Cell-Free Treatments: A New Generation of Targeted Therapies for Treatment of Ischemic Heart Disease

Document Type : Review Article

Authors

1 School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran

2 Faculty of Medicine, Graduate School of Health Science, Near East University, Nicosia, Northern Cyprus, Cyprus

3 Private Baskent Hospital, Nicosia, Northern Cyprus, Cyprus

4 Paediatric Ward, Department of Allergy and Immunology, Near East University Affiliated Hospital, Nicosia, Northern Cyprus, Cypru

5 Network of Immunity in Infection, Malignancy and Autoimmunity, Universal Scientific Education and Research Network, Tehran, Iran

6 Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran

7 Cancer Gene Therapy Research Centre, Zanjan University of Medical Sciences, Zanjan, Iran

8 Immunotherapy Research and Technology Group, Zanjan University of Medical Sciences, Zanjan, Iran

Abstract

Although recent progress in medicine has substantially reduced cardiovascular diseases (CVDs)-related
mortalities, current therapeutics have failed miserably to be beneficial for all patients with CVDs. A wide array of
evidence suggests that newly-introduced cell-free treatments (CFT) have more reliable results in the improvement
of cardiac function. The main regeneration activity of CFT protocols is based on bypassing cells and using paracrine
factors. In this article, we aim to compare various stem cell secretomes, a part of a CFT strategy, to generalize their
effective clinical outcomes for patients with CVDs. Data for this review article were collected from 70 published articles
(original, review, randomized clinical trials (RCTs), and case reports/series studies done on human and animals)
obtained from Cochrane, Science Direct, PubMed, Scopus, Elsevier, and Google Scholar) from 2015 to April 2020
using six keywords. Full-text/full-length articles, abstract, section of book, chapter, and conference papers in English
language were included. Studies with irrelevant/insufficient/data, or undefined practical methods were excluded. CFT
approaches involved in growth factors (GFs); gene-based therapies; microRNAs (miRNAs); extracellular vesicles (EVs)
[exosomes (EXs) and microvesicles (MVs)]; and conditioned media (CM). EXs and CM have shown more remarkable
results than stem cell therapy (SCT). GF-based therapies have useful results as well as side effects like pathologic
angiogenesis side effect. Cell source, cell′s aging and CM affect secretomes. Genetic manipulation of stem cells
can change the secretome’s components. Growing progression to end stage heart failure (HF), propounds CFT as
an advantageous method with practical and clinical values for replacement of injured myocardium, and induction of
neovascularization. To elucidate the secrets behind amplifying the expansion rate of cells, increasing life-expectancy,
and improving quality of life (QOL) for patients with ischemic heart diseases (IHDs), collaboration among cell biologist,
basic medical scientists, and cardiologists is highly recommended.

Keywords


1. Virani SS, Alonso A, Benjamin EJ, Bittencourt MS, Callaway CW, Carson AP, et al. Heart disease and stroke statistics-2020 update: a report from the american heart association. Circulation. 2020: 141(9): e139-e596.
2. Duan B. Concise review: harnessing iPSC-derived cells for ischemic heart disease treatment. J Transl Int Med. 2020; 8(1): 20-25.
3. Cambria E, Steiger J, Günter J, Bopp A, Wolint P, Hoerstrup SP, et al. Cardiac regenerative medicine: the potential of a new generation of stem cells. Transfus Med Hemother. 2016; 43(4): 275-281.
4. Esmaeilzadeh A, Daneshi N, Erfanmanesh M. Evaluation of methods of cultivation, processing and improving of stem cell differentiation into cardiomyocytes. Journal of Laboratory and Diagnosis. 2017; 8(34): 39-47.
5. Sasse S, Skorska A, Lux CA, Steinhoff G, David R, Gaebel R. Angiogenic potential of bone marrow derived CD133+ and CD271+ intramyocardial stem cell trans- plantation post MI. Cells. 2020; 9(1): 78.
6. Turner D, Rieger AC, Balkan W, Hare JM. Clinical-based cell therapies for heart disease-current and future state. Rambam Maimonides Med J. 2020; 11(2): e0015.
7. Sanganalmath SK, Bolli R. Cell therapy for heart failure: a comprehensive overview of experimental and clinical studies, current challenges, and future directions. Circ Res. 2013; 113(6): 810-34.
8. Singla DK. Stem cells and exosomes in cardiac repair. Curr Opin Pharmacol. 2016; 27: 19-23.
9. Wernly B, Mirna M, Rezar R, Prodinger C, Jung C, Podesser BK, et al. Regenerative cardiovascular therapies: stem cells and beyond. Int J Mol Sci. 2019; 20(6): 1420.
10. Ratajczak MZ, Kucia M, Jadczyk T, Greco NJ, Wojakowski W, Tendera M, et al. Pivotal role of paracrine effects in stem cell therapies in regenerative medicine: can we translate stem cell-secreted paracrine factors and microvesicles into better therapeutic strategies? Leukemia. 2012; 26(6): 1166-1173.
11. Prathipati P, Nandi SS, Mishra PK. Stem cell-derived exosomes, autophagy, extracellular matrix turnover, and mirnas in cardiac regeneration during stem cell therapy. Stem Cell Rev Rep. 2017; 13(1): 79-91.
12. Cambria E, Pasqualini FS, Wolint P, Günter J, Steiger J, Bopp A, et al. Translational cardiac stem cell therapy: advancing from firstgeneration to next-generation cell types. NPJ Regen Med. 2017; 2: 17.
13. Tsao CR, Liao MF, Wang MH, Cheng CM, Chen CH. Mesenchymal stem cell derived exosomes: a new hope for the treatment of cardiovascular disease? Acta Cardiol Sin. 2014; 30(5): 395-400.
14. Harrell CR, Fellabaum C, Jovicic N, Djonov V, Arsenijevic N, Volarevic V. Molecular mechanisms responsible for therapeutic potential of mesenchymal stem cell-derived secretome. Cells. 2019; 8(5): 467.
15. Khanabdali R, Rosdah AA, Dusting GJ, Lim SY. Harnessing the secretome of cardiac stem cells as therapy for ischemic heart disease. Biochem Pharmacol. 2016; 113: 1-11.
16. Mirotsou M, Jayawardena TM, Schmeckpeper J, Gnecchi M, Dzau VJ. Paracrine mechanisms of stem cell reparative and regenerative actions in the heart. J Mol Cell Cardiol. 2011; 50(2): 280-289.
17. Gallina C, Turinetto V, Giachino C. A new paradigm in cardiac regeneration: the mesenchymal stem cell secretome. Stem Cells Int. 2015; 2015: 765846.
18. Ranganath SH, Levy O, Inamdar MS, Karp JM. Harnessing the mesenchymal stem cell secretome for the treatment of cardiovascular disease. Cell Stem Cell. 2012; 10(3): 244-258.
19. Huang P, Tian X, Li Q, Yang Y. New strategies for improving stem cell therapy in ischemic heart disease. Heart Fail Rev. 2016; 21(6): 737-752.
20. Carotenuto F, Teodori L, Maccari AM, Delbono L, Orlando G, Di Nardo P. Turning regenerative technologies into treatment to repair myocardial injuries. J Cell Mol Med. 2020; 24(5): 2704-2716.
21. Toh WS, Lai RC, Zhang B, Lim SK. MSC exosome works through a protein-based mechanism of action. Biochem Soc Trans. 2018; 46(4): 843-853.
22. Liu M, Wang Y, Zhu Q, Zhao J, Wang Y, Shang M, et al. Protective effects of circulating microvesicles derived from ischemic preconditioning on myocardial ischemia/reperfusion injury in rats by inhibiting endoplasmic reticulum stress. Apoptosis. 2018; 23(7-8): 436-448.
23. Sharma S, Mishra R, Bigham GE, Wehman B, Khan MM, Xu H, et al. A deep proteome analysis identifies the complete secretome as the functional unit of human cardiac progenitor cells. Circ Res. 2017; 120(5): 816-834.
24. Yu B, Zhang X, Li X. Exosomes derived from mesenchymal stem cells. Int J Mol Sci. 2014; 15(3): 4142-4157.
25. Adamiak M, Cheng G, Bobis-Wozowicz S, Zhao L, Kedracka-Krok S, Samanta A, et al. Induced pluripotent stem cell (iPSC)-derived extracellular vesicles are safer and more effective for cardiac repair than iPSCs. Circ Res. 2018; 122(2): 296-309.
26. Chistiakov DA, Orekhov AN, Bobryshev YV. Cardiac extracellular vesicles in normal and infarcted heart. Int J Mol Sci. 2016; 17(1): 63.
27. Kishore R, Khan M. More than tiny sacks: stem cell exosomes as cell-free modality for cardiac repair. Circ Res. 2016; 118(2): 330- 343.
28. Gallet R, Dawkins J, Valle J, Simsolo E, de Couto G, Middleton R, et al. Exosomes secreted by cardiosphere-derived cells reduce scarring, attenuate adverse remodelling, and improve function in acute and chronic porcine myocardial infarction. Eur Heart J. 2017; 38(3): 201-211.
29. Ma J, Zhao Y, Sun L, Sun X, Zhao X, Sun X, et al. Exosomes derived from Akt-modified human umbilical cord mesenchymal stem cells improve cardiac regeneration and promote angiogenesis via activating platelet-derived growth factor D. Stem Cells Transl Med. 2017; 6(1): 51-59.
30. Khan M, Nickoloff E, Abramova T, Johnson J, Verma SK, Krishnamurthy P, et al. Embryonic stem cell-derived exosomes promote endogenous repair mechanisms and enhance cardiac function following myocardial infarction. Circ Res. 2015: 117(1): 52-64.
31. Mentkowski KI, Lang JK. Exosomes engineered to express a cardiomyocyte binding peptide demonstrate improved cardiac retention in vivo. Sci Rep. 2019; 9(1): 10041.
32. Yamashita T, Takahashi Y, Takakura Y. Possibility of exosomebased therapeutics and challenges in production of exosomes eligible for therapeutic application. Biol Pharm Bull. 2018; 41(6): 835-842.
33. Tian T, Zhang HX, He CP, Fan S, Zhu YL, Qi C, et al. Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials. 2018; 150: 137-149.
34. Migault M, Donnou-Fournet E, Galibert MD, Gilot D. Definition and identification of small RNA sponges: Focus on miRNA sequestration. Methods. 2017; 117: 35-47.
35. Wu G, Huang ZP, Wang DZ. MicroRNAs in cardiac regeneration and cardiovascular disease. Sci China Life Sci. 2013; 56(10): 907-913.
36. Shrestha S, Ren L, Vaidya R. miRNAs as biomarkers for diagnosis and assessment of prognosis of coronary artery disease. Open J Intern Med. 2018; 08(01): 54-63.
37. Samanta S, Balasubramanian S, Rajasingh S, Patel U, Dhanasekaran A, Dawn B, et al. MicroRNA: a new therapeutic strategy for cardiovascular diseases. Trends Cardiovasc Med. 2016; 26(5): 407-419.
38. Zhu K, Liu D, Lai H, Li J, Wang C. Developing miRNA therapeutics for cardiac repair in ischemic heart disease. J Thorac Dis. 2016; 8(9): E918-E927.
39. Wang X, Shang Y, Dai S, Wu W, Yi F, Cheng L. MicroRNA-16-5p aggravates myocardial infarction injury by targeting the expression of insulin receptor substrates 1 and mediating myocardial apoptosis and angiogenesis. Curr Neurovasc Res. 2020; 17(1): 11-7.
40. Zhao Z, Du S, Shen S, Wang L. microRNA-132 inhibits cardiomyocyte apoptosis and myocardial remodeling in myocardial infarction by targeting IL-1β. J Cell Physiol. 2020; 235(3): 2710-2721.
41. Bai Y, Bai L, Zhou J, Chen H, Zhang L. Sequential delivery of VEGF, FGF-2 and PDGF from the polymeric system enhance HUVECs angiogenesis in vitro and CAM angiogenesis. Cell Immunol. 2018; 323: 19-32.
42. Rebouças JS, Santos-Magalhães NS, Formiga FR. Cardiac regeneration using growth factors: advances and challenges. Arq Bras Cardiol. 2016; 107(3): 271-275.
43. Thavapalachandran S, Grieve SM, Hume RD, Le TYL, Raguram K, Hudson JE, et al. Platelet-derived growth factor-AB improves scar mechanics and vascularity after myocardial infarction. Sci Transl Med. 2020; 12(524): eaay2140.
44. Hajjar RJ, Hulot JS. Myocardial delivery of stromal cell-derived factor 1 in patients with ischemic heart disease: safe and promising. Circ Res. 2013; 112(5): 746-747.
45. Timmers L, Lim SK, Hoefer IE, Arslan F, Lai RC, van Oorschot AA, et al. Human mesenchymal stem cell-conditioned medium improves cardiac function following myocardial infarction. Stem Cell Res. 2011; 6(3): 206-214.
46. Gunawardena TNA, Rahman MT, Abdullah BJJ, Abu Kasim NH. Conditioned media derived from mesenchymal stem cell cultures: the next generation for regenerative medicine. J Tissue Eng Regen Med. 2019; 13(4): 569-586.
47. Yamaguchi S, Shibata R, Yamamoto N, Nishikawa M, Hibi H, Tanigawa T, et al. Dental pulp-derived stem cell conditioned medium reduces cardiac injury following ischemia-reperfusion. Sci Rep. 2015; 5: 16295.
48. Ellison-Hughes GM, Madeddu P. Exploring pericyte and cardiac stem cell secretome unveils new tactics for drug discovery. Pharmacol Ther. 2017; 171: 1-12.
49. He J, Cai Y, Luo LM, Liu HB. Hypoxic adipose mesenchymal stem cells derived conditioned medium protects myocardial infarct in rat. Eur Rev Med Pharmacol Sci. 2015; 19(22): 4397-4406.
50. Ylä-Herttuala S, Bridges C, Katz MG, Korpisalo P. Angiogenic gene therapy in cardiovascular diseases: dream or vision? Eur Heart J. 2017; 38(18): 1365-1371.
51. Eibel B, Rodrigues CG, Giusti II, Nesralla IA, Prates PRL, Sant’Anna RT, et al. Terapia gênica para cardiopatia isquêmica:
revisão de ensaios clínicos. BJCVS. 2011; 26: 635-646.
52. Lavu M, Gundewar S, Lefer DJ. Gene therapy for ischemic heart disease. J Mol Cell Cardiol. 2011; 50(5): 742-750.
53. Esmaeilzadeh A, Mohammadzadeh A, Bahmaie N. New generation of promising immunotherapeutics approaches for psoriasis dilemma; IL-35 gene as a potentiated candidate. Inflammation and Cell Signaling. 2018; 5: e1635.
54. Lin M, Liu X, Zheng H, Huang X, Wu Y, Huang A, et al. IGF-1 enhances BMSC viability, migration, and anti-apoptosis in myocardial infarction via secreted frizzled-related protein 2 pathway. Stem Cell Res Ther. 2020; 11(1): 22.
55. Su G, Liu L, Yang L, Mu Y, Guan L. Homing of endogenous bone marrow mesenchymal stem cells to rat infarcted myocardium via ultrasound-mediated recombinant SDF-1α adenovirus in microbubbles. Oncotarget. 2018; 9(1): 477-487.
56. Johnson T, Zhao L, Manuel G, Taylor H, Liu D. Approaches to therapeutic angiogenesis for ischemic heart disease. J Mol Med (Berl). 2019; 97(2): 141-151.
57. Bauzá MDR, Giménez CS, Locatelli P, De Lorenzi A, Hnatiuk A, Capogrossi MC, et al. High-dose intramyocardial HMGB1 induces long-term cardioprotection in sheep with myocardial infarction. Drug Deliv Transl Res. 2019; 9(5): 935-944.
58. Hadjizadeh A, Ghasemkhah F, Ghasemzaie N. Polymeric scaffold based gene delivery strategies to improve angiogenesis in tissue engineering: a review. Polym Rev. 2017; 57(3): 505-556.
59. Yin ZQ, Xing WH. Progress in gene therapy for chronic heart failure. Heart Surg Forum. 2018; 21(2): E075-E083.
60. Laakkonen JP, Ylä-Herttuala S. Recent advancements in cardiovascular gene therapy and vascular biology. Hum Gene Ther. 2015; 26(8): 518-524.
61. Yang L, Zhu J, Zhang C, Wang J, Yue F, Jia X, et al. Stem cellderived extracellular vesicles for myocardial infarction: a metaanalysis of controlled animal studies. Aging (Albany NY). 2019; 11(4): 1129-1150.
62. Liu C, Wang J, Hu J, Fu B, Mao Z, Zhang H, et al. Extracellular vesicles for acute kidney injury in preclinical rodent models: a meta-analysis. Stem Cell Res Ther. 2020; 11(1): 11.
63. Willis GR, Kourembanas S, Mitsialis SA. Toward exosome-based therapeutics: isolation, heterogeneity, and fit-for-purpose potency. Front Cardiovasc Med. 2017; 4: 63.
64. Meng H, Chen B, Tao Z, Xu Z, Wang L, Weizhu J, et al. Safety and efficacy of adenovirus carrying hepatocyte growth factor gene by percutaneous endocardial injection for treating post-infarct heart failure: a phase iia clinical trial. Curr Gene Ther. 2018; 18(2): 125-130.
65. Gross L, Theiss HD, Grabmaier U, Adrion C, Mansmann U, Sohn HY, et al. Combined therapy with sitagliptin plus granulocyte-colony stimulating factor in patients with acute myocardial infarction- Long-term results of the SITAGRAMI trial. Int J Cardiol. 2016; 215: 441-445.
66. Kukuła K, Urbanowicz A, Kłopotowski M, Dąbrowski M, Pręgowski J, Kądziela J, et al. Long-term follow-up and safety assessment of angiogenic gene therapy trial VIF-CAD: Transcatheter intramyocardial administration of a bicistronic plasmid expressing VEGFA165/ bFGF cDNA for the treatment of refractory coronary artery disease. Am Heart J. 2019; 215: 78-82.
67. Anttila V, Saraste A, Knuuti J, Jaakkola P, Hedman M, Svedlund S, et al. Synthetic mRNA encoding VEGF-A in patients undergoing coronary artery bypass grafting: design of a phase 2a clinical trial. Mol Ther Methods Clin Dev. 2020; 18: 464-472.
68. Greenberg B, Butler J, Felker GM, Ponikowski P, Voors AA, Desai AS, et al. Calcium upregulation by percutaneous administration of gene therapy in patients with cardiac disease (CUPID 2): a randomised, multinational, double-blind, placebo-controlled, phase 2b trial. Lancet. 2016; 387(10024): 1178-1186.
69. Chung ES, Miller L, Patel AN, Anderson RD, Mendelsohn FO, Traverse J, et al. Changes in ventricular remodelling and clinical status during the year following a single administration of stromal cellderived factor-1 non-viral gene therapy in chronic ischaemic heart failure patients: the STOP-HF randomized phase II trial. Eur Heart J. 2015; 36(33): 2228-2238.
70. Penn MS, Mendelsohn FO, Schaer GL, Sherman W, Farr M, Pastore J, et al. An open-label dose escalation study to evaluate the safety of administration of nonviral stromal cell-derived factor-1 plasmid to treat symptomatic ischemic heart failure. Circ Res. 2013; 112(5): 816-825.