Osteoimmuno-Oncology: Therapeutic Opportunities for Targeting Immune Cells in Bone Metastasis
Abstract
:1. Immuno-Oncology and Bone
1.1. Bone Microenvironment
1.2. Bone Marrow Immune Cells
1.3. Osteoimmunology
2. Immune Cells in Bone Metastasis
2.1. T Cells
2.2. NK Cells
2.3. MDSCs
2.4. Macrophages
2.5. Dendritic Cells
2.6. Neutrophils
3. Therapeutic Approaches
3.1. Bisphosphonates
3.2. RANK-Ligand Inhibition
3.3. Radiopharmaceuticals
3.4. Immunotherapies and Their Combinations in Bone Metastatic Patients
4. Concluding Remarks
Author Contributions
Funding
Conflicts of Interest
References
- Allard, B.; Aspeslagh, S.; Garaud, S.; Dupont, F.; Solinas, C.; Kok, M.; Routy, B.; Sotiriou, C.; Stagg, J.; Buisseret, L. Immuno-oncology-101: Overview of major concepts and translational perspectives. Semin. Cancer Biol. 2018, 52, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, J.; Shalabi, A.; Hubbard-Lucey, V. Comprehensive analysis of the clinical immuno-oncology landscape. Ann. Oncol. 2018, 29, 84–91. [Google Scholar] [CrossRef] [PubMed]
- Gwin, W.R., 3rd; Disis, M.L.; Ruiz-Garcia, E. Immuno-Oncology in the Era of Personalized Medicine. Adv. Exp. Med. Biol. 2019, 1168, 117–129. [Google Scholar] [CrossRef] [PubMed]
- Galon, J.; Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 2019, 18, 197–218. [Google Scholar] [CrossRef]
- Bonaventura, P.; Shekarian, T.; Alcazer, V.; Valladeau-Guilemond, J.; Valsesia-Wittmann, S.; Amigorena, S.; Caux, C.; Depil, S. Cold Tumors: A Therapeutic Challenge for Immunotherapy. Front. Immunol. 2019, 10, 168. [Google Scholar] [CrossRef] [Green Version]
- Vareki, S.M. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J. Immunother. Cancer 2018, 6, 157. [Google Scholar] [CrossRef] [PubMed]
- De Guillebon, E.; Dardenne, A.; Saldmann, A.; Séguier, S.; Tran, T.; Paolini, L.; Lebbe, C.; Tartour, E. Beyond the concept of cold and hot tumors for the development of novel predictive biomarkers and the rational design of immunotherapy combination. Int. J. Cancer 2020, 147, 1509–1518. [Google Scholar] [CrossRef] [PubMed]
- Cesano, A.; Warren, S. Bringing the Next Generation of Immuno-Oncology Biomarkers to the Clinic. Biomedicines 2018, 6, 14. [Google Scholar] [CrossRef] [Green Version]
- Marshall, H.T.; Djamgoz, M.B.A. Immuno-Oncology: Emerging Targets and Combination Therapies. Front. Oncol. 2018, 8, 315. [Google Scholar] [CrossRef]
- Pottier, C.; Wheatherspoon, A.; Roncarati, P.; Longuespée, R.; Herfs, M.; Duray, A.; Delvenne, P.; Quatresooz, P. The importance of the tumor microenvironment in the therapeutic management of cancer. Expert Rev. Anticancer Ther. 2015, 15, 943–954. [Google Scholar] [CrossRef]
- Wu, T.; Dai, Y. Tumor microenvironment and therapeutic response. Cancer Lett. 2017, 387, 61–68. [Google Scholar] [CrossRef] [PubMed]
- Zheng, J.; Gao, P. Toward Normalization of the Tumor Microenvironment for Cancer Therapy. Integr. Cancer Ther. 2019, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Suhail, Y.; Cain, M.P.; Vanaja, K.; Kurywchak, P.A.; Levchenko, A.; Kalluri, R. Kshitiz Systems Biology of Cancer Metastasis. Cell Syst. 2019, 9, 109–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pastushenko, I.; Blanpain, C. EMT Transition States during Tumor Progression and Metastasis. Trends Cell Biol. 2019, 29, 212–226. [Google Scholar] [CrossRef] [Green Version]
- Cho, E.S.; Kang, H.E.; Kim, N.H.; Yook, J.I. Therapeutic implications of cancer epithelial-mesenchymal transition (EMT). Arch. Pharmacal Res. 2019, 42, 14–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zavyalova, M.V.; Denisov, E.V.; Tashireva, L.A.; Savelieva, O.E.; Kaigorodova, E.V.; Krakhmal, N.V.; Perelmuter, V.M. Intravasation as a Key Step in Cancer Metastasis. Biochemistry 2019, 84, 762–772. [Google Scholar] [CrossRef]
- Miles, F.L.; Pruitt, F.L.; Van Golen, K.L.; Cooper, C.R. Stepping out of the flow: Capillary extravasation in cancer metastasis. Clin. Exp. Metastasis 2007, 25, 305–324. [Google Scholar] [CrossRef] [PubMed]
- Futakuchi, M.; Fukamachi, K.; Suzui, M. Heterogeneity of tumor cells in the bone microenvironment: Mechanisms and therapeutic targets for bone metastasis of prostate or breast cancer. Adv. Drug Deliv. Rev. 2016, 99, 206–211. [Google Scholar] [CrossRef] [PubMed]
- Fidler, I.J.; Kripke, M.L. The challenge of targeting metastasis. Cancer Metastasis Rev. 2015, 34, 635–641. [Google Scholar] [CrossRef] [Green Version]
- Kauffman, E.C.; Robinson, V.L.; Stadler, W.M.; Sokoloff, M.H.; Rinker-Schaeffer, C.W. Metastasis Suppression: The Evolving Role of Metastasis Suppressor Genes for Regulating Cancer Cell Growth at the Secondary Site. J. Urol. 2003, 169, 1122–1133. [Google Scholar] [CrossRef]
- Brook, N.; Brook, E.; Dharmarajan, A.; Dass, C.R.; Chan, A. Breast cancer bone metastases: Pathogenesis and therapeutic targets. Int. J. Biochem. Cell Biol. 2018, 96, 63–78. [Google Scholar] [CrossRef]
- Logothetis, C.; Morris, M.J.; Den, R.; Coleman, R.E. Current perspectives on bone metastases in castrate-resistant prostate cancer. Cancer Metastasis Rev. 2018, 37, 189–196. [Google Scholar] [CrossRef] [Green Version]
- Hernandez, R.K.; Wade, S.W.; Reich, A.; Pirolli, M.; Liede, A.; Lyman, G.H. Incidence of bone metastases in patients with solid tumors: Analysis of oncology electronic medical records in the United States. BMC Cancer 2018, 18, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fang, J.; Xu, Q. Differences of osteoblastic bone metastases and osteolytic bone metastases in clinical features and molecular characteristics. Clin. Transl. Oncol. 2014, 17, 173–179. [Google Scholar] [CrossRef] [PubMed]
- Coleman, R.E. Clinical Features of Metastatic Bone Disease and Risk of Skeletal Morbidity. Clin. Cancer Res. 2006, 12, 6243s–6249s. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, E.; Xu, H.; Wang, L.; Kryczek, I.; Wu, K.; Hu, Y.; Wang, G.; Zou, W. Bone marrow and the control of immunity. Cell. Mol. Immunol. 2011, 9, 11–19. [Google Scholar] [CrossRef] [Green Version]
- Walsh, M.C.; Takegahara, N.; Kim, H.; Choi, Y. Updating osteoimmunology: Regulation of bone cells by innate and adaptive immunity. Nat. Rev. Rheumatol. 2018, 14, 146–156. [Google Scholar] [CrossRef]
- Reinstein, Z.Z.; Pamarthy, S.; Sagar, V.; Costa, R.; Abdulkadir, S.A.; Giles, F.J.; Carneiro, B.A. Overcoming immunosuppression in bone metastases. Crit. Rev. Oncol. Hematol. 2017, 117, 114–127. [Google Scholar] [CrossRef]
- Holland, A.M.; Zakrzewski, J.L.; Tsai, J.J.; Hanash, A.M.; Dudakov, J.A.; Smith, O.M.; West, M.L.; Singer, N.V.; Brill, J.; Sun, J.C.; et al. Extrathymic development of murine T cells after bone marrow transplantation. J. Clin. Investig. 2012, 122, 4716–4726. [Google Scholar] [CrossRef] [Green Version]
- Cariappa, A.; Chase, C.; Liu, H.; Russell, P.; Pillai, S. Naive recirculating B cells mature simultaneously in the spleen and bone marrow. Blood 2006, 109, 2339–2345. [Google Scholar] [CrossRef] [Green Version]
- Ito, T.; Hamazaki, Y.; Takaori-Kondo, A.; Minato, N. Bone Marrow Endothelial Cells Induce Immature and Mature B Cell Egress in Response to Erythropoietin. Cell Struct. Funct. 2017, 42, 149–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zaretsky, A.G.; Konradt, C.; Dépis, F.; Wing, J.B.; Goenka, R.; Atria, D.G.; Silver, J.S.; Cho, S.; Wolf, A.I.; Quinn, W.J.; et al. T Regulatory Cells Support Plasma Cell Populations in the Bone Marrow. Cell Rep. 2017, 18, 1906–1916. [Google Scholar] [CrossRef] [PubMed]
- Plumb, A.W.; Sheikh, A.; Carlow, D.A.; Patton, D.T.; Ziltener, H.J.; Abraham, N. Interleukin-7 in the transition of bone marrow progenitors to the thymus. Immunol. Cell Biol. 2017, 95, 916–924. [Google Scholar] [CrossRef] [PubMed]
- Fistonich, C.; Zehentmeier, S.; Bednarski, J.J.; Miao, R.; Schjerven, H.; Sleckman, B.P.; Pereira, J.P. Cell circuits between B cell progenitors and IL-7+ mesenchymal progenitor cells control B cell development. J. Exp. Med. 2018, 215, 2586–2599. [Google Scholar] [CrossRef] [Green Version]
- Milford, T.M.; Su, R.J.; Francis, O.L.; Baez, I.; Martinez, S.; Coats, J.S.; Weldon, A.J.; Calderon, M.N.; Nwosu, M.C.; Botimer, A.R.; et al. TSLP or IL-7 provide an IL-7Rα signal that is critical for human B lymphopoiesis. Eur. J. Immunol. 2016, 46, 2155–2161. [Google Scholar] [CrossRef] [Green Version]
- Cui, G.; Hara, T.; Simmons, S.; Wagatsuma, K.; Abe, A.; Miyachi, H.; Kitano, S.; Ishii, M.; Tani-Ichi, S.; Ikuta, K. Characterization of the IL-15 niche in primary and secondary lymphoid organs in vivo. Proc. Natl. Acad. Sci. USA 2014, 111, 1915–1920. [Google Scholar] [CrossRef] [Green Version]
- Quinci, A.C.; Vitale, S.; Parretta, E.; Soriani, A.; Iannitto, M.L.; Cippitelli, M.; Fionda, C.; Bulfone-Paus, S.; Santoni, A.; Di Rosa, F. IL-15 inhibits IL-7Rα expression by memory-phenotype CD8+T cells in the bone marrow. Eur. J. Immunol. 2012, 42, 1129–1139. [Google Scholar] [CrossRef] [PubMed]
- Pelletier, M.; Girard, D. Differential Effects of IL-15 and IL-21 in Myeloid (CD11b+) and Lymphoid (CD11b−) Bone Marrow Cells. J. Immunol. 2006, 177, 100–108. [Google Scholar] [CrossRef]
- Brylka, L.J.; Schinke, T. Chemokines in Physiological and Pathological Bone Remodeling. Front. Immunol. 2019, 10, 2182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arojo, O.A.; Ouyang, X.; Liu, U.; Meng, T.; Kaech, S.M.; Pereira, J.P.; Su, B. Active mTORC2 Signaling in Naive T Cells Suppresses Bone Marrow Homing by Inhibiting CXCR4 Expression. J. Immunol. 2018, 201, 908–915. [Google Scholar] [CrossRef] [Green Version]
- Levy, E.; Reger, R.; Segerberg, F.; Lambert, M.; Leijonhufvud, C.; Baumer, Y.; Carlsten, M.; Childs, R. Enhanced Bone Marrow Homing of Natural Killer Cells Following mRNA Transfection With Gain-of-Function Variant CXCR4R334X. Front. Immunol. 2019, 10, 1262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Petty, J.M.; Lenox, C.C.; Weiss, D.J.; Poynter, M.E.; Suratt, B.T. Crosstalk between CXCR4/Stromal Derived Factor-1 and VLA-4/VCAM-1 Pathways Regulates Neutrophil Retention in the Bone Marrow. J. Immunol. 2009, 182, 604–612. [Google Scholar] [CrossRef] [Green Version]
- Schaumann, D.H.; Tuischer, J.; Ebell, W.; Manz, R.A.; Lauster, R. VCAM-1-positive stromal cells from human bone marrow producing cytokines for B lineage progenitors and for plasma cells: SDF-1, flt3L, and BAFF. Mol. Immunol. 2007, 44, 1606–1612. [Google Scholar] [CrossRef]
- Okamoto, K.; Takayanagi, H. Osteoimmunology. Cold Spring Harb. Perspect. Med. 2018, 9, a031245. [Google Scholar] [CrossRef]
- Tsukasaki, M.; Takayanagi, H. Osteoimmunology: Evolving concepts in bone–immune interactions in health and disease. Nat. Rev. Immunol. 2019, 19, 626–642. [Google Scholar] [CrossRef] [PubMed]
- Su, P.; Tian, Y.; Yang, C.; Ma, X.; Wang, X.; Pei, J.; Qian, A. Mesenchymal Stem Cell Migration during Bone Formation and Bone Diseases Therapy. Int. J. Mol. Sci. 2018, 19, 2343. [Google Scholar] [CrossRef] [Green Version]
- Morrison, S.J.; Scadden, D.T. The bone marrow niche for haematopoietic stem cells. Nat. Cell Biol. 2014, 505, 327–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Feuerbach, D.; Feyen, J.H. Expression of the cell-adhesion molecule VCAM-1 by stromal cells is necessary for osteoclastogenesis. FEBS Lett. 1997, 402, 21–24. [Google Scholar] [CrossRef] [Green Version]
- Fujii, Y.; Fujii, K.; Nakano, K.; Tanaka, Y. Crosslinking of CD44 on human osteoblastic cells upregulates ICAM-1 and VCAM-1. FEBS Lett. 2003, 539, 45–50. [Google Scholar] [CrossRef] [Green Version]
- Blin-Wakkach, C.; Rouleau, M.; Wakkach, A. Roles of osteoclasts in the control of medullary hematopoietic niches. Arch. Biochem. Biophys. 2014, 561, 29–37. [Google Scholar] [CrossRef]
- Croes, M.; Öner, F.C.; van Neerven, D.; Sabir, E.; Kruyt, M.C.; Blokhuis, T.J.; Dhert, W.J.; Alblas, J. Proinflammatory T cells and IL-17 stimulate osteoblast differentiation. Bone 2016, 84, 262–270. [Google Scholar] [CrossRef]
- Qiu, X.; Gui, Y.; Xu, Y.; Li, D.; Wang, L. DHEA promotes osteoblast differentiation by regulating the expression of osteoblast-related genes and Foxp3(+) regulatory T cells. Biosci. Trends 2015, 9, 307–314. [Google Scholar] [CrossRef] [Green Version]
- Li, S.; Li, T.; Chen, Y.; Nie, Y.; Li, C.; Liu, L.; Li, Q.; Qiu, L. Granulocyte Colony-Stimulating Factor Induces Osteoblast Inhibition by B Lymphocytes and Osteoclast Activation by T Lymphocytes during Hematopoietic Stem/Progenitor Cell Mobilization. Biol. Blood Marrow Transpl. 2015, 21, 1384–1391. [Google Scholar] [CrossRef] [Green Version]
- Panaroni, C.; Wu, J.Y. Interactions Between B Lymphocytes and the Osteoblast Lineage in Bone Marrow. Calcif. Tissue Int. 2013, 93, 261–268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cain, C.J.; Rueda, R.; McLelland, B.; Collette, N.M.; Loots, G.G.; Manilay, J.O. Absence of sclerostin adversely affects B-cell survival. J. Bone Miner. Res. 2012, 27, 1451–1461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mansour, A.; Anginot, A.; Mancini, S.J.C.; Schiff, C.; Carle, G.F.; Wakkach, A.; Blin-Wakkach, C. Osteoclast activity modulates B-cell development in the bone marrow. Cell Res. 2011, 21, 1102–1115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Panaroni, C.; Fulzele, K.; Saini, V.; Chubb, R.; Pajevic, P.D.; Wu, J.Y. PTH Signaling in Osteoprogenitors Is Essential for B-Lymphocyte Differentiation and Mobilization. J. Bone Miner. Res. 2015, 30, 2273–2286. [Google Scholar] [CrossRef] [Green Version]
- Takayanagi, H.; Ogasawara, K.; Hida, S.; Chiba, T.; Murata, S.; Sato, K.; Takaoka, A.; Yokochi, T.; Oda, H.; Tanaka, K.; et al. T-cell-mediated regulation of osteoclastogenesis by signalling cross-talk between RANKL and IFN-γ. Nature 2000, 408, 600–605. [Google Scholar] [CrossRef]
- Fumoto, T.; Takeshita, S.; Ito, M.; Ikeda, K. Physiological Functions of Osteoblast Lineage and T Cell-Derived RANKL in Bone Homeostasis. J. Bone Miner. Res. 2014, 29, 830–842. [Google Scholar] [CrossRef]
- Zhang, J.; Qiu, X.; Zhang, N.; Tang, W.; Gober, H.; Li, D.; Wang, L. Bu-Shen-Ning-Xin decoction suppresses osteoclastogenesis by modulating RANKL/OPG imbalance in the CD4+ T lymphocytes of ovariectomized mice. Int. J. Mol. Med. 2018, 42, 299–308. [Google Scholar] [CrossRef]
- Manabe, N.; Kawaguchi, H.; Chikuda, H.; Miyaura, C.; Inada, M.; Nagai, R.; Nabeshima, Y.-I.; Nakamura, K.; Sinclair, A.M.; Scheuermann, R.H.; et al. Connection Between B Lymphocyte and Osteoclast Differentiation Pathways. J. Immunol. 2001, 167, 2625–2631. [Google Scholar] [CrossRef] [PubMed]
- Francisconi, C.; Vieira, A.E.; Azevedo, M.D.C.S.; Tabanez, A.; Fonseca, A.; Trombone, A.; Letra, A.; Silva, R.; Sfeir, C.; Little, S.; et al. RANKL Triggers Treg-Mediated Immunoregulation in Inflammatory Osteolysis. J. Dent. Res. 2018, 97, 917–927. [Google Scholar] [CrossRef] [PubMed]
- Tilkeridis, K.; Kiziridis, G.; Ververidis, A.; Papoutselis, M.; Kotsianidis, I.; Kitsikidou, G.; Tousiaki, N.-E.; Drosos, G.; Kapetanou, A.; Rechova, K.V.; et al. Immunoporosis: A New Role for Invariant Natural Killer T (NKT) Cells Through Overexpression of Nuclear Factor-κB Ligand (RANKL). Med. Sci. Monit. 2019, 25, 2151–2158. [Google Scholar] [CrossRef] [PubMed]
- Horowitz, M.C.; Bothwell, A.L.M.; Hesslein, D.G.T.; Pflugh, D.L.; Schatz, D.G. B cells and osteoblast and osteoclast development. Immunol. Rev. 2005, 208, 141–153. [Google Scholar] [CrossRef]
- Fujiwara, Y.; Piemontese, M.; Liu, Y.; Thostenson, J.D.; Xiong, J.; O’Brien, C.A. RANKL (Receptor Activator of NFκB Ligand) Produced by Osteocytes Is Required for the Increase in B Cells and Bone Loss Caused by Estrogen Deficiency in Mice. J. Biol. Chem. 2016, 291, 24838–24850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.-H.; Kim, T.-S.; Choi, Y.-W.; Lorenzo, J. Osteoimmunology: Cytokines and the skeletal system. BMB Rep. 2008, 41, 495–510. [Google Scholar] [CrossRef] [Green Version]
- Coleman, R.E.; Croucher, P.I.; Padhani, A.R.; Clézardin, P.; Chow, E.; Fallon, M.; Guise, T.; Colangeli, S.; Capanna, R.; Costa, L. Bone metastases. Nat. Rev. Dis. Prim. 2020, 6, 1–28. [Google Scholar] [CrossRef]
- Owen, K.L.; Parker, B.S. Beyond the vicious cycle: The role of innate osteoimmunity, automimicry and tumor-inherent changes in dictating bone metastasis. Mol. Immunol. 2019, 110, 57–68. [Google Scholar] [CrossRef]
- Antonarakis, E.S.; Piulats, J.M.; Gross-Goupil, M.; Goh, J.; Ojamaa, K.; Hoimes, C.J.; Vaishampayan, U.; Berger, R.; Sezer, A.; Alanko, T.; et al. Pembrolizumab for Treatment-Refractory Metastatic Castration-Resistant Prostate Cancer: Multicohort, Open-Label Phase II KEYNOTE-199 Study. J. Clin. Oncol. 2020, 38, 395–405. [Google Scholar] [CrossRef]
- Luo, G.; He, Y.; Zhao, Q.; Yu, X. Immune Cells Act as Promising Targets for the Treatment of Bone Metastasis. Recent Pat. Anti-Cancer Drug Discov. 2017, 12, 221–233. [Google Scholar] [CrossRef]
- Fournier, P.; Chirgwin, J.M.; Guise, T.A. New insights into the role of T cells in the vicious cycle of bone metastases. Curr. Opin. Rheumatol. 2006, 18, 396–404. [Google Scholar] [CrossRef]
- Li, Z.; Zhang, L.-J.; Zhang, H.-R.; Tian, G.-F.; Tian, J.; Mao, X.-L.; Jia, Z.-H.; Meng, Z.-Y.; Zhao, L.-Q.; Yin, Z.-N.; et al. Tumor-derived transforming growth factor-β is critical for tumor progression and evasion from immune surveillance. Asian Pac. J. Cancer Prev. 2014, 15, 5181–5186. [Google Scholar] [CrossRef] [Green Version]
- D’Amico, L.; Roato, I. The Impact of Immune System in Regulating Bone Metastasis Formation by Osteotropic Tumors. J. Immunol. Res. 2015, 2015, 143526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, H.Y.; Zhou, J.; Zhu, K.; Riker, A.I.; Marincola, F.M.; Wang, R.-F. Identification of a Mutated Fibronectin As a Tumor Antigen Recognized by CD4+T Cells: Its Role in Extracellular Matrix Formation and Tumor Metastasis. J. Exp. Med. 2002, 195, 1397–1406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bidwell, B.N.; Slaney, C.Y.; Withana, N.P.; Forster, S.; Cao, Y.; Loi, S.; Andrews, D.; Mikeska, T.; Mangan, N.E.; Samarajiwa, S.A.; et al. Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape. Nat. Med. 2012, 18, 1224–1231. [Google Scholar] [CrossRef] [PubMed]
- Lengagne, R.; Graff-Dubois, S.; Garcette, M.; Renia, L.; Kato, M.; Guillet, J.-G.; Engelhard, V.H.; Avril, M.-F.; Abastado, J.-P.; Prévost-Blondel, A. Distinct role for CD8 T cells toward cutaneous tumors and visceral metastases. J. Immunol. 2007, 180, 130–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vidotto, T.; Saggioro, F.P.; Jamaspishvili, T.; Chesca, D.L.; De Albuquerque, C.G.P.; Reis, R.B.; Graham, C.H.; Berman, D.M.; Siemens, D.R.; Squire, J.A.; et al. PTEN-deficient prostate cancer is associated with an immunosuppressive tumor microenvironment mediated by increased expression of IDO1 and infiltrating FoxP3+ T regulatory cells. Prostate 2019, 79, 969–979. [Google Scholar] [CrossRef]
- Xiang, L.; Gilkes, D.M. The Contribution of the Immune System in Bone Metastasis Pathogenesis. Int. J. Mol. Sci. 2019, 20, 999. [Google Scholar] [CrossRef] [Green Version]
- Barnett, J.C.; Bean, S.M.; Whitaker, R.S.; Kondoh, E.; Baba, T.; Fujii, S.; Marks, J.R.; Dressman, H.K.; Murphy, S.; Berchuck, A. Ovarian cancer tumor infiltrating T-regulatory (Treg) cells are associated with a metastatic phenotype. Gynecol. Oncol. 2010, 116, 556–562. [Google Scholar] [CrossRef]
- Karavitis, J.; Hix, L.M.; Shi, Y.H.; Schultz, R.F.; Khazaie, K.; Zhang, M. Regulation of COX2 Expression in Mouse Mammary Tumor Cells Controls Bone Metastasis and PGE2-Induction of Regulatory T Cell Migration. PLoS ONE 2012, 7, e46342. [Google Scholar] [CrossRef]
- Hix, L.M.; Shi, Y.H.; Brutkiewicz, R.R.; Stein, P.L.; Wang, C.-R.; Zhang, M. CD1d-Expressing Breast Cancer Cells Modulate NKT Cell-Mediated Antitumor Immunity in a Murine Model of Breast Cancer Metastasis. PLoS ONE 2011, 6, e20702. [Google Scholar] [CrossRef]
- Liu, G.; Lu, S.; Wang, X.; Page, S.T.; Higano, C.S.; Plymate, S.R.; Greenberg, N.M.; Sun, S.; Li, Z.; Wu, J.D. Perturbation of NK cell peripheral homeostasis accelerates prostate carcinoma metastasis. J. Clin. Investig. 2013, 123, 4410–4422. [Google Scholar] [CrossRef] [PubMed]
- Mittal, D.; Vijayan, D.; Putz, E.M.; Aguilera, A.R.; Markey, K.A.; Straube, J.; Kazakoff, S.; Nutt, S.L.; Takeda, K.; Hill, G.R.; et al. Interleukin-12 from CD103+ Batf3-Dependent Dendritic Cells Required for NK-Cell Suppression of Metastasis. Cancer Immunol. Res. 2017, 5, 1098–1108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jewett, A.; Kos, J.; Fong, Y.; Ko, M.-W.; Safaei, T.; Nanut, M.P.; Kaur, K. NK cells shape pancreatic and oral tumor microenvironments; role in inhibition of tumor growth and metastasis. Semin. Cancer Biol. 2018, 53, 178–188. [Google Scholar] [CrossRef] [PubMed]
- Rautela, J.; Baschuk, N.; Slaney, C.Y.; Jayatilleke, K.M.; Xiao, K.; Bidwell, B.N.; Lucas, E.C.; Hawkins, E.; Lock, P.; Wong, C.S.; et al. Loss of Host Type-I IFN Signaling Accelerates Metastasis and Impairs NK-cell Antitumor Function in Multiple Models of Breast Cancer. Cancer Immunol. Res. 2015, 3, 1207–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Ding, Y.; Guo, N.; Wang, S. MDSCs: Key Criminals of Tumor Pre-metastatic Niche Formation. Front. Immunol. 2019, 10, 172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Souza-Fonseca-Guimaraes, F.; Young, A.; Mittal, D.; Martinet, L.; Bruedigam, C.; Takeda, K.; Andoniou, C.; Degli-Esposti, M.A.; Hill, G.; Smyth, M.J. NK cells require IL-28R for optimal in vivo activity. Proc. Natl. Acad. Sci. USA 2015, 112, E2376–E2384. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.; Yano, S.; Miki, T.; Goto, H.; Kanematsu, T.; Muguruma, H.; Uehara, H.; Sone, S. A novel bisphosphonate minodronate (YM529) specifically inhibits osteolytic bone metastasis produced by human small-cell lung cancer cells in NK-cell depleted SCID mice. Clin. Exp. Metastasis 2003, 20, 153–159. [Google Scholar] [CrossRef] [PubMed]
- Bottos, A.; Gotthardt, D.; Gill, J.W.; Gattelli, A.; Frei, A.; Tzankov, A.; Sexl, V.; Wodnar-Filipowicz, A.; Hynes, N.E. Decreased NK-cell tumour immunosurveillance consequent to JAK inhibition enhances metastasis in breast cancer models. Nat. Commun. 2016, 7, 12258. [Google Scholar] [CrossRef] [Green Version]
- Biswas, S.; Nyman, J.S.; Alvarez, J.; Chakrabarti, A.; Ayres, A.; Sterling, J.; Edwards, J.; Rana, T.; Johnson, R.; Perrien, D.S.; et al. Anti-Transforming Growth Factor ß Antibody Treatment Rescues Bone Loss and Prevents Breast Cancer Metastasis to Bone. PLoS ONE 2011, 6, e27090. [Google Scholar] [CrossRef]
- Ye, X.-Z.; Yu, S.-C.; Bian, X.-W. Contribution of myeloid-derived suppressor cells to tumor-induced immune suppression, angiogenesis, invasion and metastasis. J. Genet. Genom. 2010, 37, 423–430. [Google Scholar] [CrossRef]
- Bergenfelz, C.; Roxå, A.; Mehmeti, M.; Leandersson, K.; Larsson, A.-M. Clinical relevance of systemic monocytic-MDSCs in patients with metastatic breast cancer. Cancer Immunol. Immunother. 2020, 69, 435–448. [Google Scholar] [CrossRef] [Green Version]
- Bosiljcic, M.; Cederberg, R.A.; Hamilton, M.J.; LePard, N.E.; Harbourne, B.T.; Collier, J.L.; Halvorsen, E.C.; Shi, R.; Franks, S.E.; Kim, A.Y.; et al. Targeting myeloid-derived suppressor cells in combination with primary mammary tumor resection reduces metastatic growth in the lungs. Breast Cancer Res. 2019, 21, 103. [Google Scholar] [CrossRef]
- Wei, W.-C.; Lin, S.-Y.; Lan, C.-W.; Huang, Y.-C.; Lin, C.-Y.; Hsiao, P.-W.; Chen, Y.-R.; Yang, W.-C.; Yang, N.-S. Inhibiting MDSC differentiation from bone marrow with phytochemical polyacetylenes drastically impairs tumor metastasis. Sci. Rep. 2016, 6, 36663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cao, Y.; Slaney, C.Y.; Bidwell, B.N.; Parker, B.S.; Johnstone, C.N.; Rautela, J.; Eckhardt, B.L.; Anderson, R.L. BMP4 Inhibits Breast Cancer Metastasis by Blocking Myeloid-Derived Suppressor Cell Activity. Cancer Res. 2014, 74, 5091–5102. [Google Scholar] [CrossRef] [Green Version]
- Ma, Y.; Zhao, N.; Liu, G. Conjugate (MTC-220) of Muramyl Dipeptide Analogue and Paclitaxel Prevents Both Tumor Growth and Metastasis in Mice. J. Med. Chem. 2011, 54, 2767–2777. [Google Scholar] [CrossRef]
- Moon, E.-Y.; Ryu, Y.-K.; Lee, G.-H. Dexamethasone inhibits in vivo tumor growth by the alteration of bone marrow CD11b+ myeloid cells. Int. Immunopharmacol. 2014, 21, 494–500, Erratum in 2014, 22, 526–527. [Google Scholar] [CrossRef] [PubMed]
- Sawant, A.; Ponnazhagan, S. Myeloid-Derived Suppressor Cells as Osteoclast Progenitors: A Novel Target for Controlling Osteolytic Bone Metastasis. Cancer Res. 2013, 73, 4606–4610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martínez, V.G.; Rubio, C.; Martínez-Fernández, M.; Segovia, C.; López-Calderón, F.; Garín, M.I.; Teijeira, A.; Munera-Maravilla, E.; Varas, A.; Sacedón, R.; et al. BMP4 Induces M2 Macrophage Polarization and Favors Tumor Progression in Bladder Cancer. Clin. Cancer Res. 2017, 23, 7388–7399. [Google Scholar] [CrossRef] [Green Version]
- Niu, Z.; Shi, Q.; Zhang, W.; Shu, Y.; Yang, N.; Chen, B.; Wang, Q.; Zhao, X.; Chen, J.; Cheng, N.; et al. Caspase-1 cleaves PPARγ for potentiating the pro-tumor action of TAMs. Nat. Commun. 2017, 8, 766. [Google Scholar] [CrossRef] [Green Version]
- Buddingh, E.; Kuijjer, M.; Duim, R.A.; Bürger, H.; Agelopoulos, K.; Myklebost, O.; Serra, M.; Mertens, F.; Hogendoorn, P.; Lankester, A.C.; et al. Tumor-Infiltrating Macrophages Are Associated with Metastasis Suppression in High-Grade Osteosarcoma: A Rationale for Treatment with Macrophage Activating Agents. Clin. Cancer Res. 2011, 17, 2110–2119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takiguchi, S.; Korenaga, N.; Inoue, K.; Sugi, E.; Kataoka, Y.; Matsusue, K.; Futagami, K.; Li, Y.-J.; Kukita, T.; Teramoto, N.; et al. Involvement of CXCL14 in osteolytic bone metastasis from lung cancer. Int. J. Oncol. 2014, 44, 1316–1324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hiraoka, K.; Zenmyo, M.; Watari, K.; Iguchi, H.; Fotovati, A.; Kimura, Y.N.; Hosoi, F.; Shoda, T.; Nagata, K.; Osada, H.; et al. Inhibition of bone and muscle metastases of lung cancer cells by a decrease in the number of monocytes/macrophages. Cancer Sci. 2008, 99, 1595–1602. [Google Scholar] [CrossRef] [PubMed]
- Jones, J.; Sinder, B.; Paige, D.; Soki, F.; Koh, A.; Thiele, S.; Shiozawa, Y.; Hofbauer, L.; Daignault, S.; Roca, H.; et al. Trabectedin Reduces Skeletal Prostate Cancer Tumor Size in Association with Effects on M2 Macrophages and Efferocytosis. Neoplasia 2019, 21, 172–184. [Google Scholar] [CrossRef]
- Jing, W.; Guo, X.; Wang, G.; Bi, Y.; Han, L.; Zhu, Q.; Qiu, C.; Tanaka, M.; Zhao, Y. Breast cancer cells promote CD169+ macrophage-associated immunosuppression through JAK2-mediated PD-L1 upregulation on macrophages. Int. Immunopharmacol. 2020, 78, 106012. [Google Scholar] [CrossRef]
- Shrivastava, R.; Asif, M.; Singh, V.; Dubey, P.; Malik, S.A.; Lone, M.-U.-D.; Tewari, B.N.; Baghel, K.S.; Pal, S.; Nagar, G.K.; et al. M2 polarization of macrophages by Oncostatin M in hypoxic tumor microenvironment is mediated by mTORC2 and promotes tumor growth and metastasis. Cytokine 2019, 118, 130–143. [Google Scholar] [CrossRef]
- Wu, A.C.; He, Y.; Broomfield, A.; Paatan, N.J.; Harrington, B.S.; Tseng, H.-W.; Beaven, E.A.; Kiernan, D.M.; Swindle, P.; Clubb, A.B.; et al. CD169+macrophages mediate pathological formation of woven bone in skeletal lesions of prostate cancer. J. Pathol. 2016, 239, 218–230. [Google Scholar] [CrossRef] [Green Version]
- Tourkova, I.L.; Yamabe, K.; Chatta, G.; Shurin, G.V.; Shurin, M. NK Cells Mediate Flt3 Ligand-Induced Protection of Dendritic Cell Precursors In Vivo from the Inhibition by Prostate Carcinoma in the Murine Bone Marrow Metastasis Model. J. Immunother. 2003, 26, 468–472. [Google Scholar] [CrossRef]
- Imai, K.; Minamiya, Y.; Koyota, S.; Ito, M.; Saito, H.; Satoru, M.; Motoyama, S.; Sugiyama, T.; Ogawa, J.-I. Inhibition of dendritic cell migration by transforming growth factor-β1 increases tumor-draining lymph node metastasis. J. Exp. Clin. Cancer Res. 2012, 31, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tanaka, H.; Shinto, O.; Yashiro, M.; Yamazoe, S.; Iwauchi, T.; Muguruma, K.; Kubo, N.; Ohira, M.; Hirakawa, K. Transforming growth factor β signaling inhibitor, SB-431542, induces maturation of dendritic cells and enhances anti-tumor activity. Oncol. Rep. 2010, 24, 1637–1643. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.; Li, J.; Fan, Y.; Chang, K.; Yang, X.; Zhu, W.; Wu, X.; Pang, Y. Concurrent dendritic cell vaccine and strontium-89 radiation therapy in the management of multiple bone metastases. Ir. J. Med Sci. 2014, 184, 457–461. [Google Scholar] [CrossRef]
- Kawano, M.; Itonaga, I.; Iwasaki, T.; Tsumura, H. Enhancement of antitumor immunity by combining anti-cytotoxic T lymphocyte antigen-4 antibodies and cryotreated tumor lysate-pulsed dendritic cells in murine osteosarcoma. Oncol. Rep. 2013, 29, 1001–1006. [Google Scholar] [CrossRef] [Green Version]
- Bonavita, O.; Massara, M.; Bonecchi, R. Chemokine regulation of neutrophil function in tumors. Cytokine Growth Factor Rev. 2016, 30, 81–86. [Google Scholar] [CrossRef]
- Massara, M.; Bonavita, O.; Savino, B.; Caronni, N.; Poeta, V.M.; Sironi, M.; Setten, E.; Recordati, C.; Crisafulli, L.; Ficara, F.; et al. ACKR2 in hematopoietic precursors as a checkpoint of neutrophil release and anti-metastatic activity. Nat. Commun. 2018, 9, 676. [Google Scholar] [CrossRef] [Green Version]
- Jablonska, J.; Lang, S.; Sionov, R.V.; Granot, Z. The regulation of pre-metastatic niche formation by neutrophils. Oncotarget 2017, 8, 112132–112144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patel, S.; Fu, S.; Mastio, J.; Dominguez, G.A.; Purohit, A.; Kossenkov, A.; Lin, C.; Alicea-Torres, K.; Sehgal, M.; Nefedova, Y.; et al. Unique pattern of neutrophil migration and function during tumor progression. Nat. Immunol. 2018, 19, 1236–1247. [Google Scholar] [CrossRef] [PubMed]
- Costanzo-Garvey, D.L.; Keeley, T.; Case, A.J.; Watson, G.F.; Alsamraae, M.; Yu, Y.; Su, K.; Heim, C.E.; Kielian, T.; Morrissey, C.; et al. Neutrophils are mediators of metastatic prostate cancer progression in bone. Cancer Immunol. Immunother. 2020, 69, 1113–1130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marin-Acevedo, J.A.; Soyano, A.E.; Dholaria, B.; Knutson, K.L.; Lou, Y. Cancer immunotherapy beyond immune checkpoint inhibitors. J. Hematol. Oncol. 2018, 11, 8. [Google Scholar] [CrossRef] [PubMed]
- Stefanovic, S.; Schuetz, F.; Sohn, C.; Beckhove, P.; Domschke, C. Adoptive immunotherapy of metastatic breast cancer: Present and future. Cancer Metastasis Rev. 2013, 33, 309–320. [Google Scholar] [CrossRef] [PubMed]
- Monteiro, A.C.; Leal, A.C.; Gonçalves-Silva, T.; Mercadante, A.C.T.; Kestelman, F.; Chaves, S.; Azevedo, R.B.; Monteiro, J.P.; Bonomo, A. T Cells Induce Pre-Metastatic Osteolytic Disease and Help Bone Metastases Establishment in a Mouse Model of Metastatic Breast Cancer. PLoS ONE 2013, 8, e68171. [Google Scholar] [CrossRef] [Green Version]
- Correale, P.; Micheli, L.; Del Vecchio, M.T.; Sabatino, M.; Petrioli, R.; Pozzessere, D.; Marsili, S.; Giorgi, G.; Lozzi, L.; Neri, P.; et al. A parathyroid-hormone-related-protein (PTH-rP)-specific cytotoxic T cell response induced by in vitro stimulation of tumour-infiltrating lymphocytes derived from prostate cancer metastases, with epitope peptide-loaded autologous dendritic cells and low-dose IL-2. Br. J. Cancer 2001, 85, 1722–1730. [Google Scholar] [CrossRef]
- Domschke, C.; Ge, Y.; Bernhardt, I.; Schott, S.; Keim, S.; Juenger, S.; Bucur, M.; Mayer, L.; Blumenstein, M.; Rom, J.; et al. Long-term survival after adoptive bone marrow T cell therapy of advanced metastasized breast cancer: Follow-up analysis of a clinical pilot trial. Cancer Immunol. Immunother. 2013, 62, 1053–1060. [Google Scholar] [CrossRef] [PubMed]
- Juric, V.; O’Sullivan, C.; Stefanutti, E.; Kovalenko, M.; Greenstein, A.; Barry-Hamilton, V.; Mikaelian, I.; Degenhardt, J.; Yue, P.; Smith, V.; et al. MMP-9 inhibition promotes anti-tumor immunity through disruption of biochemical and physical barriers to T-cell trafficking to tumors. PLoS ONE 2018, 13, e0207255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brinkrolf, P.; Landmeier, S.; Altvater, B.; Chen, C.; Pscherer, S.; Rosemann, A.; Ranft, A.; Dirksen, U.; Juergens, H.; Rossig, C. A high proportion of bone marrow T cells with regulatory phenotype (CD4+CD25hiFoxP3+) in Ewing sarcoma patients is associated with metastatic disease. Int. J. Cancer 2009, 125, 879–886. [Google Scholar] [CrossRef]
- Roato, I.; Vitale, M. The Uncovered Role of Immune Cells and NK Cells in the Regulation of Bone Metastasis. Front. Endocrinol. 2019, 10, 145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Verhoeven, D.H.; de Hooge, A.S.; Mooiman, E.C.; Santos, S.J.; Dam, M.M.T.; Gelderblom, H.; Melief, C.J.; Hogendoorn, P.C.; Egeler, R.M.; van Tol, M.J.; et al. NK cells recognize and lyse Ewing sarcoma cells through NKG2D and DNAM-1 receptor dependent pathways. Mol. Immunol. 2008, 45, 3917–3925. [Google Scholar] [CrossRef]
- Zhang, H.; Vijayan, D.; Li, X.-Y.; Robson, S.C.; Geetha, N.; Teng, M.W.L.; Smyth, M.J. The role of NK cells and CD39 in the immunological control of tumor metastases. OncoImmunology 2019, 8, e1593809. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Y.; Chen, W.; Zhu, W.; Meng, H.; Chen, J.; Zhang, J. Overexpression of Interferon Regulatory Factor 7 (IRF7) Reduces Bone Metastasis of Prostate Cancer Cells in Mice. Oncol. Res. Featur. Preclin. Clin. Cancer Ther. 2017, 25, 511–522. [Google Scholar] [CrossRef]
- Miki, T.; Yano, S.; Hanibuchi, M.; Kanematsu, T.; Muguruma, H.; Sone, S. Parathyroid hormone-related protein (PTHrP) is responsible for production of bone metastasis, but not visceral metastasis, by human small cell lung cancer SBC-5 cells in natural killer cell-depleted SCID mice. Int. J. Cancer 2004, 108, 511–515. [Google Scholar] [CrossRef]
- Ogino, H.; Yano, S.; Kakiuchi, S.; Muguruma, H.; Ikuta, K.; Hanibuchi, M.; Uehara, H.; Tsuchida, K.; Sugino, H.; Sone, S. Follistatin Suppresses the Production of Experimental Multiple-Organ Metastasis by Small Cell Lung Cancer Cells in Natural Killer Cell–Depleted SCID Mice. Clin. Cancer Res. 2008, 14, 660–667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Otsuka, S.; Hanibuchi, M.; Ikuta, K.; Yano, S.; Goto, H.; Ogino, H.; Yamada, T.; Kakiuchi, S.; Nishioka, Y.; Takahashi, T.; et al. A Bone Metastasis Model With Osteolytic and Osteoblastic Properties of Human Lung Cancer ACC-LC-319/bone2 in Natural Killer Cell-Depleted Severe Combined Immunodeficient Mice. Oncol. Res. Featur. Preclin. Clin. Cancer Ther. 2009, 17, 581–591. [Google Scholar] [CrossRef]
- Arlauckas, S.P.; Garren, S.B.; Garris, C.S.; Kohler, R.H.; Oh, J.; Pittet, M.J.; Weissleder, R. Arg1 expression defines immunosuppressive subsets of tumor-associated macrophages. Theranostics 2018, 8, 5842–5854. [Google Scholar] [CrossRef]
- Jayaraman, P.; Parikh, F.; Lopez-Rivera, E.; Hailemichael, Y.; Clark, A.; Ma, G.; Cannan, D.; Ramacher, M.; Kato, M.; Overwijk, W.W.; et al. Tumor-Expressed Inducible Nitric Oxide Synthase Controls Induction of Functional Myeloid-Derived Suppressor Cells through Modulation of Vascular Endothelial Growth Factor Release. J. Immunol. 2012, 188, 5365–5376. [Google Scholar] [CrossRef] [PubMed]
- Wink, D.A.; Ridnour, L.A.; Cheng, R.; Switzer, C.W.; Glynn, S.; Ambs, S. The Oncogenic Properties Of The Redox Inflammatory Protein Inducible Nitric Oxide Synthase In ER(-) Breast Cancer. Redox Biol. 2015, 5, 413. [Google Scholar] [CrossRef] [Green Version]
- Groth, C.; Hu, X.; Weber, R.; Fleming, V.; Altevogt, P.; Utikal, J.; Umansky, V. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br. J. Cancer 2019, 120, 16–25. [Google Scholar] [CrossRef] [Green Version]
- Li, B.-H.; Garstka, M.A.; Li, Z.-F. Chemokines and their receptors promoting the recruitment of myeloid-derived suppressor cells into the tumor. Mol. Immunol. 2020, 117, 201–215. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Pang, Y.; Xie, T.; Zhu, L. CXCR4 antagonism in combination with IDO1 inhibition weakens immune suppression and inhibits tumor growth in mouse breast cancer bone metastases. OncoTargets Ther. 2019, 12, 4985–4992. [Google Scholar] [CrossRef] [Green Version]
- Danilin, S.; Merkel, A.; Johnson, J.R.; Johnson, R.W.; Edwards, J.R.; Sterling, J.A. Myeloid-derived suppressor cells expand during breast cancer progression and promote tumor-induced bone destruction. OncoImmunology 2012, 1, 1484–1494. [Google Scholar] [CrossRef] [Green Version]
- Law, A.M.K.; Valdes-Mora, F.; Gallego-Ortega, D. Myeloid-Derived Suppressor Cells as a Therapeutic Target for Cancer. Cells 2020, 9, 561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sousa, S.; Määttä, J. The role of tumour-associated macrophages in bone metastasis. J. Bone Oncol. 2016, 5, 135–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, K.; Lin, K.; Li, X.; Yuan, X.; Xu, P.; Ni, P.; Xu, D. Redefining Tumor-Associated Macrophage Subpopulations and Functions in the Tumor Microenvironment. Front. Immunol. 2020, 11, 1731. [Google Scholar] [CrossRef] [PubMed]
- Cho, H.; Seo, Y.; Loke, K.M.; Kim, S.-W.; Oh, S.-M.; Kim, J.-H.; Soh, J.; Kim, H.S.; Lee, H.; Kim, J.; et al. Cancer-Stimulated CAFs Enhance Monocyte Differentiation and Protumoral TAM Activation via IL6 and GM-CSF Secretion. Clin. Cancer Res. 2018, 24, 5407–5421. [Google Scholar] [CrossRef] [PubMed]
- Mendoza-Reinoso, V.; McCauley, L.K.; Fournier, P.G. Contribution of Macrophages and T Cells in Skeletal Metastasis. Cancers 2020, 12, 1014. [Google Scholar] [CrossRef]
- Lee, G.T.; Kwon, S.J.; Kim, J.; Kwon, Y.S.; Lee, N.; Hong, J.H.; Jamieson, C.; Kim, W.-J.; Kim, I.Y. WNT5A induces castration-resistant prostate cancer via CCL2 and tumour-infiltrating macrophages. Br. J. Cancer 2018, 118, 670–678. [Google Scholar] [CrossRef] [Green Version]
- Jiang, P.; Gao, W.; Ma, T.; Wang, R.; Piao, Y.; Dong, X.; Wang, P.; Zhang, X.; Liu, Y.; Su, W.; et al. CD137 promotes bone metastasis of breast cancer by enhancing the migration and osteoclast differentiation of monocytes/macrophages. Theranostics 2019, 9, 2950–2966. [Google Scholar] [CrossRef]
- Haegel, H.; Thioudellet, C.; Hallet, R.; Geist, M.; Menguy, T.; Le Pogam, F.; Marchand, J.-B.; Toh, M.-L.; Duong, V.; Calcei, A.; et al. A unique anti-CD115 monoclonal antibody which inhibits osteolysis and skews human monocyte differentiation from M2-polarized macrophages toward dendritic cells. mAbs 2013, 5, 736–747. [Google Scholar] [CrossRef] [Green Version]
- Kim, A.; Noh, Y.-W.; Kim, K.D.; Jang, Y.S.; Choe, Y.-K.; Lim, J.-S. Activated natural killer cell-mediated immunity is required for the inhibition of tumor metastasis by dendritic cell vaccination. Exp. Mol. Med. 2004, 36, 428–443. [Google Scholar] [CrossRef]
- Morse, M.A.; Coleman, R.E.; Akabani, G.; Niehaus, N.; Coleman, D.; Lyerly, H.K. Migration of human dendritic cells after injection in patients with metastatic malignancies. Cancer Res. 1999, 59, 56–58. [Google Scholar]
- Engblom, C.; Pfirschke, C.; Zilionis, R.; Martins, J.D.S.; Bos, S.A.; Courties, G.; Rickelt, S.; Severe, N.; Baryawno, N.; Faget, J.; et al. Osteoblasts remotely supply lung tumors with cancer-promoting SiglecFhighneutrophils. Science 2017, 358, eaal5081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, S.; Zhang, Z.; Fang, F.; Gao, X.; Sun, W.; Liu, H. The neutrophil/lymphocyte ratio is an independent prognostic indicator in patients with bone metastasis. Oncol. Lett. 2011, 2, 735–740. [Google Scholar] [CrossRef] [Green Version]
- Thio, Q.C.B.S.; Goudriaan, W.A.; Janssen, S.J.; Pereira, N.R.P.; Sciubba, D.M.; Rosovksy, R.P.; Schwab, J.H. Prognostic role of neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio in patients with bone metastases. Br. J. Cancer 2018, 119, 737–743. [Google Scholar] [CrossRef] [Green Version]
- Caliskan, B.; Korkmaz, A.N. Can Neutrophil/Lymphocyte Ratio be a Predictor for Bone Metastases of Solid Tumors? World J. Nucl. Med. 2016, 15, 196–199. [Google Scholar] [CrossRef]
- Győri, D.S.; Mócsai, A. Osteoclast Signal Transduction During Bone Metastasis Formation. Front. Cell Dev. Biol. 2020, 8, 507. [Google Scholar] [CrossRef]
- Hofbauer, L.C.; Bozec, A.; Rauner, M.; Jakob, F.; Perner, S.; Pantel, K. Novel approaches to target the microenvironment of bone metastasis. Nat. Rev. Clin. Oncol. 2021, 1–18. [Google Scholar] [CrossRef]
- Clézardin, P.; Coleman, R.; Puppo, M.; Ottewell, P.; Bonnelye, E.; Paycha, F.; Confavreux, C.B.; Holen, I. Bone metastasis: Mechanisms, therapies, and biomarkers. Physiol. Rev. 2021, 101, 797–855. [Google Scholar] [CrossRef] [PubMed]
- Von Moos, R.; Strasser, F.; Gillessen, S.; Zaugg, K. Metastatic bone pain: Treatment options with an emphasis on bisphosphonates. Support. Care Cancer 2008, 16, 1105–1115. [Google Scholar] [CrossRef] [PubMed]
- Jakob, T.; Tesfamariam, Y.M.; Macherey, S.; Kuhr, K.; Adams, A.; Monsef, I.; Heidenreich, A.; Skoetz, N. Bisphosphonates or RANK-ligand-inhibitors for men with prostate cancer and bone metastases: A network meta-analysis. Cochrane Database Syst. Rev. 2020, 12, CD013020. [Google Scholar] [CrossRef] [PubMed]
- Santini, D.; Galluzzo, S.; Zoccoli, A.; Pantano, F.; Fratto, M.; Vincenzi, B.; Lombardi, L.; Gucciardino, C.; Silvestris, N.; Riva, E.; et al. New molecular targets in bone metastases. Cancer Treat. Rev. 2010, 36, S6–S10. [Google Scholar] [CrossRef] [Green Version]
- Russell, R.G.G. Bisphosphonates: Mode of Action and Pharmacology. Pediatrics 2007, 119 (Suppl. 2), S150–S162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- George, C.N.; Canuas-Landero, V.; Theodoulou, E.; Muthana, M.; Wilson, C.; Ottewell, P. Oestrogen and zoledronic acid driven changes to the bone and immune environments: Potential mechanisms underlying the differential anti-tumour effects of zoledronic acid in pre- and post-menopausal conditions. J. Bone Oncol. 2020, 25, 100317. [Google Scholar] [CrossRef]
- Mariani, S.; Muraro, M.; Pantaleoni, F.; Fiore, F.; Nuschak, B.; Peola, S.; Foglietta, M.; Palumbo, A.; Coscia, M.; Castella, B.; et al. Effector γδ T cells and tumor cells as immune targets of zoledronic acid in multiple myeloma. Leukemia 2005, 19, 664–670. [Google Scholar] [CrossRef] [PubMed]
- Veltman, J.D.; Lambers, M.E.; van Nimwegen, M.; Hendriks, R.W.; Hoogsteden, H.C.; Hegmans, J.P.; Aerts, J.G. Zoledronic acid impairs myeloid differentiation to tumour-associated macrophages in mesothelioma. Br. J. Cancer 2010, 103, 629–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsagozis, P.; Eriksson, F.; Pisa, P. Zoledronic acid modulates antitumoral responses of prostate cancer-tumor associated macrophages. Cancer Immunol. Immunother. 2008, 57, 1451–1459. [Google Scholar] [CrossRef] [PubMed]
- Wolf, A.M.; Rumpold, H.; Tilg, H.; Gastl, G.; Gunsilius, E.; Wolf, D. The effect of zoledronic acid on the function and differentiation of myeloid cells. Haematologica 2006, 91, 1165–1171. [Google Scholar] [PubMed]
- Fowler, D.W.; Copier, J.; Dalgleish, A.G.; Bodman-Smith, M. Zoledronic acid renders human M1 and M2 macrophages susceptible to Vδ2+ γδ T cell cytotoxicity in a perforin-dependent manner. Cancer Immunol. Immunother. 2017, 66, 1205–1215. [Google Scholar] [CrossRef] [PubMed]
- Comito, G.; Segura, C.P.; Taddei, M.L.; Lanciotti, M.; Serni, S.; Morandi, A.; Chiarugi, P.; Giannoni, E. Zoledronic acid impairs stromal reactivity by inhibiting M2-macrophages polarization and prostate cancer-associated fibroblasts. Oncotarget 2016, 8, 118–132. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Wang, S.-H.; Chen, S.-C.; Chen, C.-Y.; Lin, T.-M. Zoledronic acid blocks the interaction between breast cancer cells and regulatory T-cells. BMC Cancer 2019, 19, 176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, H.; Wang, S.-H.; Chen, S.-C.; Chen, C.-Y.; Lo, J.-L.; Lin, T.-M. Immune modulation of CD4+CD25+ regulatory T cells by zoledronic acid. BMC Immunol. 2016, 17, 45. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Du, Y.; Sun, T.; Xue, H.; Jin, Z.; Tian, J. PD-1 blockade in combination with zoledronic acid to enhance the antitumor efficacy in the breast cancer mouse model. BMC Cancer 2018, 18, 669. [Google Scholar] [CrossRef] [Green Version]
- Sarhan, D.; Leijonhufvud, C.; Murray, S.; Witt, K.; Seitz, C.; Wallerius, M.; Xie, H.; Ullén, A.; Harmenberg, U.; Lidbrink, E.; et al. Zoledronic acid inhibits NFAT and IL-2 signaling pathways in regulatory T cells and diminishes their suppressive function in patients with metastatic cancer. OncoImmunology 2017, 6, e1338238. [Google Scholar] [CrossRef]
- Ubellacker, J.M.; Baryawno, N.; Severe, N.; DeCristo, M.J.; Sceneay, J.; Hutchinson, J.N.; Haider, M.-T.; Rhee, C.S.; Qin, Y.; Gregory, W.M.; et al. Modulating Bone Marrow Hematopoietic Lineage Potential to Prevent Bone Metastasis in Breast Cancer. Cancer Res. 2018, 78, 5300–5314. [Google Scholar] [CrossRef] [Green Version]
- Zysk, A.; DeNichilo, M.O.; Panagopoulos, V.; Zinonos, I.; Liapis, V.; Hay, S.; Ingman, W.; Ponomarev, V.; Atkins, G.; Findlay, D.; et al. Adoptive transfer of ex vivo expanded Vγ9Vδ2 T cells in combination with zoledronic acid inhibits cancer growth and limits osteolysis in a murine model of osteolytic breast cancer. Cancer Lett. 2017, 386, 141–150. [Google Scholar] [CrossRef] [Green Version]
- Nicol, A.J.; Tokuyama, H.; Mattarollo, S.R.; Hagi, T.; Suzuki, K.; Yokokawa, K.; Nieda, M. Clinical evaluation of autologous gamma delta T cell-based immunotherapy for metastatic solid tumours. Br. J. Cancer 2011, 105, 778–786. [Google Scholar] [CrossRef] [Green Version]
- Noguchi, A.; Kaneko, T.; Kamigaki, T.; Fujimoto, K.; Ozawa, M.; Saito, M.; Ariyoshi, N.; Goto, S. Zoledronate-activated Vγ9γδ T cell-based immunotherapy is feasible and restores the impairment of γδ T cells in patients with solid tumors. Cytotherapy 2011, 13, 92–97. [Google Scholar] [CrossRef]
- Lang, J.M.; Kaikobad, M.R.; Wallace, M.; Staab, M.J.; Horvath, D.L.; Wilding, G.; Liu, G.; Eickhoff, J.C.; McNeel, D.G.; Malkovsky, M. Pilot trial of interleukin-2 and zoledronic acid to augment γδ T cells as treatment for patients with refractory renal cell carcinoma. Cancer Immunol. Immunother. 2011, 60, 1447–1460. [Google Scholar] [CrossRef] [Green Version]
- Meraviglia, S.; Eberl, M.; Vermijlen, D.; Todaro, M.; Buccheri, S.; Cicero, G.; La Mendola, C.; Guggino, G.; D’Asaro, M.; Orlando, V.; et al. In vivo manipulation of Vγ9Vδ2 T cells with zoledronate and low-dose interleukin-2 for immunotherapy of advanced breast cancer patients. Clin. Exp. Immunol. 2010, 161, 290–297. [Google Scholar] [CrossRef] [PubMed]
- Dieli, F.; Vermijlen, D.; Fulfaro, F.; Caccamo, N.; Meraviglia, S.; Cicero, G.; Roberts, A.W.; Buccheri, S.; D’Asaro, M.; Gebbia, N.; et al. Targeting Human γδ T Cells with Zoledronate and Interleukin-2 for Immunotherapy of Hormone-Refractory Prostate Cancer. Cancer Res. 2007, 67, 7450–7457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nagamine, I.; Yamaguchi, Y.; Ohara, M.; Ikeda, T.; Okada, M. Induction of gamma delta T cells using zoledronate plus interleukin-2 in patients with metastatic cancer. Hiroshima J. Med Sci. 2009, 58, 37–44. [Google Scholar] [PubMed]
- Smith, H.A.; Kang, Y. The metastasis-promoting roles of tumor-associated immune cells. J. Mol. Med. 2013, 91, 411–429. [Google Scholar] [CrossRef] [PubMed]
- De Groot, A.; Appelman-Dijkstra, N.; van der Burg, S.; Kroep, J. The anti-tumor effect of RANKL inhibition in malignant solid tumors—A systematic review. Cancer Treat. Rev. 2018, 62, 18–28. [Google Scholar] [CrossRef] [Green Version]
- Ahern, E.; Smyth, M.J.; Dougall, W.C.; Teng, M.W.L. Roles of the RANKL–RANK axis in antitumour immunity—Implications for therapy. Nat. Rev. Clin. Oncol. 2018, 15, 676–693. [Google Scholar] [CrossRef]
- Van Dam, P.A.; Verhoeven, Y.; Trinh, X.B.; Wouters, A.; Lardon, F.; Prenen, H.; Smits, E.; Baldewijns, M.; Lammens, M. RANK/RANKL signaling inhibition may improve the effectiveness of checkpoint blockade in cancer treatment. Crit. Rev. Oncol. 2019, 133, 85–91. [Google Scholar] [CrossRef]
- Smyth, M.; Yagita, H.; McArthur, G.A. Combination Anti-CTLA-4 and Anti-RANKL in Metastatic Melanoma. J. Clin. Oncol. 2016, 34, e104–e106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Angela, Y.; Haferkamp, S.; Weishaupt, C.; Ugurel, S.; Becker, J.C.; Oberndörfer, F.; Alar, V.; Satzger, I.; Gutzmer, R. Combination of denosumab and immune checkpoint inhibition: Experience in 29 patients with metastatic melanoma and bone metastases. Cancer Immunol. Immunother. 2019, 68, 1187–1194. [Google Scholar] [CrossRef]
- Afzal, M.Z.; Shirai, K. Immune checkpoint inhibitor (anti-CTLA-4, anti-PD-1) therapy alone versus immune checkpoint inhibitor (anti-CTLA-4, anti-PD-1) therapy in combination with anti-RANKL denosumuab in malignant melanoma: A retrospective analysis at a tertiary care center. Melanoma Res. 2018, 28, 341–347. [Google Scholar] [CrossRef] [PubMed]
- Ahern, E.; Harjunpää, H.; O’Donnell, J.S.; Allen, S.; Dougall, W.C.; Teng, M.W.L.; Smyth, M.J. RANKL blockade improves efficacy of PD1-PD-L1 blockade or dual PD1-PD-L1 and CTLA4 blockade in mouse models of cancer. OncoImmunology 2018, 7, e1431088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Simatou, A.; Sarantis, P.; Koustas, E.; Papavassiliou, A.G.; Karamouzis, M.V. The Role of the RANKL/RANK Axis in the Prevention and Treatment of Breast Cancer with Immune Checkpoint Inhibitors and Anti-RANKL. Int. J. Mol. Sci. 2020, 21, 7570. [Google Scholar] [CrossRef] [PubMed]
- Gallicchio, R.; Mastrangelo, P.A.; Nardelli, A.; Mainenti, P.P.; Colasurdo, A.P.; Landriscina, M.; Guglielmi, G.; Storto, G. Radium-223 for the treatment of bone metastases in castration-resistant prostate cancer: When and why. Tumori J. 2019, 105, 367–377. [Google Scholar] [CrossRef]
- ASCO GU 2021: Randomized Phase II Study Evaluating the Addition of Pembrolizumab to Radium-223 in Metastatic Castration-Resistant Prostate Cancer. Available online: https://www.urotoday.com/conference-highlights/asco-gu-2021/prostate-cancer/127937-asco-gu-2021-randomized-phase-ii-study-evaluating-the-addition-of-pembrolizumab-to-radium-223-in-metastatic-castration-resistant-prostate-cancer.html (accessed on 13 April 2021).
- Kim, J.W.; Shin, M.S.; Kang, Y.; Kang, I.; Petrylak, D.P. Immune Analysis of Radium-223 in Patients With Metastatic Prostate Cancer. Clin. Genitourin. Cancer 2018, 16, e469–e476. [Google Scholar] [CrossRef] [Green Version]
- Giles, A.J.; Hutchinson, M.-K.N.D.; Sonnemann, H.M.; Jung, J.; Fecci, P.E.; Ratnam, N.M.; Zhang, W.; Song, H.; Bailey, R.; Davis, D.; et al. Dexamethasone-induced immunosuppression: Mechanisms and implications for immunotherapy. J. Immunother. Cancer 2018, 6, 51. [Google Scholar] [CrossRef]
- Marshall, C.H.; Fu, W.; Wang, H.; Park, J.C.; DeWeese, T.L.; Tran, P.T.; Song, D.Y.; King, S.; Afful, M.; Hurrelbrink, J.; et al. Randomized Phase II Trial of Sipuleucel-T with or without Radium-223 in Men with Bone-metastatic Castration-resistant Prostate Cancer. Clin. Cancer Res. 2021, 27, 1623–1630. [Google Scholar] [CrossRef]
- O’Sullivan, J.M.; Carles, J.; Cathomas, R.; Gomez-Iturriaga, A.; Heinrich, D.; Kramer, G.; Ost, P.; Van Oort, I.; Tombal, B. Radium-223 Within the Evolving Treatment Options for Metastatic Castration-resistant Prostate Cancer: Recommendations from a European Expert Working Group. Eur. Urol. Oncol. 2020, 3, 455–463. [Google Scholar] [CrossRef] [Green Version]
- Kähkönen, T.E.; Halleen, J.M.; Bernoulli, J. Limited data from clinical trials assessing immunotherapy effects on bone metastases. Cancer Res. 2021, in press. [Google Scholar]
- Schmid, P.; Rugo, H.S.; Adams, S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Diéras, V.; Henschel, V.; Molinero, L.; Chui, S.Y.; et al. Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): Updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2020, 21, 44–59. [Google Scholar] [CrossRef]
- Moseley, K.F.; Naidoo, J.; Bingham, C.O.; Carducci, M.A.; Forde, P.M.; Gibney, G.T.; Lipson, E.J.; Shah, A.A.; Sharfman, W.H.; Cappelli, L.C. Immune-related adverse events with immune checkpoint inhibitors affecting the skeleton: A seminal case series. J. Immunother. Cancer 2018, 6, 104. [Google Scholar] [CrossRef]
- Hilal, T.; Bansal, P.; Kelemen, K.; Slack, J. Nivolumab-associated bone marrow necrosis. Ann. Oncol. 2018, 29, 513–514. [Google Scholar] [CrossRef] [PubMed]
- Heidegger, I.; Necchi, A.; Pircher, A.; Tsaur, I.; Marra, G.; Kasivisvanathan, V.; Kretschmer, A.; Mathieu, R.; Ceci, F.; Bergh, R.C.V.D.; et al. A Systematic Review of the Emerging Role of Immune Checkpoint Inhibitors in Metastatic Castration-resistant Prostate Cancer: Will Combination Strategies Improve Efficacy? Eur. Urol. Oncol. 2020. [Google Scholar] [CrossRef] [PubMed]
Immune Cell Type | Subtype | Effect on Bone Metastases (- Anti-Tumor Effect, + Pro-Tumoral Effect) | References |
---|---|---|---|
T cell | CD4+ | (−) CD4+ cells have anti-tumor activity on primary and metastatic tumors (−) CD4+ cells prime survival of cytotoxic CD8+ cells (+) RANK/RANKL-mediated interactions with CD4+ and tumor cells increase invasion and dissemination of tumor cells and formation of metastases (+) TGF-β increases conversion of CD4+ cells to Tregs, which increases metastases | [68,71,72,73] |
CD8+ | (−) CD8+ cells have anti-tumor activity on primary and metastatic tumors (−) CD8+ cells mediate immunological cell death (−) CD8+ cells remain in bone marrow several months after metastasis formation (−) CD8+ cells are associated with metastasis-free survival in patients (+) CD8+ cells increase migration, invasion, and expression of metastasis-associated genes (+) CD8+ cells have tumor-independent effects on bone cells | [26,68,74,75,76,77] | |
Treg | (+) Tregs promote tumor cell homing to bone marrow (+) Tregs increase bone metastasis and cancer-induced bone resorption (+) Tregs are immunosuppressive and correlate with advanced stages and poor prognosis in patients | [26,78,79,80] | |
NKT | (−) NKT cells can reject tumor cells in bone marrow (−) Low number or malfunction of NKT cells results in increased metastases | [26,81] | |
NK | (−) Loss of peripheral NK cells is associated with metastatic progression (−) Deletion of NK (genetic or activity) cells increases tumor growth and formation of metastases (−) NK cells can suppress metastases via IL-17, IL-28, IFN-γ and JAK signaling | [26,68,75,78,82,83,84,85,86,87,88,89,90] | |
MDSC | (+) MDSCs are associated with increased bone metastases and decreased survival (+) The number of MDSCs is highest in bone metastases (+) MDSCs contribute to the vicious cycle of bone metastasis (+) MDSCs in bone metastases can differentiate into osteoclasts (+) MDSCs increase metastasis via CCL5, CCL15, MMPs, and chemoattractants (+) MDSCs suppress proliferation and include the apoptosis of T cells and suppress the activity of NK cells (+) MDSCs activate Tregs | [60,68,73,78,82,91,92,93,94,95,96,97,98] | |
Macrophage | TAM/ M2 | (+) TAMs cause tumor progression and the formation and growth of metastases (+) A high number of TAMs correlates with poor clinical outcome (+) TAMs secrete IL-10 and TGF-β, which decrease the activation of T cells (+) CD68+ and CD169+ macrophages increase growth of bone metastasis (+) CD169+ macrophages disable anti-tumor function of CD8+ T cells (+) Deletion of CD169+ macrophages inhibited cancer-induced new bone formation | [66,78,99,100,101,102,103,104,105,106,107] |
DC | pDC | (+) DCs are increased in bone metastases (+) DC maturation is prevented in bone metastases (+) Bone marrow DCs deactivate CD8+ T cells via TGF-β (+) DCs recruit Tregs and MDSCs into the tumor | [68,78,83,108,109,110,111,112] |
Neutrophil | TAN | (−) At early stages, neutrophils induce cytotoxic effects on cancer cells (+) Neutrophils participate in the formation of pre-metastatic niche (+) Neutrophils have immunosuppressive properties in metastases (+) N2 TANs are tumor-promoting, increasing angiogenesis, dissemination of tumor cells, and formation of bone metastases (+) N2 TANs secrete pro-metastatic CXCR4, VEGF, and MMP9 | [78,113,114,115,116,117] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kähkönen, T.E.; Halleen, J.M.; Bernoulli, J. Osteoimmuno-Oncology: Therapeutic Opportunities for Targeting Immune Cells in Bone Metastasis. Cells 2021, 10, 1529. https://doi.org/10.3390/cells10061529
Kähkönen TE, Halleen JM, Bernoulli J. Osteoimmuno-Oncology: Therapeutic Opportunities for Targeting Immune Cells in Bone Metastasis. Cells. 2021; 10(6):1529. https://doi.org/10.3390/cells10061529
Chicago/Turabian StyleKähkönen, Tiina E., Jussi M. Halleen, and Jenni Bernoulli. 2021. "Osteoimmuno-Oncology: Therapeutic Opportunities for Targeting Immune Cells in Bone Metastasis" Cells 10, no. 6: 1529. https://doi.org/10.3390/cells10061529
APA StyleKähkönen, T. E., Halleen, J. M., & Bernoulli, J. (2021). Osteoimmuno-Oncology: Therapeutic Opportunities for Targeting Immune Cells in Bone Metastasis. Cells, 10(6), 1529. https://doi.org/10.3390/cells10061529