Blood Res.  2019 Sep;54(3):165-174. 10.5045/br.2019.54.3.165.

Mesenchymal stem cells in acute myeloid leukemia: a focus on mechanisms involved and therapeutic concepts

Affiliations
  • 1Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
  • 2Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. farahzadir@tbzmed.ac.ir

Abstract

Drug resistance in cancer, especially in leukemia, creates a dilemma in treatment planning. Consequently, studies related to the mechanisms underlying drug resistance, the molecular pathways involved in this phenomenon, and alternate therapies have attracted the attention of researchers. Among a variety of therapeutic modalities, mesenchymal stem cells (MSCs) are of special interest due to their potential clinical use. Therapies involving MSCs are showing increasing promise in cancer treatment and anticancer drug screening applications; however, results have been inconclusive, possibly due to the heterogeneity of MSC populations. Most recently, the effect of MSCs on different types of cancer, such as hematologic malignancies, their mechanisms, sources of MSCs, and its advantages and disadvantages have been discussed. There are many proposed mechanisms describing the effects of MSCs in hematologic malignancies; however, the most commonly-accepted mechanism is that MSCs induce tumor cell cycle arrest. This review explains the anti-tumorigenic effects of MSCs through the suppression of tumor cell proliferation in hematological malignancies, especially in acute myeloid leukemia.

Keyword

Mesenchymal stem cells; Hematologic malignancy; Acute myeloid leukemia; Cancer therapy; Cell cycle arrest

MeSH Terms

Cell Cycle Checkpoints
Cell Proliferation
Drug Evaluation, Preclinical
Drug Resistance
Hematologic Neoplasms
Leukemia
Leukemia, Myeloid, Acute*
Mesenchymal Stromal Cells*
Population Characteristics

Figure

  • Fig. 1 Multi-lineage differentiation and cell surface markers of MSCs.

  • Fig. 2 Therapeutic implications of MSCs in the treatment of leukemia.

  • Fig. 3 Schema for the dual roles of MSCs in hematologic malignancy. MSCs have both anti-tumorigenic and pro-tumorigenic effects, as they tend to not only inhibit tumor growth but also promote tumor growth by suppressing tumor cell apoptosis.

  • Fig. 4 MSC and tumor cell interactions as MSC-based cancer therapy. The chemotactic movement of MSCs toward a tumor niche is driven by soluble factors such as EGF, IL-6, IL-8, TGF-β, and PDGF. Genetic modification of MSCs can be used to deliver a range of tumor-suppressing cargos directly into the tumor site. These cargos include growth factors and cytokines, immune-modulating agents (IFN-α, IFN-β, IL-2, IL-12, CX3CL1 etc.), and regulators of gene expression (miRNAs and other non-coding RNAs). MSCs are also capable of delivering therapeutic drugs within the tumor site. Also, micro vesicles derived from MSCs represent an alternative approach to delivering these agents.


Reference

1. Lee MW, Ryu S, Kim DS, et al. Mesenchymal stem cells in suppression or progression of hematologic malignancy: current status and challenges. Leukemia. 2019; 33:597–611.
Article
2. Fathi E, Farahzadi R. Isolation, culturing, characterization and aging of adipose tissue-derived mesenchymal stem cells: a brief overview. Braz Arch Biol Technol. 2016; 59:e16150383.
Article
3. Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991; 9:641–650.
Article
4. Varma MJ, Breuls RG, Schouten TE, et al. Phenotypical and functional characterization of freshly isolated adipose tissue-derived stem cells. Stem Cells Dev. 2007; 16:91–104.
Article
5. Mohammadian M, Shamsasenjan K, Lotfi Nezhad P, et al. Mesenchymal stem cells: new aspect in cell-based regenerative therapy. Adv Pharm Bull. 2013; 3:433–437.
6. Perrot P, Rousseau J, Bouffaut AL, et al. Safety concern between autologous fat graft, mesenchymal stem cell and osteosarcoma recurrence. PLoS One. 2010; 5:e10999.
Article
7. Fathi E, Farahzadi R. Zinc sulphate mediates the stimulation of cell proliferation of rat adipose tissue-derived mesenchymal stem cells under high intensity of EMF exposure. Biol Trace Elem Res. 2018; 184:529–535.
Article
8. Fathi E, Farahzadi R, Sheikhzadeh N. Immunophenotypic characterization, multi-lineage differentiation and aging of zebrafish heart and liver tissue-derived mesenchymal stem cells as a novel approach in stem cell-based therapy. Tissue Cell. 2019; 57:15–21.
Article
9. Farahzadi R, Fathi E, Mesbah-Namin SA, Zarghami N. Anti-aging protective effect of L-carnitine as clinical agent in regenerative medicine through increasing telomerase activity and change in the hTERT promoter CpG island methylation status of adipose tissue-derived mesenchymal stem cells. Tissue Cell. 2018; 54:105–113.
Article
10. Ren Y, Wu H, Zhou X, et al. Isolation, expansion, and differentiation of goat adipose-derived stem cells. Res Vet Sci. 2012; 93:404–411.
Article
11. Valenti MT, Mori A, Malerba G, Dalle Carbonare L. Mesenchymal stem cells: A new diagnostic tool? World J Stem Cells. 2015; 7:789–792.
Article
12. Waterman RS, Tomchuck SL, Henkle SL, Betancourt AM. A new mesenchymal stem cell (MSC) paradigm: polarization into a pro-inflammatory MSC1 or an immunosuppressive MSC2 phenotype. PLoS One. 2010; 5:e10088.
Article
13. Wong RS. Mesenchymal stem cells: angels or demons? J Biomed Biotechnol. 2011; 2011:459510.
Article
14. Efimenko A, Starostina E, Kalinina N, Stolzing A. Angiogenic properties of aged adipose derived mesenchymal stem cells after hypoxic conditioning. J Transl Med. 2011; 9:10.
Article
15. Fernandez-Garcia M, Yanez RM, Sanchez-Dominguez R, et al. Mesenchymal stromal cells enhance the engraftment of hematopoietic stem cells in an autologous mouse transplantation model. Stem Cell Res Ther. 2015; 6:165.
Article
16. Hall MN, Rosenkrantz WS, Hong JH, Griffin CE, Mendelsohn CM. Evaluation of the potential use of adipose-derived mesenchymal stromal cells in the treatment of canine atopic dermatitis: a pilot study. Vet Ther. 2010; 11:E1–E14.
17. Zimmerlin L, Donnenberg AD, Rubin JP, Basse P, Landreneau RJ, Donnenberg VS. Regenerative therapy and cancer: in vitro and in vivo studies of the interaction between adipose-derived stem cells and breast cancer cells from clinical isolates. Tissue Eng Part A. 2011; 17:93–106.
Article
18. Tian LL, Yue W, Zhu F, Li S, Li W. Human mesenchymal stem cells play a dual role on tumor cell growth in vitro and in vivo. J Cell Physiol. 2011; 226:1860–1867.
Article
19. Suzuki K, Sun R, Origuchi M, et al. Mesenchymal stromal cells promote tumor growth through the enhancement of neovascularization. Mol Med. 2011; 17:579–587.
Article
20. Lin G, Yang R, Banie L, et al. Effects of transplantation of adipose tissue-derived stem cells on prostate tumor. Prostate. 2010; 70:1066–1073.
Article
21. Wu YL, Li HY, Zhao XP, et al. Mesenchymal stem cell-derived CCN2 promotes the proliferation, migration and invasion of human tongue squamous cell carcinoma cells. Cancer Sci. 2017; 108:897–909.
Article
22. Yulyana Y, Ho IA, Sia KC, et al. Paracrine factors of human fetal MSCs inhibit liver cancer growth through reduced activation of IGF-1R/PI3K/Akt signaling. Mol Ther. 2015; 23:746–756.
Article
23. Klopp AH, Gupta A, Spaeth E, Andreeff M, Marini F 3rd. Concise review: Dissecting a discrepancy in the literature: do mesenchymal stem cells support or suppress tumor growth? Stem Cells. 2011; 29:11–19.
Article
24. Zhu N, Wang H, Wei J, et al. NR2F2 regulates bone marrow-derived mesenchymal stem cell-promoted proliferation of Reh cells. Mol Med Rep. 2016; 14:1351–1356.
Article
25. Lin HD, Fong CY, Biswas A, Choolani M, Bongso A. Human umbilical cord Wharton's jelly stem cell conditioned medium induces tumoricidal effects on lymphoma cells through hydrogen peroxide mediation. J Cell Biochem. 2016; 117:2045–2055.
Article
26. Song N, Gao L, Qiu H, et al. Mouse bone marrow-derived mesenchymal stem cells inhibit leukemia/lymphoma cell proliferation in vitro and in a mouse model of allogeneic bone marrow transplant. Int J Mol Med. 2015; 36:139–149.
Article
27. Duchi S, Sotgiu G, Lucarelli E, et al. Mesenchymal stem cells as delivery vehicle of porphyrin loaded nanoparticles: effective photoinduced in vitro killing of osteosarcoma. J Control Release. 2013; 168:225–237.
Article
28. Lee JK, Park SR, Jung BK, et al. Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS One. 2013; 8:e84256.
Article
29. Sarmadi VH, Tong CK, Vidyadaran S, Abdullah M, Seow HF, Ramasamy R. Mesenchymal stem cells inhibit proliferation of lymphoid origin haematopoietic tumour cells by inducing cell cycle arrest. Med J Malaysia. 2010; 65:209–214.
30. Fathi E, Farahzadi R, Valipour B, Sanaat Z. Cytokines secreted from bone marrow derived mesenchymal stem cells promote apoptosis and change cell cycle distribution of K562 cell line as clinical agent in cell transplantation. PLoS One. 2019; 14:e0215678.
Article
31. Russell KA, Chow NH, Dukoff D, et al. Characterization and immunomodulatory effects of canine adipose tissue- and bone marrow-derived mesenchymal stromal cells. PLoS One. 2016; 11:e0167442.
Article
32. Neuhuber B, Swanger SA, Howard L, Mackay A, Fischer I. Effects of plating density and culture time on bone marrow stromal cell characteristics. Exp Hematol. 2008; 36:1176–1185.
Article
33. Liang R, Huang GS, Wang Z, et al. Effects of human bone marrow stromal cell line (HFCL) on the proliferation, differentiation and apoptosis of acute myeloid leukemia cell lines U937, HL-60 and HL-60/VCR. Int J Hematol. 2008; 87:152–166.
Article
34. Tian K, Yang S, Ren Q, et al. p38 MAPK contributes to the growth inhibition of leukemic tumor cells mediated by human umbilical cord mesenchymal stem cells. Cell Physiol Biochem. 2010; 26:799–808.
Article
35. Li Q, Pang Y, Liu T, et al. Effects of human umbilical cord-derived mesenchymal stem cells on hematologic malignancies. Oncol Lett. 2018; 15:6982–6990.
Article
36. Peng L, Jiang D. Resveratrol eliminates cancer stem cells of osteosarcoma by STAT3 pathway inhibition. PLoS One. 2018; 13:e0205918.
Article
37. Chulpanova DS, Kitaeva KV, Tazetdinova LG, James V, Rizvanov AA, Solovyeva VV. Application of mesenchymal stem cells for therapeutic agent delivery in anti-tumor treatment. Front Pharmacol. 2018; 9:259.
Article
38. Garikipati VNS, Singh SP, Mohanram Y, Gupta AK, Kapoor D, Nityanand S. Isolation and characterization of mesenchymal stem cells from human fetus heart. PLoS One. 2018; 13:e0192244.
Article
39. Lalu MM, McIntyre L, Pugliese C, et al. Safety of cell therapy with mesenchymal stromal cells (SafeCell): a systematic review and meta-analysis of clinical trials. PLoS One. 2012; 7:e47559.
Article
40. Gao Z, Zhang L, Hu J, Sun Y. Mesenchymal stem cells: a potential targeted-delivery vehicle for anti-cancer drug, loaded nanoparticles. Nanomedicine. 2013; 9:174–184.
Article
41. Ramdasi S, Sarang S, Viswanathan C. Potential of mesenchymal stem cell based application in cancer. Int J Hematol Oncol Stem Cell Res. 2015; 9:95–103.
42. Dembinski JL, Wilson SM, Spaeth EL, et al. Tumor stroma engraftment of gene-modified mesenchymal stem cells as anti-tumor therapy against ovarian cancer. Cytotherapy. 2013; 15:20–32.
Article
43. Zhao WH, Cheng JX, Shi PF, Huang JY. Human umbilical cord mesenchymal stem cells with adenovirus-mediated interleukin 12 gene transduction inhibits the growth of ovarian carcinoma cells both in vitro and in vivo. Nan Fang Yi Ke Da Xue Xue Bao. 2011; 31:903–907.
44. Zhang X, Zhang L, Xu W, et al. Experimental therapy for lung cancer: umbilical cord-derived mesenchymal stem cell-mediated interleukin-24 delivery. Curr Cancer Drug Targets. 2013; 13:92–102.
Article
45. Yang X, Du J, Xu X, Xu C, Song W. IFN-γ-secreting-mesenchymal stem cells exert an antitumor effect in vivo via the TRAIL pathway. J Immunol Res. 2014; 2014:318098.
46. Laso-Garcia F, Ramos-Cejudo J, Carrillo-Salinas FJ, et al. Therapeutic potential of extracellular vesicles derived from human mesenchymal stem cells in a model of progressive multiple sclerosis. PLoS One. 2018; 13:e0202590.
Article
47. Rajendran RL, Gangadaran P, Bak SS, et al. Extracellular vesicles derived from MSCs activates dermal papilla cell in vitro and promotes hair follicle conversion from telogen to anagen in mice. Sci Rep. 2017; 7:15560.
Article
48. Hendijani F, Javanmard SH, Sadeghi-aliabadi H. Human Wharton's jelly mesenchymal stem cell secretome display antiproliferative effect on leukemia cell line and produce additive cytotoxic effect in combination with doxorubicin. Tissue Cell. 2015; 47:229–234.
Article
49. Otsu K, Das S, Houser SD, Quadri SK, Bhattacharya S, Bhattacharya J. Concentration-dependent inhibition of angiogenesis by mesenchymal stem cells. Blood. 2009; 113:4197–4205.
Article
50. Menge T, Gerber M, Wataha K, et al. Human mesenchymal stem cells inhibit endothelial proliferation and angiogenesis via cell-cell contact through modulation of the VE-Cadherin/β-catenin signaling pathway. Stem Cells Dev. 2013; 22:148–157.
Article
51. Kastan MB, Bartek J. Cell-cycle checkpoints and cancer. Nature. 2004; 432:316–323.
Article
52. Fonseka M, Ramasamy R, Tan BC, Seow HF. Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSC) inhibit the proliferation of K562 (human erythromyeloblastoid leukaemic cell line). Cell Biol Int. 2012; 36:793–801.
Article
53. Zhang HM, Zhang LS. Influence of human bone marrow mesenchymal stem cells on proliferation of chronic myeloid leukemia cells. Ai Zheng. 2009; 28:29–32.
54. Mattar P, Bieback K. Comparing the immunomodulatory properties of bone marrow, adipose tissue, and birth-associated tissue mesenchymal stromal cells. Front Immunol. 2015; 6:560.
Article
55. Wang Z, Tang X, Xu W, et al. The different immunoregulatory functions on dendritic cells between mesenchymal stem cells derived from bone marrow of patients with low-risk or high-risk myelodysplastic syndromes. PLoS One. 2013; 8:e57470.
Article
56. Iancu-Rubin C, Mosoyan G, Wang J, Kraus T, Sung V, Hoffman R. Stromal cell-mediated inhibition of erythropoiesis can be attenuated by Sotatercept (ACE-011), an activin receptor type II ligand trap. Exp Hematol. 2013; 41:155–166.e17.
Article
57. del Toro R, Mendez-Ferrer S. Autonomic regulation of hematopoiesis and cancer. Haematologica. 2013; 98:1663–1666.
Article
58. Arranz L, Sanchez-Aguilera A, Martin-Perez D, et al. Neuropathy of haematopoietic stem cell niche is essential for myeloproliferative neoplasms. Nature. 2014; 512:78–81.
Article
59. Lopez-Villar O, Garcia JL, Sanchez-Guijo FM, et al. Both expanded and uncultured mesenchymal stem cells from MDS patients are genomically abnormal, showing a specific genetic profile for the 5q- syndrome. Leukemia. 2009; 23:664–672.
Article
60. Abe-Suzuki S, Kurata M, Abe S, et al. CXCL12+ stromal cells as bone marrow niche for CD34+ hematopoietic cells and their association with disease progression in myelodysplastic syndromes. Lab Invest. 2014; 94:1212–1223.
Article
61. Pezeshkian B, Donnelly C, Tamburo K, Geddes T, Madlambayan GJ. Leukemia mediated endothelial cell activation modulates leukemia cell susceptibility to chemotherapy through a positive feedback loop mechanism. PLoS One. 2013; 8:e60823.
Article
62. Huang JC, Basu SK, Zhao X, et al. Mesenchymal stromal cells derived from acute myeloid leukemia bone marrow exhibit aberrant cytogenetics and cytokine elaboration. Blood Cancer J. 2015; 5:e302.
Article
63. Brenner AK, Nepstad I, Bruserud O. Mesenchymal stem cells support survival and proliferation of primary human acute myeloid leukemia cells through heterogeneous molecular mechanisms. Front Immunol. 2017; 8:106.
Article
64. Lopes MR, Pereira JK, de Melo Campos P, et al. De novo AML exhibits greater microenvironment dysregulation compared to AML with myelodysplasia-related changes. Sci Rep. 2017; 7:40707.
Article
65. Tian T, Yu S, Liu L, et al. The profile of T helper subsets in bone marrow microenvironment is distinct for different stages of acute myeloid leukemia patients and chemotherapy partly ameliorates these variations. PLoS One. 2015; 10:e0131761.
Article
66. Aliperta R, Cartellieri M, Feldmann A, et al. Bispecific antibody releasing-mesenchymal stromal cell machinery for retargeting T cells towards acute myeloid leukemia blasts. Blood Cancer J. 2015; 5:e348.
Article
67. Fracchiolla NS, Fattizzo B, Cortelezzi A. Mesenchymal stem cells in myeloid malignancies: a focus on immune escaping and therapeutic implications. Stem Cells Int. 2017; 2017:6720594.
Article
68. Chen P, Huang H, Wu J, et al. Bone marrow stromal cells protect acute myeloid leukemia cells from anti-CD44 therapy partly through regulating PI3K/Akt-p27(Kip1) axis. Mol Carcinog. 2015; 54:1678–1685.
Article
69. Bao H, Wu D. Current status of leukemia cytotherapy - exploitation with immune cells. Curr Stem Cell Res Ther. 2017; 12:188–196.
Article
70. Manabe A, Coustan-Smith E, Behm FG, Raimondi SC, Campana D. Bone marrow-derived stromal cells prevent apoptotic cell death in B-lineage acute lymphoblastic leukemia. Blood. 1992; 79:2370–2377.
Article
71. Garrido SM, Appelbaum FR, Willman CL, Banker DE. Acute myeloid leukemia cells are protected from spontaneous and drug-induced apoptosis by direct contact with a human bone marrow stromal cell line (HS-5). Exp Hematol. 2001; 29:448–457.
Article
72. Ramasamy R, Lam EW, Soeiro I, Tisato V, Bonnet D, Dazzi F. Mesenchymal stem cells inhibit proliferation and apoptosis of tumor cells: impact on in vivo tumor growth. Leukemia. 2007; 21:304–310.
Article
73. Zhu Y, Sun Z, Han Q, et al. Human mesenchymal stem cells inhibit cancer cell proliferation by secreting DKK-1. Leukemia. 2009; 23:925–933.
Article
74. Wei Z, Chen N, Guo H, et al. Bone marrow mesenchymal stem cells from leukemia patients inhibit growth and apoptosis in serum-deprived K562 cells. J Exp Clin Cancer Res. 2009; 28:141.
Article
75. Secchiero P, Zorzet S, Tripodo C, et al. Human bone marrow mesenchymal stem cells display anti-cancer activity in SCID mice bearing disseminated non-Hodgkin's lymphoma xenografts. PLoS One. 2010; 5:e11140.
Article
76. Han Y, Wang Y, Xu Z, et al. Effect of bone marrow mesenchymal stem cells from blastic phase chronic myelogenous leukemia on the growth and apoptosis of leukemia cells. Oncol Rep. 2013; 30:1007–1013.
Article
77. Yuan Y, Chen D, Chen X, Shao H, Huang S. Human umbilical cord-derived mesenchymal stem cells inhibit proliferation but maintain survival of Jurkat leukemia cells in vitro by activating Notch signaling. Nan Fang Yi Ke Da Xue Xue Bao. 2014; 34:441–447.
78. Naderi EH, Skah S, Ugland H, et al. Bone marrow stroma-derived PGE2 protects BCP-ALL cells from DNA damage-induced p53 accumulation and cell death. Mol Cancer. 2015; 14:14.
Article
79. Lee MW, Park YJ, Kim DS, et al. Human adipose tissue stem cells promote the growth of acute lymphoblastic leukemia cells in NOD/SCID mice. Stem Cell Rev Rep. 2018; 14:451–460.
Article
Full Text Links
  • BR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr