Skip to main content

MINI REVIEW article

Front. Med., 28 February 2024
Sec. Pathology

New insights into the role of mast cells as a therapeutic target in cancer through the blockade of immune checkpoint inhibitors

  • Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy

Mast cells release different anti-and pro-inflammatory agents changing their role from protective to pro-inflammatory cells involved in the progression of different pathological conditions, including autoimmune diseases and tumors. Different mediators released by mast cells are involved in their biological activities which may be anti-tumorigenic and/or pro-tumorigenic. For these reasons, tumor mast cells have been considered a novel therapeutic target to prevent tumor progression and metastatic process. Many different agents have been suggested and used in the past pre-clinical and clinical settings. Among the novel immunotherapeutic approaches to cancer treatment, different immune checkpoint inhibitors targeting PD-1/PDL-1 have been used in the treatment of many human tumors improving overall survival. In this context, inhibition of mast cell activity may be considered a novel strategy to improve the efficacy of anti-PD-1/PDL-1 therapy. The blockade of the PD-1/PD-L1 interaction may be suggested as a useful and novel therapeutic approach in the treatment of tumors in which mast cells are involved.

Tumor mast cells and related therapeutic approaches

Mast cells have multiple roles extending beyond their classical role in Ig-E-mediated allergic reactions. Mast cells release different anti-and pro-inflammatory agents changing their role from protective to pro-inflammatory cells involved in the progression of different pathological conditions, including autoimmune diseases and tumors (Table 1). Mast cells can be recruited into the tumor microenvironment by different chemotactic molecules released by tumor cells. One of the main chemoattractant factors produced by tumor cells is stem cell factor (SCF), which recruits mast cells expressing its tyrosine kinase receptor c-kit (CD117). Mast cells can exert both anti-tumorigenic and/or pro-tumorigenic roles (Table 2). Mast cells may exert detrimental effects on the host by releasing cytokines and growth factors, such as fibroblast growth factor2- (FGF-2), vascular endothelial growth factor (VEGF), nerve growth factor (NGF), and interleukin-8 (IL-8), which stimulate tumor cell expansion. Mast cells are a major source of histamine, which can induce tumor cell proliferation through H1 receptors while suppressing the immune system through H2 receptors. Mast cells produce several angiogenic factors, as well as proteases, which promote tumor vascularization and tumor invasiveness, respectively. By contrast, mast cells may promote the inhibition of tumor cell growth, tumor cell apoptosis, and inflammation.

Table 1
www.frontiersin.org

Table 1. Different types of cancer in which mast cells are involved.

Table 2
www.frontiersin.org

Table 2. Mediators released by mast cells able to stimulate or inhibit tumor growth.

by releasing cytokines such as inreleukin-1 (IL-1), IL-4, IL-6, and tumor necrosis factor alpha (TNF-α). Chondroitin sulfate may inhibit tumor cell diffusion and tryptase causes both tumor cell disruption and inflammation through activation of protease-activated receptors (PAR-1 and -2). Two mast cell phenotypes have been described called mast cell 1 and 2 (MC1 and MC2), related to pro-inflammatory and anti-inflammatory profiles, respectively. Mast cells promote tumor development by alterations in stroma-epithelial interactions, by inducing tumor angiogenesis and lymphangiogenesis, and by releasing different cytokines and growth factors. In solid and hematologic tumors, mast cells may be localized in intra-tumoral or peri-tumoral areas, with expression of favorable/unfavorable and, respectively, bed prognosis (25).

Based on the involvement of mast cells in tumor growth, these cells have been recently considered a novel therapeutic target in the control of tumor progression and metastatic capability. Many different agents have been suggested and used in pre-clinical and clinical settings. These therapeutic agents include inhibitors of c-kit (imatinib mesylate, mastinib, nilotinib, dasatinib, sunitinib, midostaurin, and ibrutinib). In this context, imatinib mesylate (Gleevec), which exerts inhibitory activity against the signaling cascade activated by CD117 (27), has been used against gastrointestinal stromal tumors (GIST) and metastatic melanoma with c-Kit mutations (28, 29). Masitinib has been used in the treatment of mastocytosis, GIST, colon cancer, prostate cancer, and pancreatic cancer (30). Sunitinib has been used in patients with imatinib-resistant GIST (31). Gabexate mesylate an inhibitor of tryptase has been used as an inhibitor of colon cancer growth with an anti-angiogenic effect (32). Cromolyn sodium, a mast cell stabilizing agent (33) that prevent cell degranulation (34), has been used in a xenograft mouse model of thyroid cancer (35). Obatoclax, which binds and blocks the anti-apoptotic activity of members of the Bcl-2 family, induces growth arrest in human neoplastic mast cells, and different mast cell lines (36), and exerts synergistic antineoplastic effects when combined with dasatinib (36).

H1 receptor antagonists reduced tumor growth, mast cell infiltration, and VEGF levels through the inhibition of hypoxia-inducible factor-1alpha (HIF-1α) in melanoma-bearing mice (37). Moreover, treatment with cimetidine, an H2 receptor antagonist, slows the growth of tumors in mice (38, 39). Chondroitin sulfate may inhibit tumor growth cell diffusion through activation of PAR-1 and-2 (40).

Immune checkpoint inhibitors

Different studies have highlighted the importance of the programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) pathway controlling inflammation degree to prevent an exacerbated immune response in tumor growth, in which PD-L1 expressed on tumor cells can inhibit the effector functions of CD8+ T cells, leading to the progression of tumors (41). Different immune checkpoint inhibitors targeting PD-1/PDL-1 have been used in the treatment of many human tumors, such as melanoma, non-small-cell lung cancer, and renal cancer, improving overall survival (42, 43). However, this therapeutic approach may be ineffective, because of the development of resistance mechanisms mediated by inflammatory cells present in the tumor microenvironment, including mast cells.

Relationship between tumor-infiltrating mast cells and response to anti-PD1/PD-L1 blockade

Human mast cells express several co-stimulatory and co-inhibitory molecules, including PDL-1 and PD-L2 (44). In the skin, mast cells express high levels of PD-L1, and in contact hypersensitivity mast cell absence abolished the PD-L1 blockade effect on CD8+T-cell activation (45). According, high levels of PDL-1 in mast cells promotes T cell immunosuppression and tumor growth in gastric cancer (46).

In high-grade serous ovarian cancer, infiltration of mast cells is associated with a decreased response to anti-PD1 blockade (47). Similarly, in a melanoma experimental model of resistance to anti-PD-1 therapy, high infiltration of mast cells predicted poor response to anti-PD1 blockade (48). An increased number of mast cells was detectable in melanoma patients after anti-PD1 therapy (49). In tumor histological sections, a co-localization of mast cells and forkhead box P3 (FOXP3)-positive Treg cells have been recognizable and associated with a down-modulation of HLA class I on tumor cells and correlated with resistance to anti-PD-1 therapy. Melanoma cells secrete chemokine (C-X-C motif) ligand 10 (CXCL10) that binds CXC motif chemokine receptor 3 (CXCR3) expressed by mast cells, favoring the recruitment of mast cells (49). Anti-PD1 treatment activates and induces expression on mast cells leading to therapeutic resistance through stimulation of angiogenesis and tumor growth (50).

Otherwise, the reduction of mast cells is associated with an improvement in the efficacy of anti-PD-1/anti-PD-L1 blockade. Combining anti-PD-1 with sunitinib or imatinib, but not PD-1 blockade alone, resulted in the depletion of mast cells and tumor regression (48). Cromolyn sodium decreases mast cell infiltration, the release of inflammatory cytokines, and improves the efficacy of anti-PD1 therapy (50). Targeting mast cells with ketotifen enhances T cells’ infiltration and cytotoxic capacity and sensitizes sarcoma cells to anti-PDL-1 therapy (51).

Concluding remarks

Mast cells play a crucial role in the control of tumor immunity and tumor growth. They can modulate the biological activity of immune and non-immune components of the tumor microenvironment through the release of a plethora of mediators, leading to different cancer-promoting and cancer-suppressive activities. The reduction of mast cell infiltration may be considered a novel therapeutic approach to cancer treatment. Mast cells can be therapeutically targeted by decreasing their number through c-Kit inhibitors; modulating mast cell activation and phenotype, and altering secreted mast cell mediators. In this context, inhibition of mast cell activity may be considered a novel strategy to improve the efficacy of anti-PD-1/PDL-1 therapy. The blockade of the PD-1/PD-L1 interaction may be suggested as a useful and novel therapeutic approach in the treatment of tumors in which mast cells are involved.

Author contributions

DR: Writing – original draft, Writing – review & editing.

Funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This work was supported by Associazione Italiana contro le Leucemie, Linfomi, e Mieloma (AIL), Bari, Italy.

Conflict of interest

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Tamma, R, Limongelli, L, Maiorano, E, Pastore, D, Cascardi, E, Tempesta, A, et al. Vascular density and inflammatory infiltrate in primary oral squamous cell carcinoma and after allogeneic hematopoietic stem cell transplantation. Ann Hematol. (2019) 98:979–86. doi: 10.1007/s00277-018-3575-3

PubMed Abstract | Crossref Full Text | Google Scholar

2. Elpek, GO, Gelen, T, Aksoy, NH, Erdoğan, A, Dertsiz, L, Demircan, A, et al. The prognostic relevance of angiogenesis and mast cells in squamous cell carcinoma of the oesophagus. J Clin Pathol. (2001) 54:940–4. doi: 10.1136/jcp.54.12.940

PubMed Abstract | Crossref Full Text | Google Scholar

3. Guidolin, D, Ruggieri, S, Annese, T, Tortorella, C, Marzullo, A, and Ribatti, D. Spatial distribution of mast cells around vessels and glands in human gastric carcinoma. Clin Exp Med. (2017) 17:531–9. doi: 10.1007/s10238-017-0452-7

PubMed Abstract | Crossref Full Text | Google Scholar

4. Ammendola, M, Patruno, R, Sacco, R, Marech, I, Sammarco, G, Zuccalà, V, et al. Mast cells positive to tryptase and tumour-associated macrophages correlate with angiogenesis in locally advanced colorectal cancer patients undergone to surgery. Expert Opin Ther Targets. (2016) 20:533–40. doi: 10.1517/14728222.2016.1158811

PubMed Abstract | Crossref Full Text | Google Scholar

5. Tamma, R, Annese, T, Ruggieri, S, Brunetti, O, Longo, V, Cascardi, E, et al. Inflammatory cells infiltrate and angiogenesis in locally advanced and metastatic cholangiocarcinoma. Eur J Clin Invest. (2019) 49:e13087. doi: 10.1111/eci.13087

PubMed Abstract | Crossref Full Text | Google Scholar

6. Longo, V, Tamma, R, Brunetti, O, Pisconti, S, Argentiero, A, Silvestris, N, et al. Mast cells and angiogenesis in pancreatic ductal adenocarcinoma. Clin Exp Med. (2018) 18:319–23. doi: 10.1007/s10238-018-0493-6

Crossref Full Text | Google Scholar

7. Tamma, R, Rutigliano, M, Lucarelli, G, Annese, T, Ruggieri, S, Cascardi, E, et al. Microvascular density, macrophages, and mast cells in human clear cell renal carcinoma with and without bevacizumab treatment. Urol Oncol. (2019) 37:355.e11–9. doi: 10.1016/j.urolonc.2019.01.025

Crossref Full Text | Google Scholar

8. Ronca, R, Tamma, R, Coltrini, D, Ruggieri, S, Presta, M, and Ribatti, D. Fibroblast growth factor modulates mast cell recruitment in a murine model of prostate cancer. Oncotarget. (2017) 8:82583–92. doi: 10.18632/oncotarget.19773

PubMed Abstract | Crossref Full Text | Google Scholar

9. Ribatti, D, Finato, N, Crivellato, E, Marzullo, A, Mangieri, D, Nico, B, et al. Neovascularization and mast cells with tryptase activity increase simultaneously with pathological progression in human endometrial cancer. Am J Obstet Gynecol. (2005) 193:1961–5. doi: 10.1016/j.ajog.2005.04.055

PubMed Abstract | Crossref Full Text | Google Scholar

10. Ribatti, D, Ennas, MG, Vacca, A, Ferreli, F, Nico, B, Orru, S, et al. Tumor vascularity and tryptase positive-mast cells correlate with a poor prognosis in melanoma. Eur J Clin Invest. (2003) 33:420–5. doi: 10.1046/j.1365-2362.2003.01152.x

PubMed Abstract | Crossref Full Text | Google Scholar

11. Ribatti, D, Nico, B, Finato, N, Crivellato, E, and Beltrami, CA. Co-localization of tryptase and cathepsin-G in mast cells in cutaneous mastocytosis. Cancer Lett. (2009) 279:209–12. doi: 10.1016/j.canlet.2009.01.039

PubMed Abstract | Crossref Full Text | Google Scholar

12. Ribatti, D, Annese, T, and Tamma, R. Controversial role of mast cells in breast cancer tumor progression and angiogenesis. Clin Breast Cancer. (2021) 21:486–91. doi: 10.1016/j.clbc.2021.08.010

PubMed Abstract | Crossref Full Text | Google Scholar

13. Sawatsubashi, M, Yamada, T, Fukushima, N, Mizokami, H, Tokunaga, O, and Shin, T. Association of vascular endothelial growth factor and mast cells with angiogenesis in laryngeal squamous cell carcinoma Virchows arch B cell Pathol. Mol Pathol. (2000) 436:243–8. doi: 10.1007/s004280050037

Crossref Full Text | Google Scholar

14. Longo, V, Catino, A, Montrone, M, Galetta, D, and Ribatti, D. Controversial role of mast cells in NSCLC tumor progression and angiogenesis. Thorac Cancer. (2022) 13:2929–34. doi: 10.1111/1759-7714.14654

PubMed Abstract | Crossref Full Text | Google Scholar

15. Ribatti, D, Vacca, A, Nico, B, Quondamatteo, F, Ria, R, Minischetti, M, et al. Bone marrow angiogenesis and mast cell density increase simultaneously with progression of human multiple myeloma. Br J Cancer. (1999) 79:451–5. doi: 10.1038/sj.bjc.6690070

PubMed Abstract | Crossref Full Text | Google Scholar

16. Molica, S, Vacca, A, Crivellato, E, Cuneo, A, and Ribatti, D. Tryptase-positive mast cells predict clinical outcome of patients with B-cell chronic lymphocytic leukemia. Eur J Haematol. (2003) 71:137–9. doi: 10.1034/j.1600-0609.2003.00110.x

PubMed Abstract | Crossref Full Text | Google Scholar

17. Ribatti, D, Polimeno, G, Vacca, A, Marzullo, A, Crivellato, E, Nico, B, et al. Correlation of bone marrow angiogenesis and mast cells with tryptase activity in myelodysplastic syndromes. Leukemia. (2002) 16:1680–4. doi: 10.1038/sj.leu.2402586

PubMed Abstract | Crossref Full Text | Google Scholar

18. Qu, Z, Kayton, RJ, Ahmadi, P, Liebler, GM, Powers, MR, Planck, SR, et al. Ultrastructural immunolocalization of basic fibroblast growth factor in mast cell secretory granules: morphological evidence for bFGF release through degranulation. J Histochem Cytochem. (1998) 46:1119–28. doi: 10.1177/002215549804601004

PubMed Abstract | Crossref Full Text | Google Scholar

19. Grutzkau, A, Kruger-Krasagakes, S, Baumeister, H, Schwarz, C, Kogel, H, Welker, P, et al. Synthesis, storage, and release of vascular endothelial growth factor/vascular permeability factor (VEGF/VPF) by human mast cells: implications for the biological significance of VEGF 206. Mol Biol Cell. (1998) 9:875–84. doi: 10.1091/mbc.9.4.875

PubMed Abstract | Crossref Full Text | Google Scholar

20. Cantarella, G, Lempereur, L, Presta, M, Ribatti, D, Lombardo, G, Lazarovici, P, et al. Nerve growth factor-endothelial cell interactions lead to angiogenesis in vitro and in vivo. FASEB J. (2002) 16:1307–9. doi: 10.1096/fj.01-1000fje

PubMed Abstract | Crossref Full Text | Google Scholar

21. Solimando, AG, Desantis, V, and Ribatti, D. Mast cells and interelukins. Int J Mol Sci. (2022) 23:14004. doi: 10.3390/ijms232214004

PubMed Abstract | Crossref Full Text | Google Scholar

22. Sorbo, J, Jakobson, A, and Norrby, K. Mast cell histamine is angiogenic through receptors for histamine 1 and histamine 2. Int J Exp Pathol. (1994) 75:43–50.

PubMed Abstract | Google Scholar

23. Ribatti, D, Ranieri, G, Nico, B, Benagiano, V, and Crivellato, E. Tryptase and chymase are angiogenic in vivo in the chorioallantoic membrane assay. Int J Dev Biol. (2011) 55:99–102. doi: 10.1387/ijdb.103138dr

PubMed Abstract | Crossref Full Text | Google Scholar

24. Galli, SJ, Tsai, M, Marichal, T, Tchougounova, E, Reber, LL, and Pejler, G. Approaches for analyzing the roles of mast cells and their proteases in vivo. Adv Immunol. (2015) 126:45–127. doi: 10.1016/bs.ai.2014.11.002

PubMed Abstract | Crossref Full Text | Google Scholar

25. Ribatti, D, and Crivellato, E. The controversial role of mast cells in tumor growth. Int Rev Cell Mol Biol. (2009) 275:89–131. doi: 10.1016/S1937-6448(09)75004-X

Crossref Full Text | Google Scholar

26. Kelly, A, Houston, SA, Sherwood, E, Casulli, J, and Travis, MA. Regulation of innate and adaptive immunity by TGFbeta. Adv Immunol. (2017) 134:137–233. doi: 10.1016/bs.ai.2017.01.001

Crossref Full Text | Google Scholar

27. Heinrich, MC, Griffith, DJ, Druker, BJ, Wait, CL, Ott, KA, and Zigler, AJ. Inhibition of c-kit receptor tyrosine kinase activity by STI 571 a selective tyrosine kinase inhibitor. Blood. (2000) 96:925–32. doi: 10.1182/blood.V96.3.925

Crossref Full Text | Google Scholar

28. Hodi, FS, Corless, CL, Giobbie-Hurder, A, Fletcher, JA, Zhu, M, Marino-Enriquez, A, et al. Imatinib for melanomas harboring mutationally activated or amplified KIT arising on mucosal, acral, and chronically sun-damaged skin. J Clin Oncol. (2013) 31:3182–90. doi: 10.1200/JCO.2012.47.7836

PubMed Abstract | Crossref Full Text | Google Scholar

29. Kitamura, Y, and Hirotab, S. Kit as a human oncogenic tyrosine kinase. Cell Mol Life Sci. (2004) 61:2924–31. doi: 10.1007/s00018-004-4273-y

Crossref Full Text | Google Scholar

30. Ribatti, D. Mast cells as therapeutic target in cancer. Eur J Pharmacol. (2016) 778:152–7. doi: 10.1016/j.ejphar.2015.02.056

Crossref Full Text | Google Scholar

31. Demetri, GD, van Oosterom, AT, Garrett, CR, Blackstein, ME, Shah, MH, Verweij, J, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet. (2006) 368:1329–38. doi: 10.1016/S0140-6736(06)69446-4

Crossref Full Text | Google Scholar

32. Yoon, WH, Jung, YJ, Kim, TD, Li, G, Park, BJ, Kim, JY, et al. Gabexate Mesilate inhibits Colon Cancer growth, invasion, and metastasis by reducing matrix metalloproteinases and angiogenesis. Clin Cancer Res. (2004) 10:4517–26. doi: 10.1158/1078-0432.CCR-04-0084

PubMed Abstract | Crossref Full Text | Google Scholar

33. Cimpean, AM, and Raica, M. The hidden side of disodium Cromolyn: from mast cell stabilizer to an Angiogenic factor and antitumor agent. Arch Immunol Ther Ex. (2016) 64:515–22. doi: 10.1007/s00005-016-0408-8

PubMed Abstract | Crossref Full Text | Google Scholar

34. Soucek, L, Lawlor, ER, Soto, D, Shchors, K, Swigart, LB, and Evan, GI. Mast cells are required for angiogenesis and macroscopic expansion of Myc-induced pancreatic islet tumors. Nat Med.. (2007) 13:1211–1218.

PubMed Abstract | Google Scholar

35. Melillo, RM, Guarino, V, Avilla, E, Galdiero, MR, Liotti, F, Prevete, N, et al. Mast cells have a protumorigenic role in human thyroid cancer. Oncogene. (2010) 29:6203–15. doi: 10.1038/onc.2010.348

PubMed Abstract | Crossref Full Text | Google Scholar

36. Peter, B, Cerny-Reiterer, S, Hadzijusufovic, E, Schuch, K, Stefanzl, G, Eisenwort, G, et al. The pan-Bcl-2blocker obatoclax promotes the expression of Puma, Noxa, and Bimm RNA and induces apoptosis in neoplastic mast cells. J Leukoc Biol. (2014) 95:95–104. doi: 10.1189/jlb.1112609

PubMed Abstract | Crossref Full Text | Google Scholar

37. Jeong, HJ, Oh, HA, Nam, SY, Han, NR, Kim, YS, Kim, JH, et al. Oh HAThe critical role of mast cell-derived hypoxia-inducible factor-1α in human and mice melanoma growth. Int J Cancer. (2013) 132:2492–501. doi: 10.1002/ijc.27937

PubMed Abstract | Crossref Full Text | Google Scholar

38. Bowrey, PF, King, J, Magarey, C, Schwartz, P, Marr, P, Bolton, E, et al. Histamine, mast cells and tumour cell proliferation in breast cancer: does preoperative cimetidine administration have an effect? Br J Cancer. (2000) 82:167–70. doi: 10.1054/bjoc.1999.0895

PubMed Abstract | Crossref Full Text | Google Scholar

39. Nordlund, JJ, and Askenase, PW. The effect of histamine, antihistamines, and a mast cell stabilizer on the growth of cloudman melanoma cells in DBA/2mice. J Invest Dermatol. (1983) 81:28–31. doi: 10.1111/1523-1747.ep12538356

Crossref Full Text | Google Scholar

40. Ribatti, D, and Crivellato, E. Mast cells, angiogenesis, and tumour growth. Biochim Biophys Acta. (2012) 1822:2–8. doi: 10.1016/j.bbadis.2010.11.010

Crossref Full Text | Google Scholar

41. Alsaab, HO, Sau, S, Alzhrani, R, Tatiparti, K, Bhise, K, Kashaw, SK, et al. PD-1 and PD-L1 checkpoint signaling inhibition for cancer immunotherapy: mechanism, combinations, and clinical outcome. Front Pharmacol. (2017) 8:561. doi: 10.3389/fphar.2017.00561

PubMed Abstract | Crossref Full Text | Google Scholar

42. Robert, C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat Commun. (2020) 11:3801. doi: 10.1038/s41467-020-17670-y

PubMed Abstract | Crossref Full Text | Google Scholar

43. Siu, LL, Ivy, SP, Dixon, EL, Gravell, AE, Reeves, SA, and Rosner, GL. Challenges and opportunities in adapting clinical trial design for immunotherapies. Clin Cancer Res. (2017) 23:4950–8. doi: 10.1158/1078-0432.CCR-16-3079

PubMed Abstract | Crossref Full Text | Google Scholar

44. Nakae, S, Suto, H, Iikura, M, Kakurai, M, Sedgwick, JD, and Tsai, M. Galli SJ mast cells enhance T cell activation: importance of mast cell costimulatory molecules and secreted TNF. J Immunol. (2006) 176:2238–48. doi: 10.4049/jimmunol.176.4.2238

PubMed Abstract | Crossref Full Text | Google Scholar

45. Hirano, T, Honda, T, Kanameishi, S, Honda, Y, Egawa, G, Kitoh, A, et al. PD-L1 on mast cells suppresses effector CD81T-cell activation in the skin in murine contact hypersensitivity. J Allergy Clin Immunol. (2021) 148:563–573.e7. doi: 10.1016/j.jaci.2020.12.654

PubMed Abstract | Crossref Full Text | Google Scholar

46. Lv, Y, Zhao, Y, Wang, X, Chen, N, Mao, F, Teng, Y, et al. Increased intratumoral mast cells foster immune suppression and gastric cancer progression through TNFα-PD-L1 pathway. J Immunother Cancer. (2019) 7:54. doi: 10.1186/s40425-019-0530-3

PubMed Abstract | Crossref Full Text | Google Scholar

47. Cao, K, Zhang, G, Zhang, X, Yang, M, Wang, Y, He, M, et al. Stromal infiltrating mast cells identify immunoevasive subtype high-grade serous ovarian cancer with poor prognosis and inferior immunotherapeutic response. Onco Targets Ther. (2021) 10:1969075. doi: 10.1080/2162402X.2021.1969075

PubMed Abstract | Crossref Full Text | Google Scholar

48. Somasundaram, R, Connelly, T, Choi, R, Choi, H, Samarkina, A, Li, L, et al. Tumor-infiltrating mast cells are associated with resistance to anti-PD-1 therapy. Nat Commun. (2021) 12:346. doi: 10.1038/s41467-020-20600-7

PubMed Abstract | Crossref Full Text | Google Scholar

49. Kuo, PT, Zeng, Z, Salim, N, Mattarollo, S, Wells, JW, and Leggatt, GR. The role of CXCR3 and its chemokine ligand in skin disease and cancer. Front Med. (2018) 5:271. doi: 10.3389/fmed.2018.00271

PubMed Abstract | Crossref Full Text | Google Scholar

50. Li, J, Peng, G, Zhu, K, Jie, X, Xu, Y, Rao, X, et al. PD-1+ mast cell enhanced by PD-1 blocking therapy associated with resistance to immunotherapy. Cancer Immunol Immunother. (2022) 72:633–45. doi: 10.1007/s00262-022-03282-6

Crossref Full Text | Google Scholar

51. Myrofora, P, Mpekris, F, Voutouri, C, Michael, C, Constantinidou, A, Martin, JD, et al. Abstract 6382: targeting mast cells restores T cell infiltration and sensitizes sarcomas to PD-L1 inhibition. Cancer Res. (2022) 82:6382. doi: 10.1158/1538-7445.AM2022-6382

Crossref Full Text | Google Scholar

Keywords: immune checkpoint inhibitors, PD-1/PDL-1, tumor growth, mast cells, tumor therapy

Citation: Ribatti D (2024) New insights into the role of mast cells as a therapeutic target in cancer through the blockade of immune checkpoint inhibitors. Front. Med. 11:1373230. doi: 10.3389/fmed.2024.1373230

Received: 19 January 2024; Accepted: 19 February 2024;
Published: 28 February 2024.

Edited by:

Luigi Tornillo, University of Basel, Switzerland

Reviewed by:

Alessandro Gambella, University of Pittsburgh, United States

Copyright © 2024 Ribatti. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Domenico Ribatti, domenico.ribatti@uniba.it

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.