The Role of TGF-β in Bone Metastases
Abstract
:1. Introduction
2. Physiology of Bone Remodeling
2.1. Bone Cells
2.1.1. Osteoclasts
2.1.2. Osteoblasts
2.1.3. Osteocytes
2.2. Bone Remodeling
3. TGF-β Signaling Mechanisms
3.1. The TGF-β Superfamily
3.2. Smad-Dependent Signaling
3.3. Smad-Independent Signaling
4. TGF-β: A Regulator of Epithelial-Mesenchymal Transition
5. Pleiotropic Role of TGF-β in Cancer Progression
6. TGF-β Expression in Human Tumors
7. TGF-β and Bone Microenvironment
8. TGF-β and Tumor-Bone Interaction in Bone Metastases
9. TGF-β and the Immune System in the Tumor Microenvironment
9.1. Myeloid Compartment
9.2. Lymphoid Compartment
10. Therapeutic Approaches to Target TGF-β Signaling
10.1. Anti–TGF-β Antibodies and TGF-β Receptor Inhibitors
10.2. TGF-β Receptor Kinase Inhibitors
10.3. Inhibitors of TGF-β Synthesis
10.4. Additional TGF-β Antagonists
11. Enhancing TGF-β Inhibitors’ Effects by Combination with Other Therapies
12. Challenges of TGF-β Targeting as Therapeutics
13. Conclusions
Author Contributions
Funding
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
TGF-β | Transforming growth factor β |
OPG | Osteoprotegerin |
PD-L1 | Programmed cell death ligand -1 |
IGF | Insulin-like growth factor |
TNFs | Tumor necrosis factors |
ILs | Interleukins |
RANKL | Receptor activator of NF-κB ligand |
IL-1 | Interleukin-1 |
MMP | Matrix metalloproteinases |
GDF | Growth differentiation factor |
ALKs | Activin-receptor like kinases |
MAP | Mitogen-activated protein |
JNK | c-Jun amino-terminal kinase |
TRAF6 | TNF receptor-associated factor 6 |
ZEB | Zinc finger E-box-binding homeobox |
DAP12 | DNAX activating protein 12 kDA |
CBP | CREB-binding protein |
FoxH1 | Forkhead box protein H1 |
Lef1 | Lymphoid enhancer factor 1 |
CAR | Chimeric antigen receptor |
PMEPA1 | Prostate transmembrane protein androgen induced 1 |
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
- Liu, D.; Kuai, Y.; Zhu, R.; Zhou, C.; Tao, Y.; Han, W.; Chen, Q. Prognosis of prostate cancer and bone metastasis pattern of patients: A SEER-based study and a local hospital based study from China. Sci. Rep. 2020, 10, 9104. [Google Scholar] [CrossRef]
- Huang, J.F.; Shen, J.; Li, X.; Rengan, R.; Silvestris, N.; Wang, M.; Derosa, L.; Zheng, X.; Belli, A.; Zhang, X.L.; et al. Incidence of patients with bone metastases at diagnosis of solid tumors in adults: A large population-based study. Ann. Transl. Med. 2020, 8, 482. [Google Scholar] [CrossRef] [PubMed]
- Yang, M.; Liu, C.; Yu, X. Skeletal-related adverse events during bone metastasis of breast cancer: Current status. Discov. Med. 2019, 27, 211–220. [Google Scholar]
- Hiraga, T. Bone metastasis: Interaction between cancer cells and bone microenvironment. J. Oral Biosci. 2019, 61, 95–98. [Google Scholar] [CrossRef]
- Coleman, R.E.; Croucher, P.I.; Padhani, A.R.; Clezardin, P.; Chow, E.; Fallon, M.; Guise, T.; Colangeli, S.; Capanna, R.; Costa, L. Bone metastases. Nat. Rev. Dis. Primers 2020, 6, 83. [Google Scholar] [CrossRef]
- Clines, G.A.; Guise, T.A. Mechanisms and treatment for bone metastases. Clin. Adv. Hematol. Oncol. 2004, 2, 295–302. [Google Scholar]
- Lipton, A. Implications of bone metastases and the benefits of bone-targeted therapy. Semin. Oncol. 2010, 37 (Suppl. S2), 15–29. [Google Scholar] [CrossRef]
- Zhang, H.; Zhu, W.; Biskup, E.; Yang, W.; Yang, Z.; Wang, H.; Qiu, X.; Zhang, C.; Hu, G.; Hu, G. Incidence, risk factors and prognostic characteristics of bone metastases and skeletal-related events (SREs) in breast cancer patients: A systematic review of the real world data. J. Bone Oncol. 2018, 11, 38–50. [Google Scholar] [CrossRef] [PubMed]
- Mundy, G.R. Metastasis to bone: Causes, consequences and therapeutic opportunities. Nat. Rev. Cancer 2002, 2, 584–593. [Google Scholar] [CrossRef] [PubMed]
- Roodman, G.D. Mechanisms of bone metastasis. N. Engl. J. Med. 2004, 350, 1655–1664. [Google Scholar] [CrossRef] [PubMed]
- Bonewald, L.F.; Mundy, G.R. Role of transforming growth factor-beta in bone remodeling. Clin. Orthop. Relat. Res. 1990, 261–276. [Google Scholar] [CrossRef]
- Sakai, R.; Eto, Y. Involvement of activin in the regulation of bone metabolism. Mol. Cell Endocrinol. 2001, 180, 183–188. [Google Scholar] [CrossRef]
- Kang, Y.; Siegel, P.M.; Shu, W.; Drobnjak, M.; Kakonen, S.M.; Cordon-Cardo, C.; Guise, T.A.; Massague, J. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 2003, 3, 537–549. [Google Scholar] [CrossRef] [Green Version]
- Khalil, N. TGF-beta: From latent to active. Microbes Infect. 1999, 1, 1255–1263. [Google Scholar] [CrossRef]
- Buijs, J.T.; Stayrook, K.R.; Guise, T.A. The role of TGF-beta in bone metastasis: Novel therapeutic perspectives. Bonekey Rep. 2012, 1, 96. [Google Scholar] [CrossRef] [Green Version]
- Ciardiello, D.; Elez, E.; Tabernero, J.; Seoane, J. Clinical development of therapies targeting TGFbeta: Current knowledge and future perspectives. Ann. Oncol. 2020, 31, 1336–1349. [Google Scholar] [CrossRef] [PubMed]
- Connolly, E.C.; Freimuth, J.; Akhurst, R.J. Complexities of TGF-beta targeted cancer therapy. Int. J. Biol. Sci. 2012, 8, 964–978. [Google Scholar] [CrossRef] [Green Version]
- Teixeira, A.F.; Ten Dijke, P.; Zhu, H.J. On-Target Anti-TGF-beta therapies are not succeeding in clinical cancer treatments: What Are remaining challenges? Front. Cell Dev. Biol. 2020, 8, 605. [Google Scholar] [CrossRef] [PubMed]
- Colak, S.; Ten Dijke, P. Targeting TGF-beta Signaling in Cancer. Trends Cancer 2017, 3, 56–71. [Google Scholar] [CrossRef]
- Jin, K.; Zhou, F.; Zhang, L. Cancer Environment Immunotherapy: Targeting TGF-beta finds its way towards tissue healing and vasculature remodeling. Signal Transduct. Target. Ther. 2021, 6, 41. [Google Scholar] [CrossRef]
- Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiao, S.; Subudhi, S.K.; Aparicio, A.; Ge, Z.; Guan, B.; Miura, Y.; Sharma, P. Differences in Tumor Microenvironment Dictate T Helper Lineage Polarization and Response to Immune Checkpoint Therapy. Cell 2019, 179, 1177–1190. [Google Scholar] [CrossRef] [PubMed]
- Ungefroren, H. Blockade of TGF-beta signaling: A potential target for cancer immunotherapy? Expert Opin. Ther. Targets 2019, 23, 679–693. [Google Scholar] [CrossRef] [PubMed]
- Hayashi, S.; Gimble, J.M.; Henley, A.; Ellingsworth, L.R.; Kincade, P.W. Differential effects of TGF-beta 1 on lymphohemopoiesis in long-term bone marrow cultures. Blood 1989, 74, 1711–1717. [Google Scholar] [CrossRef] [Green Version]
- Lin, X.; Patil, S.; Gao, Y.G.; Qian, A. The bone extracellular matrix in bone formation and regeneration. Front. Pharmacol. 2020, 11, 757. [Google Scholar] [CrossRef] [PubMed]
- Lacey, D.L.; Timms, E.; Tan, H.L.; Kelley, M.J.; Dunstan, C.R.; Burgess, T.; Elliott, R.; Colombero, A.; Elliott, G.; Scully, S.; et al. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 1998, 93, 165–176. [Google Scholar] [CrossRef] [Green Version]
- Yasuda, H.; Shima, N.; Nakagawa, N.; Yamaguchi, K.; Kinosaki, M.; Mochizuki, S.; Tomoyasu, A.; Yano, K.; Goto, M.; Murakami, A.; et al. Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL. Proc. Natl. Acad. Sci. USA 1998, 95, 3597–3602. [Google Scholar] [CrossRef] [Green Version]
- Hsu, H.; Lacey, D.L.; Dunstan, C.R.; Solovyev, I.; Colombero, A.; Timms, E.; Tan, H.L.; Elliott, G.; Kelley, M.J.; Sarosi, I.; et al. Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand. Proc. Natl. Acad. Sci. USA 1999, 96, 3540–3545. [Google Scholar] [CrossRef] [Green Version]
- Nakagawa, N.; Kinosaki, M.; Yamaguchi, K.; Shima, N.; Yasuda, H.; Yano, K.; Morinaga, T.; Higashio, K. RANK is the essential signaling receptor for osteoclast differentiation factor in osteoclastogenesis. Biochem. Biophys. Res. Commun. 1998, 253, 395–400. [Google Scholar] [CrossRef] [PubMed]
- Nakashima, T.; Hayashi, M.; Fukunaga, T.; Kurata, K.; Oh-Hora, M.; Feng, J.Q.; Bonewald, L.F.; Kodama, T.; Wutz, A.; Wagner, E.F.; et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat. Med. 2011, 17, 1231–1234. [Google Scholar] [CrossRef]
- Xiong, J.; Onal, M.; Jilka, R.L.; Weinstein, R.S.; Manolagas, S.C.; O’Brien, C.A. Matrix-embedded cells control osteoclast formation. Nat. Med. 2011, 17, 1235–1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakashima, T.; Hayash, M.; Takayanagi, H. Regulation of bone resorption by osteocytes. Clin. Calcium 2012, 22, 685–696. [Google Scholar] [PubMed]
- Simonet, W.S.; Lacey, D.L.; Dunstan, C.R.; Kelley, M.; Chang, M.S.; Luthy, R.; Nguyen, H.Q.; Wooden, S.; Bennett, L.; Boone, T.; et al. Osteoprotegerin: A novel secreted protein involved in the regulation of bone density. Cell 1997, 89, 309–319. [Google Scholar] [CrossRef] [Green Version]
- Blair, J.M.; Zhou, H.; Seibel, M.J.; Dunstan, C.R. Mechanisms of Disease: Roles of OPG, RANKL and RANK in the pathophysiology of skeletal metastasis. Nat. Clin. Pract. Oncol. 2006, 3, 41–49. [Google Scholar] [CrossRef]
- Blair, J.M.; Zheng, Y.; Dunstan, C.R. RANK ligand. Int. J. Biochem. Cell Biol. 2007, 39, 1077–1081. [Google Scholar] [CrossRef]
- Anderson, D.M.; Maraskovsky, E.; Billingsley, W.L.; Dougall, W.C.; Tometsko, M.E.; Roux, E.R.; Teepe, M.C.; DuBose, R.F.; Cosman, D.; Galibert, L. A homologue of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function. Nature 1997, 390, 175–179. [Google Scholar] [CrossRef]
- Wong, B.R.; Rho, J.; Arron, J.; Robinson, E.; Orlinick, J.; Chao, M.; Kalachikov, S.; Cayani, E.; Bartlett, F.S., 3rd; Frankel, W.N.; et al. TRANCE is a novel ligand of the tumor necrosis factor receptor family that activates c-Jun N-terminal kinase in T cells. J. Biol. Chem. 1997, 272, 25190–25194. [Google Scholar] [CrossRef] [Green Version]
- Clarke, B. Normal bone anatomy and physiology. Clin. J. Am. Soc. Nephrol. 2008, 3 (Suppl. S3), 131–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eriksen, E.F. Cellular mechanisms of bone remodeling. Rev. Endocr. Metab. Disord. 2010, 11, 219–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kular, J.; Tickner, J.; Chim, S.M.; Xu, J. An overview of the regulation of bone remodelling at the cellular level. Clin. Biochem. 2012, 45, 863–873. [Google Scholar] [CrossRef] [PubMed]
- Franz-Odendaal, T.A.; Hall, B.K.; Witten, P.E. Buried alive: How osteoblasts become osteocytes. Dev. Dyn. 2006, 235, 176–190. [Google Scholar] [CrossRef] [PubMed]
- Noble, B.S. The osteocyte lineage. Arch. Biochem. Biophys. 2008, 473, 106–111. [Google Scholar] [CrossRef]
- Buckwalter, J.A.; Cooper, R.R. Bone structure and function. Instr. Course Lect. 1987, 36, 27–48. [Google Scholar]
- Hill, P.A. Bone remodelling. Br. J. Orthod. 1998, 25, 101–107. [Google Scholar] [CrossRef]
- Vaananen, H.K.; Horton, M. The osteoclast clear zone is a specialized cell-extracellular matrix adhesion structure. J. Cell Sci. 1995, 108, 2729–2732. [Google Scholar] [CrossRef]
- Nakamura, I.; Pilkington, M.F.; Lakkakorpi, P.T.; Lipfert, L.; Sims, S.M.; Dixon, S.J.; Rodan, G.A.; Duong, L.T. Role of alpha(v)beta(3) integrin in osteoclast migration and formation of the sealing zone. J. Cell Sci. 1999, 112, 3985–3993. [Google Scholar] [CrossRef]
- Blair, H.C.; Teitelbaum, S.L.; Ghiselli, R.; Gluck, S. Osteoclastic bone resorption by a polarized vacuolar proton pump. Science 1989, 245, 855–857. [Google Scholar] [CrossRef]
- Minkin, C. Bone acid phosphatase: Tartrate-resistant acid phosphatase as a marker of osteoclast function. Calcif. Tissue Int. 1982, 34, 285–290. [Google Scholar] [CrossRef] [PubMed]
- Lorget, F.; Kamel, S.; Mentaverri, R.; Wattel, A.; Naassila, M.; Maamer, M.; Brazier, M. High extracellular calcium concentrations directly stimulate osteoclast apoptosis. Biochem. Biophys. Res. Commun. 2000, 268, 899–903. [Google Scholar] [CrossRef] [PubMed]
- Marie, P.J. Transcription factors controlling osteoblastogenesis. Arch. Biochem. Biophys. 2008, 473, 98–105. [Google Scholar] [CrossRef]
- Del Fattore, A.; Teti, A.; Rucci, N. Bone cells and the mechanisms of bone remodelling. Front. Biosci. 2012, 4, 2302–2321. [Google Scholar] [CrossRef]
- Massague, J.; Blain, S.W.; Lo, R.S. TGFbeta signaling in growth control, cancer, and heritable disorders. Cell 2000, 103, 295–309. [Google Scholar] [CrossRef] [Green Version]
- Kubiczkova, L.; Sedlarikova, L.; Hajek, R.; Sevcikova, S. TGF-beta—An excellent servant but a bad master. J. Transl. Med. 2012, 10, 183. [Google Scholar] [CrossRef] [Green Version]
- Grant, C.; Oh, U.; Yao, K.; Yamano, Y.; Jacobson, S. Dysregulation of TGF-beta signaling and regulatory and effector T-cell function in virus-induced neuroinflammatory disease. Blood 2008, 111, 5601–5609. [Google Scholar] [CrossRef] [Green Version]
- David, C.J.; Massague, J. Contextual determinants of TGFbeta action in development, immunity and cancer. Nat. Rev. Mol. Cell Biol. 2018, 19, 419–435. [Google Scholar] [CrossRef]
- Massague, J. TGFbeta in Cancer. Cell 2008, 134, 215–230. [Google Scholar] [CrossRef] [Green Version]
- Vander Ark, A.; Cao, J.; Li, X. TGF-beta receptors: In and beyond TGF-beta signaling. Cell Signal 2018, 52, 112–120. [Google Scholar] [CrossRef]
- Heldin, C.H.; Moustakas, A. Signaling Receptors for TGF-beta Family Members. Cold Spring Harb. Perspect. Biol. 2016, 8, a022053. [Google Scholar] [CrossRef] [Green Version]
- Shi, M.; Zhu, J.; Wang, R.; Chen, X.; Mi, L.; Walz, T.; Springer, T.A. Latent TGF-beta structure and activation. Nature 2011, 474, 343–349. [Google Scholar] [CrossRef] [Green Version]
- Derynck, R.; Zhang, Y.E. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature 2003, 425, 577–584. [Google Scholar] [CrossRef] [PubMed]
- Hinck, A.P. Structural studies of the TGF-betas and their receptors—Insights into evolution of the TGF-beta superfamily. FEBS Lett. 2012, 586, 1860–1870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miyazono, K. TGF-beta signaling by Smad proteins. Cytokine Growth Factor Rev. 2000, 11, 15–22. [Google Scholar] [CrossRef]
- Ikushima, H.; Komuro, A.; Isogaya, K.; Shinozaki, M.; Hellman, U.; Miyazawa, K.; Miyazono, K. An Id-like molecule, HHM, is a synexpression group-restricted regulator of TGF-beta signalling. EMBO J. 2008, 27, 2955–2965. [Google Scholar] [CrossRef] [Green Version]
- Ikushima, H.; Miyazono, K. TGFbeta signalling: A complex web in cancer progression. Nat. Rev. Cancer 2010, 10, 415–424. [Google Scholar] [CrossRef]
- Wakefield, L.M.; Hill, C.S. Beyond TGFbeta: Roles of other TGFbeta superfamily members in cancer. Nat. Rev. Cancer 2013, 13, 328–341. [Google Scholar] [CrossRef]
- Yan, X.; Liu, Z.; Chen, Y. Regulation of TGF-beta signaling by Smad7. Acta Biochim. Biophys. Sin. 2009, 41, 263–272. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.E. Non-Smad pathways in TGF-beta signaling. Cell Res. 2009, 19, 128–139. [Google Scholar] [CrossRef] [Green Version]
- Mu, Y.; Gudey, S.K.; Landstrom, M. Non-Smad signaling pathways. Cell Tissue Res. 2012, 347, 11–20. [Google Scholar] [CrossRef] [PubMed]
- Yamashita, M.; Fatyol, K.; Jin, C.; Wang, X.; Liu, Z.; Zhang, Y.E. TRAF6 mediates Smad-independent activation of JNK and p38 by TGF-beta. Mol. Cell 2008, 31, 918–924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nieto, M.A.; Huang, R.Y.; Jackson, R.A.; Thiery, J.P. Emt: 2016. Cell 2016, 166, 21–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gonzalez, D.M.; Medici, D. Signaling mechanisms of the epithelial-mesenchymal transition. Sci. Signal. 2014, 7, re8. [Google Scholar] [CrossRef] [Green Version]
- Luo, M.; Li, Z.; Wang, W.; Zeng, Y.; Liu, Z.; Qiu, J. Long non-coding RNA H19 increases bladder cancer metastasis by associating with EZH2 and inhibiting E-cadherin expression. Cancer Lett. 2013, 333, 213–221. [Google Scholar] [CrossRef]
- Lee, Y.H.; Albig, A.R.; Regner, M.; Schiemann, B.J.; Schiemann, W.P. Fibulin-5 initiates epithelial-mesenchymal transition (EMT) and enhances EMT induced by TGF-beta in mammary epithelial cells via a MMP-dependent mechanism. Carcinogenesis 2008, 29, 2243–2251. [Google Scholar] [CrossRef]
- Zaravinos, A. The Regulatory Role of MicroRNAs in EMT and Cancer. J. Oncol. 2015, 2015, 865816. [Google Scholar] [CrossRef] [Green Version]
- Vo, B.T.; Morton, D., Jr.; Komaragiri, S.; Millena, A.C.; Leath, C.; Khan, S.A. TGF-beta effects on prostate cancer cell migration and invasion are mediated by PGE2 through activation of PI3K/AKT/mTOR pathway. Endocrinology 2013, 154, 1768–1779. [Google Scholar] [CrossRef]
- Lee, P.S.; Chang, C.; Liu, D.; Derynck, R. Sumoylation of Smad4, the common Smad mediator of transforming growth factor-beta family signaling. J. Biol. Chem. 2003, 278, 27853–27863. [Google Scholar] [CrossRef] [Green Version]
- Hata, A.; Chen, Y.G. TGF-beta Signaling from Receptors to Smads. Cold Spring Harb. Perspect. Biol. 2016, 8. [Google Scholar] [CrossRef]
- Massague, J.; Chen, Y.G. Controlling TGF-beta signaling. Genes Dev. 2000, 14, 627–644. [Google Scholar] [PubMed]
- Pardali, E.; Goumans, M.J.; ten Dijke, P. Signaling by members of the TGF-beta family in vascular morphogenesis and disease. Trends Cell Biol. 2010, 20, 556–567. [Google Scholar] [CrossRef] [PubMed]
- Sun, N.; Taguchi, A.; Hanash, S. Switching Roles of TGF-beta in Cancer Development: Implications for Therapeutic Target and Biomarker Studies. J. Clin. Med. 2016, 5, 109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lebrun, J.J. The dual role of TGFbeta in human cancer: From tumor suppression to cancer metastasis. ISRN Mol. Biol. 2012, 2012, 381428. [Google Scholar] [CrossRef] [Green Version]
- Yeh, H.W.; Lee, S.S.; Chang, C.Y.; Lang, Y.D.; Jou, Y.S. A New Switch for TGFbeta in Cancer. Cancer Res. 2019, 79, 3797–3805. [Google Scholar] [CrossRef] [Green Version]
- Gong, C.; Qu, S.; Liu, B.; Pan, S.; Jiao, Y.; Nie, Y.; Su, F.; Liu, Q.; Song, E. MiR-106b expression determines the proliferation paradox of TGF-beta in breast cancer cells. Oncogene 2015, 34, 84–93. [Google Scholar] [CrossRef] [PubMed]
- Chu, I.M.; Lai, W.C.; Aprelikova, O.; El Touny, L.H.; Kouros-Mehr, H.; Green, J.E. Expression of GATA3 in MDA-MB-231 triple-negative breast cancer cells induces a growth inhibitory response to TGFss. PLoS ONE 2013, 8, e61125. [Google Scholar] [CrossRef]
- Rivlin, N.; Brosh, R.; Oren, M.; Rotter, V. Mutations in the p53 Tumor Suppressor Gene: Important Milestones at the Various Steps of Tumorigenesis. Genes Cancer 2011, 2, 466–474. [Google Scholar] [CrossRef] [Green Version]
- Her, N.G.; Jeong, S.I.; Cho, K.; Ha, T.K.; Han, J.; Ko, K.P.; Park, S.K.; Lee, J.H.; Lee, M.G.; Ryu, B.K.; et al. PPARdelta promotes oncogenic redirection of TGF-beta1 signaling through the activation of the ABCA1-Cav1 pathway. Cell Cycle 2013, 12, 1521–1535. [Google Scholar] [CrossRef] [Green Version]
- Leight, J.L.; Wozniak, M.A.; Chen, S.; Lynch, M.L.; Chen, C.S. Matrix rigidity regulates a switch between TGF-beta1-induced apoptosis and epithelial-mesenchymal transition. Mol. Biol. Cell 2012, 23, 781–791. [Google Scholar] [CrossRef]
- Larocca, C.; Cohen, J.R.; Fernando, R.I.; Huang, B.; Hamilton, D.H.; Palena, C. An autocrine loop between TGF-beta1 and the transcription factor brachyury controls the transition of human carcinoma cells into a mesenchymal phenotype. Mol. Cancer Ther. 2013, 12, 1805–1815. [Google Scholar] [CrossRef] [Green Version]
- Vrana, J.A.; Stang, M.T.; Grande, J.P.; Getz, M.J. Expression of tissue factor in tumor stroma correlates with progression to invasive human breast cancer: Paracrine regulation by carcinoma cell-derived members of the transforming growth factor beta family. Cancer Res. 1996, 56, 5063–5070. [Google Scholar]
- Travers, M.T.; Barrett-Lee, P.J.; Berger, U.; Luqmani, Y.A.; Gazet, J.C.; Powles, T.J.; Coombes, R.C. Growth factor expression in normal, benign, and malignant breast tissue. Br. Med. J. 1988, 296, 1621–1624. [Google Scholar] [CrossRef] [Green Version]
- Mizukami, Y.; Nonomura, A.; Yamada, T.; Kurumaya, H.; Hayashi, M.; Koyasaki, N.; Taniya, T.; Noguchi, M.; Nakamura, S.; Matsubara, F. Immunohistochemical demonstration of growth factors, TGF-alpha, TGF-beta, IGF-I and neu oncogene product in benign and malignant human breast tissues. Anticancer Res. 1990, 10, 1115–1126. [Google Scholar]
- De Jong, J.S.; van Diest, P.J.; van der Valk, P.; Baak, J.P. Expression of growth factors, growth-inhibiting factors, and their receptors in invasive breast cancer. II: Correlations with proliferation and angiogenesis. J. Pathol. 1998, 184, 53–57. [Google Scholar] [CrossRef]
- Kong, F.M.; Anscher, M.S.; Murase, T.; Abbott, B.D.; Iglehart, J.D.; Jirtle, R.L. Elevated plasma transforming growth factor-beta 1 levels in breast cancer patients decrease after surgical removal of the tumor. Ann. Surg. 1995, 222, 155–162. [Google Scholar] [CrossRef]
- Ghellal, A.; Li, C.; Hayes, M.; Byrne, G.; Bundred, N.; Kumar, S. Prognostic significance of TGF beta 1 and TGF beta 3 in human breast carcinoma. Anticancer Res. 2000, 20, 4413–4418. [Google Scholar] [PubMed]
- Morton, D.M.; Barrack, E.R. Modulation of transforming growth factor beta 1 effects on prostate cancer cell proliferation by growth factors and extracellular matrix. Cancer Res. 1995, 55, 2596–2602. [Google Scholar] [PubMed]
- Wikstrom, P.; Stattin, P.; Franck-Lissbrant, I.; Damber, J.E.; Bergh, A. Transforming growth factor beta1 is associated with angiogenesis, metastasis, and poor clinical outcome in prostate cancer. Prostate 1998, 37, 19–29. [Google Scholar] [CrossRef]
- Kim, I.Y.; Ahn, H.J.; Lang, S.; Oefelein, M.G.; Oyasu, R.; Kozlowski, J.M.; Lee, C. Loss of expression of transforming growth factor-beta receptors is associated with poor prognosis in prostate cancer patients. Clin. Cancer Res. 1998, 4, 1625–1630. [Google Scholar]
- Divella, R.; Daniele, A.; Savino, E.; Palma, F.; Bellizzi, A.; Giotta, F.; Simone, G.; Lioce, M.; Quaranta, M.; Paradiso, A.; et al. Circulating levels of transforming growth factor-betaeta (TGF-beta) and chemokine (C-X-C motif) ligand-1 (CXCL1) as predictors of distant seeding of circulating tumor cells in patients with metastatic breast cancer. Anticancer Res. 2013, 33, 1491–1497. [Google Scholar] [PubMed]
- Buck, M.B.; Fritz, P.; Dippon, J.; Zugmaier, G.; Knabbe, C. Prognostic significance of transforming growth factor beta receptor II in estrogen receptor-negative breast cancer patients. Clin. Cancer Res. 2004, 10, 491–498. [Google Scholar] [CrossRef] [Green Version]
- Richardsen, E.; Uglehus, R.D.; Johnsen, S.H.; Busund, L.T. Immunohistochemical expression of epithelial and stromal immunomodulatory signalling molecules is a prognostic indicator in breast cancer. BMC Res. Notes 2012, 5, 110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reis, S.T.; Pontes-Junior, J.; Antunes, A.A.; Sousa-Canavez, J.M.; Abe, D.K.; Cruz, J.A.; Dall’oglio, M.F.; Crippa, A.; Passerotti, C.C.; Ribeiro-Filho, L.A.; et al. Tgf-beta1 expression as a biomarker of poor prognosis in prostate cancer. Clinics 2011, 66, 1143–1147. [Google Scholar] [CrossRef]
- De Kruijf, E.M.; Dekker, T.J.A.; Hawinkels, L.; Putter, H.; Smit, V.; Kroep, J.R.; Kuppen, P.J.K.; van de Velde, C.J.H.; Ten Dijke, P.; Tollenaar, R.; et al. The prognostic role of TGF-beta signaling pathway in breast cancer patients. Ann. Oncol. 2013, 24, 384–390. [Google Scholar] [CrossRef]
- Tang, S.Y.; Alliston, T. Regulation of postnatal bone homeostasis by TGFbeta. Bonekey Rep. 2013, 2, 255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alliston, T.; Choy, L.; Ducy, P.; Karsenty, G.; Derynck, R. TGF-beta-induced repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and inhibits osteoblast differentiation. EMBO J. 2001, 20, 2254–2272. [Google Scholar] [CrossRef] [Green Version]
- Tang, Y.; Wu, X.; Lei, W.; Pang, L.; Wan, C.; Shi, Z.; Zhao, L.; Nagy, T.R.; Peng, X.; Hu, J.; et al. TGF-beta1-induced migration of bone mesenchymal stem cells couples bone resorption with formation. Nat. Med. 2009, 15, 757–765. [Google Scholar] [CrossRef] [Green Version]
- Iqbal, J.; Sun, L.; Zaidi, M. Coupling bone degradation to formation. Nat. Med. 2009, 15, 729–731. [Google Scholar] [CrossRef]
- Grafe, I.; Alexander, S.; Peterson, J.R.; Snider, T.N.; Levi, B.; Lee, B.; Mishina, Y. TGF-beta Family Signaling in Mesenchymal Differentiation. Cold Spring Harb. Perspect. Biol. 2018, 10, a022202. [Google Scholar] [CrossRef]
- Borton, A.J.; Frederick, J.P.; Datto, M.B.; Wang, X.F.; Weinstein, R.S. The loss of Smad3 results in a lower rate of bone formation and osteopenia through dysregulation of osteoblast differentiation and apoptosis. J. Bone Miner. Res. 2001, 16, 1754–1764. [Google Scholar] [CrossRef]
- Fox, S.W.; Lovibond, A.C. Current insights into the role of transforming growth factor-beta in bone resorption. Mol. Cell Endocrinol. 2005, 243, 19–26. [Google Scholar] [CrossRef]
- Kaneda, T.; Nojima, T.; Nakagawa, M.; Ogasawara, A.; Kaneko, H.; Sato, T.; Mano, H.; Kumegawa, M.; Hakeda, Y. Endogenous production of TGF-beta is essential for osteoclastogenesis induced by a combination of receptor activator of NF-kappa B ligand and macrophage-colony-stimulating factor. J. Immunol. 2000, 165, 4254–4263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mohammad, K.S.; Chen, C.G.; Balooch, G.; Stebbins, E.; McKenna, C.R.; Davis, H.; Niewolna, M.; Peng, X.H.; Nguyen, D.H.; Ionova-Martin, S.S.; et al. Pharmacologic inhibition of the TGF-beta type I receptor kinase has anabolic and anti-catabolic effects on bone. PLoS ONE 2009, 4, e5275. [Google Scholar] [CrossRef] [Green Version]
- Dole, N.S.; Mazur, C.M.; Acevedo, C.; Lopez, J.P.; Monteiro, D.A.; Fowler, T.W.; Gludovatz, B.; Walsh, F.; Regan, J.N.; Messina, S.; et al. Osteocyte-Intrinsic TGF-beta signaling regulates bone quality through perilacunar/canalicular remodeling. Cell Rep. 2017, 21, 2585–2596. [Google Scholar] [CrossRef] [Green Version]
- Weilbaecher, K.N.; Guise, T.A.; McCauley, L.K. Cancer to bone: A fatal attraction. Nat. Rev. Cancer 2011, 11, 411–425. [Google Scholar] [CrossRef] [PubMed]
- Esposito, M.; Guise, T.; Kang, Y. The Biology of Bone Metastasis. Cold Spring Harb. Perspect. Med. 2018, 8, a031252. [Google Scholar] [CrossRef]
- Patel, L.R.; Camacho, D.F.; Shiozawa, Y.; Pienta, K.J.; Taichman, R.S. Mechanisms of cancer cell metastasis to the bone: A multistep process. Future Oncol. 2011, 7, 1285–1297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guise, T.A. The vicious cycle of bone metastases. J. Musculoskelet. Neuronal Interact. 2002, 2, 570–572. [Google Scholar]
- Kang, Y.; He, W.; Tulley, S.; Gupta, G.P.; Serganova, I.; Chen, C.R.; Manova-Todorova, K.; Blasberg, R.; Gerald, W.L.; Massague, J. Breast cancer bone metastasis mediated by the Smad tumor suppressor pathway. Proc. Natl. Acad. Sci. USA 2005, 102, 13909–13914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sterling, J.A.; Edwards, J.R.; Martin, T.J.; Mundy, G.R. Advances in the biology of bone metastasis: How the skeleton affects tumor behavior. Bone 2011, 48, 6–15. [Google Scholar] [CrossRef]
- Mishra, S.; Tang, Y.; Wang, L.; deGraffenried, L.; Yeh, I.T.; Werner, S.; Troyer, D.; Copland, J.A.; Sun, L.Z. Blockade of transforming growth factor-beta (TGFbeta) signaling inhibits osteoblastic tumorigenesis by a novel human prostate cancer cell line. Prostate 2011, 71, 1441–1454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fournier, P.G.; Juarez, P.; Jiang, G.; Clines, G.A.; Niewolna, M.; Kim, H.S.; Walton, H.W.; Peng, X.H.; Liu, Y.; Mohammad, K.S.; et al. The TGF-beta Signaling Regulator PMEPA1 Suppresses Prostate Cancer Metastases to Bone. Cancer Cell 2015, 27, 809–821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ruscetti, F.W.; Akel, S.; Bartelmez, S.H. Autocrine transforming growth factor-beta regulation of hematopoiesis: Many outcomes that depend on the context. Oncogene 2005, 24, 5751–5763. [Google Scholar] [CrossRef] [Green Version]
- Bierie, B.; Moses, H.L. Tumour microenvironment: TGFbeta: The molecular Jekyll and Hyde of cancer. Nat. Rev. Cancer 2006, 6, 506–520. [Google Scholar] [CrossRef] [PubMed]
- Flavell, R.A.; Sanjabi, S.; Wrzesinski, S.H.; Licona-Limon, P. The polarization of immune cells in the tumour environment by TGFbeta. Nat. Rev. Immunol. 2010, 10, 554–567. [Google Scholar] [CrossRef] [Green Version]
- Sanjabi, S.; Oh, S.A.; Li, M.O. Regulation of the Immune Response by TGF-beta: From Conception to Autoimmunity and Infection. Cold Spring Harb. Perspect. Biol. 2017, 9, a022236. [Google Scholar] [CrossRef] [Green Version]
- Wahl, S.M.; Hunt, D.A.; Wakefield, L.M.; McCartney-Francis, N.; Wahl, L.M.; Roberts, A.B.; Sporn, M.B. Transforming growth factor type beta induces monocyte chemotaxis and growth factor production. Proc. Natl. Acad. Sci. USA 1987, 84, 5788–5792. [Google Scholar] [CrossRef] [Green Version]
- Ohura, K.; Katona, I.M.; Wahl, L.M.; Chenoweth, D.E.; Wahl, S.M. Co-expression of chemotactic ligand receptors on human peripheral blood monocytes. J. Immunol. 1987, 138, 2633–2639. [Google Scholar]
- Gong, D.; Shi, W.; Yi, S.J.; Chen, H.; Groffen, J.; Heisterkamp, N. TGFbeta signaling plays a critical role in promoting alternative macrophage activation. BMC Immunol. 2012, 13, 31. [Google Scholar] [CrossRef] [Green Version]
- Zhang, F.; Wang, H.; Wang, X.; Jiang, G.; Liu, H.; Zhang, G.; Wang, H.; Fang, R.; Bu, X.; Cai, S.; et al. TGF-beta induces M2-like macrophage polarization via SNAIL-mediated suppression of a pro-inflammatory phenotype. Oncotarget 2016, 7, 52294–52306. [Google Scholar] [CrossRef] [Green Version]
- Strobl, H.; Knapp, W. TGF-beta1 regulation of dendritic cells. Microbes Infect. 1999, 1, 1283–1290. [Google Scholar] [CrossRef]
- De Saint-Vis, B.; Fugier-Vivier, I.; Massacrier, C.; Gaillard, C.; Vanbervliet, B.; Ait-Yahia, S.; Banchereau, J.; Liu, Y.J.; Lebecque, S.; Caux, C. The cytokine profile expressed by human dendritic cells is dependent on cell subtype and mode of activation. J. Immunol. 1998, 160, 1666–1676. [Google Scholar] [PubMed]
- Jakob, T.; Saitoh, A.; Udey, M.C. E-cadherin-mediated adhesion involving Langerhans cell-like dendritic cells expanded from murine fetal skin. J. Immunol. 1997, 159, 2693–2701. [Google Scholar]
- Zermati, Y.; Fichelson, S.; Valensi, F.; Freyssinier, J.M.; Rouyer-Fessard, P.; Cramer, E.; Guichard, J.; Varet, B.; Hermine, O. Transforming growth factor inhibits erythropoiesis by blocking proliferation and accelerating differentiation of erythroid progenitors. Exp. Hematol. 2000, 28, 885–894. [Google Scholar] [CrossRef]
- Gao, X.; Lee, H.Y.; da Rocha, E.L.; Zhang, C.; Lu, Y.F.; Li, D.; Feng, Y.; Ezike, J.; Elmes, R.R.; Barrasa, M.I.; et al. TGF-beta inhibitors stimulate red blood cell production by enhancing self-renewal of BFU-E erythroid progenitors. Blood 2016, 128, 2637–2641. [Google Scholar] [CrossRef] [PubMed]
- Kuhikar, R.; Khan, N.; Philip, J.; Melinkeri, S.; Kale, V.; Limaye, L. Transforming growth factor beta1 accelerates and enhances in vitro red blood cell formation from hematopoietic stem cells by stimulating mitophagy. Stem Cell Res. Ther. 2020, 11, 71. [Google Scholar] [CrossRef]
- Sandra, G.; Robertson, A.; Ahamed, J. Megakaryocyte-Derived TGF-β1 Negatively Regulates Megakaryopoiesis in Mice. Blood 2020, 136, 36. [Google Scholar] [CrossRef]
- Hestdal, K.; Jacobsen, S.E.; Ruscetti, F.W.; Longo, D.L.; Boone, T.C.; Keller, J.R. Increased granulopoiesis after sequential administration of transforming growth factor-beta 1 and granulocyte-macrophage colony-stimulating factor. Exp. Hematol. 1993, 21, 799–805. [Google Scholar]
- Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Topalian, S.L.; Drake, C.G.; Pardoll, D.M. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr. Opin. Immunol. 2012, 24, 207–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Canellas, A.; Hernando-Momblona, X.; et al. TGFbeta drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [Green Version]
- Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel, E.E., III; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef]
- Zhang, Z.; Ma, L.; Goswami, S.; Ma, J.; Zheng, B.; Duan, M.; Liu, L.; Zhang, L.; Shi, J.; Dong, L.; et al. Landscape of infiltrating B cells and their clinical significance in human hepatocellular carcinoma. Oncoimmunology 2019, 8, e1571388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Z.; Zhu, Y.; Wang, Z.; Zhang, T.; Wu, P.; Huang, J. Yin-yang effect of tumor infiltrating B cells in breast cancer: From mechanism to immunotherapy. Cancer Lett. 2017, 393, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Ladanyi, A. Prognostic and predictive significance of immune cells infiltrating cutaneous melanoma. Pigment. Cell Melanoma Res. 2015, 28, 490–500. [Google Scholar] [CrossRef]
- Kehrl, J.H.; Roberts, A.B.; Wakefield, L.M.; Jakowlew, S.; Sporn, M.B.; Fauci, A.S. Transforming growth factor beta is an important immunomodulatory protein for human B lymphocytes. J. Immunol. 1986, 137, 3855–3860. [Google Scholar] [PubMed]
- Kehrl, J.H.; Thevenin, C.; Rieckmann, P.; Fauci, A.S. Transforming growth factor-beta suppresses human B lymphocyte Ig production by inhibiting synthesis and the switch from the membrane form to the secreted form of Ig mRNA. J. Immunol. 1991, 146, 4016–4023. [Google Scholar]
- Huai, G.; Markmann, J.F.; Deng, S.; Rickert, C.G. TGF-beta-secreting regulatory B cells: Unsung players in immune regulation. Clin. Transl. Immunol. 2021, 10, e1270. [Google Scholar] [CrossRef] [PubMed]
- Palumbo, J.S.; Talmage, K.E.; Massari, J.V.; La Jeunesse, C.M.; Flick, M.J.; Kombrinck, K.W.; Jirouskova, M.; Degen, J.L. Platelets and fibrin(ogen) increase metastatic potential by impeding natural killer cell-mediated elimination of tumor cells. Blood 2005, 105, 178–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Viel, S.; Marcais, A.; Guimaraes, F.S.; Loftus, R.; Rabilloud, J.; Grau, M.; Degouve, S.; Djebali, S.; Sanlaville, A.; Charrier, E.; et al. TGF-beta inhibits the activation and functions of NK cells by repressing the mTOR pathway. Sci. Signal. 2016, 9, ra19. [Google Scholar] [CrossRef] [PubMed]
- Trotta, R.; Dal Col, J.; Yu, J.; Ciarlariello, D.; Thomas, B.; Zhang, X.; Allard, J., 2nd; Wei, M.; Mao, H.; Byrd, J.C.; et al. TGF-beta utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and antibody-dependent cellular cytotoxicity in human NK cells. J. Immunol. 2008, 181, 3784–3792. [Google Scholar] [CrossRef] [Green Version]
- Donatelli, S.S.; Zhou, J.M.; Gilvary, D.L.; Eksioglu, E.A.; Chen, X.; Cress, W.D.; Haura, E.B.; Schabath, M.B.; Coppola, D.; Wei, S.; et al. TGF-beta-inducible microRNA-183 silences tumor-associated natural killer cells. Proc. Natl. Acad. Sci. USA 2014, 111, 4203–4208. [Google Scholar] [CrossRef] [Green Version]
- Haque, S.; Morris, J.C. Transforming growth factor-beta: A therapeutic target for cancer. Hum. Vaccin. Immunother. 2017, 13, 1741–1750. [Google Scholar] [CrossRef] [PubMed]
- Huynh, L.K.; Hipolito, C.J.; Ten Dijke, P. A Perspective on the Development of TGF-beta Inhibitors for Cancer Treatment. Biomolecules 2019, 9, 743. [Google Scholar] [CrossRef] [Green Version]
- Teicher, B.A. TGFbeta-Directed Therapeutics: 2020. Pharmacol. Ther. 2021, 217, 107666. [Google Scholar] [CrossRef]
- Lenferink, A.E.; Magoon, J.; Pepin, M.C.; Guimond, A.; O’Connor-McCourt, M.D. Expression of TGF-beta type II receptor antisense RNA impairs TGF-beta signaling in vitro and promotes mammary gland differentiation in vivo. Int. J. Cancer 2003, 107, 919–928. [Google Scholar] [CrossRef]
- Muraoka, R.S.; Dumont, N.; Ritter, C.A.; Dugger, T.C.; Brantley, D.M.; Chen, J.; Easterly, E.; Roebuck, L.R.; Ryan, S.; Gotwals, P.J.; et al. Blockade of TGF-beta inhibits mammary tumor cell viability, migration, and metastases. J. Clin. Investig. 2002, 109, 1551–1559. [Google Scholar] [CrossRef] [PubMed]
- Nam, J.S.; Terabe, M.; Mamura, M.; Kang, M.J.; Chae, H.; Stuelten, C.; Kohn, E.; Tang, B.; Sabzevari, H.; Anver, M.R.; et al. An anti-transforming growth factor beta antibody suppresses metastasis via cooperative effects on multiple cell compartments. Cancer Res. 2008, 68, 3835–3843. [Google Scholar] [CrossRef] [Green Version]
- Smith, A.L.; Robin, T.P.; Ford, H.L. Molecular pathways: Targeting the TGF-beta pathway for cancer therapy. Clin. Cancer Res. 2012, 18, 4514–4521. [Google Scholar] [CrossRef] [Green Version]
- Morris, J.C.; Tan, A.R.; Olencki, T.E.; Shapiro, G.I.; Dezube, B.J.; Reiss, M.; Hsu, F.J.; Berzofsky, J.A.; Lawrence, D.P. Phase I study of GC1008 (fresolimumab): A human anti-transforming growth factor-beta (TGFbeta) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PLoS ONE 2014, 9, e90353. [Google Scholar] [CrossRef]
- Zhu, H.; Gu, X.; Xia, L.; Zhou, Y.; Bouamar, H.; Yang, J.; Ding, X.; Zwieb, C.; Zhang, J.; Hinck, A.P.; et al. A Novel TGFbeta Trap Blocks Chemotherapeutics-Induced TGFbeta1 Signaling and Enhances Their Anticancer Activity in Gynecologic Cancers. Clin. Cancer Res. 2018, 24, 2780–2793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, H.; Wang, J.; Koteliansky, V.E.; Gotwals, P.J.; Hauer-Jensen, M. Recombinant soluble transforming growth factor beta type II receptor ameliorates radiation enteropathy in mice. Gastroenterology 2000, 119, 1286–1296. [Google Scholar] [CrossRef]
- Nemunaitis, J.; Dillman, R.O.; Schwarzenberger, P.O.; Senzer, N.; Cunningham, C.; Cutler, J.; Tong, A.; Kumar, P.; Pappen, B.; Hamilton, C.; et al. Phase II study of belagenpumatucel-L, a transforming growth factor beta-2 antisense gene-modified allogeneic tumor cell vaccine in non-small-cell lung cancer. J. Clin. Oncol. 2006, 24, 4721–4730. [Google Scholar] [CrossRef]
- Inman, G.J.; Nicolas, F.J.; Callahan, J.F.; Harling, J.D.; Gaster, L.M.; Reith, A.D.; Laping, N.J.; Hill, C.S. SB-431542 is a potent and specific inhibitor of transforming growth factor-beta superfamily type I activin receptor-like kinase (ALK) receptors ALK4, ALK5, and ALK7. Mol. Pharmacol. 2002, 62, 65–74. [Google Scholar] [CrossRef]
- Petersen, M.; Thorikay, M.; Deckers, M.; van Dinther, M.; Grygielko, E.T.; Gellibert, F.; de Gouville, A.C.; Huet, S.; ten Dijke, P.; Laping, N.J. Oral administration of GW788388, an inhibitor of TGF-beta type I and II receptor kinases, decreases renal fibrosis. Kidney Int. 2008, 73, 705–715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tojo, M.; Hamashima, Y.; Hanyu, A.; Kajimoto, T.; Saitoh, M.; Miyazono, K.; Node, M.; Imamura, T. The ALK-5 inhibitor A-83-01 inhibits Smad signaling and epithelial-to-mesenchymal transition by transforming growth factor-beta. Cancer Sci. 2005, 96, 791–800. [Google Scholar] [CrossRef] [PubMed]
- Ehata, S.; Hanyu, A.; Fujime, M.; Katsuno, Y.; Fukunaga, E.; Goto, K.; Ishikawa, Y.; Nomura, K.; Yokoo, H.; Shimizu, T.; et al. Ki26894, a novel transforming growth factor-beta type I receptor kinase inhibitor, inhibits in vitro invasion and in vivo bone metastasis of a human breast cancer cell line. Cancer Sci. 2007, 98, 127–133. [Google Scholar] [CrossRef]
- Dunn, L.K.; Mohammad, K.S.; Fournier, P.G.; McKenna, C.R.; Davis, H.W.; Niewolna, M.; Peng, X.H.; Chirgwin, J.M.; Guise, T.A. Hypoxia and TGF-beta drive breast cancer bone metastases through parallel signaling pathways in tumor cells and the bone microenvironment. PLoS ONE 2009, 4, e6896. [Google Scholar] [CrossRef] [Green Version]
- Xu, Q.; Zhang, X.M.; Duan, K.Z.; Gu, X.Y.; Han, M.; Liu, B.L.; Zhao, Z.Q.; Zhang, Y.Q. Peripheral TGF-beta1 signaling is a critical event in bone cancer-induced hyperalgesia in rodents. J. Neurosci. 2013, 33, 19099–19111. [Google Scholar] [CrossRef] [Green Version]
- Crooke, S.T. Progress in antisense technology. Annu. Rev. Med. 2004, 55, 61–95. [Google Scholar] [CrossRef] [PubMed]
- Bennett, C.F.; Swayze, E.E. RNA targeting therapeutics: Molecular mechanisms of antisense oligonucleotides as a therapeutic platform. Annu. Rev. Pharmacol. Toxicol. 2010, 50, 259–293. [Google Scholar] [CrossRef] [PubMed]
- Yingling, J.M.; Blanchard, K.L.; Sawyer, J.S. Development of TGF-beta signalling inhibitors for cancer therapy. Nat. Rev. Drug Discov. 2004, 3, 1011–1022. [Google Scholar] [CrossRef] [PubMed]
- Muraoka-Cook, R.S.; Kurokawa, H.; Koh, Y.; Forbes, J.T.; Roebuck, L.R.; Barcellos-Hoff, M.H.; Moody, S.E.; Chodosh, L.A.; Arteaga, C.L. Conditional overexpression of active transforming growth factor beta1 in vivo accelerates metastases of transgenic mammary tumors. Cancer Res. 2004, 64, 9002–9011. [Google Scholar] [CrossRef] [Green Version]
- Huang, C.Y.; Chung, C.L.; Hu, T.H.; Chen, J.J.; Liu, P.F.; Chen, C.L. Recent progress in TGF-beta inhibitors for cancer therapy. Biomed. Pharmacother. 2021, 134, 111046. [Google Scholar] [CrossRef]
- Juarez, P.; Fournier, P.G.J.; Mohammad, K.S.; McKenna, R.C.; Davis, H.W.; Peng, X.H.; Niewolna, M.; Mauviel, A.; Chirgwin, J.M.; Guise, T.A. Halofuginone inhibits TGF-beta/BMP signaling and in combination with zoledronic acid enhances inhibition of breast cancer bone metastasis. Oncotarget 2017, 8, 86447–86462. [Google Scholar] [CrossRef] [Green Version]
- Juarez, P.; Mohammad, K.S.; Yin, J.J.; Fournier, P.G.; McKenna, R.C.; Davis, H.W.; Peng, X.H.; Niewolna, M.; Javelaud, D.; Chirgwin, J.M.; et al. Halofuginone inhibits the establishment and progression of melanoma bone metastases. Cancer Res. 2012, 72, 6247–6256. [Google Scholar] [CrossRef] [Green Version]
- Chiechi, A.; Waning, D.L.; Stayrook, K.R.; Buijs, J.T.; Guise, T.A.; Mohammad, K.S. Role of TGF-beta in breast cancer bone metastases. Adv. Biosci. Biotechnol. 2013, 4, 15–30. [Google Scholar] [CrossRef] [Green Version]
- Cui, Q.; Lim, S.K.; Zhao, B.; Hoffmann, F.M. Selective inhibition of TGF-beta responsive genes by Smad-interacting peptide aptamers from FoxH1, Lef1 and CBP. Oncogene 2005, 24, 3864–3874. [Google Scholar] [CrossRef] [Green Version]
- Zhao, B.M.; Hoffmann, F.M. Inhibition of transforming growth factor-beta1-induced signaling and epithelial-to-mesenchymal transition by the Smad-binding peptide aptamer Trx-SARA. Mol. Biol. Cell 2006, 17, 3819–3831. [Google Scholar] [CrossRef] [Green Version]
- Gadir, N.; Jackson, D.N.; Lee, E.; Foster, D.A. Defective TGF-beta signaling sensitizes human cancer cells to rapamycin. Oncogene 2008, 27, 1055–1062. [Google Scholar] [CrossRef] [Green Version]
- Filyak, Y.; Filyak, O.; Stoika, R. Transforming growth factor beta-1 enhances cytotoxic effect of doxorubicin in human lung adenocarcinoma cells of A549 line. Cell Biol. Int. 2007, 31, 851–855. [Google Scholar] [CrossRef]
- Formenti, S.C.; Lee, P.; Adams, S.; Goldberg, J.D.; Li, X.; Xie, M.W.; Ratikan, J.A.; Felix, C.; Hwang, L.; Faull, K.F.; et al. Focal Irradiation and Systemic TGFbeta Blockade in Metastatic Breast Cancer. Clin. Cancer Res. 2018, 24, 2493–2504. [Google Scholar] [CrossRef] [Green Version]
- Formenti, S.C.; Hawtin, R.E.; Dixit, N.; Evensen, E.; Lee, P.; Goldberg, J.D.; Li, X.; Vanpouille-Box, C.; Schaue, D.; McBride, W.H.; et al. Baseline T cell dysfunction by single cell network profiling in metastatic breast cancer patients. J. Immunother. Cancer 2019, 7, 177. [Google Scholar] [CrossRef]
- Wang, K.; Gu, Y.; Liao, Y.; Bang, S.; Donnelly, C.R.; Chen, O.; Tao, X.; Mirando, A.J.; Hilton, M.J.; Ji, R.R. PD-1 blockade inhibits osteoclast formation and murine bone cancer pain. J. Clin. Investig. 2020, 130, 3603–3620. [Google Scholar] [CrossRef] [PubMed]
- Derynck, R.; Turley, S.J.; Akhurst, R.J. TGFbeta biology in cancer progression and immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18, 9–34. [Google Scholar] [CrossRef] [PubMed]
- Chang, Z.L.; Lorenzini, M.H.; Chen, X.; Tran, U.; Bangayan, N.J.; Chen, Y.Y. Rewiring T-cell responses to soluble factors with chimeric antigen receptors. Nat. Chem. Biol. 2018, 14, 317–324. [Google Scholar] [CrossRef]
- Hou, A.J.; Chang, Z.L.; Lorenzini, M.H.; Zah, E.; Chen, Y.Y. TGF-beta-responsive CAR-T cells promote anti-tumor immune function. Bioeng. Transl. Med. 2018, 3, 75–86. [Google Scholar] [CrossRef]
- Bhola, N.E.; Balko, J.M.; Dugger, T.C.; Kuba, M.G.; Sanchez, V.; Sanders, M.; Stanford, J.; Cook, R.S.; Arteaga, C.L. TGF-beta inhibition enhances chemotherapy action against triple-negative breast cancer. J. Clin. Investig. 2013, 123, 1348–1358. [Google Scholar] [CrossRef]
- Ahmadi, A.; Najafi, M.; Farhood, B.; Mortezaee, K. Transforming growth factor-beta signaling: Tumorigenesis and targeting for cancer therapy. J. Cell Physiol. 2019, 234, 12173–12187. [Google Scholar] [CrossRef] [PubMed]
- Van den Bulk, J.; de Miranda, N.; Ten Dijke, P. Therapeutic targeting of TGF-beta in cancer: Hacking a master switch of immune suppression. Clin. Sci. 2021, 135, 35–52. [Google Scholar] [CrossRef]
- Hengst, V.; Oussoren, C.; Kissel, T.; Storm, G. Bone targeting potential of bisphosphonate-targeted liposomes. Preparation, characterization and hydroxyapatite binding in vitro. Int. J. Pharm. 2007, 331, 224–227. [Google Scholar] [CrossRef] [PubMed]
- Wang, G.; Mostafa, N.Z.; Incani, V.; Kucharski, C.; Uludag, H. Bisphosphonate-decorated lipid nanoparticles designed as drug carriers for bone diseases. J. Biomed. Mater. Res. A 2012, 100, 684–693. [Google Scholar] [CrossRef] [PubMed]
- Ganesh, K.; Massague, J. Targeting metastatic cancer. Nat. Med. 2021, 27, 34–44. [Google Scholar] [CrossRef] [PubMed]
- Bai, X.; Yi, M.; Jiao, Y.; Chu, Q.; Wu, K. Blocking TGF-beta signaling to enhance the efficacy of immune checkpoint inhibitor. OncoTargets Ther. 2019, 12, 9527–9538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lind, H.; Gameiro, S.R.; Jochems, C.; Donahue, R.N.; Strauss, J.; Gulley, J.M.; Palena, C.; Schlom, J. Dual targeting of TGF-beta and PD-L1 via a bifunctional anti-PD-L1/TGF-betaRII agent: Status of preclinical and clinical advances. J. Immunother. Cancer 2020, 8, e000433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Groeneveldt, C.; van Hall, T.; van der Burg, S.H.; Ten Dijke, P.; van Montfoort, N. Immunotherapeutic Potential of TGF-beta Inhibition and Oncolytic Viruses. Trends Immunol. 2020, 41, 406–420. [Google Scholar] [CrossRef]
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
Trivedi, T.; Pagnotti, G.M.; Guise, T.A.; Mohammad, K.S. The Role of TGF-β in Bone Metastases. Biomolecules 2021, 11, 1643. https://doi.org/10.3390/biom11111643
Trivedi T, Pagnotti GM, Guise TA, Mohammad KS. The Role of TGF-β in Bone Metastases. Biomolecules. 2021; 11(11):1643. https://doi.org/10.3390/biom11111643
Chicago/Turabian StyleTrivedi, Trupti, Gabriel M. Pagnotti, Theresa A. Guise, and Khalid S. Mohammad. 2021. "The Role of TGF-β in Bone Metastases" Biomolecules 11, no. 11: 1643. https://doi.org/10.3390/biom11111643
APA StyleTrivedi, T., Pagnotti, G. M., Guise, T. A., & Mohammad, K. S. (2021). The Role of TGF-β in Bone Metastases. Biomolecules, 11(11), 1643. https://doi.org/10.3390/biom11111643