Dendritic cells in therapy of breast adenocarcinoma


Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

Dendritic cells (DC), as professional antigen-presenting cells with functional plasticity, are widely studied in preclinical and clinical research of vaccines against various types of malignant tumors. Some of the studies DС have proved their efficiency. However, all over the world, there is no officially approved DC vaccine for the treatment of breast cancer. Knowledge and understanding of fundamental functions of DC, as well as the development of methods enhancing the effect of such vaccines, may contribute to the progress in this direction. In this review, we consider the features of the main types of DC and their role in the pathogenesis of breast cancer. Also, we describe the mechanisms of antigen uptake, processing, and presentation. In addition, considerable attention is paid to the current research of DC-based treatment of breast adenocarcinoma as DC are studied both as monotherapy or in combination with other therapeutic strategies.

Full Text

Restricted Access

About the authors

A. A Chernysheva

Voronezh State University

Email: 79202208606@yandex.ru
Universitetskaya pl., 1, Voronezh, 394018, Russian Federation

I. V Chekhonin

V.P. Serbsky National Medical Research Centre for Psychiatry and Narcology

Email: 79202208606@yandex.ru
Kropotkinskiy bystreet, 23, Moscow, 119034, Russian Federation

O. I Gurina

V.P. Serbsky National Medical Research Centre for Psychiatry and Narcology

Email: 79202208606@yandex.ru
Kropotkinskiy bystreet, 23, Moscow, 119034, Russian Federation

I. I Shepeleva

V.P. Serbsky National Medical Research Centre for Psychiatry and Narcology; N.I. Pirogov Russian National Research Medical University

Email: 79202208606@yandex.ru
23, Moscow, 119034, Russian Federation; Ostrovitianova str., 1, Moscow, 117997, Russian Federation

T. N Popova

Voronezh State University

Email: 79202208606@yandex.ru
Universitetskaya pl., 1, Voronezh, 394018, Russian Federation

References

  1. Каприн А.Д., Старинский В.В., Петрова Г.В. Злокачественные новообразования в России в 2017 г. (заболеваемость и смертность). М.: МНИОИ им. П.А. Герцена - филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2018; 250
  2. Bray F., Ferlay J., Soerjomataram I., Siegel R.L., Torre L.A., Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J. for Clinicians. 2018; 68 (6): 394-424. https:// doi.org/10.3322/caac.21492
  3. Министерство здравоохранения РФ. Клинические рекомендации: рак молочной железы (http://oncology-asso-ciation.ru/docs/rak_molochnoy_zhelezy. pdf)
  4. Ju J., Zhu A-J., Yuan P Progress in targeted therapy for breast cancer. Chronic Diseases and Translational Medicine. 2018; 4 (3): 164-75. https://doi.org/10.10Wj. cdtm.2018.04.002
  5. Baklaushev V.P., Grinenko N.F., Yusubalieva G.M., Abakumov M.A., Gubskii I.L., Cherepanov S.A., Kashparov I.A., Burenkov M.S., Rabinovich E.Z., Ivanova N.V., Antonova O.M., Chekhonin V.P. Modeling and Integral X-Ray, Optical, and MRI Visualization of Multiorgan Metastases of Orthotopic 4T1 Breast Carcinoma in BALB/c Mice. Bulletin of Experimental Biology and Medicine. 2015; 158 (4): 581-8. https://doi. org/10.1007/s10517-015-2810-3
  6. Wu L., Galy A. The development of dendritic cells from hematopoietic precursors. In: Dendritic Cells (Second Edition): Biology and Clinical Applications. (M.T. Lotze, A.W Thomson). Academic Press Co. 2001; 3-12.
  7. Roberts E.W, Broz M.L., Binnewies M., Headley M.B., Nelson A.E., Wolf D.M., Kaisho T., Bogunovic D., Bhardwaj N., Krummel M.F. Critical Role for CD103+/CD141 + Dendritic Cells Bearing CCR7 for Tumor Antigen Trafficking and Priming of T Cell Immunity in Melanoma. Cancer Cell. 2016; 30 (2): 324-36. https://doi.org/10.10Wj. ccell.2016.06.003
  8. Krishnaswamy J.K., Gowthaman U., Zhang B., Mattsson J., Szeponik L., Liu D., Wu R., White T, Calabro S., Xu L., Collet M.A., Yurieva M., Alsen S., Fogelstrand P, Walter A., Heath W.R., Mueller S.N., Yrlid U., Williams A., Eisenbarth S.C. Migratory CD11b+ conventional dendritic cells induce T follicular helper cell-dependent antibody responses. Science Immunology. 2017; 2 (18): eaam9169. https://doi.org/10.1126/ sciimmunol.aam9169
  9. Yang G-X., Lian Z-X., Kikuchi K., Moritoki Y, Ansari A.A., Liu Y-J., Ikehara S., Gershwin M.E. Plasmacytoid Dendritic Cells of Different Origins Have Distinct Characteristics and Function: Studies of Lymphoid Progenitors versus Myeloid Progenitors. J. Immunol. 2005; 175 (11): 7281-7. https:// doi.org/10.4049/jimmunol.175.11.7281
  10. Zhang H., Gregorio J.D., Iwahori T, Zhang X., Choi O., Tolentino L.L., Prestwood T., Carmi Y, Engleman E.G. A distinct subset of plasmacytoid dendritic cells induces activation and differentiation of B and T lymphocytes. Proc. Natl. Acad. Sci USA. 2017; 114 (8): 1988-93. https://doi.org/10.1073/ pnas.1610630114
  11. Sawant A., Ponnazhagan S. Role of plasmacytoid dendritic cells in breast cancer bone dissemination. Oncoimmunology, 2013; 2 (2): e22983. https://doi.org/10.4161/ onci.22983
  12. Wu J., Li S., Yang Y, Zhu S., Zhang M., Qiao Y, Liu Y-J., Chen J. TLR-activated plasmacytoid dendritic cells inhibit breast cancer cell growth in vitro and in vivo. Oncotarget. 2017; 8: 11708-18. https://doi.org/10.18632/ oncotarget.14315
  13. Polak M.E., Newell L., Taraban V.Y., Pickard C., Healy E., Friedmann PS., Al-Shamkhani A., Ardern-Jones M.R. CD70-CD27 Interaction Augments CD8+ T-Cell Activation by Human Epidermal Langer-hans Cells. J. Invest Dermatol. 2012; 132 (6): 1636-44. https://doi.org/10.1038/ jid.2012.26
  14. Tsuge T, Yamakawa M., Tsukamoto M. Infiltrating dendritic/Langerhans cells in primary breast cancer. Breast Cancer Res Treat. 2000; 59 (2): 141-52. https://doi. org/10.1023/A:1006396216933
  15. Amorim K.N.S., Chagas D.C.G., Sulczewski F.B., Boscardin S.B. Dendritic Cells and Their Multiple Roles during Malaria Infection. J. of Immunology Research. 2016; 2016: 2926436. https://doi. org/10.1155/2016/2926436
  16. Liu Z., Roche P.A. Macropinocytosis in phagocytes: regulation of MHC class-II-restricted antigen presentation in dendritic cells. Front. Physiol. 2015; 6: 1. https://doi. org/10.3389/fphys.2015.00001
  17. Moreau H.D., Blanch-Mercader C., Attia R., Alraies Z., Maurin M., Bousso P, Joanny J-F, Voituriez R., Piel M., Lennon-Dumenil A-M. Macropinocytosis overcomes directional bias due to hydraulic resistance to enhance space exploration by dendritic cells. bioRxiv. 2018; 272682. https://doi. org/10.1101/272682
  18. Chapman H.A. Endosomal proteases in antigen presentation. Curr Opin Immunol. 2006; 18 (1): 78-84. https://doi. org/10.1016/j.coi.2005.11.011
  19. Schmidt M., Lill J.R. MHC class I presented antigens from malignancies: A perspective on analytical characterization & immuno-genicity. J. of Proteomics. 2019; 191: 48-57. https://doi.org/10.10Wj.jprot.2018.04.021
  20. Basha G., Lizee G., Reinicke A.T., Seipp R.P, Omilusik K.D., Jefferies W.A. MHC Class I Endosomal and Lysosomal Trafficking Coincides with Exogenous Antigen Loading in Dendritic Cells. PLoS One. 2008; 3 (9): e3247. https://doi.org/10.1371/journal. pone.0003247
  21. Santambrogio L., Sato A.K., Carven G.J., Belyanskaya S.L., Strominger J.L., Stern L.J. Extracellular antigen processing and presentation by immature dendritic cells. Proc. Natl. Acad. Sci. USA. 1999; 96 (26): 15056-61. https://doi.org/10.1073/ pnas.96.26.15056
  22. Талаев В.Ю., Плеханова М.В., Матвеичев А.В. Экспериментальные модели, пригодные для оценки влияния компонентов новых разрабатываемых вакцин на дифференцировку дендритных клеток. Медиаль. 2014; 2 (12): 135-53.
  23. Constantino J., Gomes C., Falcao A., Cruz M.T., Neves B.M. Antitumor dendritic cell-based vaccines: lessons from 20 years of clinical trials and future perspectives. Transl Res. 2016; 168: 74-95. https://doi. org/10.1016/j.trsl.2015.07.008
  24. Maj T., Slawek A., Chelmonska-Soyta A. CD80 and CD86 Costimulatory Molecules Differentially Regulate OT-II CD4+ T Lymphocyte Proliferation and Cytokine Response in Cocultures with Antigen-Presenting Cells Derived from Pregnant and Pseudopregnant Mice. Mediators Inflamm. 2014; 2014: 769239. https://doi. org/10.1155/2014/769239
  25. Gong J., Avigan D., Chen D., Wu Z., Koido S., Kashiwaba M., Kufe D. Activation of antitumor cytotoxic T lymphocytes by fusions of human dendritic cells and breast carcinoma cells. Proc. Natl. Acad. Sci. USA. 2000; 97 (6): 2715-8. https://doi. org/10.1073/pnas.050587197
  26. Neidhardt-Berard E-M., Berard F, Banchereau J., Palucka A.K. Dendritic cells loaded with killed breast cancer cells induce differentiation of tumor-specific cytotoxic T lymphocytes. Breast Cancer Res. 2004; 6: R322. https://doi.org/10.1186/ bcr794
  27. Gervais A., Leveque J., Bouet-Toussaint F, Burtin F, Lesimple T, Sulpice L., Patard J.-J., Genetet N., Catros-Quemener V Dendritic cells are defective in breast cancer patients: a potential role for polyamine in this immunodeficiency. Breast Cancer Res. 2005; 7 (3): 326-35. https://doi.org/10.1186/ bcr1001
  28. Чехонин И.В., Гурина О.И., Черепанов С.А., Абакумов М.А., Ионова К.П., Жигарев Д.К., Макаров А.В., Чехонин В.П. Сенсибилизированные дендритные клетки для терапии экспериментальной глиомы. Бюллетень экспериментальной биологии и медицины. 2016; 161 (6): 747-52.
  29. El Deeb N.M., Mehanna R.A. Assessment of maturation status of tumor-infiltrating dendritic cells in invasive ductal carcinoma of the breast: relation with vascular endothelial growth factor expression. Turk Patoloji Derg. 2013; 29 (3): 193-200. https:// doi.org/10.5146/tjpath.2013.01186.
  30. Bohnenkamp H.R., Coleman J., Burchell J.M., Taylor-Papadimitriou J., Noll T. Breast carcinoma cell lysate-pulsed dendritic cells cross-prime MUC1-specific CD8+ T cells identified by peptide-MHC-class-I tetramers. Cell Immunol. 2004; 231 (1-2): 112-25. https://doi.org/10.10Wj.cel-limm.2004.12.007
  31. Brossart P, Wirths S., Stuhler G., Reichardt V.L., Kanz L., Brugger W. Induction of cytotoxic T-lymphocyte responses in vivo after vaccinations with peptide-pulsed dendritic cells. Blood. 2000; 96: 3102-8.
  32. Lowenfeld L., Mick R., Datta J., Xu S., Fitzpatrick E., Fisher C.S., Fox K.R., DeMichele A., Zhang P.J., Weinstein S.P., Roses R.E., Czerniecki B.J. Dendritic Cell Vaccination Enhances Immune Responses and Induces Regression of HER2pos Ductal Carcinoma In Situ Independent of Route: Results of Randomized Selection Design Trial. Clin. Cancer Res. 2017; 23 (12): 2961-71. https://doi.org/10.1158/1078-0432.CCR-16-1924
  33. Cui H., Zhang W, Hu W, Liu K., Wang T., Ma N., Liu X., Liu Y, Jiang Y Recombinant Mammaglobin A Adenovirus-Infected Dendritic Cells Induce Mammaglobin A-Specific CD8+ Cytotoxic T Lymphocytes against Breast Cancer Cells In Vitro. PLoS One. 2013; 8 (5): e63055. https://doi. org/10.1371/journal.pone.o063055
  34. Delirezh N., Moazzeni S.M., Shokri F, Shokr-gozar M.A., Atri M., Kokhaei P Autologous dendritic cells loaded with apoptotic tumor cells induce T cell-mediated immune responses against breast cancer in vitro. Cell. Immunol. 2009; 257 (1-2): 23-31. htt-ps://doi.org/10.1016/j.cellimm.2009.02.002
  35. Zheng J., Liu Q., Yang J., Ren Q., Cao W, Yang J., Yu Z., Yu F, Wu Y, Shi H., Liu W Co-culture of apoptotic breast cancer cells with immature dendritic cells: a novel approach for DC based vaccination in breast cancer. Brazilian Journal of Medical and Biological Research. 2012; 45 (6): 510-5. https://doi.org/10.1590/S0100-879X2012007500061
  36. Avigan D., Vasir B., Gong J., Borges V, Wu Z., Uhl L., Atkins M., Mier J., McDermott D., Smith T, Giallambardo N., Stone C., Schadt K., Dolgoff J., Tetreault J.C., Villarroel M., Kufe D. Fusion Cell Vaccination of Patients with Metastatic Breast and Renal Cancer Induces Immunological and Clinical Responses. Clin. Cancer Res. 2004; 10 (14): 4699-708. https://doi.org/10.1158/1078-0432.CCR-04-0347
  37. Zhang P, Yi S., Li X., Liu R., Jiang H., Huang Z., Liu Y, Wu J., Huang Y Preparation of Triple-Negative Breast Cancer Vaccine through Electrofusion with Day-3 Dendritic Cells. PLoS One. 2014; 9 (7): e102197. htt-ps://doi.org/10.1371/journal.pone.0102197
  38. Das M., Law S. Role of Tumor Microenvironment in Cancer Stem Cell Chemoresist-ance and recurrence. Int J. Biochem Cell Biol. 2018; 103 (2018): 115-24. https://doi. org/10.1016/j.biocel.2018.08.011
  39. Dashti A., Ebrahimi M., Hadjati J., Memarnejadian A., Moazzeni S.M. Dendritic cell based immunotherapy using tumor stem cells mediates potent antitumor immune responses. Cancer Lett. 2016;(1): 175-85. https://doi.org/10.10Wj. canlet.2016.01.021
  40. Nguyen S.T, Nguyen H.L., Pham V.Q., Nguyen G.T, Tran C.D-T, Phan N.K., Pham PV Targeting specificity of dendritic cells on breast cancer stem cells: in vitro and in vivo evaluations. Onco Targets Ther. 2015; 8: 323-34. https://doi.org/10.2147/OTT.S77554
  41. Pham P.V., Le H.T., Vu B.T., Pham V.Q., Le P.M., Phan N.L., Trinh N.V., Nguyen H.T., Nguyen S.T, Nguyen T.L., Phan N.K. Targeting breast cancer stem cells by dendritic cell vaccination in humanized mice with breast tumor: preliminary results. Onco Targets Ther. 2016; 2016 (9): 4441-51. https:// doi.org/10.2147/OTT.S105239
  42. Bryson PD., Han X., Truong N., Wang P Breast cancer vaccines delivered by dendritic cell targeted lentivectors induce potent antitumor immune responses and protect mice from mammary tumor growth. Vaccine. 2017; 35 (43): 5842-9. https://doi. org/10.1016/j.vaccine.2017.09.017
  43. Tang M., Liu Y, Zhang Q-C., Zhang P, Wu J-K., Wang J-N., Ruan Y, Huang Y. Antitumor efficacy of the Runx2-dendritic cell vaccine in triple-negative breast cancer in vitro. Oncology Lett. 2018; 16 (3): 2813-22. https://doi.org/10.3892/ol.2018.9001
  44. Rathinaraj P, Al-Jumaily A., Huh D.S. Internalization: acute apoptosis of breast cancer cells using herceptin-immobilized gold nanoparticles. Breast Cancer: Targets and Therapy 2015; 2015 (7): 51-8. https:// doi.org/10.2147/BCTT.S69834
  45. Acharya S., Fahima D., Sahoo S.K. Targeted epidermal growth factor receptor nanoparticle bioconjugates for breast cancer therapy Biomaterials. 2009; 30 (29): 5737-50. https://doi.org/10.1016/j.bioma-terials.2009.07.008
  46. Iranpour S., Nejati V, Delirezh N., Biparva P, Shirian S. Enhanced stimulation of antibreast cancer T cells responses by dendritic cells loaded with poly lactic-co-glycolic acid (PLGA) nanoparticle encapsulated tumor antigens. J. Exp. Clin. Cancer Res. 2016; 35: 168. https://doi.org/10.1186/ s13046-016-0444-6
  47. Kim S., Park S., Cho M.S., Lim W., Moon B-I., Sung S.H. Strong Correlation of Indoleam-ine 2,3-Dioxygenase 1 Expression with Basal-Like Phenotype and Increased Lymphocytic Infiltration in Triple-Negative Breast Cancer. J. of Cancer. 2017; 8 (1): 124-30. https://doi.org/10.7150/jca.17437
  48. Smith C., Chang M-Y, Parker K., Beury D., DuHadaway J.B., Flick H.E., Boulden J., Sutanto-Ward E., Soler A.P, Laury-Kleintop L. D., Mandik-Nayak L., Metz R., Ostrand-Rosenberg S., Prendergast G.C., Muller A.J. IDO Is a Nodal Pathogenic Driver of Lung Cancer and Metastasis Development. Cancer Discov. 2012; 2 (8): 722-35. https:// doi.org/10.1158/2159-8290.CD-12-0014
  49. Soliman H., Khambati F, Han H.S., Ismail-Khan R., Bui M.M., Sullivan D.M., Antonia S. A phase-1/2 study of adenovirus-p53 transduced dendritic cell vaccine in combination with indoximod in metastatic solid tumors and invasive breast cancer. Oncotarget. 2018; 9 (11): 10110-7. https:// doi.org/10.18632/oncotarget.24118
  50. Jadidi-Niaragh F, Atyabi F, Rastegari A., Kheshtchin N., Arab S., Hassannia H., Ajami M. , Mirsanei Z., Habibi S., Masoumi F, Noor-bakhsh F., Shokri F., Hadjati J. CD73 specific siRNA loaded chitosan lactate nanoparticles potentiate the antitumor effect of a dendritic cell vaccine in 4T1 breast cancer bearing mice. J. Control Release. 2017;
  51. Gall V.A., Philips A.V, Qiao N., Clise-Dwyer K., Perakis A.A., Zhang M., Clifton G.T., Sukhumalchandra P, Ma Q., Reddy S.M., Yu D., Molldrem J.J., Peoples G.E., Alatrash G., Mittendorf E.A. Trastuzumab Increases HER2 Uptake and Cross-Presentation by Dendritic Cells. Cancer Res. 2017; 77 (19): 5374-83. https://doi.org/10.1158/0008-5472.CAN-16-2774

Supplementary files

Supplementary Files
Action
1. JATS XML

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies