Shinoda, K.; Hirahara, K.; Iinuma, T.; Ichikawa, T.; Suzuki, A.S.; Sugaya, K.; Tumes, D.J.; Yamamoto, H.; Hara, T.; Tani-Ichi, S.; et al. Thy1+IL-7+ lymphatic endothelial cells in iBALT provide a survival niche for memory T-helper cells in allergic airway inflammation. Proc. Natl. Acad. Sci. USA 2016, 113, E2842–E2851.
  59. Sharma, P.U.; Khosla, R.; David, P.; Rastogi, A.; Vyas, A.; Singh, D.; Bhardwaj, A.; Sahney, A.; Maiwall, R.; Sarin, S.K.; et al. CD4+CD25+CD127low Regulatory T Cells Play Predominant Anti-Tumor Suppressive Role in Hepatitis B Virus-Associated Hepatocellular Carcinoma. Front. Immunol. 2015, 6, 49.
  60. Bennett, C.; Christie, J.; Ramsdell, F.; Brunkow, M.E.; Ferguson, P.J.; Whitesell, L.; Kelly, T.E.; Saulsbury, F.T.; Chance, P.F.; Ochs, H.D. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP. Nat. Genet. 2001, 27, 20–21.
  61. Cinier, J.; Hubert, M.; Besson, L.; Di Roio, A.; Rodriguez, C.; Lombardi, V.; Caux, C.; Ménétrier-Caux, C. Recruitment and Expansion of Tregs Cells in the Tumor Environment—How to Target Them? Cancers 2021, 13, 1850.
  62. Berlato, C.; Khan, M.N.; Schioppa, T.; Thompson, R.; Maniati, E.; Montfort, A.; Jangani, M.; Canosa, M.; Kulbe, H.; Hagemann, U.B.; et al. A CCR4 antagonist reverses the tumor-promoting microenvironment of renal cancer. J. Clin. Investig. 2017, 127, 801–813.
  63. Chang, D.-K.; Peterson, E.; Sun, J.; Goudie, C.; Drapkin, R.I.; Liu, J.F.; Matulonis, U.; Zhu, Q.; Marasco, W.A. Anti-CCR4 monoclonal antibody enhances antitumor immunity by modulating tumor-infiltrating Tregs in an ovarian cancer xenograft humanized mouse model. OncoImmunology 2015, 5, e1090075.
  64. Ishida, T.; Ueda, R. CCR4 as a novel molecular target for immunotherapy of cancer. Cancer Sci. 2006, 97, 1139–1146.
  65. Kurose, K.; Ohue, Y.; Wada, H.; Iida, S.; Ishida, T.; Kojima, T.; Doi, T.; Suzuki, S.; Isobe, M.; Funakoshi, T.; et al. Phase Ia Study of FoxP3+ CD4 Treg Depletion by Infusion of a Humanized Anti-CCR4 Antibody, KW-0761, in Cancer Patients. Clin. Cancer Res. 2015, 21, 4327–4336.
  66. Lee, J.H.; Cho, Y.-S.; Lee, J.Y.; Kook, M.C.; Park, J.-W.; Nam, B.-H.; Bae, J.-M. The Chemokine Receptor CCR4 is Expressed and Associated With a Poor Prognosis in Patients With Gastric Cancer. Ann. Surg. 2009, 249, 933–941.
  67. Li, J.-Y.; Ou, Z.-L.; Yu, S.-J.; Gu, X.-L.; Yang, C.; Chen, A.-X.; Di, G.-H.; Shen, Z.-Z.; Shao, Z.-M. The chemokine receptor CCR4 promotes tumor growth and lung metastasis in breast cancer. Breast Cancer Res. Treat. 2011, 131, 837–848.
  68. Liu, W.; Wei, X.; Li, L.; Wu, X.; Yan, J.; Yang, H.; Song, F. CCR4 mediated chemotaxis of regulatory T cells suppress the activation of T cells and NK cells via TGF-β pathway in human non-small cell lung cancer. Biochem. Biophys. Res. Commun. 2017, 488, 196–203.
  69. Maeda, S.; Murakami, K.; Inoue, A.; Yonezawa, T.; Matsuki, N. CCR4 Blockade Depletes Regulatory T Cells and Prolongs Survival in a Canine Model of Bladder Cancer. Cancer Immunol. Res. 2019, 7, 1175–1187.
  70. Maolake, A.; Izumi, K.; Shigehara, K.; Natsagdorj, A.; Iwamoto, H.; Kadomoto, S.; Takezawa, Y.; Machioka, K.; Narimoto, K.; Namiki, M.; et al. Tumor-associated macrophages promote prostate cancer migration through activation of the CCL22-CCR4 axis. Oncotarget 2016, 8, 9739–9751.
  71. Olkhanud, P.B.; Baatar, D.; Bodogai, M.; Hakim, F.; Gress, R.; Anderson, R.; Deng, J.; Xu, M.; Briest, S.; Biragyn, A. Breast Cancer Lung Metastasis Requires Expression of Chemokine Receptor CCR4 and Regulatory T Cells. Cancer Res. 2009, 69, 5996–6004.
  72. Inngjerdingen, M.; Damaj, B.; Maghazachi, A. Human NK Cells Express CC Chemokine Receptors 4 and 8 and Respond to Thymus and Activation-Regulated Chemokine, Macrophage-Derived Chemokine, and I. J. Immunol. 2000, 164, 4048–4054.
  73. Islam, S.A.; Chang, D.S.; Colvin, R.A.; Byrne, M.H.; McCully, M.L.; Moser, B.; Lira, S.A.; Charo, I.F.; Luster, A.D. Mouse CCL8, a CCR8 agonist, promotes atopic dermatitis by recruiting IL-5+ TH2 cells. Nat. Immunol. 2011, 12, 167–177.
  74. Zhou, W.-J.; Hou, X.-X.; Wang, X.-Q.; Li, D.-J. The CCL17-CCR4 axis between endometrial stromal cells and macrophages contributes to the high levels of IL-6 in ectopic milieu. Am. J. Reprod. Immunol. 2017, 78, e12644.
  75. Shono, Y.; Suga, H.; Kamijo, H.; Fujii, H.; Oka, T.; Miyagaki, T.; Shishido-Takahashi, N.; Sugaya, M.; Sato, S. Expression of CCR3 and CCR4 Suggests a Poor Prognosis in Mycosis Fungoides and Sézary Syndrome. Acta Derm. Venereol. 2019, 99, 809–812.
  76. Wang, L.; Zhang, M.; Zhu, Y.; Zhang, X.; Yang, Y.; Wang, C. CCR4 Expression Is Associated With Poor Prognosis in Patients With Early Stage (pN0) Oral Tongue Cancer. J. Oral Maxillofac. Surg. 2018, 77, 426–432.
  77. 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.
  78. Yonekura, K.; Kanzaki, T.; Gunshin, K.; Kawakami, N.; Takatsuka, Y.; Nakano, N.; Tokunaga, M.; Kubota, A.; Takeuchi, S.; Kanekura, T.; et al. Effect of anti-CCR4 monoclonal antibody (mogamulizumab) on adult T-cell leukemia-lymphoma: Cutaneous adverse reactions may predict the prognosis. J. Dermatol. 2014, 41, 239–244.
  79. Freeman, C.M.; Chiu, B.-C.; Stolberg, V.R.; Hu, J.; Zeibecoglou, K.; Lukacs, N.W.; Lira, S.A.; Kunkel, S.L.; Chensue, S.W. CCR8 Is Expressed by Antigen-Elicited, IL-10-Producing CD4+CD25+ T Cells, Which Regulate Th2-Mediated Granuloma Formation in Mice. J. Immunol. 2005, 174, 1962–1970.
  80. Roos, R.S.; Loetscher, M.; Legler, D.F.; Clark-Lewis, I.; Baggiolini, M.; Moser, B. Identification of CCR8, the Receptor for the Human CC Chemokine I. J. Biol. Chem. 1997, 272, 17251–17254.
  81. Inngjerdingen, M.; Damaj, B.; Maghazachi, A.A. Expression and regulation of chemokine receptors in human natural killer cells. Blood 2001, 97, 367–375.
  82. Chensue, S.W.; Lukacs, N.W.; Yang, T.-Y.; Shang, X.; Frait, K.A.; Kunkel, S.L.; Kung, T.; Wiekowski, M.T.; Hedrick, J.A.; Cook, D.; et al. Aberrant in Vivo T Helper Type 2 Cell Response and Impaired Eosinophil Recruitment in Cc Chemokine Receptor 8 Knockout Mice. J. Exp. Med. 2001, 193, 573–584.
  83. Soler, D.; Chapman, T.R.; Poisson, L.R.; Wang, L.; Cote-Sierra, J.; Ryan, M.; McDonald, A.; Badola, S.; Fedyk, E.; Coyle, A.J.; et al. CCR8 expression identifies CD4 memory T cells enriched for FOXP3+ regulatory and Th2 effector lymphocytes. J. Immunol. 2006, 177, 6940–6951.
  84. Islam, S.A.; Ling, M.; Leung, J.; Shreffler, W.G.; Luster, A.D. Identification of human CCR8 as a CCL18 receptor. J. Exp. Med. 2013, 210, 1889–1898.
  85. Biber, K.; Zuurman, M.W.; Homan, H.; Boddeke, H.W.G.M. Expression of L-CCR in HEK 293 cells reveals functional responses to CCL2, CCL5, CCL7, and CCL. J. Leukoc. Biol. 2003, 74, 243–251.
  86. Howard, O.M.; Dong, H.F.; Shirakawa, A.K.; Oppenheim, J.J. LEC induces chemotaxis and adhesion by interacting with CCR1. CCR Blood 2000, 96, 840–845.
  87. Strasly, M.; Doronzo, G.; Capello, P.; Valdembri, D.; Arese, M.; Mitola, S.; Moore, P.; Alessandri, G.; Giovarelli, M.; Bussolino, F. CCL16 activates an angiogenic program in vascular endothelial cells. Blood 2004, 103, 40–49.
  88. Barsheshet, Y.; Wildbaum, G.; Levy, E.; Vitenshtein, A.; Akinseye, C.; Griggs, J.; Lira, S.A.; Karin, N. CCR8+FOXp3+ Treg cells as master drivers of immune regulation. Proc. Natl. Acad. Sci. USA 2017, 114, 6086–6091.
  89. Plitas, G.; Konopacki, C.; Wu, K.; Bos, P.D.; Morrow, M.; Putintseva, E.V.; Chudakov, D.M.; Rudensky, A.Y. Regulatory T Cells Exhibit Distinct Features in Human Breast Cancer. Immunity 2016, 45, 1122–1134.
  90. Wang, T.; Zhou, Q.; Zeng, H.; Zhang, H.; Liu, Z.; Shao, J.; Wang, Z.; Xiong, Y.; Wang, J.; Bai, Q.; et al. CCR8 blockade primes anti-tumor immunity through intratumoral regulatory T cells destabilization in muscle-invasive bladder cancer. Cancer Immunol. Immunother. 2020, 69, 1855–1867.
  91. Villarreal, D.O.; L’Huillier, A.; Armington, S.; Mottershead, C.; Filippova, E.V.; Coder, B.D.; Petit, R.G.; Princiotta, M.F. Targeting CCR8 Induces Protective Antitumor Immunity and Enhances Vaccine-Induced Responses in Colon Cancer. Cancer Res. 2018, 78, 5340–5348.
  92. Eruslanov, E.; Stoffs, T.L.; Kim, W.-J.; Daurkin, I.; Gilbert, S.M.; Su, L.-M.; Vieweg, J.; Daaka, Y.; Kusmartsev, S. Expansion of CCR8+ Inflammatory Myeloid Cells in Cancer Patients with Urothelial and Renal Carcinomas. Clin. Cancer Res. 2013, 19, 1670–1680.
  93. Rytlewski, J.; Milhem, M.M.; Monga, V. Turning ‘Cold’ tumors ‘Hot’: Immunotherapies in sarcoma. Ann. Transl. Med. 2021, 9, 1039.
  94. Yarmarkovich, M.; Maris, J.M. When Cold Is Hot: Immune Checkpoint Inhibition Therapy for Rhabdoid Tumors. Cancer Cell 2019, 36, 575–576.
  95. Galon, J.; Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 2019, 18, 197–218.
  96. Warming “Cold” Melanoma with TLR9 Agonists. Cancer Discov. 2018, 8, 670.
More
Upload a video for this entry
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 : Nathan Karin
View Times: 856
Revisions: 2 times (View History)
Update Date: 29 Mar 2022
Notice
You are not a member of the advisory board for this topic. If you want to update advisory board member profile, please contact office@encyclopedia.pub.
OK
Confirm
Only members of the Encyclopedia advisory board for this topic are allowed to note entries. Would you like to become an advisory board member of the Encyclopedia?
Yes
No
${ textCharacter }/${ maxCharacter }
Submit
Cancel
There is no comment~
${ textCharacter }/${ maxCharacter }
Submit
Cancel
${ selectedItem.replyTextCharacter }/${ selectedItem.replyMaxCharacter }
Submit
Cancel
Confirm
Are you sure to Delete?
Yes No
Academic Video Service