Generic placeholder image

Recent Patents on Drug Delivery & Formulation

Editor-in-Chief

ISSN (Print): 1872-2113
ISSN (Online): 2212-4039

Review Article

Scientific Papers and Patents on Substances with Unproven Effects. Part 2

Author(s): Sergei V. Jargin*

Volume 13, Issue 3, 2019

Page: [160 - 173] Pages: 14

DOI: 10.2174/1872211313666190819124752

Abstract

Several examples are discussed in this review, where substances without proven effects were proposed for practical use within the scope of evidence-based medicines. The following is discussed here: generalizations of the hormesis concept and its use in support of homeopathy; phytoestrogens and soy products potentially having feminizing effects; glycosaminoglycans for the treatment of osteoarthritis and possibilities of their replacement by diet modifications; flavonoids recommended for the treatment of chronic venous insufficiency and varicose veins; acetylcysteine as a mucolytic agent and its questionable efficiency especially by an oral intake; stem cells and cell therapies. In conclusion, placebo therapies can be beneficial and ethically justifiable but it is not a sufficient reason to publish biased information. Importantly, placebo must be devoid of adverse effects, otherwise, it is named pseudo-placebo. Therapeutic methods with unproven effects should be tested in high-quality research shielded from the funding bias. Some issues discussed in this review are not entirely clear, and the arguments provided here can initiate a constructive discussion.

Keywords: Placebo, hormesis, nutrition, phytoestrogens, soy, osteoarthritis, acetylcysteine.

Graphical Abstract
[1]
Jargin SV. Scientific papers and patents on substances with unproven effects. Recent Pat Drug Deliv Formul 2019; 13: 37-45.
[http://dx.doi.org/10.2174/1872211313666190307162041] [PMID: 30848224]
[2]
Mattson MP, Calabrese EJ. Hormesis A Revolution in Biology, Toxicology and Medicine. Springer New York 2010.
[3]
Jargin SV. Hormesis and homeopathy: The artificial twins. J Intercult Ethnopharmacol 2015; 4(1): 74-7.
[http://dx.doi.org/10.5455/jice.20140929114417] [PMID: 26401389]
[4]
Calabrese EJ, Calabrese V, Giordano J. The role of hormesis in the functional performance and protection of neural systems. Brain Circ 2017; 3(1): 1-13.
[PMID: 30276298]
[5]
Jargin SV. Hormetic use of stress in gerontological interventions requires a cautious approach. Biogerontology 2016; 17(2): 417-20.
[http://dx.doi.org/10.1007/s10522-015-9630-8] [PMID: 26712317]
[6]
Calabrese EJ, Iavicoli I, Calabrese V. Hormesis: Its impact on medicine and health. Hum Exp Toxicol 2013; 32(2): 120-52.
[http://dx.doi.org/10.1177/0960327112455069] [PMID: 23060412]
[7]
Calabrese EJ, Iavicoli I, Calabrese V. Hormesis: Why it is important to biogerontologists. Biogerontology 2012; 13(3): 215-35.
[http://dx.doi.org/10.1007/s10522-012-9374-7] [PMID: 22270337]
[8]
Calabrese EJ. Hormesis: Principles and applications. Homeopathy 2015; 104(2): 69-82.
[http://dx.doi.org/10.1016/j.homp.2015.02.007] [PMID: 25869971]
[9]
Weihrauch TR. Placebo treatment is effective differently in different diseases- but is it also harmless? A brief synopsis. Sci Eng Ethics 2004; 10(1): 151-5.
[http://dx.doi.org/10.1007/s11948-004-0072-y] [PMID: 14986781]
[10]
Moffett JR. Miasmas, germs, homeopathy and hormesis: Commentary on the relationship between homeopathy and hormesis. Hum Exp Toxicol 2010; 29(7): 539-43.
[http://dx.doi.org/10.1177/0960327110369855] [PMID: 20558603]
[11]
Nebera SA. Homeopathic preparation for the treatment of myopia. RU2203674C1 (2003)
[12]
Ostreikovskii IE. Treatment method of acute pneumonia. RU2063224C1 (1996).
[13]
Chernozubov IE. The medications and method of tuberculosis treatment. RU2119338C1 (1996).
[14]
Epshtein OI. The medications and treatment method of pathological syndrome. RU2192888C1 (2002).
[15]
Luchkin VA. Method of treatment of degenerative - Dystrophic diseases of locomotor system. RU2015147929A (2016)
[16]
Patisaul HB. Endocrine disruption by dietary phyto-oestrogens: Impact on dimorphic sexual systems and behaviours. Proc Nutr Soc 2017; 76(2): 130-44.
[http://dx.doi.org/10.1017/S0029665116000677] [PMID: 27389644]
[17]
Rietjens IMCM, Louisse J, Beekmann K. The potential health effects of dietary phytoestrogens. Br J Pharmacol 2017; 174(11): 1263-80.
[http://dx.doi.org/10.1111/bph.13622] [PMID: 27723080]
[18]
Gencel VB, Benjamin MM, Bahou SN, Khalil RA. Vascular effects of phytoestrogens and alternative menopausal hormone therapy in cardiovascular disease. Mini Rev Med Chem 2012; 12(2): 149-74.
[http://dx.doi.org/10.2174/138955712798995020] [PMID: 22070687]
[19]
Taku K, Melby MK, Kronenberg F, Kurzer MS, Messina M. Extracted or synthesized soybean isoflavones reduce menopausal hot flash frequency and severity: Systematic review and meta-analysis of randomized controlled trials. Menopause 2012; 19(7): 776-90.
[http://dx.doi.org/10.1097/gme.0b013e3182410159] [PMID: 22433977]
[20]
Baber R. Phytoestrogens and post reproductive health. Maturitas 2010; 66(4): 344-9.
[http://dx.doi.org/10.1016/j.maturitas.2010.03.023] [PMID: 20409653]
[21]
Gold EB, Leung K, Crawford SL, Huang MH, Waetjen LE, Greendale GA. Phytoestrogen and fiber intakes in relation to incident vasomotor symptoms: Results from the study of women’s health across the nation. Menopause 2013; 20(3): 305-14.
[PMID: 23435028]
[22]
Lethaby AE, Brown J, Marjoribanks J, Kronenberg F, Roberts H, Eden J. Phytoestrogens for vasomotor menopausal symptoms. Cochrane Database Syst Rev 2007; 4 CD001395 PMID: 17943751.
[23]
Krebs EE, Ensrud KE, MacDonald R, Wilt TJ. Phytoestrogens for treatment of menopausal symptoms: A systematic review. Obstet Gynecol 2004; 104(4): 824-36.
[http://dx.doi.org/10.1097/01.AOG.0000140688.71638.d3] [PMID: 15458907]
[24]
Cheema D, Coomarasamy A, El-Toukhy T. Non-hormonal therapy of post-menopausal vasomotor symptoms: A structured evidence-based review. Arch Gynecol Obstet 2007; 276(5): 463-9.
[http://dx.doi.org/10.1007/s00404-007-0390-9] [PMID: 17593379]
[25]
Al-Azzawi F, Wahab M. Effectiveness of phytoestrogens in climacteric medicine. Ann N Y Acad Sci 2010; 1205: 262-7.
[http://dx.doi.org/10.1111/j.1749-6632.2010.05678.x] [PMID: 20840282]
[26]
Villaseca P. Non-estrogen conventional and phytochemical treatments for vasomotor symptoms: What needs to be known for practice. Climacteric 2012; 15(2): 115-24.
[http://dx.doi.org/10.3109/13697137.2011.624214] [PMID: 22148909]
[27]
Roberts H, Lethaby A. Phytoestrogens for menopausal vasomotor symptoms: A cochrane review summary. Maturitas 2014; 78(2): 79-81.
[http://dx.doi.org/10.1016/j.maturitas.2014.04.004] [PMID: 24768128]
[28]
Sirtori CR, Arnoldi A, Johnson SK. Phytoestrogens: End of a tale? Ann Med 2005; 37(6): 423-38.
[http://dx.doi.org/10.1080/07853890510044586] [PMID: 16203615]
[29]
Davis SR. Phytoestrogen therapy for menopausal symptoms? BMJ 2001; 323(7309): 354-5.
[http://dx.doi.org/10.1136/bmj.323.7309.354] [PMID: 11509412]
[30]
Coxam V. Phyto-oestrogens and bone health. Proc Nutr Soc 2008; 67(2): 184-95.
[http://dx.doi.org/10.1017/S0029665108007027] [PMID: 18412992]
[31]
Ye CF, Pan YM, Zhou H. Regulation of vitamin D receptor and Genistein on bone metabolism in mouse osteoblasts and the molecular mechanism of osteoporosis. J Biol Regul Homeost Agents 2018; 32(3): 497-505.
[PMID: 29921374]
[32]
Lagari VS, Levis S. Phytoestrogens in the prevention of postmenopausal bone loss. J Clin Densitom 2013; 16(4): 445-9.
[http://dx.doi.org/10.1016/j.jocd.2013.08.011] [PMID: 24090647]
[33]
Arcoraci V, Atteritano M, Squadrito F, et al. Antiosteoporotic activity of genistein aglycone in postmenopausal women: Evidence from a post-hoc analysis of a multicenter randomized controlled trial. Nutrients 2017; 9(2): 1-7.
[http://dx.doi.org/10.3390/nu9020179] [PMID: 28241420]
[34]
Tit DM, Bungau S, Iovan C, et al. Effects of the hormone replacement therapy and of soy isoflavones on bone resorption in postmenopause. J Clin Med 2018; 7(10): 1-13.
[http://dx.doi.org/10.3390/jcm7100297] [PMID: 30248918]
[35]
This P, de Cremoux P, Leclercq G, Jacquot Y. A critical view of the effects of phytoestrogens on hot flashes and breast cancer risk. Maturitas 2011; 70(3): 222-6.
[http://dx.doi.org/10.1016/j.maturitas.2011.07.001] [PMID: 21813250]
[36]
Haimov-Kochman R, Brzezinski A, Hochner-Celnikier D. Herbal remedies for menopausal symptoms: Are we cautious enough? Eur J Contracept Reprod Health Care 2008; 13(2): 133-7.
[http://dx.doi.org/10.1080/13625180801920131] [PMID: 18465474]
[37]
Wilson S, Blaschek K, de Mejia E. Allergenic proteins in soybean: Processing and reduction of P34 allergenicity. Nutr Rev 2005; 63(2): 47-58.
[http://dx.doi.org/10.1111/j.1753-4887.2005.tb00121.x] [PMID: 15762088]
[38]
de Villiers TJ, Bagratee JS, Dalmeyer JP, Davey MR, Davis CP, Guidozzi F. South African Menopause Society Council revised consensus position statement on menopausal hormone therapy. S Afr Med J 2007; 97: 354-7.
[39]
Leclercq G, de Cremoux P, This P, Jacquot Y. Lack of sufficient information on the specificity and selectivity of commercial phytoestrogens preparations for therapeutic purposes. Maturitas 2011; 68(1): 56-64.
[http://dx.doi.org/10.1016/j.maturitas.2010.10.003] [PMID: 21074339]
[40]
Usui T. Pharmaceutical prospects of phytoestrogens. Endocr J 2006; 53(1): 7-20.
[http://dx.doi.org/10.1507/endocrj.53.7] [PMID: 16543667]
[41]
Setchell KD, Clerici C. Equol: History, chemistry, and formation. J Nutr 2010; 40(7): 1355S-62S.
[http://dx.doi.org/10.3945/jn.109.119776] [PMID: 20519412]
[42]
Rizzo G, Baroni L. Soy, soy foods and their role in vegetarian diets. Nutrients 2018; 10(1): 1-51.
[http://dx.doi.org/10.3390/nu10010043] [PMID: 29304010]
[43]
van Duursen MBM. Modulation of estrogen synthesis and metabolism by phytoestrogens in vitro and the implications for women’s health. Toxicol Res (Camb) 2017; 6(6): 772-94.
[http://dx.doi.org/10.1039/C7TX00184C] [PMID: 30090542]
[44]
Czuczwar P, Paszkowski T, Lisiecki M, Woźniak S, Stępniak A. The safety and tolerance of phytotherapies in menopausal medicine - A review of the literature. Przegl Menopauz 2017; 16(1): 8-11.
[http://dx.doi.org/10.5114/pm.2017.67365] [PMID: 28546801]
[45]
Adgent MA, Daniels JL, Edwards LJ, Siega-Riz AM, Rogan WJ. Early-life soy exposure and gender-role play behavior in children. Environ Health Perspect 2011; 119(12): 1811-6.
[http://dx.doi.org/10.1289/ehp.1103579] [PMID: 21813368]
[46]
Martinez J, Lewi JE. An unusual case of gynecomastia associated with soy product consumption. Endocr Pract 2008; 14(4): 415-8.
[http://dx.doi.org/10.4158/EP.14.4.415] [PMID: 18558591]
[47]
Jacobsen BK, Jaceldo-Siegl K, Knutsen SF, Fan J, Oda K, Fraser GE. Soy isoflavone intake and the likelihood of ever becoming a mother: The adventist health study-2. Int J Womens Health 2014; 6: 377-84.
[http://dx.doi.org/10.2147/IJWH.S57137] [PMID: 24741329]
[48]
Adgent M, Daniels JL, Rogan WJ, et al. Early-life soy exposure and age at menarche. Paediatr Perinat Epidemiol 2012; 26(2): 163-75.
[http://dx.doi.org/10.1111/j.1365-3016.2011.01244.x] [PMID: 22324503]
[49]
Jefferson W, Williams CJ. Circulating levels of genistein in the neonate, apart from dose and route, predict future adverse female reproductive outcomes. Reprod Toxicol 2011; 31(3): 272-9.
[http://dx.doi.org/10.1016/j.reprotox.2010.10.001] [PMID: 20955782]
[50]
Testa I, Salvatori C, Di Cara G, et al. Soy-based infant formula: Are phyto-oestrogens still in doubt? Front Nutr 2018; 5: 110.
[http://dx.doi.org/10.3389/fnut.2018.00110] [PMID: 30533415]
[51]
Whitten PL, Lewis C, Russell E, Naftolin F. Potential adverse effects of phytoestrogens. J Nutr 1995; 125(3): 771S-6S.
[PMID: 7884563]
[52]
Hashem NM, Abo-Elsoud MA, Nour El-Din ANM, Kamel KI, Hassan GA. Prolonged exposure of dietary phytoestrogens on semen characteristics and reproductive performance of rabbit bucks. Domest Anim Endocrinol 2018; 64: 84-92.
[http://dx.doi.org/10.1016/j.domaniend.2018.03.003] [PMID: 29754011]
[53]
Dean M, Murphy BT, Burdette JE. Phytosteroids beyond estrogens: Regulators of reproductive and endocrine function in natural products. Mol Cell Endocrinol 2017; 442: 98-105.
[http://dx.doi.org/10.1016/j.mce.2016.12.013] [PMID: 27986590]
[54]
Yang J, Wen L, Jiang Y, Yang B. Natural estrogen receptor modulators and their heterologous biosynthesis. Trends Endocrinol Metab 2019; 30(1): 66-76.
[http://dx.doi.org/10.1016/j.tem.2018.11.002] [PMID: 30527917]
[55]
Beszterda M, Frański R. Endocrine disruptor compounds in environment: As a danger for children health. Pediatr Endocrinol Diabetes Metab 2018; 24(2): 88-95.
[http://dx.doi.org/10.18544/PEDM-24.02.0107] [PMID: 30300430]
[56]
Saayman BD. The use of alternative lipid emulsions in paediatric and neonatal parenteral nutrition. South Afr J Clin Nutr 2011; 24: S32-4.
[http://dx.doi.org/10.1080/16070658.2011.11734378]
[57]
Messina M. Soybean isoflavone exposure does not have feminizing effects on men: A critical examination of the clinical evidence. Fertil Steril 2010; 93(7): 2095-104.
[http://dx.doi.org/10.1016/j.fertnstert.2010.03.002] [PMID: 20378106]
[58]
Nikitina NA, Sobenin IA, Myasoedova VA, et al. Antiatherogenic effect of grape flavonoids in an ex vivo model. Bull Exp Biol Med 2006; 141(6): 712-5.
[http://dx.doi.org/10.1007/s10517-006-0260-7] [PMID: 17364057]
[59]
Orekhov AN, Sobenin IA, Revin VV, Bobryshev YV. Development of antiatherosclerotic drugs on the basis of natural products using cell model approach. Oxid Med Cell Longev 2015; 2015 463797
[http://dx.doi.org/10.1155/2015/463797] [PMID: 26347804]
[60]
Jargin SV. Phytoestrogens and other botanicals: On the problems of evidence-based evaluation. Recent Pat Cardiovasc Drug Discov 2013; 8(1): 67-71.
[http://dx.doi.org/10.2174/18722083113079990009] [PMID: 23514086]
[61]
Jargin SV. Development of antiatherosclerotic drugs on the basis of cell models: A comment. Int J Pharmacol Phytochem Ethnomed 2015; 1: 10-4.
[http://dx.doi.org/10.18052/www.scipress.com/IJPPE.1.10]
[62]
Kuznetsova IV, Kirpikov AS, Khovrina EA. Method of treating benign hyperplastic processes of female reproductive system. RU2471485C1 (2013).
[63]
Bruyère O, Cooper C, Pelletier JP, et al. A consensus statement on the European Society for Clinical and Economic Aspects of Osteoporosis and Osteoarthritis (ESCEO) algorithm for the management of knee osteoarthritis-From evidence-based medicine to the real-life setting. Semin Arthritis Rheum 2016; 45(4): S3-S11.
[http://dx.doi.org/10.1016/j.semarthrit.2015.11.010] [PMID: 26806188]
[64]
Jones IA, Togashi R, Wilson ML, Heckmann N, Vangsness CT. Intra-articular treatment options for knee osteoarthritis. Nat Rev Rheumatol 2019; 15(2): 77-90.
[http://dx.doi.org/10.1038/s41584-018-0123-4] [PMID: 30498258]
[65]
Vista ES, Lau CS. What about supplements for osteoarthritis? A critical and evidenced-based review. Int J Rheum Dis 2011; 14(2): 152-8.
[http://dx.doi.org/10.1111/j.1756-185X.2011.01619.x] [PMID: 21518314]
[66]
Wandel S, Jüni P, Tendal B, et al. Effects of glucosamine, chondroitin, or placebo in patients with osteoarthritis of hip or knee: Network meta-analysis. BMJ 2010; 341: c4675.
[http://dx.doi.org/10.1136/bmj.c4675] [PMID: 20847017]
[67]
Singh JA, Noorbaloochi S, MacDonald R, Maxwell LJ. Chondroitin for osteoarthritis. Cochrane Database Syst Rev 2015; 1 CD005614
[PMID: 25629804]
[68]
Vincent P. Intra-articular hyaluronic acid in the symptomatic treatment of knee osteoarthritis: A meta-analysis of single-injection products. Curr Ther Res Clin Exp 2019; 90: 39-51.
[http://dx.doi.org/10.1016/j.curtheres.2019.02.003] [PMID: 31289603]
[69]
Wehling P, Evans C, Wehling J, Maixner W. Effectiveness of intra-articular therapies in osteoarthritis: A literature review. Ther Adv Musculoskelet Dis 2017; 9(8): 183-96.
[http://dx.doi.org/10.1177/1759720X17712695] [PMID: 28835778]
[70]
Zhang B, Thayaparan A, Horner N, Bedi A, Alolabi B, Khan M. Outcomes of hyaluronic acid injections for glenohumeral osteoarthritis: A systematic review and meta-analysis. J Shoulder Elbow Surg 2019; 28(3): 596-606.
[http://dx.doi.org/10.1016/j.jse.2018.09.011] [PMID: 30502030]
[71]
Zhu X, Sang L, Wu D, Rong J, Jiang L. Effectiveness and safety of glucosamine and chondroitin for the treatment of osteoarthritis: A meta-analysis of randomized controlled trials. J Orthop Surg Res 2018; 13(1): 170.
[http://dx.doi.org/10.1186/s13018-018-0871-5] [PMID: 29980200]
[72]
Simental-Mendía M, Sánchez-García A, Vilchez-Cavazos F, Acosta-Olivo CA, Peña-Martínez VM, Simental-Mendía LE. Effect of glucosamine and chondroitin sulfate in symptomatic knee osteoarthritis: A systematic review and meta-analysis of randomized placebo-controlled trials. Rheumatol Int 2018; 38(8): 1413-28.
[http://dx.doi.org/10.1007/s00296-018-4077-2] [PMID: 29947998]
[73]
Black C, Clar C, Henderson R, MacEachern C, McNamee P, Quayyum Z. The clinical effectiveness of glucosamine and chondroitin supplements in lowing or arresting progression of osteoarthritis of the knee: A systematic review and economic evaluation Health echnol Assess (Rockv) 2009; 13: 1-148.
[74]
Volpi N. Quality of different chondroitin sulfate preparations in relation to their therapeutic activity. J Pharm Pharmacol 2009; 61(10): 1271-80.
[http://dx.doi.org/10.1211/jpp.61.10.0002] [PMID: 19814858]
[75]
Arrich J, Piribauer F, Mad P, Schmid D, Klaushofer K, Müllner M. Intra-articular hyaluronic acid for the treatment of osteoarthritis of the knee: Systematic review and meta-analysis. CMAJ 2005; 172(8): 1039-43.
[http://dx.doi.org/10.1503/cmaj.1041203] [PMID: 15824412]
[76]
Rutjes AW, Jüni P, da Costa BR, Trelle S, Nüesch E, Reichenbach S. Viscosupplementation for osteoarthritis of the knee: A systematic review and meta-analysis. Ann Intern Med 2012; 157(3): 180-91.
[http://dx.doi.org/10.7326/0003-4819-157-3-201208070-00473] [PMID: 22868835]
[77]
Nguyen C, Lefèvre-Colau MM, Poiraudeau S, Rannou F. Evidence and recommendations for use of intra-articular injections for knee osteoarthritis. Ann Phys Rehabil Med 2016; 59(3): 184-9.
[http://dx.doi.org/10.1016/j.rehab.2016.02.008] [PMID: 27103055]
[78]
Witteveen AG, Hofstad CJ, Kerkhoffs GM. Hyaluronic acid and other conservative treatment options for osteoarthritis of the ankle. Cochrane Database Syst Rev 2015; 10 CD010643
[http://dx.doi.org/10.1002/14651858.CD010643.pub2] [PMID: 26475434]
[79]
Lohmander LS, Dalén N, Englund G, et al. Hyaluronan multicentre trial group. Intra-articular hyaluronan injections in the treatment of osteoarthritis of the knee: A randomised, double blind, placebo controlled multicentre trial. Ann Rheum Dis 1996; 55(7): 424-31.
[http://dx.doi.org/10.1136/ard.55.7.424] [PMID: 8774159]
[80]
Brandt KD, Smith GN, Simon LS. Intraarticular injection of hyaluronan as treatment for knee osteoarthritis: What is the evidence? Arthritis Rheum 2000; 43(6): 1192-203.
[http://dx.doi.org/10.1002/1529-0131(200006)43:6<1192:AID-ANR2>3.0.CO;2-L] [PMID: 10857778]
[81]
Machado RC, Capela S, Rocha FAC. Polysaccharides as viscosupplementation agents: Structural molecular characteristics but not rheology appear crucial to the therapeutic response. Front Med (Lausanne) 2017; 4: 82.
[http://dx.doi.org/10.3389/fmed.2017.00082] [PMID: 28674692]
[82]
Uebelhart D. Clinical review of chondroitin sulfate in osteoarthritis. Osteoarthritis Cartilage 2008; 16(Suppl. 3): S19-21.
[http://dx.doi.org/10.1016/j.joca.2008.06.006] [PMID: 18674931]
[83]
Leighton R, Fitzpatrick J, Smith H, Crandall D, Flannery CR, Conrozier T. Systematic clinical evidence review of NASHA (Durolane hyaluronic acid) for the treatment of knee osteoarthritis. Open Access Rheumatol 2018; 10: 43-54.
[http://dx.doi.org/10.2147/OARRR.S162127] [PMID: 29849470]
[84]
Kulikov VG, Shusharin AG, Makhotin AA, Shevela AI. Method of treating coxarthrosis. RU2396961C1 ( 2010).
[85]
Migliore A, Anichini S. Intra-articular therapy in hip osteoarthritis. Clin Cases Miner Bone Metab 2017; 14(2): 179-81.
[http://dx.doi.org/10.11138/ccmbm/2017.14.1.179] [PMID: 29263729]
[86]
Maheu E, Bannuru RR, Herrero-Beaumont G, Allali F, Bard H, Migliore A. Why we should definitely include intra-articular hyaluronic acid as a therapeutic option in the management of knee osteoarthritis: Results of an extensive critical literature review. Semin Arthritis Rheum 2019; 48(4): 563-72.
[http://dx.doi.org/10.1016/j.semarthrit.2018.06.002] [PMID: 30072113]
[87]
Altman R, Hackel J, Niazi F, Shaw P, Nicholls M. Efficacy and safety of repeated courses of hyaluronic acid injections for knee osteoarthritis: A systematic review. Semin Arthritis Rheum 2018; 48(2): 168-75.
[http://dx.doi.org/10.1016/j.semarthrit.2018.01.009] [PMID: 29496227]
[88]
Xing D, Wang B, Liu Q, et al. Intra-articular hyaluronic acid in treating knee osteoarthritis: A PRISMA-compliant systematic review of overlapping meta-analysis. Sci Rep 2016; 6: 32790.
[http://dx.doi.org/10.1038/srep32790] [PMID: 27616273]
[89]
Jargin SV. Supplementation of glycosaminoglycans and their precursors in osteoarthritis versus diet modification. Int J Rheum Dis 2012; 15(3): e45-6.
[http://dx.doi.org/10.1111/j.1756-185X.2012.01707.x] [PMID: 22709499]
[90]
Tikhonov VP, Sidliarov DP, Zaveshchevskaia TL. Agent for care of peripheral joints and backbone area. RU2376011C1 (2009)
[91]
Bhathal A, Spryszak M, Louizos C, Frankel G. Glucosamine and chondroitin use in canines for osteoarthritis: A review. Open Vet J 2017; 7(1): 36-49.
[http://dx.doi.org/10.4314/ovj.v7i1.6] [PMID: 28331832]
[92]
Mansilha A, Sousa J. Pathophysiological mechanisms of chronic venous disease and implications for venoactive drug therapy. Int J Mol Sci 2018; 19(6): 1-21.
[http://dx.doi.org/10.3390/ijms19061669] [PMID: 29874834]
[93]
Katsenis K. Micronized Purified Flavonoid Fraction (MPFF): A review of its pharmacological effects, therapeutic efficacy and benefits in the management of chronic venous insufficiency. Curr Vasc Pharmacol 2005; 3(1): 1-9.
[http://dx.doi.org/10.2174/1570161052773870] [PMID: 15641940]
[94]
Danielsson G, Jungbeck C, Peterson K, Norgren L. A randomised controlled trial of micronised purified flavonoid fraction vs placebo in patients with chronic venous disease. Eur J Vasc Endovasc Surg 2002; 23(1): 73-6.
[http://dx.doi.org/10.1053/ejvs.2001.1531] [PMID: 11748952]
[95]
Bush R, Comerota A, Meissner M, Raffetto JD, Hahn SR, Freeman K. Recommendations for the medical management of chronic venous disease: The role of Micronized Purified Flavanoid Fraction (MPFF). Phlebology 2017; 32(1): 3-19.
[http://dx.doi.org/10.1177/0268355517692221] [PMID: 28211296]
[96]
Martinez-Zapata MJ, Vernooij RW, Uriona TSM, et al. Phlebotonics for venous insufficiency. Cochrane Database Syst Rev 2016; 4(4) CD003229
[PMID: 27048768]
[97]
Kharkevich DA. The principles of action and use of phlebotropic agents. Klin Med (Mosk) 2004; 82(11): 4-10.
[PMID: 15656390]
[98]
Bevan JA. Some bases of differences in vascular response to sympathetic activity. Circ Res 1979; 45(2): 161-71.
[http://dx.doi.org/10.1161/01.RES.45.2.161] [PMID: 36236]
[99]
Nicolaides AN. Cardiovascular disease educational and research trust european society of vascular surgery the international angiology scientific activity congress organization international union of angiology union internationale de phlebologie at the abbaye des vaux de cernay. investigation of chronic venous insufficiency: A consensus statement (France, March 5-9, 1997). Circulation 2000; 102(20): E126-63.
[http://dx.doi.org/10.1161/01.CIR.102.20.e126] [PMID: 11076834]
[100]
Perrin M, Ramelet AA. Pharmacological treatment of primary chronic venous disease: Rationale, results and unanswered questions. Eur J Vasc Endovasc Surg 2011; 41(1): 117-25.
[http://dx.doi.org/10.1016/j.ejvs.2010.09.025] [PMID: 21126890]
[101]
Duarte J, Pérez-Vizcaíno F, Zarzuelo A, Jiménez J, Tamargo J. Vasodilator effects of quercetin in isolated rat vascular smooth muscle. Eur J Pharmacol 1993; 239(1-3): 1-7.
[http://dx.doi.org/10.1016/0014-2999(93)90968-N] [PMID: 8223884]
[102]
Schmitt M, Blackman DJ, Middleton GW, Cockcroft JR, Frenneaux MP. Assessment of venous capacitance. Radionuclide plethysmography: Methodology and research applications. Br J Clin Pharmacol 2002; 54(6): 565-76.
[http://dx.doi.org/10.1046/j.1365-2125.2002.t01-7-01689.x] [PMID: 12492602]
[103]
Aellig WH. Clinical pharmacology, physiology and pathophysiology of superficial veins-2. Br J Clin Pharmacol 1994; 38(4): 289-305.
[http://dx.doi.org/10.1111/j.1365-2125.1994.tb04357.x] [PMID: 7833218]
[104]
Ivanov IS, Sidekhmenova AV, Nosarev AV, Tyukavkina NA, Plotnikov MB. Effect of dihydroquercetin on the tone of isolated rat veins. Bull Exp Biol Med 2013; 155(1): 65-6.
[http://dx.doi.org/10.1007/s10517-013-2080-x] [PMID: 23667873]
[105]
Savineau JP, Marthan R. Diosmin-induced increase in sensitivity to Ca2+ of the smooth muscle contractile apparatus in the rat isolated femoral vein. Br J Pharmacol 1994; 111(4): 978-80.
[http://dx.doi.org/10.1111/j.1476-5381.1994.tb14838.x] [PMID: 8032624]
[106]
Testai L, Calderone V. Nutraceutical value of citrus flavanones and their implications in cardiovascular disease. Nutrients 2017; 9(5): 502.
[http://dx.doi.org/10.3390/nu9050502] [PMID: 28509871]
[107]
Takumi H, Nakamura H, Simizu T, et al. Bioavailability of orally administered water-dispersible hesperetin and its effect on peripheral vasodilatation in human subjects: Implication of endothelial functions of plasma conjugated metabolites. Food Funct 2012; 3(4): 389-98.
[http://dx.doi.org/10.1039/c2fo10224b] [PMID: 22307524]
[108]
Scallon C, Bell-Syer SE, Aziz Z. Flavonoids for treating venous leg ulcers. Cochrane Database Syst Rev 2013; 5 CD006477
[PMID: 23728661]
[109]
Aziz Z, Tang WL, Chong NJ, Tho LY. A systematic review of the efficacy and tolerability of hydroxyethylrutosides for improvement of the signs and symptoms of chronic venous insufficiency. J Clin Pharm Ther 2015; 40(2): 177-85.
[http://dx.doi.org/10.1111/jcpt.12247] [PMID: 25630350]
[110]
Kakkos SK, Nicolaides AN. Efficacy of micronized purified flavonoid fraction (Daflon®) on improving individual symptoms, signs and quality of life in patients with chronic venous disease: A systematic review and meta-analysis of randomized double-blind placebo-controlled trials. Int Angiol 2018; 37(2): 143-54.
[PMID: 29385792]
[111]
Geroulakos G, Nicolaides AN. Controlled studies of Daflon 500 mg in chronic venous insufficiency. Angiology 1994; 45(6 Pt 2): 549-53.
[PMID: 8203786]
[112]
Duchene MP, Amiel M, Barbe R. Evaluation of the clinical pharmacological activity of a phlebotonic agent. Application to the study of Daflon 500 mg. Int Angiol 1988; 7(2): 25-32.
[PMID: 3053940]
[113]
Allaert FA. Meta-analysis of the impact of the principal venoactive drugs agents on malleolar venous edema. Int Angiol 2012; 31(4): 310-5.
[PMID: 22801396]
[114]
Rabe E, Agus GB, Roztocil K. Analysis of the effects of micronized purified flavonoid fraction versus placebo on symptoms and quality of life in patients suffering from chronic venous disease: From a prospective randomized trial. Int Angiol 2015; 34(5): 428-36.
[PMID: 25972136]
[115]
Giorgio M. Oxidative stress and the unfulfilled promises of antioxidant agents. Ecancermedicalscience 2015; 9: 556.
[http://dx.doi.org/10.3332/ecancer.2015.556] [PMID: 26284120]
[116]
Jargin SV. On the use of carnosine and antioxidants: A letter from Russia. J Intercult Ethnopharmacol 2016; 5(3): 317-9.
[http://dx.doi.org/10.5455/jice.20160409010229] [PMID: 27366359]
[117]
Abu Raihan SM. Effect of plant flavonoids on mosquito larvae. Nat Univ J Sci 2014; 1: 27-30.
[118]
Goszcz K, Deakin SJ, Duthie GG, et al. Antioxidants in cardiovascular therapy: Panacea or false hope? Front Cardiovasc Med 2015; 2: 29.
[http://dx.doi.org/10.3389/fcvm.2015.00029] [PMID: 26664900]
[119]
Duda-Chodak A. The inhibitory effect of polyphenols on human gut microbiota. J Physiol Pharmacol 2012; 63(5): 497-503.
[PMID: 23211303]
[120]
Lichota A, Gwozdzinski L, Gwozdzinski K. Therapeutic potential of natural compounds in inflammation and chronic venous insufficiency. Eur J Med Chem 2019; 176: 68-91.
[http://dx.doi.org/10.1016/j.ejmech.2019.04.075] [PMID: 31096120]
[121]
Gattuso G, Barreca D, Gargiulli C, Leuzzi U, Caristi C. Flavonoid composition of Citrus juices. Molecules 2007; 12(8): 1641-73.
[http://dx.doi.org/10.3390/12081641] [PMID: 17960080]
[122]
Jargin SV. Grapefruit: Some perspectives in pharmacology and nutrition. J Intercult Ethnopharmacol 2017; 6(3): 339-41.
[http://dx.doi.org/10.5455/jice.20170511061624] [PMID: 28894634]
[123]
Sathe NA, Krishnaswami S, Andrews J, Ficzere C, McPheeters ML. Pharmacologic agents that promote airway clearance in hospitalized subjects: A systematic review. Respir Care 2015; 60(7): 1061-70.
[http://dx.doi.org/10.4187/respcare.04086] [PMID: 25944943]
[124]
Rogers DF. Mucoactive agents for airway mucus hypersecretory diseases. Respir Care 2007; 52(9): 1176-93.
[PMID: 17716385]
[125]
Otu A, Langridge P, Denning DW. Nebulised N-acetylcysteine for unresponsive bronchial obstruction in allergic brochopulmonary aspergillosis: A case series and review of the literature. J Fungi (Basel) 2018; 4(4): 1-13.
[http://dx.doi.org/10.3390/jof4040117] [PMID: 30326585]
[126]
Konrad F, Schoenberg MH, Wiedmann H, Kilian J, Georgieff M. The application of n-acetylcysteine as an antioxidant and mucolytic in mechanical ventilation in intensive care patients. A prospective, randomized, placebo-controlled, double-blind study. Anaesthesist 1995; 44(9): 651-8.
[http://dx.doi.org/10.1007/s001010050200] [PMID: 7485927]
[127]
Cazzola M, Calzetta L, Page C, et al. Influence of N-acetylcysteine on chronic bronchitis or COPD exacerbations: A meta-analysis. Eur Respir Rev 2015; 24(137): 451-61.
[http://dx.doi.org/10.1183/16000617.00002215] [PMID: 26324807]
[128]
Decramer M, Rutten-van Mölken M, Dekhuijzen PN, et al. Effects of N-acetylcysteine on outcomes in chronic obstructive pulmonary disease (Bronchitis Randomized on NAC Cost-Utility Study, BRONCUS): A randomised placebo-controlled trial. Lancet 2005; 365(9470): 1552-60.
[http://dx.doi.org/10.1016/S0140-6736(05)66456-2] [PMID: 15866309]
[129]
Rubin BK. Aerosol medications for treatment of mucus clearance disorders. Respir Care 2015; 60(6): 825-9.
[http://dx.doi.org/10.4187/respcare.04087] [PMID: 26070577]
[130]
Tam J, Nash EF, Ratjen F, Tullis E, Stephenson A. Nebulized and oral thiol derivatives for pulmonary disease in cystic fibrosis. Cochrane Database Syst Rev 2013; 7 CD007168
[http://dx.doi.org/10.1002/14651858.CD007168.pub3] [PMID: 23852992]
[131]
Black PN, Morgan-Day A, McMillan TE, Poole PJ, Young RP. Randomised, controlled trial of N-acetylcysteine for treatment of acute exacerbations of chronic obstructive pulmonary disease. ISRCTN21676344 BMC Pulm Med 2004; 4: 13.
[http://dx.doi.org/10.1186/1471-2466-4-13] [PMID: 15581425]
[132]
Rubin BK. Mucolytics, expectorants, and mucokinetic medications. Respir Care 2007; 52(7): 859-65.
[PMID: 17594730]
[133]
Tarrant BJ, Maitre CL, Romero L, et al. Mucoactive agents for adults with acute lung conditions: A systematic review. Heart Lung 2019; 48(2): 141-7.
[http://dx.doi.org/10.1016/j.hrtlng.2018.09.010] [PMID: 30409442]
[134]
Tarrant BJ, Le Maitre C, Romero L, et al. Mucoactive agents for chronic, non-cystic fibrosis lung disease: A systematic review and meta-analysis. Respirology 2017; 22(6): 1084-92.
[http://dx.doi.org/10.1111/resp.13047] [PMID: 28397992]
[135]
Ehre C, Rushton ZL, Wang B, et al. An improved inhaled mucolytic to treat airway muco-obstructive diseases. Am J Respir Crit Care Med 2019; 199(2): 171-80.
[http://dx.doi.org/10.1164/rccm.201802-0245OC] [PMID: 30212240]
[136]
Poole PJ, Black PN. Oral mucolytic drugs for exacerbations of chronic obstructive pulmonary disease: Systematic review. BMJ 2001; 322(7297): 1271-4.
[http://dx.doi.org/10.1136/bmj.322.7297.1271] [PMID: 11375228]
[137]
Gerrits CM, Herings RM, Leufkens HG, Lammers JW. N-acetylcysteine reduces the risk of re-hospitalisation among patients with chronic obstructive pulmonary disease. Eur Respir J 2003; 21(5): 795-8.
[http://dx.doi.org/10.1183/09031936.03.00063402] [PMID: 12765423]
[138]
Ernst P, Suissa S. N-acetylcysteine is unlikely to reduce hospitalisation for chronic obstructive pulmonary disease. Eur Respir J 2003; 22(5): 865.
[http://dx.doi.org/10.1183/09031936.03.00063403] [PMID: 14621098]
[139]
Sun T, Liu J, Zhao W. Zhao de W. Efficacy of N-acetylcysteine in idiopathic pulmonary fibrosis: A systematic review and meta-analysis. Medicine (Baltimore) 2016; 95(19) e3629
[http://dx.doi.org/10.1097/MD.0000000000003629] [PMID: 27175674]
[140]
Bridgeman MM, Marsden M, MacNee W, Flenley DC, Ryle AP. Cysteine and glutathione concentrations in plasma and bronchoalveolar lavage fluid after treatment with N-acetylcysteine. Thorax 1991; 46(1): 39-42.
[http://dx.doi.org/10.1136/thx.46.1.39] [PMID: 1871695]
[141]
Cotgreave IA, Eklund A, Larsson K, Moldéus PW. No penetration of orally administered N-acetylcysteine into bronchoalveolar lavage fluid. Eur J Respir Dis 1987; 70(2): 73-7.
[PMID: 3817074]
[142]
Rodenstein D, DeCoster A, Gazzaniga A. Pharmacokinetics of oral acetylcysteine: Absorption, binding and metabolism in patients with respiratory disorders. Clin Pharmacokinet 1978; 3(3): 247-54.
[http://dx.doi.org/10.2165/00003088-197803030-00005] [PMID: 657688]
[143]
Millar AB, Pavia D, Agnew JE, Lopez-Vidriero MT, Lauque D, Clarke SW. Effect of oral N-acetylcysteine on mucus clearance. Br J Dis Chest 1985; 79(3): 262-6.
[http://dx.doi.org/10.1016/0007-0971(85)90046-4] [PMID: 3893512]
[144]
Holdiness MR. Clinical pharmacokinetics of N-acetylcysteine. Clin Pharmacokinet 1991; 20(2): 123-34.
[http://dx.doi.org/10.2165/00003088-199120020-00004] [PMID: 2029805]
[145]
Pintucci JP, Corno S, Garotta M. Biofilms and infections of the upper respiratory tract. Eur Rev Med Pharmacol Sci 2010; 14(8): 683-90.
[PMID: 20707288]
[146]
Dinicola S, De Grazia S, Carlomagno G, Pintucci JP. N-acetylcysteine as powerful molecule to destroy bacterial biofilms. A systematic review. Eur Rev Med Pharmacol Sci 2014; 18(19): 2942-8.
[PMID: 25339490]
[147]
Smith A, Buchinsky FJ, Post JC. Eradicating chronic ear, nose, and throat infections: A systematically conducted literature review of advances in biofilm treatment. Otolaryngol Head Neck Surg 2011; 144(3): 338-47.
[http://dx.doi.org/10.1177/0194599810391620] [PMID: 21493193]
[148]
Bulut F, Meric F, Yorgancilar E, et al. Effects of N-acetyl-cysteine and acetylsalicylic acid on the tonsil bacterial biofilm tissues by light and electron microscopy. Eur Rev Med Pharmacol Sci 2014; 18(23): 3720-5.
[PMID: 25535147]
[149]
Blasi F, Page C, Rossolini GM, et al. The effect of N-acetylcysteine on biofilms: Implications for the treatment of respiratory tract infections. Respir Med 2016; 117: 190-7.
[http://dx.doi.org/10.1016/j.rmed.2016.06.015] [PMID: 27492531]
[150]
Moon JH, Choi YS, Lee HW, Heo JS, Chang SW, Lee JY. Antibacterial effects of N-acetylcysteine against endodontic pathogens. J Microbiol 2016; 54(4): 322-9.
[http://dx.doi.org/10.1007/s12275-016-5534-9] [PMID: 27033208]
[151]
Onger ME, Gocer H, Emir D, Kaplan S. N-acetylcysteine eradicates Pseudomonas aeruginosa biofilms in bone cement. Scanning 2016; 38(6): 766-70.
[http://dx.doi.org/10.1002/sca.21326] [PMID: 27186786]
[152]
Pei Y, Liu H, Yang Y, et al. Biological activities and potential oral applications of N-acetylcysteine: progress and prospects. Oxid Med Cell Longev 2018; 2018 2835787
[http://dx.doi.org/10.1155/2018/2835787] [PMID: 29849877]
[153]
Livingstone CR, Andrews MA, Jenkins SM, Marriott C. Model systems for the evaluation of mucolytic drugs: Acetylcysteine and S-carboxymethylcysteine. J Pharm Pharmacol 1990; 42(2): 73-8.
[http://dx.doi.org/10.1111/j.2042-7158.1990.tb05357.x] [PMID: 1972405]
[154]
Johnson K, McEvoy CE, Naqvi S, et al. High-dose oral N-acetylcysteine fails to improve respiratory health status in patients with chronic obstructive pulmonary disease and chronic bronchitis: A randomized, placebo-controlled trial. Int J Chron Obstruct Pulmon Dis 2016; 11: 799-807.
[PMID: 27143871]
[155]
Zheng JP, Wen FQ, Bai CX, et al. PANTHEON study group. Twice daily N-acetylcysteine 600 mg for exacerbations of chronic obstructive pulmonary disease (PANTHEON): A randomised, double-blind placebo-controlled trial. Lancet Respir Med 2014; 2(3): 187-94.
[http://dx.doi.org/10.1016/S2213-2600(13)70286-8] [PMID: 24621680]
[156]
Ziment I. Acetylcysteine: A drug that is much more than a mucokinetic. Biomed Pharmacother 1988; 42(8): 513-9.
[PMID: 3066412]
[157]
Tse HN, Tseng CZ. Update on the pathological processes, molecular biology, and clinical utility of N-acetylcysteine in chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 2014; 9: 825-36.
[http://dx.doi.org/10.2147/COPD.S51057] [PMID: 25125976]
[158]
Calzetta L, Matera MG, Rogliani P, Cazzola M. Multifaceted activity of N-acetyl-l-cysteine in chronic obstructive pulmonary disease. Expert Rev Respir Med 2018; 12(8): 693-708.
[http://dx.doi.org/10.1080/17476348.2018.1495562] [PMID: 29972340]
[159]
Lau A, Kennedy BK, Kirkland JL, Tullius SG. Mixing old and young: Enhancing rejuvenation and accelerating aging. J Clin Invest 2019; 129(1): 4-11.
[http://dx.doi.org/10.1172/JCI123946] [PMID: 30601138]
[160]
Neves J, Sousa-Victor P, Jasper H. Rejuvenating strategies for stem cell-based therapies in aging. Cell Stem Cell 2017; 20(2): 161-75.
[http://dx.doi.org/10.1016/j.stem.2017.01.008] [PMID: 28157498]
[161]
Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: Past, present, and future. Stem Cell Res Ther 2019; 10(1): 68.
[http://dx.doi.org/10.1186/s13287-019-1165-5] [PMID: 30808416]
[162]
Blum B, Benvenisty N. The tumorigenicity of diploid and aneuploid human pluripotent stem cells. Cell Cycle 2009; 8(23): 3822-30.
[http://dx.doi.org/10.4161/cc.8.23.10067] [PMID: 19887907]
[163]
Mancuso P, Raman S, Glynn A, Barry F, Murphy JM. Mesenchymal stem cell therapy for osteoarthritis: The critical role of the cell secretome. Front Bioeng Biotechnol 2019; 7: 9.
[http://dx.doi.org/10.3389/fbioe.2019.00009] [PMID: 30761298]
[164]
Castro-Viñuelas R, Sanjurjo-Rodríguez C, Piñeiro-Ramil M, et al. Induced pluripotent stem cells for cartilage repair: Current status and future perspectives. Eur Cell Mater 2018; 36: 96-109.
[http://dx.doi.org/10.22203/eCM.v036a08] [PMID: 30204229]
[165]
Rashid H, Kwoh CK. Should platelet-rich plasma or stem cell therapy be used to treat osteoarthritis? Rheum Dis Clin North Am 2019; 45(3): 417-38.
[http://dx.doi.org/10.1016/j.rdc.2019.04.010] [PMID: 31277753]
[166]
Harrell CR, Markovic BS, Fellabaum C, Arsenijevic A, Volarevic V. Mesenchymal stem cell-based therapy of osteoarthritis: Current knowledge and future perspectives. Biomed Pharmacother 2019; 109: 2318-26.
[http://dx.doi.org/10.1016/j.biopha.2018.11.099] [PMID: 30551490]
[167]
Wang AT, Feng Y, Jia HH, Zhao M, Yu H. Application of mesenchymal stem cell therapy for the treatment of osteoarthritis of the knee: A concise review. World J Stem Cells 2019; 11(4): 222-35.
[http://dx.doi.org/10.4252/wjsc.v11.i4.222] [PMID: 31110603]
[168]
Berardis S, Dwisthi SP, Najimi M, Sokal EM. Use of mesenchymal stem cells to treat liver fibrosis: Current situation and future prospects. World J Gastroenterol 2015; 21(3): 742-58.
[http://dx.doi.org/10.3748/wjg.v21.i3.742] [PMID: 25624709]
[169]
Kwak KA, Cho HJ, Yang JY, Park YS. Current perspectives regarding stem cell-based therapy for liver cirrhosis. Can J Gastroenterol Hepatol 2018; 2018 4197857
[http://dx.doi.org/10.1155/2018/4197857] [PMID: 29670867]
[170]
Guo Y, Chen B, Chen LJ, Zhang CF, Xiang C. Current status and future prospects of mesenchymal stem cell therapy for liver fibrosis. J Zhejiang Univ Sci B 2016; 17(11): 831-41.
[http://dx.doi.org/10.1631/jzus.B1600101] [PMID: 27819130]
[171]
Marsh SE, Blurton-Jones M. Neural stem cell therapy for neurodegenerative disorders: The role of neurotrophic support. Neurochem Int 2017; 106: 94-100.
[http://dx.doi.org/10.1016/j.neuint.2017.02.006] [PMID: 28219641]
[172]
Walker PA, Shah SK, Harting MT, Cox CS. Progenitor cell therapies for traumatic brain injury: Barriers and opportunities in translation. Dis Model Mech 2009; 2(1-2): 23-38.
[http://dx.doi.org/10.1242/dmm.001198] [PMID: 19132123]
[173]
Fleifel D, Rahmoon MA, AlOkda A, Nasr M, Elserafy M, El-Khamisy SF. Recent advances in stem cells therapy: A focus on cancer, Parkinson’s and Alzheimer’s. J Genet Eng Biotechnol 2018; 16(2): 427-32.
[http://dx.doi.org/10.1016/j.jgeb.2018.09.002] [PMID: 30733756]
[174]
Scudellari M. How iPS cells changed the world. Nature 2016; 534(7607): 310-2.
[http://dx.doi.org/10.1038/534310a] [PMID: 27306170]
[175]
Tartaglione AM, Popoli P, Calamandrei G. Regenerative medicine in Huntington’s disease: Strengths and weaknesses of preclinical studies. Neurosci Biobehav Rev 2017; 77: 32-47.
[http://dx.doi.org/10.1016/j.neubiorev.2017.02.017] [PMID: 28223129]
[176]
Duncan T, Valenzuela M. Alzheimer’s disease, dementia, and stem cell therapy. Stem Cell Res Ther 2017; 8(1): 111.
[http://dx.doi.org/10.1186/s13287-017-0567-5] [PMID: 28494803]
[177]
Bardelli S, Moccetti M. Stem and progenitor cells in human cardiopulmonary development and regeneration. Stem Cells Int 2017; 2017 2653142
[http://dx.doi.org/10.1155/2017/2653142] [PMID: 29075297]
[178]
Fisher SA, Zhang H, Doree C, Mathur A, Martin-Rendon E. Stem cell treatment for acute myocardial infarction. Cochrane Database Syst Rev 2015; 9 CD006536
[http://dx.doi.org/10.1002/14651858.CD006536.pub4] [PMID: 26419913]
[179]
Fisher SA, Doree C, Mathur A, Taggart DP, Martin-Rendon E. Stem cell therapy for chronic ischaemic heart disease and congestive heart failure. Cochrane Database Syst Rev 2016; 12 CD007888
[http://dx.doi.org/10.1002/14651858.CD007888.pub3] [PMID: 28012165]
[180]
Tang JN, Cores J, Huang K, et al. Concise review: Is cardiac cell therapy dead? Embarrassing trial outcomes and new directions for the future. Stem Cells Transl Med 2018; 7(4): 354-9.
[http://dx.doi.org/10.1002/sctm.17-0196] [PMID: 29468830]
[181]
Yanamandala M, Zhu W, Garry DJ, et al. Overcoming the roadblocks to cardiac cell therapy using tissue engineering. J Am Coll Cardiol 2017; 70(6): 766-75.
[http://dx.doi.org/10.1016/j.jacc.2017.06.012] [PMID: 28774384]
[182]
Bareeqa SB, Bibi F, Ahmed SI, Samar SS. Advancement in stem cell therapy for ischemic myocardial cell: A systematic review. Int J Hematol Oncol Stem Cell Res 2018; 12(4): 282-90.
[http://dx.doi.org/10.18502/ijhoscr.v12i4.106] [PMID: 30774828]
[183]
Wernly B, Mirna M, Rezar R, et al. Regenerative cardiovascular therapies: Stem cells and beyond. Int J Mol Sci 2019; 20(6) E1420
[http://dx.doi.org/10.3390/ijms20061420] [PMID: 30901815]
[184]
Toyserkani NM, Jørgensen MG, Tabatabaeifar S, Jensen CH, Sheikh SP, Sørensen JA. Concise review: A safety assessment of adipose-derived cell therapy in clinical trials: A systematic review of reported adverse events. Stem Cells Transl Med 2017; 6(9): 1786-94.
[http://dx.doi.org/10.1002/sctm.17-0031] [PMID: 28722289]
[185]
A futile cycle in cell therapy. Nat Biotechnol 2017; 35(4): 291.
[http://dx.doi.org/10.1038/nbt.3857] [PMID: 28398319]
[186]
Comella K, Parcero J, Bansal H, et al. Effects of the intramyocardial implantation of stromal vascular fraction in patients with chronic ischemic cardiomyopathy. J Transl Med 2016; 14(1): 158.
[http://dx.doi.org/10.1186/s12967-016-0918-5] [PMID: 27255774]
[187]
van den Akker F, de Jager SC, Sluijter JP. Mesenchymal stem cell therapy for cardiac inflammation: Immunomodulatory properties and the influence of toll-like receptors. Mediators Inflamm 2013; 2013 181020
[http://dx.doi.org/10.1155/2013/181020] [PMID: 24391353]
[188]
Jeong H, Yim HW, Park HJ, et al. Mesenchymal stem cell therapy for ischemic heart disease: Systematic review and meta-analysis. Int J Stem Cells 2018; 11(1): 1-12.
[http://dx.doi.org/10.15283/ijsc17061] [PMID: 29482311]
[189]
Ullah M, Sun Z. Stem cells and anti-aging genes: Double-edged sword-do the same job of life extension. Stem Cell Res Ther 2018; 9(1): 3.
[http://dx.doi.org/10.1186/s13287-017-0746-4] [PMID: 29321045]
[190]
Terzic A, Behfar A. Posology for regenerative therapy. Circ Res 2017; 121(11): 1213-5.
[http://dx.doi.org/10.1161/CIRCRESAHA.117.312074] [PMID: 29122941]
[191]
Tasso R, Pennesi G. When stem cells meet immunoregulation. Int Immunopharmacol 2009; 9(5): 596-8.
[http://dx.doi.org/10.1016/j.intimp.2009.01.014] [PMID: 19539568]
[192]
Suncion VY, Ghersin E, Fishman JE, et al. Does transendocardial injection of mesenchymal stem cells improve myocardial function locally or globally?: An analysis from the Percutaneous Stem Cell Injection Delivery Effects on Neomyogenesis (POSEIDON) randomized trial. Circ Res 2014; 114(8): 1292-301.
[http://dx.doi.org/10.1161/CIRCRESAHA.114.302854] [PMID: 24449819]
[193]
Kirillov AM, Fatkhudinov TK, Dyachkov AV, Koroteev AV, Goldshtein DV, Bochkov NP. Transplantation of allogenic cells in the therapy of patients with dilated cardiomyopathy. Bull Exp Biol Med 2007; 144(4): 635-9.
[http://dx.doi.org/10.1007/s10517-007-0392-4] [PMID: 18642728]
[194]
Schoenhard JA, Hatzopoulos AK. Stem cell therapy: Pieces of the puzzle. J Cardiovasc Transl Res 2010; 3(1): 49-60.
[http://dx.doi.org/10.1007/s12265-009-9148-z] [PMID: 20119487]
[195]
Bilgimol JC, Ragupathi S, Vengadassalapathy L, et al. Stem cells: An eventual treatment option for heart diseases. World J Stem Cells 2015; 7(8): 1118-26.
[http://dx.doi.org/10.4252/wjsc.v7.i8.1118] [PMID: 26435771]
[196]
Jargin SV. Scientific misconduct and related topics. Am J Exp Clin Res 2017; 4(1): 197-201.
[197]
Jargin SV. Invasive procedures with questionable indications. Ann Med Surg (Lond) 2014; 3(4): 126-9.
[http://dx.doi.org/10.1016/j.amsu.2014.06.003] [PMID: 25568799]
[198]
Shomina EA, Yarygin NV, Yarygin KN. Unproven stem-cell therapy ban. Nature 2018; 454: 923.
[199]
Rossbauer M. Unproven stem-cell therapy ban. Nature 2008; 454(7207): 923.
[http://dx.doi.org/10.1038/454923a] [PMID: 18719550]
[200]
Qiu J. China clamps down on controversial therapies. Lancet 2009; 373(9678): 1834-5.
[http://dx.doi.org/10.1016/S0140-6736(09)61007-2] [PMID: 19504665]
[201]
Foddy B. A duty to deceive: Placebos in clinical practice. Am J Bioeth 2009; 9(12): 4-12.
[http://dx.doi.org/10.1080/15265160903318350] [PMID: 20013484]
[202]
Hróbjartsson A. Clinical placebo interventions are unethical, unnecessary, and unprofessional. J Clin Ethics 2008; 19(1): 66-9.
[PMID: 18552056]
[203]
Linde K, Fässler M, Meissner K. Placebo interventions, placebo effects and clinical practice. Philos Trans R Soc Lond B Biol Sci 2011; 366(1572): 1905-12.
[http://dx.doi.org/10.1098/rstb.2010.0383] [PMID: 21576148]
[204]
Jargin SV. Urological concern after nuclear accidents. Urol Ann 2018; 10(3): 240-2.
[http://dx.doi.org/10.4103/0974-7796.236525] [PMID: 30089979]
[205]
Jargin SV. Renal biopsy research with implications for therapy of glomerulonephritis. Curr Drug Ther 2012; 7(4): 263-7.
[http://dx.doi.org/10.2174/157488512804999055]
[206]
Smith R. Research misconduct: The poisoning of the well. J R Soc Med 2006; 99(5): 232-7.
[http://dx.doi.org/10.1177/014107680609900514] [PMID: 16672756]
[207]
Jargin SV. Pathology in the former Soviet Union: Scientific misconduct and related phenomena. Dermatol Pract Concept 2011; 1(1): 75-81.
[http://dx.doi.org/10.5826/dpc.0101a16] [PMID: 24396725]
[208]
Jargin SV. Discussion of Evaluation of cholesterol-lowering and antioxidant properties of sugar cane policosanols in hamsters and humans. Appl Physiol Nutr Metab 2009; 34(1): 75.
[http://dx.doi.org/10.1139/H08-141] [PMID: 19234588]
[209]
Jargin SV. Calf hemodialysate hypothesis: Nutritive value plus placebo effect. Med Hypotheses 2017; 109: 95-6.
[http://dx.doi.org/10.1016/j.mehy.2017.09.029] [PMID: 29150305]

© 2024 Bentham Science Publishers | Privacy Policy