Phase 1 Study of a Sulforaphane-Containing Broccoli Sprout Homogenate for Sickle Cell Disease

Jennifer F Doss, Jude C Jonassaint, Melanie E Garrett, Allison E Ashley-Koch, Marilyn J Telen, Jen-Tsan Chi, Jennifer F Doss, Jude C Jonassaint, Melanie E Garrett, Allison E Ashley-Koch, Marilyn J Telen, Jen-Tsan Chi

Abstract

Sickle cell disease (SCD) is the most common inherited hemoglobinopathy worldwide. Our previous results indicate that the reduced oxidative stress capacity of sickle erythrocytes may be caused by decreased expression of NRF2 (Nuclear factor (erythroid-derived 2)-like 2), an oxidative stress regulator. We found that activation of NRF2 with sulforaphane (SFN) in erythroid progenitors significantly increased the expression of NRF2 targets HMOX1, NQO1, and HBG1 (subunit of fetal hemoglobin) in a dose-dependent manner. Therefore, we hypothesized that NRF2 activation with SFN may offer therapeutic benefits for SCD patients by restoring oxidative capacity and increasing fetal hemoglobin concentration. To test this hypothesis, we performed a Phase 1, open-label, dose-escalation study of SFN, contained in a broccoli sprout homogenate (BSH) that naturally contains SFN, in adults with SCD. The primary and secondary study endpoints were safety and physiological response to NRF2 activation, respectively. We found that BSH was well tolerated, and the few adverse events that occurred during the trial were not likely related to BSH consumption. We observed an increase in the mean relative whole blood mRNA levels for the NRF2 target HMOX1 (p = 0.02) on the last day of BSH treatment, compared to pre-treatment. We also observed a trend toward increased mean relative mRNA levels of the NRF2 target HBG1 (p = 0.10) from baseline to end of treatment, but without significant changes in HbF protein. We conclude that BSH, in the provided doses, is safe in stable SCD patients and may induce changes in gene expression levels. We therefore propose investigation of more potent NRF2 inducers, which may elicit more robust physiological changes and offer clinical benefits to SCD patients. Trial registration: ClinicalTrials.gov NCT01715480.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. CONSORT flow diagram enrollment summary
Fig 1. CONSORT flow diagram enrollment summary
Fig 2. Treatment of erythroid progenitors with…
Fig 2. Treatment of erythroid progenitors with sulforaphane induces NRF2 target expression.
Relative mRNA expression of in vitro differentiating erythroid progenitors 48 hours after beginning treatment, relative to untreated for A) NQO1, B) HMOX1, and C) HBG1/(HBB+HBG1) (n = 3). Cells were treated starting on Day 8 of differentiation, and were treated once every 24 hours, with a total of two treatments for the listed concentration. TBHQ was used as a positive control. All expression was set relative to pre-treatment values, was normalized with GAPDH, and all statistical analyses were performed using an unpaired t-test (* = p<0.05, ** = p<0.01, *** = p<0.001).
Fig 3. Effect of BSH ingestion on…
Fig 3. Effect of BSH ingestion on whole blood mRNA in SCD subjects.
The composite of the mean for relative mRNA expression of A) NQO1, B) HMOX1, and C) HBG1/(HBB+HBG1) for all patients during the study period. All expression was set relative to pre-treatment values. Expression was normalized with GAPDH, and all statistical analyses were performed using Friedman’s test (* = p

Fig 4. Fetal hemoglobin levels during study…

Fig 4. Fetal hemoglobin levels during study period.

Fetal hemoglobin levels were assessed with hemoglobin…

Fig 4. Fetal hemoglobin levels during study period.
Fetal hemoglobin levels were assessed with hemoglobin electrophoresis for the A) 50g, B) 100g, and C) 150g cohorts. Each individual patient is designated with letter, symbol, and color codes; several patients participated in multiple cohorts. Patient F was diagnosed with HbSß° thalassemia; all other patients were diagnosed with HbSS. Patients taking hydroxyurea during the study period are denoted with an asterisk.
Fig 4. Fetal hemoglobin levels during study…
Fig 4. Fetal hemoglobin levels during study period.
Fetal hemoglobin levels were assessed with hemoglobin electrophoresis for the A) 50g, B) 100g, and C) 150g cohorts. Each individual patient is designated with letter, symbol, and color codes; several patients participated in multiple cohorts. Patient F was diagnosed with HbSß° thalassemia; all other patients were diagnosed with HbSS. Patients taking hydroxyurea during the study period are denoted with an asterisk.

References

    1. Prevention CfDCa. Sickle Cell Disease: Data & Statistics 16 September 2011 Accessed: 18 April 2015.
    1. Pauling L, Itano HA, Singer SJ, Wells IC. Sickle cell anemia, a molecular disease. Science. 1949;109(2835):443 .
    1. Kaul DK, Tsai HM, Liu XD, Nakada MT, Nagel RL, Coller BS. Monoclonal antibodies to alphaVbeta3 (7E3 and LM609) inhibit sickle red blood cell-endothelium interactions induced by platelet-activating factor. Blood. 2000;95(2):368–74. .
    1. Zennadi R, Moeller BJ, Whalen EJ, Batchvarova M, Xu K, Shan S, et al. Epinephrine-induced activation of LW-mediated sickle cell adhesion and vaso-occlusion in vivo. Blood. 2007;110(7):2708–17. 10.1182/blood-2006-11-056101
    1. Elmariah H, Garrett ME, De Castro LM, Jonassaint JC, Ataga KI, Eckman JR, et al. Factors associated with survival in a contemporary adult sickle cell disease cohort. American journal of hematology. 2014;89(5):530–5. 10.1002/ajh.23683
    1. Rees DC, Williams TN, Gladwin MT. Sickle-cell disease. Lancet. 2010;376(9757):2018–31. Epub 2010/12/07. 10.1016/S0140-6736(10)61029-X .
    1. Nur E, Biemond BJ, Otten HM, Brandjes DP, Schnog JJ, Group CS. Oxidative stress in sickle cell disease; pathophysiology and potential implications for disease management. American journal of hematology. 2011;86(6):484–9. Epub 2011/05/06. 10.1002/ajh.22012 .
    1. Kato GJ, McGowan V, Machado RF, Little JA, Taylor JT, Morris CR, et al. Lactate dehydrogenase as a biomarker of hemolysis-associated nitric oxide resistance, priapism, leg ulceration, pulmonary hypertension, and death in patients with sickle cell disease. Blood. 2006;107(6):2279–85. 10.1182/blood-2005-06-2373
    1. Schaer DJ, Buehler PW, Alayash AI, Belcher JD, Vercellotti GM. Hemolysis and free hemoglobin revisited: exploring hemoglobin and hemin scavengers as a novel class of therapeutic proteins. Blood. 2013;121(8):1276–84. Epub 2012/12/25. 10.1182/blood-2012-11-451229
    1. Hebbel RP, Eaton JW, Balasingam M, Steinberg MH. Spontaneous oxygen radical generation by sickle erythrocytes. The Journal of clinical investigation. 1982;70(6):1253–9.
    1. Schacter L, Warth JA, Gordon EM, Prasad A, Klein BL. Altered amount and activity of superoxide dismutase in sickle cell anemia. Faseb J. 1988;2(3):237–43. .
    1. Morris CR, Suh JH, Hagar W, Larkin S, Bland DA, Steinberg MH, et al. Erythrocyte glutamine depletion, altered redox environment, and pulmonary hypertension in sickle cell disease. Blood. 2008;111(1):402–10. Epub 2007/09/13. 10.1182/blood-2007-04-081703
    1. Jaiswal AK. Nrf2 signaling in coordinated activation of antioxidant gene expression. Free radical biology & medicine. 2004;36(10):1199–207. Epub 2004/04/28. 10.1016/j.freeradbiomed.2004.02.074 .
    1. Venugopal R, Jaiswal AK. Nrf1 and Nrf2 positively and c-Fos and Fra1 negatively regulate the human antioxidant response element-mediated expression of NAD(P)H:quinone oxidoreductase1 gene. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(25):14960–5. Epub 1996/12/10.
    1. Sangokoya C, Telen MJ, Chi JT. microRNA miR-144 modulates oxidative stress tolerance and associates with anemia severity in sickle cell disease. Blood. 2010;116(20):4338–48. 10.1182/blood-2009-04-214817
    1. Lee JM, Chan K, Kan YW, Johnson JA. Targeted disruption of Nrf2 causes regenerative immune-mediated hemolytic anemia. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(26):9751–6. .
    1. Charache S, Barton FB, Moore RD, Terrin ML, Steinberg MH, Dover GJ, et al. Hydroxyurea and sickle cell anemia. Clinical utility of a myelosuppressive "switching" agent. The Multicenter Study of Hydroxyurea in Sickle Cell Anemia. Medicine. 1996;75(6):300–26. .
    1. Steinberg MH, Barton F, Castro O, Pegelow CH, Ballas SK, Kutlar A, et al. Effect of hydroxyurea on mortality and morbidity in adult sickle cell anemia: risks and benefits up to 9 years of treatment. Jama. 2003;289(13):1645–51. .
    1. Akinsheye I, Alsultan A, Solovieff N, Ngo D, Baldwin CT, Sebastiani P, et al. Fetal hemoglobin in sickle cell anemia. Blood. 2011;118(1):19–27. Epub 2011/04/15. 10.1182/blood-2011-03-325258
    1. Voskaridou E, Christoulas D, Bilalis A, Plata E, Varvagiannis K, Stamatopoulos G, et al. The effect of prolonged administration of hydroxyurea on morbidity and mortality in adult patients with sickle cell syndromes: results of a 17-year, single-center trial (LaSHS). Blood. 2010;115(12):2354–63. Epub 2009/11/12. 10.1182/blood-2009-05-221333 .
    1. Macari ER, Lowrey CH. Induction of human fetal hemoglobin via the NRF2 antioxidant response signaling pathway. Blood. 2011;117(22):5987–97. 10.1182/blood-2010-10-314096
    1. Cipolla BG, Mandron E, Lefort JM, Coadou Y, Della Negra E, Corbel L, et al. Effect of Sulforaphane in Men with Biochemical Recurrence after Radical Prostatectomy. Cancer prevention research. 2015. Epub 2015/05/15. 10.1158/1940-6207.CAPR-14-0459 .
    1. Gao B, Doan A, Hybertson BM. The clinical potential of influencing Nrf2 signaling in degenerative and immunological disorders. Clinical pharmacology: advances and applications. 2014;6:19–34. Epub 2014/02/13. 10.2147/CPAA.S35078
    1. Pergola PE, Raskin P, Toto RD, Meyer CJ, Huff JW, Grossman EB, et al. Bardoxolone methyl and kidney function in CKD with type 2 diabetes. The New England journal of medicine. 2011;365(4):327–36. 10.1056/NEJMoa1105351 .
    1. Zhang Y, Talalay P, Cho CG, Posner GH. A major inducer of anticarcinogenic protective enzymes from broccoli: isolation and elucidation of structure. Proceedings of the National Academy of Sciences of the United States of America. 1992;89(6):2399–403.
    1. Riedl MA, Saxon A, Diaz-Sanchez D. Oral sulforaphane increases Phase II antioxidant enzymes in the human upper airway. Clinical immunology. 2009;130(3):244–51. Epub 2008/11/26. 10.1016/j.clim.2008.10.007
    1. Clarke JD, Hsu A, Riedl K, Bella D, Schwartz SJ, Stevens JF, et al. Bioavailability and inter-conversion of sulforaphane and erucin in human subjects consuming broccoli sprouts or broccoli supplement in a cross-over study design. Pharmacological research: the official journal of the Italian Pharmacological Society. 2011;64(5):456–63. 10.1016/j.phrs.2011.07.005
    1. Noah TL, Zhang H, Zhou H, Glista-Baker E, Muller L, Bauer RN, et al. Effect of broccoli sprouts on nasal response to live attenuated influenza virus in smokers: a randomized, double-blind study. PloS one. 2014;9(6):e98671 Epub 2014/06/10. 10.1371/journal.pone.0098671
    1. Meyer M, Kesic MJ, Clarke J, Ho E, Simmen RC, Diaz-Sanchez D, et al. Sulforaphane induces SLPI secretion in the nasal mucosa. Respiratory medicine. 2013;107(3):472–5. 10.1016/j.rmed.2012.11.006
    1. Shapiro TA, Fahey JW, Wade KL, Stephenson KK, Talalay P. Chemoprotective glucosinolates and isothiocyanates of broccoli sprouts: metabolism and excretion in humans. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2001;10(5):501–8. .
    1. Atwell LL, Hsu A, Wong CP, Stevens JF, Bella D, Yu TW, et al. Absorption and chemopreventive targets of sulforaphane in humans following consumption of broccoli sprouts or a myrosinase-treated broccoli sprout extract. Molecular nutrition & food research. 2015;59(3):424–33. Epub 2014/12/19. 10.1002/mnfr.201400674
    1. Okpala I. The intriguing contribution of white blood cells to sickle cell disease—a red cell disorder. Blood reviews. 2004;18(1):65–73. Epub 2003/12/20. .
    1. Nath KA, Hebbel RP. Sickle cell disease: renal manifestations and mechanisms. Nature reviews Nephrology. 2015;11(3):161–71. Epub 2015/02/11. 10.1038/nrneph.2015.8 .
    1. Pergola PE, Raskin P, Toto RD, Meyer CJ, Huff JW, Grossman EB, et al. Bardoxolone Methyl and Kidney Function in CKD with Type 2 Diabetes. N Engl J Med. 2011. Epub 2011/06/28. 10.1056/NEJMoa1105351 .
    1. Pergola PE, Krauth M, Huff JW, Ferguson DA, Ruiz S, Meyer CJ, et al. Effect of bardoxolone methyl on kidney function in patients with T2D and Stage 3b-4 CKD. American journal of nephrology. 2011;33(5):469–76. Epub 2011/04/22. 10.1159/000327599 .
    1. de Zeeuw D, Akizawa T, Audhya P, Bakris GL, Chin M, Christ-Schmidt H, et al. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. The New England journal of medicine. 2013;369(26):2492–503. 10.1056/NEJMoa1306033 .
    1. Macari ER, Schaeffer EK, West RJ, Lowrey CH. Simvastatin and t-butylhydroquinone suppress KLF1 and BCL11A gene expression and additively increase fetal hemoglobin in primary human erythroid cells. Blood. 2013;121(5):830–9. Epub 2012/12/12. 10.1182/blood-2012-07-443986
    1. Promsote W, Makala L, Li B, Smith SB, Singh N, Ganapathy V, et al. Monomethylfumarate induces gamma-globin expression and fetal hemoglobin production in cultured human retinal pigment epithelial (RPE) and erythroid cells, and in intact retina. Investigative ophthalmology & visual science. 2014;55(8):5382–93. 10.1167/iovs.14-14179
    1. Wong CP, Hsu A, Buchanan A, Palomera-Sanchez Z, Beaver LM, Houseman EA, et al. Effects of sulforaphane and 3,3'-diindolylmethane on genome-wide promoter methylation in normal prostate epithelial cells and prostate cancer cells. PloS one. 2014;9(1):e86787 Epub 2014/01/28. 10.1371/journal.pone.0086787
    1. Gibbs A, Schwartzman J, Deng V, Alumkal J. Sulforaphane destabilizes the androgen receptor in prostate cancer cells by inactivating histone deacetylase 6. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(39):16663–8. Epub 2009/10/07. 10.1073/pnas.0908908106
    1. Singh K, Connors SL, Macklin EA, Smith KD, Fahey JW, Talalay P, et al. Sulforaphane treatment of autism spectrum disorder (ASD). Proceedings of the National Academy of Sciences of the United States of America. 2014;111(43):15550–5. 10.1073/pnas.1416940111
    1. Probst BL, Trevino I, McCauley L, Bumeister R, Dulubova I, Wigley WC, et al. RTA 408, A Novel Synthetic Triterpenoid with Broad Anticancer and Anti-Inflammatory Activity. PloS one. 2015;10(4):e0122942 Epub 2015/04/22. 10.1371/journal.pone.0122942 .
    1. Goldman DC, Alexeev V, Lash E, Guha C, Rodeck U, Fleming WH. The Triterpenoid RTA 408 is a Robust Mitigator of Hematopoietic Acute Radiation Syndrome in Mice. Radiation research. 2015;183(3):338–44. Epub 2015/03/05. 10.1667/RR13900.1 .

Source: PubMed

3
订阅