Take a look at the Recent articles

Putting a recombinant extracellular peptide of human Fzd7 into the context of triple negative breast cancer

Zhaoli Zhou

Shanghai Key Laboratory for Molecular Imaging, Collaborative Scientific Research Center. Shanghai University of Medicine & Health Science, Shanghai, China

Department of Pharmacology, Faculty School of Pharmacy, Shanghai University of Medicine & Health Science, Shanghai, China.

Wei Xie

Shanghai Key Laboratory for Molecular Imaging, Collaborative Scientific Research Center. Shanghai University of Medicine & Health Science, Shanghai, China

Department of Pharmacology, Faculty School of Pharmacy, Shanghai University of Medicine & Health Science, Shanghai, China.

Yihan Qian

Faculty School of Pharmacy, Fudan University, Shanghai, China

Jin Wang

Department of Pharmacology, Faculty School of Pharmacy, Shanghai University of Medicine & Health Science, Shanghai, China.

Gang Huang

Shanghai Key Laboratory for Molecular Imaging, Collaborative Scientific Research Center. Shanghai University of Medicine & Health Science, Shanghai, China

DOI: 10.15761/JCMM.1000122

Article
Article Info
Author Info
Figures & Data

Abstract

Triple negative breast cancer (TNBC) is one of those intractable malignancies, featured by dysregulated activation of Wnt/β-catenin signaling pathway. A recent study conducted by Shanghai University of Medicine & Health Science shows that a novel recombinant human Frizzled-7 protein (rhFzd7) has potential efficacy against triple negative breast cancer by inhibiting Wnt/β-catenin pathway, which give insightful meaning for the development of Fzd7-targeted drugs against TNBC. rhFzd7 may serve as a pharmacological probe to explore further the function of FZD7 in cancer biology and develop FZD7-based antibody.

Keywords

human Fzd7, breast cancer, Fzd7-targeted drugs, FZD7-based antibody, Triple negative breast cancer

Background

Frizzled receptors (FZDs) are a family of seven-transmembrane-spanning proteins belonging to the superfamily of G protein-coupled receptors [1]. They are comprised of ten members (FZD1–FZD10), which anchor in cell membrane and function as receptors of Wingless-type (WNT) signaling pathway. Wnt/β-catenin signaling pathway through FZDs play key roles in both normal development and tumorigenesis, especially they have been demonstrated to be involved in a large variety of cancer-related processes, such as proliferation, migration, invasion, angiogenesis, stemness, and chemoresistance after cancer recurrence. FZD7 is the most studied member in the FZDs family. Those findings highlight the biological role of FZD7 in cancer and indicate FZD7 being an attractive pharmacological target.

Different approaches have been designed to block FZD7, including small molecule inhibitors, small peptides, antibodies, and soluble FZD7 [2]. A recent review, titled as “Frizzled Receptors as Potential Therapeutic Targets in Human Cancers”, give a list of those FZD7 antibodies/inhibitors under development, including two small molecule compounds SRI37892 and FJ9, two small interfering peptides RHPDs and dFz7-21 [3], two antibodies scFvs and OMP-18R5, and one soluble FZD7. OMP-18R5 (Vantictumab) is the only one currently in phase I clinical evaluation. The cancer type for those inhibitors are Wilms’ tumor, breast cancer, pancreatic cancer, and hepatocellular carcinoma, et al.

Triple negative breast cancer (TNBC) is a type of breast cancer whose expression of ER, PR, and HER2 is negative. Clinically it is characterized as more aggressive and less responsive to standard treatment with a poorer overall patient prognosis. Recent studies indicate that Wnt/β-catenin signaling is particularly activated in TNBC. FZD7 is frequently upregulated in TNBC cell lines and tissues. Knockdown of FZD7 with siRNA, or using scFvs against FZD7, can suppress tumor transformation and inhibit tumor growth of TNBC. One condition of the clinical trial of Vantictumab is for locally recurrent or metastatic breast cancer (In combination with Paclitaxel, NCT01973309).

In sum, targeting Wnt signal pathway, especially targeting Fzd7, is a promising strategy against TNBC. Those inhibitors under development will be helpful for us to clarify the pathway, such as their role in cancer, and the redundancy of each components in this pathway. They are also promising agents against the intractable cancer TNBC.

Results and Conclusion

rhFzd7 (around 27kd) was generated using E. coli system, with the codons optimized by the preference of E. coli. This novel rhFzd7 was demonstrated to have specificity and affinity to Wnt3a, antagonize Wnt/β-catenin pathway, and exhibit anti-tumor and anti-angiogenesis activity against TNBC. When used in combination with standard chemotherapeutic agent Docetaxel, rhFzd7 sensitized TNBC to Docetaxel significantly. These results give insightful meaning for the development of Fzd7-targeted drugs against TNBC. The peptide described here represents a promising small ligand that specifically targets FZD7, and it serves as a pharmacological probe to explore further the function of FZD7 in cancer biology and develop FZD7-based antibody.

References

  1. Eisenbarth GS (1986) Type I diabetes mellitus. A chronic autoimmune disease. N Engl J Med 314: 1360-1368. [Crossref]
  2. Tisch R, McDevitt H (1996) Insulin-dependent diabetes mellitus. Cell 85: 291-297. [Crossref]
  3. Libby P, Nathan DM, Abraham K, Brunzell JD, Fradkin JE, et al. (2005) Report of the National Heart, Lung, and Blood Institute-National Institute of Diabetes and Digestive and Kidney Diseases Working Group on Cardiovascular Complications of Type 1 Diabetes Mellitus. Circulation 111: 3489-3493. [Crossref]
  4. Zhang J, Yuen BH, Currie WD, Leung PC (1991) Suppression of luteinizing hormone secretion by atrial and brain natriuretic peptides in ovariectomized rats. Endocrinology 129: 801-806. [Crossref]
  5. Czerkinsky C, Anjuere F, McGhee JR, George-Chandy A, Holmgren J, et al. (1999) Mucosal immunity and tolerance: relevance to vaccine development. Immunol Rev 170: 197-222. [Crossref]
  6. Arakawa T, Yu J, Chong DK, Hough J, Engen PC, et al. (1998) A plant-based cholera toxin B subunit-insulin fusion protein protects against the development of autoimmune diabetes. Nat Biotechnol 16: 934-938. [Crossref]
  7. Arakawa T, Chong DK, Langridge WH (1998) Efficacy of a food plant-based oral cholera toxin B subunit vaccine. Nat Biotechnol 16: 292-297. [Crossref]
  8. Arakawa T, Yu J, Langridge WH (1999) Food plant-delivered cholera toxin B subunit for vaccination and immunotolerization. Adv Exp Med Biol 464: 161-178. [Crossref]
  9. Bergerot I, Ploix C, Petersen J, Moulin V, Rask C, et al. (1997) A cholera toxoid-insulin conjugate as an oral vaccine against spontaneous autoimmune diabetes. Proc Natl Acad Sci U S A 94: 4610-4614. [Crossref]
  10. Shreedhar VK, Kelsall BL, Neutra MR (2003) Cholera toxin induces migration of dendritic cells from the subepithelial dome region to T- and B-cell areas of Peyer's patches. Infect Immun 71: 504-509. [Crossref]
  11. Sun JB, Holmgren J, Czerkinsky C (1994) Cholera toxin B subunit: an efficient transmucosal carrier-delivery system for induction of peripheral immunological tolerance. Proc Natl Acad Sci U S A 91: 10795-10799. [Crossref]
  12. Carter JE 3rd, Yu J, Choi NW, Hough J, Henderson D, et al. (2006) Bacterial and plant enterotoxin B subunit-autoantigen fusion proteins suppress diabetes insulitis. Mol Biotechnol 32: 1-15. [Crossref]
  13. Banchereau J, Steinman RM (1998) Dendritic cells and the control of immunity. Nature 392: 245-252. [Crossref]
  14. Pulendran B, Smith JL, Caspary G, Brasel K, Pettit D, et al. (1999) Distinct dendritic cell subsets differentially regulate the class of immune response in vivo. Proc Natl Acad Sci U S A 96: 1036-1041. [Crossref]
  15. Pulendran B, Kumar P, Cutler CW, Mohamadzadeh M, Van Dyke T, et al. (2001) Lipopolysaccharides from distinct pathogens induce different classes of immune responses in vivo. J Immunol 167: 5067-5076. [Crossref]
  16. Itano AA, McSorley SJ, Reinhardt RL, Ehst BD, Ingulli E, et al. (2003) Distinct dendritic cell populations sequentially present antigen to CD4 T cells and stimulate different aspects of cell-mediated immunity. Immunity 19: 47-57. [Crossref]
  17. Kapsenberg ML (2003) Dendritic-cell control of pathogen-driven T-cell polarization. Nat Rev Immunol 3: 984-993. [Crossref]
  18. Piccinni MP, Beloni L, Livi C, Maggi E, Scarselli G, et al. (1998) Defective production of both leukemia inhibitory factor and type 2 T-helper cytokines by decidual T cells in unexplained recurrent abortions. Nat Med 4: 1020-1024. [Crossref]
  19. Odumosu O, Payne K, Baez I, Jutzy J, Wall N, et al. (2010) Suppression of dendritic cell activation by diabetes autoantigens linked to the cholera toxin B subunit. Immunobiology 216: 447-456. [Crossref]
  20. Ebert LM, MacRaild SE, Davis ID, Cebon J, Chen W (2012) A novel method for detecting antigen-specific human regulatory T cells. J Immunol Methods 377: 56-61. [Crossref]
  21. Valitutti S, Lanzavecchia A (1997) Serial triggering of TCRs: a basis for the sensitivity and specificity of antigen recognition. Immunol Today 18: 299-304. [Crossref]
  22. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M (1995) Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 155: 1151-64. [Crossref]
  23. Steinman RM, Turley S, Mellman I, Inaba K (2000) The induction of tolerance by dendritic cells that have captured apoptotic cells. J Exp Med 191: 411-416. [Crossref]
  24. Wakkach A, Fournier N, Brun V, Breittmayer JP, Cottrez F, et al. (2003) Characterization of dendritic cells that induce tolerance and T regulatory 1 cell differentiation in vivo. Immunity18: 605-617. [Crossref]
  25. Gong Z, Long X, Pan L, Le Y, Liu Q, et al. (2009) Cloning, expression, purification and characterization of the cholera toxin B subunit and triple glutamic acid decarboxylase epitopes fusion protein in Escherichia coli. Protein Expr Purif 66: 191-197. [Crossref]
  26. Gong Z, Jin Y, Zhang Y (2007) Suppression of diabetes in non-obese diabetic (NOD) mice by oral administration of a cholera toxin B subunit-insulin B chain fusion protein vaccine produced in silkworm. Vaccine 25: 1444-1451. [Crossref]
  27. Denes B, Krausova V, Fodor N, Timiryasova T, Henderson D, et al. (2005) Protection of NOD mice from type 1 diabetes after oral inoculation with vaccinia viruses expressing adjuvanted islet autoantigens. J Immunother 28: 438-448. [Crossref]
  28. Battaglia M, Stabilini A, Draghici E, Migliavacca B, Gregori S, et al. (2006) Induction of tolerance in type 1 diabetes via both CD4+CD25+ T regulatory cells and T regulatory type 1 cells. Diabetes 55: 1571-1580. [Crossref]
  29. Roncarolo MG, Gregori S, Battaglia M, Bacchetta R, Fleischhauer K, et al. (2006) Interleukin-10-secreting type 1 regulatory T cells in rodents and humans. Immunol Rev 212: 28-50. [Crossref]
  30. Levings MK, Gregori S, Tresoldi E, Cazzaniga S, Bonini C, et al. (2005) Differentiation of Tr1 cells by immature dendritic cells requires IL-10 but not CD25+CD4+ Tr cells. Blood 105: 1162-1169. [Crossref]
  31. Khattri R, Cox T, Yasayko SA, Ramsdell F (2003) An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat Immunol 4: 337-342. [Crossref]
  32. Hori S, Nomura T, Sakaguchi S (2003) Control of regulatory T cell development by the transcription factor Foxp3. Science 299: 1057-1061. [Crossref]
  33. Fontenot JD, Gavin MA, Rudensky AY (2003) Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 4: 330-336. [Crossref]
  34. Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, et al. (2006) CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med 203: 1701-1711.
  35. Yudoh K, Matsuno H, Nakazawa F, Yonezawa T, Kimura T (2000) Reduced expression of the regulatory CD4+ T cell subset is related to Th1/Th2 balance and disease severity in rheumatoid arthritis. Arthritis Rheum 43: 617-627. [Crossref]
  36. Hall AM, Ward FJ, Vickers MA, Stott LM, Urbaniak SJ, et al. (2002) Interleukin-10-mediated regulatory T-cell responses to epitopes on a human red blood cell autoantigen. Blood 100: 4529-4536. [Crossref]
  37. Fujino S, Andoh A, Bamba S, Ogawa A, Hata K, et al. (2003) Increased expression of interleukin 17 in inflammatory bowel disease. Gut 52: 65-70. [Crossref]
  38. Nakae S, Nambu A, Sudo K, Iwakura Y (2003) Suppression of immune induction of collagen-induced arthritis in IL-17-deficient mice. J Immunol 171: 6173-6177. [Crossref]
  39. Komiyama Y, Nakae S, Matsuki T, Nambu A, Ishigame H, et al. (2006) IL-17 plays an important role in the development of experimental autoimmune encephalomyelitis. J Immunol 177: 566-573. [Crossref]
  40. Tzartos JS, Friese MA, Craner MJ, Palace J, Newcombe J, et al. (2008) Interleukin-17 production in central nervous system-infiltrating T cells and glial cells is associated with active disease in multiple sclerosis. Am J Pathol 172: 146-155. [Crossref]
  41. Wong CK, Ho CY, Li EK, Lam CW (2000) Elevation of proinflammatory cytokine (IL-18, IL-17, IL-12) and Th2 cytokine (IL-4) concentrations in patients with systemic lupus erythematosus. Lupus 9: 589-593. [Crossref]
  42. Hofstetter HH, Ibrahim SM, Koczan D, Kruse N, Weishaupt A, et al. (2005) Therapeutic efficacy of IL-17 neutralization in murine experimental autoimmune encephalomyelitis. Cell Immunol 237: 123-130. [Crossref]
  43. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, et al. (2006) Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441: 235-238. [Crossref]
  44. Mangan PR, Harrington LE, O'Quinn DB, Helms WS, Bullard DC, et al. (2006) Transforming growth factor-beta induces development of the T(H)17 lineage. Nature 441: 231-234. [Crossref]
  45. Yokota S, Imagawa T, Mori M, Miyamae T, Aihara Y, et al. (2008) Efficacy and safety of tocilizumab in patients with systemic-onset juvenile idiopathic arthritis: a randomised, double-blind, placebo-controlled, withdrawal phase III trial. Lancet 371: 998-1006. [Crossref]
  46. Nishimoto N, Terao K, Mima T, Nakahara H, Takagi N, et al. (2008) Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease. Blood 112: 3959-3964. [Crossref]

Editorial Information

Editor-in-Chief

Huasong Zeng
Guangzhou Medical University

Article Type

Short Communication

Publication history

Received date: November 28, 2018
Accepted date: December 10, 2018
Published date: December 12, 2018

Copyright

© 2018 Zhou Z. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Citation

Zhou Z, Xie W, Qian Y, Wang J, Huang G (2018) Putting a recombinant extracellular peptide of human Fzd7 into the context of triple negative breast cancer. J Clin Mol Med 1: DOI: 10.15761/JCMM.1000122

Corresponding author

Gang Huang

Shanghai Key Laboratory for Molecular Imaging, Collaborative Research Center. Shanghai University of Medicine & Health Science, Shanghai, P.R. China

E-mail : bhuvaneswari.bibleraaj@uhsm.nhs.uk

No Figures.