Scolaris Content Display Scolaris Content Display

Hidroxiurea (hidroxicarbamida) para la anemia de células falciformes

This is not the most recent version

Collapse all Expand all

References

BABY HUG 2011 {published data only}

Adams RJ, Barredo J, Bonds DR, Brown C, Casella J, Daner L, et al. TCD in infants: A report from the BABY HUG trial. Blood 2005;106(11). [Abstract no: 952]CENTRAL
Adams RJ, Luden J, Miller S, Wang W, Rees R, Li D, et al. TCD in infants: a report from the Baby Hug study. 28th Annual Meeting of the National Sickle Cell Disease Program; 2005 Apr 9‐13; Cincinnati, Ohio. 2005:105. CENTRAL
Alvarez O, Miller ST, Wang WC, Luo Z, McCarville MB, Schwartz GJ, et al. Effect of hydroxyurea treatment on renal function parameters: results from the multi‐center placebo‐controlled BABY HUG clinical trial for infants with sickle cell anemia. Pediatric Blood & Cancer 2012;59(4):668‐74. [CENTRAL: 848700; CRS: 5500125000000525; PUBMED: 22294512]CENTRAL
Armstrong FD, Elkin TD, Brown RC, Glass P, Rana S, Casella JF, et al. Developmental function in toddlers with sickle cell anemia. Pediatrics 2013;131(2):e406‐14. [CENTRAL: 853612; CRS: 5500125000000530; PUBMED: 23296434]CENTRAL
Armstrong FD, Elkin TD, Brown RC, Glass P, Rees RC, Wang WC, et al. Neurodevelopment in infants with sickle cell anemia: baseline data from the Baby HUG trial. Blood 2008;112(11):713. CENTRAL
Armstrong FD, Rees RC, Li D, Bonner M, Elkin D, Strouse JJ, et al. Baseline developmental function by age for children in the pediatric hydroxyurea phase 3 clinical trial (Baby Hug). 28th Annual Meeting of the National Sickle Cell Disease Program; 2005 Apr 9‐13; Cincinnati, Ohio. 2005:137. CENTRAL
Casella JF, Wang WC, Rogers ZR, Iyer RV, Rana S, Driscoll MC, et al. Progress of the multicenter trial of hydroxyurea in infants with sickle cell anemia (BABY HUG) and assessment of baseline splenic and renal function. Pediatric Academic Societies Annual Meeting. 2005; Vol. 57:1111. CENTRAL
Kalpatthi R, Thompson B, Lu M, Wang WC, Patel N, Kutlar A, et al. Comparison of hematologic measurements between local and central laboratories: data from the BABY HUG trial. Clinical Biochemistry 2013;46(3):278‐81. [CENTRAL: 977455; CRS: 5500125000000527; PUBMED: 23123915]CENTRAL
Lebensburger JD, Miller ST, Howard TH, Casella JF, Brown RC, Lu M, et al. Influence of hemoglobin level on clinical findings in infants with sickle cell anemia; data from BABY HUG. 52nd ASH Meeting and Exposition; 2010 Dec 4‐7; Orlando. 2010. [Abstract no: 1631]CENTRAL
Lebensburger JD, Miller ST, Howard TH, Casella JF, Brown RC, Lu M, et al. Influence of severity of anemia on clinical findings in infants with sickle cell anemia: analyses from the BABY HUG study. Pediatric Blood & Cancer 2012;59(4):675‐8. [CENTRAL: 854381; CRS: 5500125000000528; PUBMED: 22190441]CENTRAL
Lederman HM, Connolly MA, Kalpatthi R, Ware RE, Wang WC, Luchtman‐Jones L, et al. Immunologic effects of hydroxyurea in sickle cell anemia. Pediatrics 2014;134(4):686‐95. [CENTRAL: 1053679; CRS: 5500135000000943; PUBMED: 25180279]CENTRAL
Lederman HM, Connolly MA, Ware RE, Luchtman‐Jones L, Goldsmith JC. Effects of hydroxyurea (HU) on lymphocyte subsets and the immune response to pneumococcal, measles, mumps and rubella vaccination in the pediatric hydroxyurea phase III clinical trial ‐ BABY HUG ‐ (ClinicalTrials.gov Identifier: NCT00006400). Blood 2012;120(21). [Abstract no: 243; CENTRAL: 977456; CRS: 5500125000000532]CENTRAL
McCarville MB, Luo Z, Huang X, Rees RC, Rogers ZR, Miller ST, et al. Abdominal ultrasound with scintigraphic and clinical correlates in infants with sickle cell anemia: baseline data from the BABY HUG trial. AJR American Journal of Roentgenology 2011;196(6):1399‐404. CENTRAL
McCarville MB, Rees RC, Rogers ZR, Kalpatthi R, Miller ST, Wang WC, et al. Adbominal ultrasound findings in infants with sickle cell anemia; baseline data from the BABY HUG Trial. 3rd Annual Sickle Cell Disease Research and Educational Symposium and Annual Sickle Cell Disease Scientific Meeting; 2009 Feb 18‐20. 2009. [Abstract no: 212]CENTRAL
McGann PT, Flanagan JM, Howard TA, Dertinger SD, He J, Kulharya AS, et al. Genotoxicity associated with hydroxyurea exposure in infants with sickle cell anemia: results from BABY‐HUG phase III clinical trial. 53rd ASH Annual Meeting and Exposition; 2011 Dec 10‐13; San Diego, California. 2011. [Abstract no: 8]CENTRAL
McGann PT, Flanagan JM, Howard TA, Dertinger SD, He J, Kulharya AS, et al. Genotoxicity associated with hydroxyurea exposure in infants with sickle cell anemia: results from the BABY‐HUG Phase III Clinical Trial. Pediatric Blood & Cancer 2012;59(2):254‐7. [CENTRAL: 854422; CRS: 5500125000000524; PUBMED: 22012708]CENTRAL
Miller ST, Barredo J, Brown C, Bonds DR, Casella JF, Li D, et al. Renal concentrating ability in infants with sickle cell anemia; baseline data from Baby Hug, a multicenter trial. 29th Annual Meeting of the National Sickle Cell Disease Program; 2006 Apr 8‐12; Memphis, USA. 2006. [Abstract no: 141]CENTRAL
Miller ST, Rey K, He J, Flanagan J, Fish BJ, Rogers ZR, et al. Massive accidental overdose of hydroxyurea in a young child with sickle cell anemia. Pediatric Blood & Cancer 2012;59(1):170‐2. CENTRAL
Miller ST, Wang WC, Iyer R, Rana S, Lane P, Ware RE, et al. Urine concentrating ability in infants with sickle cell disease: baseline data from the phase III trial of hydroxyurea (BABY HUG). Pediatric Blood & Cancer2010; Vol. 54, issue 2:265‐8. CENTRAL
Miller ST, Wang WC, Iyer RV, Rana SR, Lane PA, Ware RE, et al. Urine concentrating ability in infants with sickle cell anemia: baseline data from the Baby HUG trial. Blood 2008;112(11):1413. CENTRAL
Miller ST, Ware RE, Kutlar A, Alvarez OA, Iyer RV, Sarnaik SA, et al. Serum cystatin‐C levels in infants with sickle cell anemia: baseline data from the BABY HUG trial. Blood 2008;112(11):4791. CENTRAL
Pavlakis SG, Rees RC, Huang X, Brown RC, Casella JF, Iyer RV, et al. Transcranial doppler ultrasonography (TCD) in infants with sickle cell anemia: baseline data from the BABY HUG trial. Pediatric Blood & Cancer2010; Vol. 54, issue 2:256‐9. CENTRAL
Rana S, Houston PE, Wang WC, Iyer RV, Goldsmith J, Casella JF, et al. Hydroxyurea and growth in young children with sickle cell disease. Pediatrics 2014;134(3):465‐472. Supplemental information. http://pediatrics.aappublications.org/content/134/3/465.supplemental. [CRS: 5500135000001451]CENTRAL
Rana S, Houston PE, Wang WC, Iyer RV, Goldsmith J, Casella JF, et al. Hydroxyurea and growth in young children with sickle cell disease. Pediatrics 2014;134(3):465‐72. [CRS: 5500135000000945; PUBMED: 25157002]CENTRAL
Rogers ZR, Capparelli EV, Thompson B, Ware RE, Wang WC, Iyer RV, et al. Pharmacokinetics of hydroxyurea in young children with sickle cell anemia: a report from the Baby Hug trial. 29th Annual Meeting of the National Sickle Cell Disease Program; 2006 Apr 8‐12; Memphis, USA. 2006:157. CENTRAL
Rogers ZR, Rees RC, Files B, Iyer RV, Shulkin BL, Shalaby‐Rana E, et al. Spleen function in infants with sickle cell anemia : baseline data from the BABY HUG trial. Blood 2008;112(11):1416. CENTRAL
Rogers ZR, Rees RC, Files B, Iyer RV, Shulkin BL, Shalaby‐Rana E, et al. Spleen function in infants with sickle cell anemia: baseline data from the Baby Hug trial. 3rd Annual Sickle Cell Disease Research and Educational Symposium and Annual Sickle Cell Disease Scientific Meeting; 2009 Feb 18‐20. 2009. [Abstract no: 199]CENTRAL
Rogers ZR, Rees RR, Wang WC, Li D, Iyer RV, Rana S, et al. Evaluation of splenic function in infants with sickle cell anemia in the Baby Hug trial. 28th Annual Meeting of the National Sickle Cell Disease Program; 2005 Apr 9‐13; Cincinnati, Ohio. 2005:106. CENTRAL
Rogers ZR, Thompson B, Ware RE, Wang WC, Iyer RV, Miller ST, et al. Pharmacokinetics of hydroxyurea in young children with sickle cell anemia: a report from the BABY HUG trial. Blood. 2005, issue 11. [Abstract no:: 3184]CENTRAL
Sheehan VA, Luo Z, Flanagan JM, Howard TA, Thompson BW, Wang WC, et al. Genetic modifiers of sickle cell anemia in the BABY HUG cohort: influence on laboratory and clinical phenotypes. American Journal of Hematology 2013;88(7):571‐6. [CENTRAL: 983421; CRS: 5500125000000710; PUBMED: 23606168]CENTRAL
Thompson BW, Miller ST, Rogers ZR, Rees RC, Ware RE, Waclawiw MA, et al. The pediatric hydroxyurea phase III clinical trial (BABY HUG): challenges of study design. Pediatric Blood & Cancer2010; Vol. 54, issue 2:250‐5. CENTRAL
Thompson BW, Wang WC, Miller ST, Rogers ZR, Ware RE, Thornburg CD, et al. The physiological and clinical effects of interrupting a treatment regimen of hydroxyurea in young children with sickle cell anemia (SCA). 53rd ASH Annual Meeting and Exposition; 2011 Dec 10‐13; San Diego. 2011. [Abstract no: 2134]CENTRAL
Thornburg CD, Files BA, Luo Z, Miller ST, Kalpatthi R, Iyer R, et al. Impact of hydroxyurea on clinical events in the BABY HUG trial. Blood 2012;120(22):4304‐10; quiz 4448. [CENTRAL: 853818; CRS: 5500125000000526; PUBMED: 22915643]CENTRAL
Thornburg CD, Rogers ZR, Jeng MR, Rana SR, Iyer RV, Faughnan L, et al. Adherence to study medication and visits: data from the BABY HUG trial. Pediatric Blood & Cancer2010; Vol. 54, issue 2:260‐4. CENTRAL
Thornburg CD, Rogers ZR, Wang W, Jeng M, Rana SR, Iyer RV, et al. Study drug and visit adherence: data from the Baby HUG trial. Blood 2008;112(11):1275. CENTRAL
Wang W, Luo Z, Alvarez O, Fixler J, Miller S, Ware RE, et al. Effects of hydroxyurea in asymptomatic infants with sickle cell anemia: Analysis F from the BABY HUG trial. American Journal of Hematology 2012;7:E20‐E21. CENTRAL
Wang W, Rees RC, Miller ST, Brown RC, Casella JF, Iyer RV, et al. Transcranial doppler (TCD) ultrasonography in infants with sickle cell anemia: baseline data from the BABY HUG trial. Blood 2008;112(11):1436. CENTRAL
Wang WC, Oyeku SO, Luo Z, Boulet SL, Miller ST, Casella JF, et al. Hydroxyurea is associated with lower costs of care of young children with sickle cell anemia. Pediatrics 2013;132(4):677‐83. [CENTRAL: 962768; CRS: 5500125000000529; PUBMED: 23999955]CENTRAL
Wang WC, Ware RE, Miller ST, Iyer RV, Casella JF, Minniti CP, et al. Hydroxycarbamide in very young children with sickle‐cell anaemia: a multicentre, randomised, controlled trial (BABY HUG). Lancet 2011;377(9778):1663‐72. CENTRAL
Wang WC, Yeku SO, Luo Z, Boulet SL, Miller ST, Fish B, et al. Costs associated with the care of very young children with sickle cell anemia (SCA): analysis from the BABY HUG study. 53rd ASH Annual Meeting and Exposition; 2011 Dec 10‐13; San Diego. 2011. [Abstract no: 171]CENTRAL
Ware RE, Rees RC, Sarnaik SA, Iyer RV, Alvarez OA, Casella JF, et al. Renal function in infants with sickle cell anemia: baseline data from the BABY HUG trial. Blood 2008;112(11):1414. CENTRAL
Ware RE, Rees RC, Sarnaik SA, Iyer RV, Alvarez OA, Casella JF, et al. Renal function in infants with sickle cell anemia: baseline data from the BABY HUG trial. Journal of Pediatrics2010; Vol. 156, issue 1:66‐70. CENTRAL
Wynn L, Debenham E, Faughnan L, Martin B, Kelly T, Reed C, et al. Recruitment in the Baby Hug pediatric hydroxyurea phase 3 clinical trial. 35th Anniversary Convention of the National Sickle Cell Disease Program; 2007 Sep 17‐22; Washington DC, USA. 2007:245. CENTRAL
Wynn L, Miller S, Faughnan L, Luo Z, Debenham E, Adix L, et al. Recruitment of infants with sickle cell anemia to a Phase III trial: data from the BABY HUG study. Contemporary Clinical Trials2010; Vol. 31, issue 6:558‐63. CENTRAL
Wynn LW, Faughnan L, Li D, Wang W, Martin B, Kelly T, et al. Recruitment of infants with sickle cell anemia to a phase III trials: data from the BABY HUG study. Blood 2008;112(11):1429. CENTRAL

Belgian Study 1996 {published data only}

Ferster A, Vermylen C, Cornu G, Buyse M, Corazza F, Devalck C, et al. Hydroxyurea for treatment of severe sickle cell anemia: a pediatric clinical trial. Blood 1996;88(6):1960‐4. CENTRAL

CHAMPS 2011 {published data only}

Wang W, Brugnara C, Snyder C, Wynn L, Rogers Z, Kalinyak K, et al. The effects of hydroxycarbamide and magnesium on haemoglobin SC disease: results of the multi‐centre CHAMPS trial. British Journal of Haematology 2011;152(6):771‐6. [CENTRAL: 801819; CRS: 5500125000000521; PUBMED: 21275961]CENTRAL
Wang WC, Snyder C, Brugnara C, Telen MJ, Steinberg MH, Wynn LW, et al. Effects of hydroxyurea (HU) and magnesium pidolate (Mg) in hemoglobin SC disease (HbSC): the "CHAMPS" trial. Blood2009; Vol. 22. [Abstract no: 819]CENTRAL

Jain 2012 {published data only}

Jain D. Low dose hydroxyurea in children severely affected with sickle cell disease: hospital based randomized controlled study. 4th Annual Sickle Cell Disease Research and Educational Symposium & Grant Writing Institute AND Annual Sickle Cell Disease Scientific Meeting; 2010 Feb 14‐19; Hollywood, Florida. 2010. [Abstract no: 076]CENTRAL
Jain DL, Sarathi V, Desai S, Bhatnagar M, Lodha A. Low fixed‐dose hydroxyurea in severely affected Indian children with sickle cell disease. Hemoglobin 2012;36(4):323‐32. [CENTRAL: 879848; CRS: 5500125000000522; PUBMED: 22734586]CENTRAL

MSH 1995 {published data only}

Armstrong FD, Steinberg MH, Ballas SK, Ataga KI, Waclawiw MA, Kutlar A, et al. Development outcomes of offspring of adults treated with hydroxyurea in the multicenter study of hydroxyurea. Blood2009, issue 22. [Abstract no: 1543]CENTRAL
Ballas SK, Barton F, Castro O, Bellevue R, Investigators of the multicenter study of hydroxyurea in sickle cell anemia. Narcotic analgesia use among adult patients with sickle cell anemia. Blood 1995;86(10 Suppl 1):642a. CENTRAL
Ballas SK, Barton F, Castro O, Koshy M, Bellevue R. Pattern of narcotic analgesic consumption among adult patients with sickle cell anemia. National Sickle Cell Disease Program 21st Annual Meeting; 1996 Mar. 1996:63. CENTRAL
Ballas SK, Barton FB, Waclawiw MA, Swerdlow P, Eckman JR, Pegelow CH, et al. Hydroxyurea and sickle cell anemia: effect on quality of life. Health and Quality of Life Outcomes 2006;4:59. CENTRAL
Ballas SK, Bauserman RL, McCarthy WF, Castro OL, Smith WR, Waclawiw MA. Utilization of analgesics in the multicenter study of hydroxyurea in sickle cell anemia: Effect of sex,age, and geographical location. American Journal of Hematology 2010;85(8):613‐6. CENTRAL
Ballas SK, Bauserman RL, McCarthy WF, Castro OL, Smith WR, Waclawiw MA, IoftheMSofHinSCA. Hydroxyurea and acute painful crises in sickle cell anemia: effects on hospital length of stay and opioid utilization during hospitalization, outpatient acute care contacts, and at home. Journal of Pain and Symptom Management 2010;40(6):870‐82. [CENTRAL: 779191; CRS: 5500135000001680; PUBMED: 20864308]CENTRAL
Ballas SK, Bauserman RL, McCarthy WF, Waclawiw MA. The impact of hydroxyurea on career and employment of patients with sickle cell anemia. JAMA2010; Vol. 102, issue 11:993‐9. CENTRAL
Ballas SK, Bauserman RL, McCarthy WF, Waclawiw MA, Barton BA. Impact of hydroxyurea on employment among patients with sickle cell anemia. Blood2009; Vol. 114, issue 22. [Abstract no: 2485]CENTRAL
Ballas SK, Marcolina MJ, Dover GJ, Barton FB. Erythropoietic activity in patients with sickle cell anaemia before and after treatment with hydroxyurea. British Journal of Haematology 1999;105(2):491‐6. CENTRAL
Ballas SK, Marcolina MJ, Investigators of the multicenter study of hydroxyurea in sickle cell anemia. In vivo RBC survival and ferrokinetic data in patients with sickle cell anemia before and after treatment with hydroxyurea. Blood 1995;86(10 Suppl 1):140a. CENTRAL
Ballas SK, McCarthy WF, Bauseman RI, Castro OL, Swerdlow PS, Smith W, et al. Patterns of analgesic utilization in the multicenter study of hydroxyurea (MSH). Blood2009; Vol. 114, issue 22. [Abstract no: 2577]CENTRAL
Ballas SK, McCarthy WF, Bauserman RL, Castro OL, Waclawiw MA, Barton BA. Sickle cell genetic markers: geographic distribution and relation to pain outcomes in multicenter study of hydroxyurea in sickle cell anemia. Blood2009; Vol. 114, issue 22. [Abstract no: 2582]CENTRAL
Ballas SK, McCarthy WF, Bauserman RL, Valafar F, Waclawiw M, Barton BA, Kutlar A. Definition of the responder to hydroxyurea therapy: revisited. Blood 2009;114(22). [Abstract no: 1513]CENTRAL
Ballas SK, McCarthy WF, Guo N, Brugnara C, Kling G, Bauserman R, et al. Early detection of responders to hydroxyurea therapy. 4th Annual Sickle Cell Disease Research and Educational Symposium & Grant Writing Institute AND Annual Sickle Cell Disease Scientific Meeting; 2010 Feb 14‐19; Hollywood, Florida. 2010; Vol. 26. [Abstract no: 030]CENTRAL
Ballas SK, McCarthy WF, Guo N, DeCastro L, Bellevue R, Barton BA, et al. Exposure to hydroxyurea and pregnancy outcomes in patients with sickle cell anemia. Journal of the National Medical Association 2009;101(10):1046‐51. [CENTRAL: 730469; CRS: 5500050000000074; PUBMED: 19860305]CENTRAL
Barton F, Terrin M, Moore R, McMahon RP, Charache S. Ascertainment of the primary end point in the Multicenter Study of Hydroxyrea in Sickle Cell Anemia (MSH). The MSH Investigators. Controlled Clinical Trials 1996;17(2 Suppl):67S. CENTRAL
Brandon AE, McCarthy WF, Barton FB, Terrin ML. Vital status determination of patients' lost to follow‐up in the multicenter study of hydroxyurea in sickle cell anemia (MSH) patients' follow‐up study. Clinical Trials 2004;2:209. CENTRAL
Charache S. Effects of hydroxyurea therapy in patients with sickle cell anemia. Australian and New Zealand Journal of Medicine 1996;26:326. CENTRAL
Charache S. Experimental therapy of sickle cell disease. Use of hydroxyurea. American Journal of Pediatric Hematology/Oncology 1994;16(1):62‐6. CENTRAL
Charache S. Mechanism of action of hydroxyurea in the management of sickle cell anemia in adults. Seminars in Hematology 1997;34(3 Suppl 3):15‐21. CENTRAL
Charache S. Preventing pain in sickle cell anemia (HB SS): baseline data from patients in a hydroxyurea trial. Blood 1993;82(10 Suppl):356a. CENTRAL
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. Medicine 1996;75(6):300‐26. CENTRAL
Charache S, Terrin M, Moore RD, Dover GJ, Barton FB, Eckert SV, et al. Effect of hydroxyurea on the frequency of painful crisis in sickle cell anemia. New England Journal of Medicine 1995;332(20):1317‐22. CENTRAL
Charache S, Terrin ML, Moore RD, Dover GJ, McMahon RP, Barton FB, et al. Design of the multicenter study of hydroxyurea in sickle cell anemia. Controlled Clinical Trials 1995;16(6):432‐46. CENTRAL
Hackney AC, Heizer W, Hoffman E, Jones S, Strayhorn D, Orringer EP. Effect of hydroxyurea (HU) administration on the body weight, body composition and exercise performance of patients with sickle cell anemia. Blood 1995;86(10 Suppl 1):141a. CENTRAL
Hackney AC, Hezier W, Gulledge TP, Jones S, Strayhorn D, Busby M, et al. Effects of hydroxyurea administration on the body weight, body composition and exercise performance of patients with sickle‐cell anaemia. Clinical Science 1997;92(5):481‐6. CENTRAL
Handy C, Barton F, Moore R, McMahon R, Eckert S, Terrin M. Dose titration in the multicentre study of hydroxyurea in sickle cell anemia (MSH). Controlled Clinical Trials 1996;17(Suppl 2):92S. CENTRAL
Heizer WD, Hackney AC, Busby M, Gulledge T, Jones S, Strayhorn G, et al. The composition and etiology of weight gain in sickle cell patients receiving hydroxyurea (HU): An ancillary study to the multicentre study of hydroxyurea (MSH). National Sickle Cell Disease Program 18th Annual Meeting; 1993 May. 1993:117a. CENTRAL
Kutlar A, Barton F, Terrin M, Steinberg MH. Effect of hydroxyurea on hematologic and biochemical laboratory values in sickle cell disease: The MSH at 7‐8 years follow‐up. National Sickle Cell Disease Program 25th Annual Meeting; 2001 Apr. 2001. [Abstract no: #126]CENTRAL
McCarthy WF, Bauserman RL, Barton BA, Guo N, Ballas SK, Smith W. Time series analysis of the pain diary data obtained during the multicenter study for hydroxyurea (MSH) clinical trial. Blood 2006;11. [Abstract no: 3807]CENTRAL
McMahon RP, Waclawiw MA, Geller NL, Barton FB, Terrin ML, Bonds DR. An extension of stochastic curtailment for incompletely reported events: the multicenter study of hydroxyurea in sickle cell anemia (MSH). Controlled Clinical Trials 1997;18(5):420‐30. CENTRAL
Moore RD, Charache S, Terrin M, Barton FB, Ballas SK. Cost‐effectiveness of hydroxyurea in sickle cell anemia. National Sickle Cell Disease Program 23rd Meeting; 1999 Mar. 1999:210. CENTRAL
Moore RD, Charache S, Terrin ML, Barton FB, Ballas SK, and the investigators of the MSH study of hydroxyurea in sickle cell anemia. Cost‐effectiveness of hydroxyurea in sickle cell anemia. American Journal of Hematology 2000;64(1):26‐31. CENTRAL
Orringer EP, Jones S, Strayhorn D, Hoffman E, Parker J, Greenberg C. The effect of hydroxyurea (HU) administration on circulating D‐dimer levels in patients with sickle cell anemia (HbSS). National Sickle Cell Disease Program 21st Meeting; 1996 Mar. 1996:131. CENTRAL
Orringer EP, Jones S, Strayhorn D, Hoffman E, Parker J, Greenberg CS. The effect of hydroxyurea (HU) administration on circulating d‐dimer levels in patients with sickle cell anemia. Blood 1996;88(10 Suppl 1):496a. CENTRAL
Smith WR, Ballas SK, McCarthy WF, Bauserman RL, Swerdlow PS, Steinberg MH. The association between hydroxyurea treatment and pain intensity, analgesic use,  and utilization in ambulatory sickle cell anemia patients. Pain Medicine 2011;12(5):697‐705. CENTRAL
Smith WR, Bauseman RL, McCarthy WF, Barton BA, Ballas SK. Effect of geography and climate on pain frequency in patients enrolled in the multicenter study of hydroxyurea in sickle cell anemia. 3rd Annual Sickle Cell Disease Research and Educational Symposium AND Annual Sickle Cell Disease Scientific Meeting; 2009 Feb 18‐20. 2009. [Abstract no: 253]CENTRAL
Steinberg MH. Determinants of fetal hemoglobin response to hydroxyurea. Seminars in Hematology 1997;3(Suppl 3):8‐14. CENTRAL
Steinberg MH. Mortality at 3‐5 years: The multicenter study of hydroxyurea in sickle cell anemia (MSH). National Sickle Cell Disease Program Annual Meeting; 1997 Sep. 1997:68. CENTRAL
Steinberg MH, Ballas S, Barton F, Terrin M, the MSH. Mortality at 4‐5 years: results from the multicenter study of hydroxyurea in sickle cell anemia (MSH). Blood 1997;90(10 Suppl 1 Pt 1):444a. CENTRAL
Steinberg MH, Barton F, Castro O, Koshy M, Eckman J, Terrin M. Risks and benefits of hydroxyurea (HU) in adult sickle cell anaemia. Effects at 6‐ to 7‐ years. Blood 1999;94(10 Suppl 1 Pt 1):644a‐5a. CENTRAL
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. CENTRAL
Steinberg MH, Barton F, Castro O, Ramirez G, Bellevue R, Terrin M, et al. Hydroxyurea (HU) is associated with reduced mortality in adults with sickle cell anemia. Blood 2000;96(11 Pt 1):485a. CENTRAL
Steinberg MH, Castro O, Ballas SK, Barton F, Terrin M. The multicenter study of hydroxyurea in sickle cell anemia (MSH): mortality at 5‐6 years. Blood 1998;92(10 Suppl 1 Pt 1):496a. CENTRAL
Steinberg MH, Lu ZH, Barton FB, Terrin ML, Charache S, Dover GJ. Fetal hemoglobin in sickle cell anemia: determinants of response to hydroxyurea. Blood 1997;89(3):1078‐8. CENTRAL
Steinberg MH, Lu ZH, Barton M, Terrin S, Charache S, Dover G, et al. Fetal hemoglobin (Hb F) in sickle cell anemia (HbSS): Determinents of response to hydroxyurea (HU). Blood 1995;86(10 Suppl 1):418a. CENTRAL
Steinberg MH, McCarthy WF, Castro O, Ballas SK, Armstrong FD, Smith W, et al. The risks and benefits of long‐term use of hydroxyurea in sickle cell anemia: A 17.5 year follow‐up. American Journal of Hematolology2010; Vol. 85, issue 6:403‐8. CENTRAL
Terrin ML, Barton FB, Bonds D, Ballas SK, Swerdlow P, Pegelow CH, et al. Effect of hydroxyurea on quality of life: 2‐year results from the multicenter study of hydroxyurea in sickle cell anemia. National Sickle Cell Disease Program 23rd Annual Meeting; 1999 Mar. 1999:161. CENTRAL

SCATE 2015 {published data only}

Hankins JS, McCarville MB, Rankine‐Mullings A, Reid ME, Lobo CL, Moura PG, et al. Prevention of conversion to abnormal transcranial Doppler with hydroxyurea in sickle cell anemia: a phase III international randomized clinical trial. American Journal of Hematology 2015;90(12):1099‐105. [DOI: 10/1002/ajh.24198]CENTRAL
NCT01531387. Sparing Conversion to Abnormal TCD (Transcranial Doppler) Elevation (SCATE). http://clinicaltrials.gov/show/NCT01531387. CENTRAL

SWiTCH 2012 {published data only}

Alvarez O, Yovetich NA, Scott JP, Owen W, Miller ST, Schultz W, et al. Pain and other non‐neurological adverse events in children with sickle cell anemia and previous stroke who received hydroxyurea and phlebotomy or chronic transfusions and chelation: results from the SWiTCH clinical trial. American Journal of Hematology 2013;88(11):932‐8. [CENTRAL: 963136; CRS: 5500125000000520; PUBMED: 23861242]CENTRAL
Aygun B, Mortier NA, Kesler K, Lockhart A, Schultz WH, Cohen AR, et al. Therapeutic phlebotomy is safe in children with sickle cell anaemia and can be effective treatment for transfusional iron overload. British Journal of Haematology 2015;169(2):262‐6. [CRS: 5500135000001418; PUBMED: 25612463]CENTRAL
Aygun B, Mortier NA, Kesler K, Schultz WH, Alvarez OA, Rogers ZR, et al. Therapeutice phlebotomy in children with sickle cell anemia, stroke, and iron overload: the SWiTCH experience. 53rd ASH Annual Meeting and Exposition; 2011 Dec 10‐13; San Diego, California. 2011. [Abstract no: 1044]CENTRAL
Kwiatkowski JL, Cohen AR, Garro J, Alvarez O, Nagasubramanian R, Sarnaik S, et al. Transfusional iron overload in children with sickle cell anemia on chronic transfusion therapy for secondary stroke prevention. American Journal of Hematology 2012;87(2):221‐3. [CENTRAL: 864015; CRS: 5500100000011226; PUBMED: 22120913]CENTRAL
NCT00122980. Stroke with transfusions changing to hydroxyurea. clinicaltrials.gov/show/NCT00122980 Date first received: 20 July 2005. CENTRAL
Sheehan VA, Howard TA, Sabo A, Nagasaswamy U, Crosby JR, Davis B, et al. Genetic predictors of hemoglobin F response to hydroxyurea in sickle cell anemia. Blood 2012;120(21). [Abstract no: 241; CENTRAL: 977454; CRS: 5500125000000531]CENTRAL
Ware RE, Helms RW. Stroke With Transfusions Changing to Hydroxyurea (SWiTCH). Blood 2012;119(17):3925‐32. [CENTRAL: 849022; CRS: 5500100000004120]CENTRAL
Ware RE, Helms RW. Stroke with transfusions changing to hydroxyurea (SWiTCH): a phase 3 randomised clinical trial for treatment of children with sickle cell anemia. 52nd ASH Meeting and Exposisiton; 2010 Dec 4‐7; Orlando, Florida. 2010. [Abstract no: 844]CENTRAL
Ware RE, McMurray MA, Schultz WH, Alvarez OA, Aygun B, Cavalier ME, et al. Academic community standards for chronic transfusion therapy in children with sickle cell anemia and stroke. Blood 2006;108(11):Abst 1213. CENTRAL
Ware RE, Schultz WH, Yovetich N, Mortier NA, Alvarez O, Hilliard L, et al. Stroke with transfusions changing to hydroxyurea (SWiTCH): A phase III randomized clinical trial for treatment of children with sickle cell anemia, stroke, and iron overload. Pediatric Blood & Cancer 2011;57(6):1011‐7. [CENTRAL: 806683; CRS: 5500100000004123]CENTRAL

TWiTCH 2016 {published data only}

Aygun B,  Wruck LM,  Schultz WH,  Mueller BU,  Brown C,  Luchtman‐Jones L,  et al. Chronic transfusion practices for prevention of primary stroke in children with sickle cell anemia and abnormal TCD velocities. American Journal of Hematology 2012;87(4):428‐30. CENTRAL
Helton KJ, Roberts D, Schultz WH, Davis BR, Kalfa TA, Pressel SL, et al. Effects of chronic transfusion therapy on MRI and MRA in children with sickle cell anemia. Blood 2014;124(21):4052. [CENTRAL: 1261883; CRS: 5500135000001683]CENTRAL
Imran H, Aygun B, Davis BR, Pressel SL, Herbert Schultz W, Jackson SM, et al. Effects of chronic transfusion therapy on transcranial doppler ultrasonography velocities in children with sickle cell anemia at risk for primary stroke: baseline findings from the Twitch trial. Blood 2014;124(21):87. [CENTRAL: 1261882; CRS: 5500135000001682]CENTRAL
Ware RE, Davis BR, Schultz WH, Brown RC, Aygun B, Sarnaik S, et al. Hydroxycarbamide versus chronic transfusion for maintenance of transcranial doppler flow velocities in children with sickle cell anaemia—TCD With Transfusions Changing to Hydroxyurea (TWiTCH): a multicentre, open‐label, phase 3, non‐inferiority trial. Lancet 2016;387:661‐70. CENTRAL
Wood JC, Cohen A, Aygun B, Imran H, Luchtman‐Jones L, Thompson. Extrahepatic iron deposition in chronically transfused children with sickle cell anemia‐ baseline findings from the Twitch trial. Blood 2013;122(21):2238. [CENTRAL: 1261881; CRS: 5500135000001681]CENTRAL
Wood JC, Cohen AR, Pressel SL, Aygun B, Imran H, Luchtman‐Jones L. Organ iron accumulation in chronically transfused children with sickle cell anaemia: baseline results from the TWiTCH trial. British Journal of Haematology 2016;172(1):122‐30. CENTRAL
Wood JC, Pressel S, Rogers ZR, Odame I, Kwiatkowski JL, Lee MT, et al. Liver iron concentration measurements by MRI in chronically transfused children with sickle cell anemia: baseline results from the TWiTCH trial. American Journal of Hematology 2015;90(9):806‐10. CENTRAL
Wood JC, Pressel S, Rogers ZR, Odame I, Kwiatkowski JL, Lee MT, et al. Liver iron concentration measurements by MRI in chronically transfused children with sickle cell anemia: Baseline results from the TWiTCH trial. American Journal of Hematology 2015;90(9):806‐10. [CENTRAL: 1090263; EMBASE: 2015311270; CRS: 5500050000000271]CENTRAL

Al‐Nood 2011 {published data only}

Al‐Nood HA, Al‐Khawlani MM, Al‐Akwa A. Fetal hemoglobin response to hydroxyurea in Yemeni sickle cell disease patients. Hemoglobin2011, issue 1:13‐21. CENTRAL

De Montalembert 2006 {published data only}

De Montalembert M, Bachir D, Hulin A, Gimeno L, Mogenet A, Bresson JL, et al. A phase 1 pharmacokinetics (PK) study of hydroxyurea (HU) 1,000 mg coated breakable tablets and 500mg capsules in pediatric and adult patients with sickle cell disease. Blood 2005;106(11):Abst 3194. [Abstract no: 3194]CENTRAL
de Montalembert M, Bachir D, Hulin A, Gimeno L, Mogenet A, Bresson JL, et al. Pharmacokinetics of hydroxyurea 1,000 mg coated breakable tablets and 500 mg capsules in pediatric and adult patients with sickle cell disease. Haematologica 2006;91(12):1685‐8. CENTRAL

George 2013 {published data only}

George A, Aygun B, Mortier N, Sparreboom A, Ware R. A randomized controlled trial of a dose‐prediction equation to determine maximum tolerated dose of hydroxyurea in children with sickle cell anemia. Pediatric Blood & Cancer 2013;60 Suppl. [Abstract no: 588; CENTRAL: 1007869; CRS: 5500050000000224; EMBASE: 71047876]CENTRAL

NCT00004492 {published data only}

NCT00004492. Phase I/II Randomized Study of Hydroxyurea With or Without Clotrimazole in Patients With Sickle Cell Anemia. https://clinicaltrials.gov/show/NCT00004492. CENTRAL

NCT01848925 {published data only}

NCT01848925. A Phase I Open‐label, Randomized, Safety and Efficacy Study of SANGUINATE™ at Two Doses Levels Versus Hydroxyurea in Sickle Cell Disease (SCD) Patients. clinicaltrials.gov/show/NCT01848925 (date first received 23 April 2013). CENTRAL

NCT01960413 {published data only}

NCT01960413. Phase 2 Study of Montelukast for the Treatment of Sickle Cell Anemia. https://clinicaltrials.gov/show/NCT01960413. CENTRAL

NCT02149537 {published data only}

NCT02149537. Risk Stratification for Clinical Severity of Sickle Cell Disease in Nigeria and Assessment of Efficacy and Safety During Treatment With Hydroxyurea. https://clinicaltrials.gov/show/NCT02149537. CENTRAL

Pushi 2000 {published data only}

Pushi A. Hydroxyurea ameliorates importantly the clinical course of sickle cell disease reducing the frequency of painful crises. Hematology Journal 2000;1(Suppl. 1):34, Abstract no: 130. CENTRAL

Silva‐Pinto 2007 {published data only}

Silva‐Pinto AC, Carrara RC, Oliveira VC, Palma PV, Campos AD, Zago MA, et al. Hydroxyurea treatment of sickle cell diseases causes megaloblastic transformation of the bone marrow that is responsible for the increase of the mean corpuscular volume. Haematologica 2007;Suppl 1:298. CENTRAL

Silva‐Pinto 2014 {published data only}

Silva‐Pinto AC, Dias‐Carlos C, Saldanha‐Araujo F, Ferreira FI, Palma PV, Araujo AG, et al. Hydroxycarbamide modulates components involved in the regulation of adenosine levels in blood cells from sickle‐cell anemia patients. Annals of Hematology 2014;93(9):1457‐65. [CENTRAL: 1000846; CRS: 5500131000000084; JID:: 9107334; PUBMED: 24696091]CENTRAL

Vichinsky 2013 {published data only}

Vichinsky E, Torres M, Glass J, Minniti CP, Barrette S, Habr D, et al. A randomized phase II study evaluating the efficacy and safety of deferasirox versus deferoxamine in patients with sickle cell disease (SCD0: 2‐year results including pharmacokinetics (PK) and safety of deferasirox with concomitant hydroxyurea therapy. Blood 2011;118(21). [Abstract no: 1082]CENTRAL
Vichinsky E, Torres M, Minniti CP, Barrette S, Habr D, Zhang Y, et al. Efficacy and safety of deferasirox compared with deferoxamine in sickle cell disease: two‐year results including pharmacokinetics and concomitant hydroxyurea. American Journal of Hematology 2013;88(12):1068‐73. [CRS: 5500125000000523; PUBMED: 23946212]CENTRAL

Voskaridou 2005 {published data only}

Voskaridou E, Terpos E, Margeli A, Hantzi E, Meletis J, Papassotiriou I, et al. Renal dysfunction and osteodystrophy in patients with sickle cell thalassaemia under long‐term treatment with hydroxyurea. 10th Congress of the European Hematology Association; 2005 June 2‐5; Stockholm International Fairs, Sweden. 2005. [Abstract no: 0174]CENTRAL

Anyanwu 2016 {published data only}

Anyanwu JN, Williams O, Sautter CL, Kasirye P, Hume H, Opoka RO, Latham T, Ndugwa C, Ware RE, John CC. Novel Use of Hydroxyurea in an African Region With Malaria: Protocol for a Randomized Controlled Clinical Trial. JMIR research protocols 2016;5(2):e110. CENTRAL

NCT02560935 {published data only}

NCT02560935. Primary Prevention of Stroke in Children With SCD in Sub‐Saharan Africa II (SPRING). clinicaltrials.gov/show/NCT02560935 (first received 02 September 2015). CENTRAL

NCT02675790 {published data only}

NCT02675790. Low Dose Hydroxyurea for Secondary Stroke Prevention in Children With Sickle Cell Disease in Sub‐Saharan Africa (SPRINT). clinicaltrials.gov/ct2/show/NCT02675790 (first received 08 January 2016). CENTRAL

NCT01389024 {published data only}

NCT01389024. Hydroxyurea to Prevent Brain Injury in Sickle Cell Disease (HUPrevent). clinicaltrials.gov/ct2/show/NCT01389024 (first received 30 June 2011). CENTRAL

Aumont 2015

Aumont C, Driss F, Lazure T, Picard V, Creidy R, De Botton S, et al. Myelodysplastic syndrome with clonal cytogenetic abnormalities followed by fatal erythroid leukemia after 14 years of exposure to hydroxyurea for sickle cell anemia. American Journal of Hematology 2015;90(7):E131‐2.

Ballas 1996

Ballas SK, Barton F, Castro O, Koshy M, Bellevue R. Pattern of narcotic analgesic consumption among adult patients with sickle cell anemia. National Sickle Cell Disease Program 21st Annual Meeting; 1996 Mar. 1996:63.

Ballas 2006

Ballas SK, Barton FB, Waclawiw MA, Swerdlow P, Eckman JR, Pegelow CH, et al. Hydroxyurea and sickle cell anemia: effect on quality of life. Health and Quality of Life Outcomes 2006;4:59.

Ballas 2009

Ballas SK, Guo N, DeCastro L, Bellevue R. Exposure to hydroxyurea and pregnancy outcomes in patients with sickle cell anemia. JAMA 2009;101(10):1046.

Barton 1996

Barton F, Terrin M, Moore R, McMahon RP, Charache S. Ascertainment of the primary end point in the Multicenter Study of Hydroxyrea in Sickle Cell Anemia (MSH). The MSH Investigators. Controlled Clinical Trials 1996;17:67S.

Baz 2012

Baz W, Najfeld V, Yotsuya M, Talwar J, Terjanian T, Forte F. Development of myelodysplastic syndrome and acute myeloid leukemia 15 years after hydroxyurea use in a patient with sickle cell anemia. Clinical Medicine Insights. Oncology 2012;6:149‐52.

Berk 1995

Berk PD, Wasserman LR, Fruchtman SM, Goldberg JD. Treatment of polycythemia vera: a summary of clinical trials conducted by the Polycythemia Vera Study Group. In: Wasserman LR BP, Berlin NI editor(s). Polycythemia Vera and the Myeloproliferation Disorders. Philadelphia: W B Saunders, 1995:166‐94.

Bernaudin 2016

Bernaudin F, Verlhac S, Arnaud C, Kamdem A, Hau I, Leveillé E, et al. Long‐term treatment follow‐up of children with sickle cell disease monitored with abnormal transcranial Doppler velocities. Blood 2016;127(15):1814‐22. [10.1182/blood‐2015‐10‐675231. Epub 2016 Feb 5.]

Berthaut 2008

Berthaut I, Guignedoux G, Kirsch‐Noir F, de Larouziere V, Ravel C, Bachir D, et al. Influence of sickle cell disease and treatment with hydroxyurea on sperm parameters and fertility of human males. Haematologica 2008;93(7):988‐93.

Brousseau 2010

Brousseau DC, A Panepinto J, Nimmer M, Hoffmann RG. The number of people with sickle‐cell disease in the United States: national and state estimates. American journal of hematology 2010 Jan 1;85(1):77‐8.

Brozovic 1987

Brozovic M, Davies SC. Management of sickle cell disease. Postgraduate Medical Journal 1987;63:605‐9.

Chakravorty 2015

Chakravorty S, Williams TN. Sickle cell disease: a neglected chronic disease of increasing global health importance. Archives of Disease in Childhood 2015;100(1):48‐53.

Charache 1995

Charache S, Terrin TL, Moore RD, Dover GJ, Barton FB, Eckert SV, et al. Effect of hydroxyurea on the frequency of painful crises in sickle cell anaemia. New England Journal of Medicine 1995;332:1317‐22.

Davies 1997a

Davies SC, Roberts‐Harewood M. Blood transfusion in sickle cell disease. Blood Reviews 1997;11:57‐71.

Davies 1997b

Davies SC, Oni L. The management of patients with sickle cell disease. British Medical Journal 1997;315:656‐60.

Elbourne 2002

Elbourne DR, Altman DG, Higgins JPT, Curtin F, Worthington HV, Vail A. Meta‐analysis involving cross‐over trials: methodological issues. International Journal of Epidemiology 2002;31(1):140‐9.

Fruchtman 1994

Fruchtman SM, Kaplan ME, Peterson P, Mack K, Berk PD, Wasserman LR. Acute leukemia (AL), hydroxyurea (HU), and polycythemia vera (PV): an analysis of risk from the Polycythemia Vera Study Group. Blood 1994;84(Suppl 1):518a.

Fuggle 1996

Fuggle P, Shand PAX, Gill LJ, Davies SC. Pain, quality of life, and coping in sickle cell disease. Archives of Disease in Childhood 1996;75:199‐203.

Gardner 2016

Gardner K, Douiri A, Drasar E, Allman M, Mwirigi A, Awogbade M, et al. Survival in adults with sickle cell disease in a high income setting. Blood 2016;128(10):1436‐1438.

Gray 1991

Gray A, Anionwu EN, Davies SC, Brozovic M. Mortality in sickle cell disease: the experience of a British centre. Journal of Clinical Pathology 1991;44:459‐63.

Green 2014

Green NS, Barral S. Emerging science of hydroxyurea therapy for pediatric sickle cell disease. Pediatric research 2013;75(1‐2):196‐204.

Grosse 2011

Grosse SD, Odame I, Atrash HK, Amendah DD, Piel FB, Williams TN. Sickle cell disease in Africa: a neglected cause of early childhood mortality. American journal of preventive medicine 2011 Dec 31;41(6):S398‐405.

Handy 1996

Handy C, Barton F, Moore R, McMahon R, Eckert S, Terrin M. Dose titration in the Multicentre Study of Hydroxyurea in Sickle Cell Anemia (MSH). Controlled Clinical Trials 1996;17(Suppl 2):92S.

Hankins 2014

Hankins JS, Aygun B, Nottage K, Thornburg C, Smeltzer MP, Ware RE, et al. From infancy to adolescence: fifteen years of continuous treatment with hydroxyurea in sickle cell anemia. Medicine (Baltimore) 2014;93(28):e215. [DOI: 10.1097/MD.0000000000000215]

Hassell 2010

Hassell KL. Population estimates of sickle cell disease in the US. American journal of preventive medicine 2010 Apr 30;38(4):S512‐21.

Higgins 2003

Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analysis. BMJ 2003;327:557‐60.

Higgins 2011

Higgins JPT, Altman DG, Sterne JAC (editors). Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from www.cochrane‐handbook.org..

Howard 2015

Howard J, Telfer P. Sickle Cell Disease in Clinical Practice. London 2015: Springer‐Verlag, 2015.

Jones 2009

Jones AP, Riley RD, Williamson PR, Whitehead A. Meta‐analysis of individual patient data versus aggregate data from longitudinal clinical trials. Clinical Trials 2009;6(1):16‐27.

Lanzkron 2013

Lanzkron S, Carroll CP, Haywood Jr C. Mortality Rates and Age at Death from Sickle Cell Disease: US. 1979‐2005. Public Health Reports 2013;128:110‐116.

Luchtman‐Jones 2016

Luchtman‐Jones L, Pressel S, Hilliard L, Brown RC, Smith MG, Thompson AA, et al. Effects of hydroxyurea treatment for patients with hemoglobin SC disease. American Journal of Hematology 2016;91(2):238‐42.

Manci 2003

Manci EA,  Culberson DE,  Yang YM,  Gardner TM,  Powell R,  Haynes J, et al. Causes of death in sickle cell disease: an autopsy study. British Journal of Haematology 2003;123(2):359‐65.

McGann 2011

McGann PT, Howard TA, Flanagan JM, Lahti JM, Ware RE. Chromosome damage and repair in children with sickle cell anaemia and long‐term hydroxycarbamide exposure. British Journal of Haematology 2011;154(1):134‐40.

Najean 1997

Najean Y, Rain JD. Treatment of polycythemia vera: the use of hydroxyurea and pipobroman in 292 patients under the age of 65 years. Blood 1997;90(9):3370‐7.

National Toxicology Program 2008

National Toxicology Program. NTP‐CERHR monograph on the potential human reproductive and developmental effects of hydroxyurea. NTP CERHR MON2008; Vol. Oct, issue 21:vii‐viii, v, ix‐III1. [NIH Publication no: 08‐5993]

NCEPOD 2008

NCEPOD. ‘Sickle: A sickle crisis?’ A report of the national confidential enquiry into patient outcome and death. 2008. www.ncepod.org.uk.2008sc.html (accessed 19 September 2016).

NICE 2012

National Insititute of Clinical Excellence (NICE). Sickle cell acute painful episode: management of an acute painful sickle cell episode in hospital. http://www.nice.org.uk/guidance/cg143 (accessed 06 March 2017)2012.

NIH 2014

Evidence Based Management of Sickle Cell Disease. Expert Panel Report. National Institute of Health. 2014. www.nhlbi.nih.gov/sites/www.nhlbi.nih.gov/files/sickle‐cell‐disease‐report%20020816.pdf (accessed 19 September 2016).

Perrine 1978

Perrine RP, Pembrey ME, John P, Perrine S, Shoup F. Natural history of sickle cell anaemia in Saudi Arabs: a study of 270 subjects. Annals of Internal Medicine 1978;88:1‐6.

Perronne 2002

Perronne V,  Roberts‐Harewood M,  Bachir D,  Roudot‐Thoraval F,  Delord JM,  Thuret I, et al. Patterns of mortality in sickle cell disease in adults in France and England. Haematology Journal 2002;3(1):56‐60.

Piel 2013

Piel FB, Patil AP, Howes RE, Nyangiri OA, Gething PW, Dewi M, et al. Global epidemiology of sickle haemoglobin in neonates: a contemporary geostatistical model‐based map and population estimates. Lancet 2013;381(9861):142‐51.

Platt 1984

Platt OS, Orkin SH, Dover GJ, Beardsley GP, Miller B, Nathan DG. Hydroxyurea increases fetal hemoglobin production in sickle cell anemia. Transactions of the Association of American Physicians 1984;97:268‐74.

Platt 1994

Platt OS, Brambilla DJ, Rosse WF, Milner PF, Castro O, Steinberg MH, et al. Mortality in sickle cell disease ‐ life expectancy and risk factors for early death. New England Journal of Medicine 1994;330:1639‐43.

Powars 1983

Powars D, Overturf G, Turner E. Is there an increased risk of Haemophilus influenzae septicemia in sickle cell anaemia?. Pediatrics 1983;71:927‐31.

Quinn 2010

Quinn CT, Rogers ZR, McCavit TL, Buchanan GR. Improved survival of children and adolescents with sickle cell disease. Blood 2010;115:3447‐52.

Rogers 1978

Rogers DW, Clarke JM, Cupidore L, Ramlal A, Sparke BR, Serjeant GR. Early deaths in Jamaican children with sickle cell disease. British Medical Journal 1978;1:1515‐6.

Steinberg 1999

Steinberg MH. The Management of Sickle Cell Disease. New England Journal of Medicine 1999;340:1021‐30.

Steinberg 2003

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.

Steinberg 2010

Steinberg MH, McCarthy WF, Castro O, Ballas SK, Armstrong FD, Smith W, et al. The risks and benefits of long‐term use of hydroxyurea in sickle cell anemia: A 17.5 year follow‐up. American Journal of Hematolology2010; Vol. 85, issue 6:403‐8.

Sterkers 1998

Sterkers Y, Preudhomme C, Lai JL, Demory JL, Caulier MT, Wattel E, et al. Acute myeloid leukemia and myelodysplastic syndromes following essential thrombocythemia treated with hydroxyurea: high proportion of cases with 17p deletion. Blood 1998;91(2):616‐22.

STOP 2006

Lee MT, Piomelli S, Granger S, et al. Stroke Prevention Trial in Sickle Cell Anemia (STOP): extended follow‐up and final results. Blood 2006;108(3):847‐52.

Strouse 2008

Strouse JJ, Lanzkron S, Beach MC, Haywood C, Park H, Witkop C, et al. Hydroxyurea for sickle cell disease: a systematic review for efficacy and toxicity in children. Pediatrics 2008;122(6):1332‐43.

Telfer 2007

Telfer P,  Coen P,  Chakravorty S,  Wilkey O,  Evans J,  Newell H,  et al. Clinical outcomes in children with sickle cell disease living in England: a neonatal cohort in East London.. Haematologica 2007;92(7):905‐12.

Terrin 1999

Terrin ML, Barton FB, Bonds D, Ballas SK, Swerdlow P, Pegelow CH, et al. Effect of hydroxyurea on quality of life: 2‐year results from the multicenter study of hydroxyurea in sickle cell anemia. National Sickle Cell Disease Program 23rd Annual Meeting; 1999 Mar. 1999:161.

Triadou 1994

Triadou P, Maier‐Redelsperger M, Krishnamoorty R, Deschamps A, Casadevall N, Dunda O, et al. Fetal haemoglobin variations following hydroxyurea treatment in patients with cyanotic congenital heart disease. Nouvelle Revue Francaise d Hematologie 1994;36(5):367‐72.

van der Plas 2011

van der Plas EM,  van den Tweel XW,  Geskus RB,  Heijboer H,  Biemond BJ,  Peters M,  et al. Mortality and causes of death in children with sickle cell disease in the Netherlands, before the introduction of neonatal screening. British Journal of Haematology 2011;155(1):106‐10.

Veith 1985

Veith R, Galanello R, Papayannopoulou T, Stamatoyannopoulos G. Stimulation of F‐cell production in patients with sickle cell anaemia treated with cytarabine or hydroxyurea. New England Journal of Medicine 1985;313:1571‐5.

Voskaridou 2010

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‐centre trial (LaSHS). Blood 2010;115:2354‐63.

Ware 2006

Ware RE, McMurray MA, Schultz WH, Alvarez OA, Aygun B, Cavalier ME, et al. Academic community standards for chronic transfusion therapy in children with sickle cell anemia and stroke. Blood 2006;108(11):Abst 1213.

Ware 2010

Ware RE. How I use hydroxyurea to treat young patients with sickle cell anemia. Blood 2010;115:5300‐11.

Wierenga 2001

Wierenga KJ,  Hambleton IR,  Lewis NA. Survival estimates for patients with homozygous sickle‐cell disease in Jamaica: a clinic‐based population study. Lancet 2001;357(9257):680‐3.

Zimmerman 2004

Zimmerman SA, Schultz WH, Davis JS, Pickens CV, Mortier NA, Howard TA, et al. Sustained long‐term hematologic efficacy of hydroxyurea at maximum tolerated dose in children with sickle cell disease. Blood 2004;103(6):2039‐45.

References to other published versions of this review

Jones 2001

Jones AP, Davies SC, Olujohungbe A. Hydroxyurea for sickle cell disease. Cochrane Database of Systematic Reviews 2001, Issue 2. [DOI: 10.1002/14651858.CD002202]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

BABY HUG 2011

Methods

Randomised, double blind (participant, investigator), placebo control, parallel assignment, efficacy study conducted at 13 centres in the USA.

Participants

Children aged 9 months to 18 months with SCD (HbSS or HbSߺ‐thalassemia) irrespective of clinical severity. Exclusion criteria were transfusion within 2 months, height, weight or head circumference below the 5th percentile, mental development index less than 70 or abnormal TCD ultrasound velocity.

193 randomised, 96 participants to hydroxyurea and 97 to placebo.

Mean (SD) age: hydroxyurea group : 13.6 (2.7) months, placebo group: 13.5 (2.8) months

187 (97%) with HbSS genotype, 109 (56%) females

Interventions

Hydroxyurea (20 mg/kg/day) versus placebo for 24 months.

Outcomes

Primary outcomes: spleen function (decline in splenic uptake); and glomerular filtration rate with 99mTc‐diethyl‐enetriaminepentaacetic acid plasma clearance.

Secondary outcomes: ratio of nuclear decay counts in the spleen and liver; proportion of red blood cells containing pits or Howell‐Jolly bodies; renal function; growth; and development (including neuro‐developmental assessment).

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

The telephone randomisation schedule was developed by the medical co‐ordinating centre.

Allocation concealment (selection bias)

Low risk

Centralised telephone randomisation was used.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Some secondary outcomes reported for only the individuals who completed the study but ITT approach taken for primary outcomes (via multiple imputation) and safety outcomes so risk of bias judged to be low.

Selective reporting (reporting bias)

Low risk

All outcomes well defined in the methods and reported well in the results.

Other bias

Low risk

None identified.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants, caregivers and medical coordinating centre staff were masked to treatment allocation via packaging and treatment of the same appearance.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

An unmasked "primary endpoint person" monitored laboratory values and assisted clinical management. Other outcome assessors (e.g. those reading the liver‐spleen scans).

Belgian Study 1996

Methods

Randomised placebo‐controlled cross‐over study conducted in a single centre in Belgium.

Participants

25 children with HbSS genotype, age 2 ‐ 22 years (median 9 years) with > 3 vaso‐occlusive events reported in preceding 12 months and/or history of CVA off transfusion (severe alloimmunisation or compliance), ACS, ASS.

Interventions

Hydroxyurea 20 mg/kg/day rising to 25 (unless toxic cytopenia) versus placebo. Treatment period was for 6 months.

Outcomes

Number of hospitalisations.
Number of inpatient days.
FBC.
HbF%.

Notes

Data are not presented in a way that results can be included in the review so results are presented narratively.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Described as "drawing sealed envelopes, patients were randomly allocated to one of the following treatment sequences", therefore the generation of the treatment sequence are not clear from this statement.

Allocation concealment (selection bias)

Unclear risk

Described as "drawing sealed envelopes, patients were randomly allocated to one of the following treatment sequences", therefore the allocation of the treatment sequence are not clear from this statement.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

3 participants who were excluded from the analysis due to their failure to attend the monthly evaluation at 4 ‐ 5 months. There was no discussion of whether or not an ITT analysis was used so the risk is unclear.

Selective reporting (reporting bias)

High risk

A planned outcome (number of days in pain) was dropped from analysis due to difficulty in obtaining information to inform this analysis from participants

Other bias

Low risk

Unclear if a washout period was used in this cross‐over study but tests for period effects and carry‐over effect were not significant so the risk of bias in the cross‐over design is low.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The hospital pharmacy provided the treatment and placebo for each participant and both were described as "indistinguishable," however the physician was aware of the treatment schedule because of the difficulty of blinding the attending physician to the treatment received. Unclear if this could have influenced results.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided.

CHAMPS 2011

Methods

Randomised multicentre, phase II, double blind placebo‐controlled study.

Participants

Eligible participants over the age of 5 years with HbSC and at least 1 vaso‐occlusive event in the previous 12 months (but none in the 4 weeks prior to study entry).

44 participants randomised; 11 to each treatment group (see 'Interventions').

Mean age: 13.6 years (range 5 ‐ 53 years), 43% females.

Interventions

Participants randomised to 1 of 4 arms in a factorial design:

1. hydroxyurea (20 mg/kg/day) and magnesium (0.6 mmol/kg/day in 2 doses);

2. hydroxyurea (20 mg/kg/day) and placebo;

3. placebo and magnesium (0.6 mmol/kg/day in 2 doses);

4. placebo and placebo.

Outcomes

Primary outcome: proportion of hyperdense red blood cells at 8 weeks.

Secondary outcomes: central laboratory evaluations (including measurements of red cell density, HbF, red cell cation content, KCl co‐transport and Gardos channel activity, cell adhesion to endothelial cells and laminin, and erythrocyte membrane phosphatidyl serine (PS exposure)) at baseline (twice) and weeks 8, 16, 24, and 44.

Participants were evaluated at 2 or 4 week intervals for 11 months (15 visits).

Notes

The study was not designed to measure efficacy and all analyses were considered exploratory.

The study was terminated early due to low enrolment after 44 participants had been randomised (target 188).

Due to factorial design, no data can be entered into analysis and results for hydroxyurea groups (groups 1 and 2) compared to no hydroxyurea groups are summarised narratively.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A sequential allocation algorithm (i.e. minimisation) was used due to small numbers within each strata (site and age group).

Allocation concealment (selection bias)

Unclear risk

No information provided.

Incomplete outcome data (attrition bias)
All outcomes

High risk

36 out of 44 participants completed 8 weeks and 22 out of 44 completed 44 weeks. Only those who completed the follow‐up time were included in analysis (8 weeks for primary endpoint and 44 weeks for secondary endpoints). This is not an ITT approach.

Selective reporting (reporting bias)

Low risk

Outcome defined in the methods section well described in the results section. Study was not designed to measure efficacy.

Other bias

Low risk

Study terminated early after only 44 of planned sample size of 188 were recruited. However, the study was not designed to measure efficacy and performed only exploratory analyses therefore the early termination is unlikely to have introduced bias.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All tablets were 'over‐capsulated' to disguise appearance.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

No information provided.

Jain 2012

Methods

Double‐blind randomised controlled study conducted in a tertiary hospital in Nagpur City, India.

Participants

Indian children between the ages of 5 and 18 years with severe manifestations (defined as 3 or more blood transfusions or VOC requiring hospitalisation per year) despite high HbF.

Exclusion criteria included seropositivity for HIV or chronic illness.

60 participants randomised; 30 to each treatment group.

53% females (16 females per group).

Mean (SD) age ‐ hydroxyurea group: 12.73 (4.4) years, placebo group: 11.73 (4.08) years.

Interventions

Hydroxyurea (fixed dose 10 mg/mg/day) compared to placebo for 18 months.

Outcomes

Primary outcome: frequency of VOC per participant per year.

Secondary outcomes: frequency of blood transfusions, hospitalisations and HbF levels.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomised using randomisation tables.

Allocation concealment (selection bias)

Unclear risk

No information provided.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants completed 18‐months follow‐up and were included in analysis.

Selective reporting (reporting bias)

Low risk

No protocol available, outcomes defined in the methods reported well in the results.

Other bias

Low risk

None identified.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants were blinded with placebo tablets of identical appearance. The clinician who assessed participants were not aware of treatment arm.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The laboratory technician and the clinician who assessed participants were not aware of treatment arm.

MSH 1995

Methods

Randomised multicentre parallel, double‐blind placebo‐controlled study. Conducted in the USA and Canada across 21 sites.

Participants

Adults over 18 years of age with HbSS genotype who had reported more than 3 'crises' to treating physician in the preceding 12 months and who had < 15% HbA. Exclusions included: HbA > 15%, pregnancy, opiate addiction, other potent anti‐sickling agents, cytopenia, CVA in the preceding 6 years, HIV antibody +, prior hydroxyurea therapy

152 randomised to hydroxyurea and 147 given placebo.

Interventions

Hydroxyurea starting at 15 mg/kg/day rising 12‐weekly by 5 mg/kg/day unless marrow depression (then cessation of drug until recovery and restarted at 2.5 mg/kg/day lower, i.e. MTD) compared to placebo for 2 years.

Outcomes

Primary outcome: pain events ‐ attending hospital > 4 hours & parenteral opiate treatment.
Secondary outcomes: 'crises' including ACS, CVA, priapism etc, daily pain charts, time to first and second crisis, FBC, F cells & Hb F%, dense cells.

Notes

93% follow‐up at 2 years, treatment stopped in 14 hydroxyurea and 6 placebo participants.

Due to beneficial treatment effects, the study was stopped at a mean follow‐up of 21 months, before the planned 24 months of treatment had been completed for all participants.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Treatment assignments were made centrally using a computer programme to generate separate, randomised block assignment schedules for each clinic.

Allocation concealment (selection bias)

Unclear risk

There was no clear discussion on allocation concealment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Withdrawals from the study were well documented, all participants included in analysis in an ITT approach in the primary publications but later reported outcomes (such as quality of life) reported only for those who contributed data.

Selective reporting (reporting bias)

Low risk

All outcomes well defined in the methods and reported well in the results.

Other bias

Low risk

None identified.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Stated as double‐blind (physician and participant), where treatment was assigned in combinations of identically appearing capsules.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

There was no clear discussion on blinding of outcome assessors.

SCATE 2015

Methods

Phase III randomised, partially masked (outcome assessors) multicentre study conducted in 3 centres in the USA, Jamaica and Brazil between May 2012 and August 2013.

Participants

Children with SCA and conditional TCD ultrasound velocities (170 cm ‐ 199 cm per second).

Exclusion criteria were prior abnormal TCD velocities or clinical stroke, red blood cell transfusion within 2 months of enrolment, concurrent use of another anti‐sickling medication or contraindication to hydroxyurea therapy (allergy, pregnancy, renal insufficiency).

22 participants randomised, 11 to each treatment group.

Mean age (SD): hydroxyurea group: 6.2 (2.4) years, observation group: 6.6 (1.5) years.

64% females, 7 per group. 21 participants with HbSS and 1 with HbSβº‐thalassemia.

Interventions

Hydroxyurea (starting at 20 mg/kg/day escalated to a maximum of 35 mg/kg/day) compared to standard treatment (observation).

Outcomes

Primary outcome: conversion to abnormal maximum abnormal TAMV.

Secondary outcomes: changes in serial TCD velocities.

Indicence of acute events including stroke.

Health‐related quality of life (planned but not recorded, see 'Selective reporting' below).

Notes

The planned length of follow‐up was 30 months but the study was terminated early after 15 months of follow‐up due to slow participant accrual and the unlikelihood of meeting the trial recruitment target (100) and the primary endpoint.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was performed using an adaptive blocked algorithm.

Allocation concealment (selection bias)

Low risk

Central pharmacy distribution of allocations.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Numbers screened, randomised and number receiving randomised treatment reported. Analysis conducted using an ITT approach.

Selective reporting (reporting bias)

High risk

A planned outcome (health‐related quality of life) was not analysed or presented as the outcome was not sufficiently collected due to early study termination.

Other bias

High risk

Study terminated early after only 22 of planned sample size of 100 were recruited therefore study is likely to be statistically underpowered.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants and personnel could not be masked to treatment allocation by design (hydroxyurea compared to observation only). The primary outcome (conversion to abnormal TAMV) was objective and determined by masked outcome assessors so lack of blinding of participants and personnel is unlikely to have affected results.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All TCD site examiners, central reviewers were masked and site clinicians were masked to participants TCD results.

SWiTCH 2012

Methods

Phase III, multicentre, single masked (outcome assessors), non‐inferiority study conducted across 26 paediatric sites in the USA.

Participants

Participants with previous clinical stroke, aged 7 to 17 years, PRBC transfusions for at least 18 months and transfusional iron overload.

No specific exclusion criteria stated.

133 participants were randomised; 67 randomised to hydroxyurea treatment and 66 to standard treatment.

Mean (SD) age at study enrolment in years: hydroxyurea group 13.0 (4.0) years, standard treatment group 13.3 (3.8) years.

132 out of 133 participants with HbSS genotype, 61 females (46%).

Interventions

Hydroxyurea starting at 20 mg/kg/day escalated to MTD and phlebotomy compared to standard treatment (transfusions and chelation) for 30 months.

Outcomes

The primary outcome of the trial was a composite outcome. This involved occurrence of a secondary stroke and quantitative liver iron level change from baseline.

Secondary outcomes included quality of life, non‐stroke neurological events, other SCD‐related events, growth and development, functional evaluations, neurocognitive evaluations, transfusion related complications, chelation related complications, hydroxyurea‐related complications, phlebotomy related complications, liver biopsy related complications and adverse and serious adverse events.

Notes

Numerous secondary outcomes were not reported in the main paper or any subsequent papers.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Study described as randomised, no further details given.

Allocation concealment (selection bias)

Unclear risk

No details were given on allocation concealment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Withdrawals from treatment reported, all individuals included in analysis in an ITT approach.

Selective reporting (reporting bias)

High risk

Many of the secondary outcomes (such as growth and development, functional evaluations, neurocognitive evaluations) have not yet been reported, If these results can be included at a later date then this judgement will be reconsidered.

Other bias

Low risk

None identified.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants and personnel could not be masked to treatment allocation by design (hydroxyurea compared to transfusion). The primary outcome (secondary stroke and quantitative liver iron level change from baseline) was objective and determined by masked outcome assessors so lack of blinding of participants and personnel is unlikely to have affected results.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The primary outcome (secondary stroke and quantitative liver iron level change from baseline) were determined by a group of treatment masked neurologists and neuroradiologists.

TWiTCH 2016

Methods

Multicentre phase III randomised open‐label (partially masked) non‐inferiority study conducted at 26 paediatric hospitals and health centres in the USA and Canada.

Participants

Children aged 4 ‐ 16 years with SCA and abnormal TCD ultrasound velocities (> 200 cm per second) if they had received 12 months of chronic transfusions.

Exclusion criteria were documented clinical stroke, TIA or severe vasculopathy.

121 participants were randomised; 60 randomised to hydroxyurea treatment and 61 to standard treatment.

Mean (SD) age at study enrolment in years: hydroxyurea group 9.5 (2.6) years, standard treatment group 9.7 (3.2) years.

119 out of 121 participants with HbSS genotype, 73 females (60%).

Interventions

Hydroxyurea starting at 20 mg/kg/day escalated to MTD and phlebotomy compared to standard treatment (transfusions and chelation) for 24 months.

Outcomes

Primary outcome: maximum TCD time averaged mean velocity on the index side (i.e. the cerebral hemisphere with the higher mean arterial velocity at baseline assessment).

Secondary outcomes: TCD velocity on the non‐index side, neurological events, new brain lesions, hepatic iron overload, SCD‐related events, treatment‐related complication (reported in this publication) neuropsychological status, quality of life and growth and (to be reported in future publications).

Notes

Study was terminated early at the first interim analysis when non‐inferiority was demonstrated, target sample size was met at early termination.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Centralised block randomisation with block size four, with stratification by site and balanced by baseline age and TCD velocity.

Allocation concealment (selection bias)

Low risk

Central randomisation and treatment allocation.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Numbers discontinuing the interventions stated, all participants included in analysis in an ITT analysis.

Selective reporting (reporting bias)

High risk

Outcomes of neuropsychological status, quality of life and growth were measured but results are not yet published. If these results can be included at a later date then this judgement will be reconsidered.

Other bias

Low risk

Study was terminated early at the first interim analysis when non‐inferiority was demonstrated, target sample size was met at early termination so the study is adequately powered to detect differences.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Participants and personnel could not be masked to treatment allocation by design (hydroxyurea compared to transfusion). The primary outcome (maximum TCD time averaged mean velocity) was objective and determined by masked outcome assessors so lack of blinding of participants and personnel is unlikely to have affected results.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All TCD examinations were read centrally by observers blinded to treatment allocation and previous TCD results.

ACS: acute chest syndrome
ASS: acute splenic sequestration
CVA: cerebro‐vascular accident
Cytopenia: refers to either neutropenia or thrombocytopenia, anaemia is also a risk but was not reported
FBC: full blood count
HbA: adult haemoglobin
HbF: fetal haemoglobin
HbSβº: haemoglobin Sβºthalassaemia genotype
HbSC: haemoglobin SC genotype
HbSS: haemoglobin SS genotype
ITT: intention‐to‐treat
KCl: potassium chloride
MTD: maximum tolerated dose
PRBC: packed red blood cells
SCA: sickle cell anaemia
SCD: sickle cell disease
SD: standard deviation
TAMV: time averaged mean velocity
TCD: transcranial doppler
TIA: transient ischaemic attack
VOC: vaso‐occlusive crisis

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Al‐Nood 2011

This is not a randomised study.

De Montalembert 2006

This study does not fulfil the inclusion criteria. The length of treatment was 8 days, the inclusion criteria state that treatment should be at least 1 month.

George 2013

This does not make a randomised comparison of hydroxyurea and placebo or standard treatment (the randomised comparison is dosing schedules of hydroxyurea).

NCT00004492

This does not make a randomised comparison of hydroxyurea and placebo or standard treatment.

NCT01848925

This study does not fulfil the inclusion criteria. The length of treatment was 7 days, the inclusion criteria state that treatment should be at least 1 month.

NCT01960413

This does not make a randomised comparison of hydroxyurea and placebo or standard treatment.

NCT02149537

This is not an appropriate design to measure the effectiveness of hydroxyurea (cross‐over design of low‐dose hydroxyurea compared to no treatment to monitor those at increased risk of infection).

Pushi 2000

This is not a randomised study.

Silva‐Pinto 2007

This is not a randomised study.

Silva‐Pinto 2014

This is not a randomised study.

Vichinsky 2013

This does not make a randomised comparison of hydroxyurea and placebo or standard treatment.

Voskaridou 2005

This is not a randomised study.

Characteristics of studies awaiting assessment [ordered by study ID]

Anyanwu 2016

Methods

Prospective, randomised, placebo‐controlled, double‐blinded phase III trial (NOHARM trial)

Participants

Study participants will be recruited from the Mulago Hospital Sickle Cell Clinic (MHSCC) in Kampala, Uganda.

Children aged 1 to 4 years with documented HbSS living in an area of meso‐endemic malaria transmission.

Interventions

Hydroxyurea 20 ± 2.5 mg/kg/day compared to placebo

Outcomes

Primary outcome: malaria incidence, defined as episodes of clinical malaria occurring over the 1‐year randomised study treatment period.

Secondary outcomes: frequency of haematologic toxicities and AEs, relationships between hydroxyurea treatment and fetal haemoglobin, soluble intracellular adhesion molecule‐1, and nitric oxide levels, and between levels of these factors and risk of subsequent malaria.

Notes

Currently, only a protocol is available for the NOHARM study.

We are unsure if this study meets the inclusion criteria of the review of 'Type of Participants' due to the study objectives around determining the incidence of malaria in SCA individuals. We will make an assessment of the eligibility of the population when study results are available.

NCT02560935

Methods

Randomised, double‐blind, parallel group, dose‐controlled study (SPRING)

Participants

Participants with HbSS or HbSβº‐thalassemia, S variant with baseline haemoglobin less than 10 g/dL or other sickle cell syndromes apart from HbSC between the ages of 5 and 12 years, living in sub‐Saharan Africa, without prior overt stroke.

Inclusion criteria for a 'non‐elevated TCD group' (participants who are not eligible to receive hydroxyurea therapy but are willing to be following for a minimum of 3 years).

Interventions

Hydroxyurea (moderate dose): 20 mg/kg/day (range 17.5 ‐ 26 mg/kg/day) for 24 months.

Hydroxyurea (low dose): 10 mg/kg/day (range 7 ‐ 15 mg/kg/day) for 24 months.

Outcomes

Primary outcome: efficacy of moderate vs low‐dose hydroxyurea therapy for primary stroke prevention.

Secondary outcome: incidence of all‐cause hospitalizations.

Secondary outcome: long‐term safety of hydroxyurea therapy.

Notes

Unclear if this is an appropriate design (dose‐control) and population for the review ('non‐elevated TCD group').

Trial is ongoing (estimated completion date ‐ December 2021), we will make an assessment of the eligibility of the design and population when trial results are available.

NCT02675790

Methods

Randomised, double‐blind, parallel group, dose‐controlled study (SPRINT).

Participants

Participants between the age of 1 and 18 years with SCA and a history of stroke up to 30 days before entry into the study, living in sub‐Saharan Africa.

Interventions

Hydroxyurea (moderate dose): 20 mg/kg/day (range 17.5 ‐ 26 mg/kg/day) for 24 months.

Hydroxyurea (low dose): 10 mg/kg/day (range 7 ‐ 15 mg/kg/day) for 24 months.

Outcomes

Primary outcome: rate of clinical stroke recurrence.

Secondary outcome: incidence of all‐cause hospitalisation.

Notes

Unclear if this is an appropriate design (dose‐control) and population for the review (participants can have already received hydroxyurea treatment).

Study is ongoing (estimated completion date July 2019), we will make an assessment of the eligibility of the design and population when study results are available.

AEs: adverse events
HbSβº: haemoglobin Sβºthalassaemia genotype
HbSC: haemoglobin SC genotype
HbSS: haemoglobin SS genotype
TCD: transcranial doppler

Characteristics of ongoing studies [ordered by study ID]

NCT01389024

Trial name or title

NCT01389024: Hydroxyurea to Prevent Brain Injury in Sickle Cell Disease (HUPrevent)

Methods

Randomised, double‐blind, parallel design, phase 2 study.

Participants

Participants with HbSS or HbSβº‐thalassemia aged between 9 and 48 months of age, with or without central nervous system complications.

Interventions

Hydroxyurea 20 mg/kg/day increased by 5 mg/kg every 8 weeks to maximum of 35 mg/kg/day;

placebo (sucrose) 0.2 mL/kg/day increased to max of 0.35 mL/kg/day.

Outcomes

Primary outcome: central nervous system complications (a composite of abnormally elevated cerebral blood flow velocity as measured by TCD ultrasound, SCI, or stroke)

Secondary outcome: proportion of participants with severe adverse events attributed to study procedures.

Starting date

October 2011

Contact information

Johns Hopkins University

Diane Weiss, BA ([email protected])

James F. Casella, MD ([email protected])

Notes

Estimated completion date October 2017 (final data collection date for primary outcome measure)

HbSβº: haemoglobin Sβºthalassaemia genotype
HbSC: Haemoglobin SC genotype
HbSS: Haemoglobin SS genotype
SCI: silent cerebral infarct
TCD: transcranial doppler

Data and analyses

Open in table viewer
Comparison 1. Hydroxyurea versus placebo for participants with sickle cell disease

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain crises Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.1

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 1 Pain crises.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 1 Pain crises.

1.1 Mean annual crisis rate at 2 years (all crises)

1

Mean Difference (IV, Fixed, 95% CI)

‐2.80 [‐4.74, ‐0.86]

1.2 Mean annual crisis rate at 2 years (all crises requiring hospitalisation)

1

Mean Difference (IV, Fixed, 95% CI)

‐1.50 [‐2.58, ‐0.42]

1.3 Number of vaso‐occlusive crises after 18 months of treatment

1

Mean Difference (IV, Fixed, 95% CI)

‐9.6 [‐10.86, ‐8.34]

2 Proportion experiencing pain Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.2

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 2 Proportion experiencing pain.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 2 Proportion experiencing pain.

3 Proportion experiencing life threatening events during study Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 3 Proportion experiencing life threatening events during study.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 3 Proportion experiencing life threatening events during study.

3.1 Acute chest syndrome

2

492

Risk Ratio (M‐H, Fixed, 95% CI)

0.43 [0.29, 0.63]

3.2 Hepatic sequestration

1

299

Risk Ratio (M‐H, Fixed, 95% CI)

0.32 [0.03, 3.06]

3.3 Stroke

2

492

Risk Ratio (M‐H, Fixed, 95% CI)

0.54 [0.12, 2.53]

3.4 Patients transfused

2

492

Risk Ratio (M‐H, Fixed, 95% CI)

0.66 [0.52, 0.82]

3.5 Splenic sequestration

1

193

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.36, 2.23]

4 Number of life‐threatening events during study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.4

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 4 Number of life‐threatening events during study.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 4 Number of life‐threatening events during study.

4.1 Blood transfusions

1

Mean Difference (IV, Fixed, 95% CI)

‐1.85 [‐2.18, ‐1.52]

4.2 Hospitalisations

1

Mean Difference (IV, Fixed, 95% CI)

‐8.89 [‐10.04, ‐7.74]

4.3 Duration of hospitalisation (days)

1

Mean Difference (IV, Fixed, 95% CI)

‐2.00 [‐4.87, ‐3.13]

5 Deaths during the study Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.5

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 5 Deaths during the study.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 5 Deaths during the study.

5.1 All deaths

3

552

Risk Ratio (M‐H, Fixed, 95% CI)

0.39 [0.08, 1.96]

5.2 Deaths related to SCD

3

552

Risk Ratio (M‐H, Fixed, 95% CI)

0.48 [0.09, 2.60]

6 Change from baseline in fetal haemoglobin (HbF %) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.6

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 6 Change from baseline in fetal haemoglobin (HbF %).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 6 Change from baseline in fetal haemoglobin (HbF %).

7 Fetal haemoglobin (HbF %) after treatment Show forest plot

2

359

Mean Difference (IV, Fixed, 95% CI)

4.07 [2.95, 5.18]

Analysis 1.7

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 7 Fetal haemoglobin (HbF %) after treatment.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 7 Fetal haemoglobin (HbF %) after treatment.

8 Change from baseline in absolute neutrophil count (x10³ per μL) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.8

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 8 Change from baseline in absolute neutrophil count (x10³ per μL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 8 Change from baseline in absolute neutrophil count (x10³ per μL).

9 Neutrophil response (10⁹/L) after treatment Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.9

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 9 Neutrophil response (10⁹/L) after treatment.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 9 Neutrophil response (10⁹/L) after treatment.

9.1 Neutrophils (x10 9/l) at 10 weeks

1

Mean Difference (IV, Fixed, 95% CI)

‐1.90 [‐2.51, ‐1.29]

9.2 Neutrophils (x10 9/l) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐1.50 [‐2.01, ‐0.99]

10 Change from baseline in haemoglobin (g/L) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.10

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 10 Change from baseline in haemoglobin (g/L).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 10 Change from baseline in haemoglobin (g/L).

11 Change from baseline in m corpuscular volume (fL) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.11

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 11 Change from baseline in m corpuscular volume (fL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 11 Change from baseline in m corpuscular volume (fL).

12 Change from baseline in white blood cells (x10³ per μL) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.12

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 12 Change from baseline in white blood cells (x10³ per μL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 12 Change from baseline in white blood cells (x10³ per μL).

13 Change from baseline in absolute reticulocyte count (x10³ per μL) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.13

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 13 Change from baseline in absolute reticulocyte count (x10³ per μL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 13 Change from baseline in absolute reticulocyte count (x10³ per μL).

14 Change from baseline in reticulocytes (%) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.14

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 14 Change from baseline in reticulocytes (%).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 14 Change from baseline in reticulocytes (%).

15 Change from baseline in total bilirubin (mg/L) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.15

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 15 Change from baseline in total bilirubin (mg/L).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 15 Change from baseline in total bilirubin (mg/L).

16 Change from baseline in platelet count (x10³ per μL) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.16

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 16 Change from baseline in platelet count (x10³ per μL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 16 Change from baseline in platelet count (x10³ per μL).

17 Haemoglobin (g/dL) Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.17

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 17 Haemoglobin (g/dL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 17 Haemoglobin (g/dL).

17.1 At 10 weeks

1

299

Mean Difference (IV, Fixed, 95% CI)

0.5 [0.19, 0.81]

17.2 At the end of the study

2

359

Mean Difference (IV, Fixed, 95% CI)

1.04 [0.82, 1.25]

18 Mean corpuscular volume (fL) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.18

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 18 Mean corpuscular volume (fL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 18 Mean corpuscular volume (fL).

18.1 At 10 weeks

1

Mean Difference (IV, Fixed, 95% CI)

12.30 [9.69, 14.91]

18.2 At 2 years

1

Mean Difference (IV, Fixed, 95% CI)

10.0 [7.34, 12.66]

19 Total bilirubin (mg/L) Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

Analysis 1.19

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 19 Total bilirubin (mg/L).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 19 Total bilirubin (mg/L).

19.1 At the end of the study

2

359

Mean Difference (IV, Fixed, 95% CI)

‐1.56 [‐1.90, ‐1.23]

20 Reticulocytes Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.20

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 20 Reticulocytes.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 20 Reticulocytes.

20.1 Reticulocytes (10⁵/mm³) at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

‐0.66 [‐0.90, ‐0.42]

20.2 Reticulocytes (10⁹/L) at 10 weeks

1

Mean Difference (IV, Fixed, 95% CI)

‐130.0 [‐152.17, ‐107.83]

20.3 Reticulocytes (10⁹/L) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐69.0 [‐91.56, ‐46.44]

21 Platelet count Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.21

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 21 Platelet count.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 21 Platelet count.

21.1 Platelet count (10³/mm³) at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

‐0.05 [‐0.16, 0.06]

21.2 Platelet count (x10⁹/L) at 10 weeks

1

Mean Difference (IV, Fixed, 95% CI)

‐35.0 [‐75.19, 5.19]

21.3 Platelet count (x10⁹/L) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐24.0 [‐51.88, 3.88]

22 Packed cell volume Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.22

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 22 Packed cell volume.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 22 Packed cell volume.

22.1 Packed cell volume (%) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

1.90 [0.85, 2.95]

23 F reticulocytes Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.23

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 23 F reticulocytes.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 23 F reticulocytes.

23.1 F reticulocytes at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

2.0 [0.18, 3.82]

24 F cells Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.24

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 24 F cells.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 24 F cells.

24.1 F cells (%) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

13.0 [8.33, 17.67]

25 Red blood count Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.25

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 25 Red blood count.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 25 Red blood count.

25.1 Red blood count (10⁶/mm³) at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

‐1.13 [‐1.24, ‐1.02]

26 White blood cells Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.26

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 26 White blood cells.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 26 White blood cells.

26.1 White blood cells (109/L) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐2.30 [‐2.97, ‐1.63]

27 Dense cells Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.27

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 27 Dense cells.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 27 Dense cells.

27.1 Dense cells (%) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐2.0 [‐3.48, ‐0.52]

28 Leucocytes Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.28

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 28 Leucocytes.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 28 Leucocytes.

28.1 Leucocytes (10³/mm³) at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

‐0.84 [‐3.07, 1.39]

29 Creatinine Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.29

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 29 Creatinine.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 29 Creatinine.

29.1 Creatinine (mg/dL) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [‐0.11, 0.11]

30 Aspartate aminotransferase Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.30

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 30 Aspartate aminotransferase.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 30 Aspartate aminotransferase.

30.1 Aspartate aminotransferase at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐4.0 [‐9.40, 1.40]

31 Alkaline phosphatase Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.31

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 31 Alkaline phosphatase.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 31 Alkaline phosphatase.

31.1 Alkaline phosphatase at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐2.0 [‐15.78, 11.78]

32 Change from baseline in growth Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.32

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 32 Change from baseline in growth.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 32 Change from baseline in growth.

32.1 Height (cm)

1

Mean Difference (Fixed, 95% CI)

‐0.2 [1.00, 0.60]

32.2 Weight (kg)

1

Mean Difference (Fixed, 95% CI)

0.10 [‐0.20, 0.40]

32.3 Head circumference (cm)

1

Mean Difference (Fixed, 95% CI)

‐0.2 [‐0.60, 0.20]

33 Quality of life: general health perception Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.33

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 33 Quality of life: general health perception.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 33 Quality of life: general health perception.

33.1 General health perception at 6 months

1

Mean Difference (IV, Fixed, 95% CI)

0.30 [‐0.54, 1.14]

33.2 General health perception at 1 year

1

Mean Difference (IV, Fixed, 95% CI)

0.60 [‐0.18, 1.38]

33.3 General health perception at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

0.9 [0.08, 1.72]

33.4 General health perception at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

0.4 [‐0.51, 1.31]

34 Quality of life: pain recall Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.34

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 34 Quality of life: pain recall.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 34 Quality of life: pain recall.

34.1 Pain recall at 6 months

1

Mean Difference (IV, Fixed, 95% CI)

0.40 [‐0.13, 0.93]

34.2 Pain recall at 1 year

1

Mean Difference (IV, Fixed, 95% CI)

0.40 [‐0.18, 0.98]

34.3 Pain recall at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

0.70 [0.11, 1.29]

34.4 Pain recall at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

0.3 [‐0.30, 0.90]

35 Quality of life: social function Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.35

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 35 Quality of life: social function.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 35 Quality of life: social function.

35.1 Social function at 6 months

1

Mean Difference (IV, Fixed, 95% CI)

0.4 [‐0.15, 0.95]

35.2 Social function at 1 year

1

Mean Difference (IV, Fixed, 95% CI)

0.2 [‐0.36, 0.76]

35.3 Social function at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

0.40 [‐0.21, 1.01]

35.4 Social function at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

0.3 [‐0.31, 0.91]

36 Changes in 'Ladder of Life' Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 1.36

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 36 Changes in 'Ladder of Life'.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 36 Changes in 'Ladder of Life'.

36.1 Changes in 'Ladder of Life' at 6 months

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [‐0.52, 0.52]

36.2 Changes in 'Ladder of Life' at 1 year

1

Mean Difference (IV, Fixed, 95% CI)

0.4 [‐0.15, 0.95]

36.3 Changes in 'Ladder of Life' at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

0.30 [‐0.22, 0.82]

36.4 Changes in 'Ladder of Life' at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

0.3 [‐0.23, 0.83]

37 Proportion of participants with signs of organ damage Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.37

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 37 Proportion of participants with signs of organ damage.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 37 Proportion of participants with signs of organ damage.

37.1 New leg ulcers

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.85 [0.44, 1.64]

37.2 Aseptic necrosis (humerus or femur)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.39, 2.37]

37.3 Decreased spleen function at exit (compared to baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.72 [0.44, 1.16]

38 Signs of organ damage ‐ change from baseline in DTPA GFR Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.38

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 38 Signs of organ damage ‐ change from baseline in DTPA GFR.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 38 Signs of organ damage ‐ change from baseline in DTPA GFR.

39 Signs of organ damage ‐ change from baseline in Howell‐Jolley body (per 106 red blood cells) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.39

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 39 Signs of organ damage ‐ change from baseline in Howell‐Jolley body (per 106 red blood cells).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 39 Signs of organ damage ‐ change from baseline in Howell‐Jolley body (per 106 red blood cells).

40 Signs of organ damage ‐ change from baseline in pitted cells (%) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.40

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 40 Signs of organ damage ‐ change from baseline in pitted cells (%).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 40 Signs of organ damage ‐ change from baseline in pitted cells (%).

41 Signs of organ damage ‐ change from baseline in spleen: liver ratio of counts Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.41

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 41 Signs of organ damage ‐ change from baseline in spleen: liver ratio of counts.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 41 Signs of organ damage ‐ change from baseline in spleen: liver ratio of counts.

42 Signs of organ damage ‐ change from baseline in spleen volume (cm3) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.42

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 42 Signs of organ damage ‐ change from baseline in spleen volume (cm3).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 42 Signs of organ damage ‐ change from baseline in spleen volume (cm3).

43 Signs of organ damage ‐ change from baseline in creatinine (mg/L) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.43

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 43 Signs of organ damage ‐ change from baseline in creatinine (mg/L).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 43 Signs of organ damage ‐ change from baseline in creatinine (mg/L).

44 Signs of organ damage ‐ change from baseline in Schwartz GFR Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.44

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 44 Signs of organ damage ‐ change from baseline in Schwartz GFR.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 44 Signs of organ damage ‐ change from baseline in Schwartz GFR.

44.1 Schwartz glomerular filtration rate (mL/min per 1.73m2)

1

Mean Difference (Fixed, 95% CI)

‐8.0 [‐35.00, 21.00]

45 Signs of organ damage ‐ change from baseline in cystatin C Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.45

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 45 Signs of organ damage ‐ change from baseline in cystatin C.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 45 Signs of organ damage ‐ change from baseline in cystatin C.

46 Signs of organ damage ‐ change from baseline in urine osmolality Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.46

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 46 Signs of organ damage ‐ change from baseline in urine osmolality.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 46 Signs of organ damage ‐ change from baseline in urine osmolality.

47 Signs of organ damage ‐ change from baseline in urine pH Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.47

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 47 Signs of organ damage ‐ change from baseline in urine pH.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 47 Signs of organ damage ‐ change from baseline in urine pH.

48 Signs of organ damage ‐ change from baseline in urine‐specific gravity Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.48

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 48 Signs of organ damage ‐ change from baseline in urine‐specific gravity.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 48 Signs of organ damage ‐ change from baseline in urine‐specific gravity.

49 Signs of organ damage ‐ change from baseline in total kidney volume Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.49

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 49 Signs of organ damage ‐ change from baseline in total kidney volume.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 49 Signs of organ damage ‐ change from baseline in total kidney volume.

50 Signs of organ damage ‐ change from baseline in TCD ultrasound velocity (time‐averaged mean maximum velocity) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.50

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 50 Signs of organ damage ‐ change from baseline in TCD ultrasound velocity (time‐averaged mean maximum velocity).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 50 Signs of organ damage ‐ change from baseline in TCD ultrasound velocity (time‐averaged mean maximum velocity).

51 Signs of organ damage ‐ change from baseline in CNS measures Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

Analysis 1.51

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 51 Signs of organ damage ‐ change from baseline in CNS measures.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 51 Signs of organ damage ‐ change from baseline in CNS measures.

51.1 Bayley Mental Development Index

1

Mean Difference (Fixed, 95% CI)

3.0 [0.00, 8.00]

51.2 Bayley motor performance development index

1

Mean Difference (Fixed, 95% CI)

2.0 [‐1.00, 7.00]

52 Proportion of participants experiencing adverse events and toxicity Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 1.52

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 52 Proportion of participants experiencing adverse events and toxicity.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 52 Proportion of participants experiencing adverse events and toxicity.

52.1 hair loss at 1 or 2 visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.25 [0.69, 2.26]

52.2 hair loss at 3 or more visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.61 [0.31, 1.21]

52.3 skin rash at 1 or 2 visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.62, 1.51]

52.4 skin rash at 3 or more visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.58, 1.60]

52.5 fever at 1 or 2 visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.55, 1.69]

52.6 fever at 3 or more visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.68, 1.17]

52.7 Gastroinestinal disturbance at 1 or 2 visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.83 [0.50, 1.36]

52.8 Gastrointestinal disturbance at 3 or more visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.74, 1.31]

52.9 Other abnormalities at 1 or 2 visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.60 [0.33, 1.11]

52.10 Other abnormalities at 3 or more visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.83, 1.40]

52.11 Hospitalisation (for any reason)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.83 [0.72, 0.96]

52.12 Dactylitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.34 [0.20, 0.58]

52.13 Priapism

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.52 [0.26, 8.87]

52.14 Sepsis or bacteraemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.40 [0.08, 2.03]

52.15 Splenomegaly

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.61, 1.32]

52.16 Absolute Neutrophil Count < 500

1

Risk Ratio (M‐H, Fixed, 95% CI)

2.53 [0.50, 12.71]

52.17 Absolute Neutrophil 500 ‐ 1250

1

Risk Ratio (M‐H, Fixed, 95% CI)

2.53 [1.58, 4.03]

52.18 Thrombocytopaenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.59 [0.64, 3.92]

52.19 Alanine transaminase > 150 U/L

1

Risk Ratio (M‐H, Fixed, 95% CI)

2.02 [0.19, 21.92]

52.20 Severe anaemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.51 [0.05, 5.48]

52.21 Bilirubin

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.34 [0.01, 8.17]

52.22 Creatinine

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

52.23 Skin and subcutaneous disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.91 [0.75, 1.10]

52.24 Splenic sequestration

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.36, 2.23]

52.25 Gastroenteritis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.44 [0.28, 0.71]

52.26 Nausea

1

Risk Ratio (M‐H, Fixed, 95% CI)

5.0 [0.25, 99.95]

52.27 Skin Rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

7.0 [0.38, 129.93]

Open in table viewer
Comparison 2. Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion experiencing pain Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.1

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 1 Proportion experiencing pain.

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 1 Proportion experiencing pain.

1.1 Vaso‐occlusive or sickle cell‐related pain (all)

1

133

Risk Ratio (M‐H, Fixed, 95% CI)

1.03 [0.81, 1.30]

1.2 Vaso‐occlusive or sickle cell‐related pain (serious)

2

254

Risk Ratio (M‐H, Fixed, 95% CI)

3.37 [1.59, 7.11]

2 Proportion experiencing life‐threatening events during study Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.2

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 2 Proportion experiencing life‐threatening events during study.

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 2 Proportion experiencing life‐threatening events during study.

2.1 Stroke (secondary)

1

133

Risk Ratio (M‐H, Fixed, 95% CI)

14.78 [0.86, 253.66]

2.2 Transient ischaemic attack (primary)

1

121

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.21, 4.84]

2.3 Transient ischaemic attack (secondary)

1

133

Risk Ratio (M‐H, Fixed, 95% CI)

0.66 [0.25, 1.74]

2.4 Other neurological event (primary)

1

121

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.07, 15.88]

2.5 Acute chest syndrome

2

254

Risk Ratio (M‐H, Fixed, 95% CI)

2.84 [1.25, 6.42]

2.6 Infections and Infestations

2

254

Risk Ratio (M‐H, Fixed, 95% CI)

3.65 [1.05, 12.76]

2.7 Splenic sequestration or splenectomy

2

254

Risk Ratio (M‐H, Fixed, 95% CI)

0.34 [0.01, 8.16]

2.8 Hepatobiliary disease

1

121

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.07, 15.88]

2.9 Total with serious adverse events

2

254

Risk Ratio (M‐H, Fixed, 95% CI)

1.93 [1.17, 3.20]

2.10 Total with sickle cell related, non‐neurological adverse events

1

133

Risk Ratio (M‐H, Fixed, 95% CI)

3.10 [1.42, 6.75]

3 Deaths during the study Show forest plot

2

254

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.06, 15.42]

Analysis 2.3

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 3 Deaths during the study.

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 3 Deaths during the study.

4 Change from baseline in fetal haemoglobin (HbF %) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.4

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 4 Change from baseline in fetal haemoglobin (HbF %).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 4 Change from baseline in fetal haemoglobin (HbF %).

5 Change from baseline in absolute neutrophil count (x10⁹/L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.5

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 5 Change from baseline in absolute neutrophil count (x10⁹/L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 5 Change from baseline in absolute neutrophil count (x10⁹/L).

6 Change from baseline in mean corpuscular volume (fL) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.6

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 6 Change from baseline in mean corpuscular volume (fL).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 6 Change from baseline in mean corpuscular volume (fL).

7 Change from baseline in sickle haemoglobin (%) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.7

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 7 Change from baseline in sickle haemoglobin (%).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 7 Change from baseline in sickle haemoglobin (%).

8 Change from baseline in haemoglobin (g/L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.8

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 8 Change from baseline in haemoglobin (g/L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 8 Change from baseline in haemoglobin (g/L).

9 Change from baseline in absolute reticulocyte count (10⁹ / L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.9

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 9 Change from baseline in absolute reticulocyte count (10⁹ / L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 9 Change from baseline in absolute reticulocyte count (10⁹ / L).

10 Change from baseline in white blood count (10⁹ / L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.10

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 10 Change from baseline in white blood count (10⁹ / L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 10 Change from baseline in white blood count (10⁹ / L).

11 Change from baseline in platelets (10⁹ / L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.11

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 11 Change from baseline in platelets (10⁹ / L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 11 Change from baseline in platelets (10⁹ / L).

12 Change from baseline in total bilirubin (mg/L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.12

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 12 Change from baseline in total bilirubin (mg/L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 12 Change from baseline in total bilirubin (mg/L).

13 Change from baseline in liver iron concentration Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.13

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 13 Change from baseline in liver iron concentration.

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 13 Change from baseline in liver iron concentration.

14 Change from baseline in serum ferritin (ng/mL) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.14

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 14 Change from baseline in serum ferritin (ng/mL).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 14 Change from baseline in serum ferritin (ng/mL).

15 Change from baseline in lactate dehydrogenase (U/L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.15

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 15 Change from baseline in lactate dehydrogenase (U/L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 15 Change from baseline in lactate dehydrogenase (U/L).

16 Signs of organ damage ‐ CNS measures at the end of the study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 2.16

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 16 Signs of organ damage ‐ CNS measures at the end of the study.

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 16 Signs of organ damage ‐ CNS measures at the end of the study.

16.1 Final TCD ultrasound velocity

1

Mean Difference (IV, Fixed, 95% CI)

‐5.0 [‐9.16, ‐0.84]

17 Proportion of participants experiencing non‐neurological adverse events and toxicity Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 2.17

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 17 Proportion of participants experiencing non‐neurological adverse events and toxicity.

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 17 Proportion of participants experiencing non‐neurological adverse events and toxicity.

17.1 Infections and infestations

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.24 [0.89, 1.72]

17.2 Gastrointestinal disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.16 [0.74, 1.80]

17.3 Fever

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.66, 1.75]

17.4 Musculoskelatal disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.14 [0.59, 2.20]

17.5 Immune system disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.12 [0.02, 0.96]

17.6 Cholelithiasis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.74 [0.17, 3.17]

17.7 Cholecystitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.97 [0.18, 21.21]

17.8 Asthma

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.74 [0.17, 3.17]

17.9 Acute chest syndrome

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.72 [0.53, 5.61]

17.10 Renal or urinary disorder

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.66 [0.11, 3.80]

17.11 Priapism

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.97 [0.18, 21.21]

17.12 Cathether‐related complications

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.48 [0.26, 8.56]

17.13 Cardiac disorder

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.06, 15.42]

17.14 Hyderbilirubianemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.49 [0.24, 1.02]

17.15 Alanine transaminase increase

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.55, 2.01]

17.16 Aspartate transaminase increase

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.38 [0.66, 2.88]

17.17 Serum creatinine increase

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.97 [0.51, 7.55]

17.18 Thrombocytopenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

6.90 [0.87, 54.51]

17.19 Reticuloctopenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

6.90 [2.16, 22.02]

17.20 Neutropenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

9.85 [1.30, 74.80]

17.21 Anaemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

6.57 [2.05, 21.05]

17.22 Sickle cell pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.29 [0.92, 1.82]

17.23 Sickle cell‐related events

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.03 [0.81, 1.30]

17.24 All adverse events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.92, 1.05]

Open in table viewer
Comparison 3. Hydroxyurea compared to observation for participants with SCD and risk of stroke

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion experiencing life‐threatening events during the study Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.1

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 1 Proportion experiencing life‐threatening events during the study.

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 1 Proportion experiencing life‐threatening events during the study.

1.1 Vaso‐occlusive events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.4 [0.10, 1.64]

1.2 Acute splenic sequestration

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.07, 14.05]

1.3 Blood transfusions required

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.04, 2.73]

2 Signs of organ damage ‐ proportion of participants with a change in CNS measures Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.2

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 2 Signs of organ damage ‐ proportion of participants with a change in CNS measures.

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 2 Signs of organ damage ‐ proportion of participants with a change in CNS measures.

2.1 Participants converting from conditional to abnormal TCD ultrasound velocity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.2 [0.03, 1.45]

3 Signs of organ damage ‐ change from baseline in CNS measures Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

Analysis 3.3

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 3 Signs of organ damage ‐ change from baseline in CNS measures.

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 3 Signs of organ damage ‐ change from baseline in CNS measures.

3.1 TCD ultrasound velocity (time‐averaged mean maximum velocity)

1

Mean Difference (IV, Fixed, 95% CI)

‐25.7 [‐45.38, ‐6.02]

4 Adverse events and toxicity Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

Analysis 3.4

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 4 Adverse events and toxicity.

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 4 Adverse events and toxicity.

4.1 Dizziness

1

Risk Ratio (M‐H, Fixed, 95% CI)

3.0 [0.14, 66.53]

4.2 Headaches

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.07, 14.05]

4.3 Transient neutropenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

3.0 [0.14, 66.53]

4.4 Reticulocytopenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.07, 14.05]

4.5 Parasite infection

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.07, 14.05]

Study flow diagram.
Figures and Tables -
Figure 1

Study flow diagram.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figures and Tables -
Figure 2

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 1 Pain crises.
Figures and Tables -
Analysis 1.1

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 1 Pain crises.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 2 Proportion experiencing pain.
Figures and Tables -
Analysis 1.2

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 2 Proportion experiencing pain.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 3 Proportion experiencing life threatening events during study.
Figures and Tables -
Analysis 1.3

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 3 Proportion experiencing life threatening events during study.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 4 Number of life‐threatening events during study.
Figures and Tables -
Analysis 1.4

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 4 Number of life‐threatening events during study.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 5 Deaths during the study.
Figures and Tables -
Analysis 1.5

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 5 Deaths during the study.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 6 Change from baseline in fetal haemoglobin (HbF %).
Figures and Tables -
Analysis 1.6

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 6 Change from baseline in fetal haemoglobin (HbF %).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 7 Fetal haemoglobin (HbF %) after treatment.
Figures and Tables -
Analysis 1.7

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 7 Fetal haemoglobin (HbF %) after treatment.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 8 Change from baseline in absolute neutrophil count (x10³ per μL).
Figures and Tables -
Analysis 1.8

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 8 Change from baseline in absolute neutrophil count (x10³ per μL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 9 Neutrophil response (10⁹/L) after treatment.
Figures and Tables -
Analysis 1.9

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 9 Neutrophil response (10⁹/L) after treatment.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 10 Change from baseline in haemoglobin (g/L).
Figures and Tables -
Analysis 1.10

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 10 Change from baseline in haemoglobin (g/L).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 11 Change from baseline in m corpuscular volume (fL).
Figures and Tables -
Analysis 1.11

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 11 Change from baseline in m corpuscular volume (fL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 12 Change from baseline in white blood cells (x10³ per μL).
Figures and Tables -
Analysis 1.12

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 12 Change from baseline in white blood cells (x10³ per μL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 13 Change from baseline in absolute reticulocyte count (x10³ per μL).
Figures and Tables -
Analysis 1.13

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 13 Change from baseline in absolute reticulocyte count (x10³ per μL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 14 Change from baseline in reticulocytes (%).
Figures and Tables -
Analysis 1.14

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 14 Change from baseline in reticulocytes (%).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 15 Change from baseline in total bilirubin (mg/L).
Figures and Tables -
Analysis 1.15

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 15 Change from baseline in total bilirubin (mg/L).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 16 Change from baseline in platelet count (x10³ per μL).
Figures and Tables -
Analysis 1.16

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 16 Change from baseline in platelet count (x10³ per μL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 17 Haemoglobin (g/dL).
Figures and Tables -
Analysis 1.17

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 17 Haemoglobin (g/dL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 18 Mean corpuscular volume (fL).
Figures and Tables -
Analysis 1.18

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 18 Mean corpuscular volume (fL).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 19 Total bilirubin (mg/L).
Figures and Tables -
Analysis 1.19

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 19 Total bilirubin (mg/L).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 20 Reticulocytes.
Figures and Tables -
Analysis 1.20

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 20 Reticulocytes.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 21 Platelet count.
Figures and Tables -
Analysis 1.21

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 21 Platelet count.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 22 Packed cell volume.
Figures and Tables -
Analysis 1.22

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 22 Packed cell volume.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 23 F reticulocytes.
Figures and Tables -
Analysis 1.23

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 23 F reticulocytes.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 24 F cells.
Figures and Tables -
Analysis 1.24

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 24 F cells.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 25 Red blood count.
Figures and Tables -
Analysis 1.25

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 25 Red blood count.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 26 White blood cells.
Figures and Tables -
Analysis 1.26

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 26 White blood cells.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 27 Dense cells.
Figures and Tables -
Analysis 1.27

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 27 Dense cells.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 28 Leucocytes.
Figures and Tables -
Analysis 1.28

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 28 Leucocytes.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 29 Creatinine.
Figures and Tables -
Analysis 1.29

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 29 Creatinine.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 30 Aspartate aminotransferase.
Figures and Tables -
Analysis 1.30

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 30 Aspartate aminotransferase.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 31 Alkaline phosphatase.
Figures and Tables -
Analysis 1.31

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 31 Alkaline phosphatase.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 32 Change from baseline in growth.
Figures and Tables -
Analysis 1.32

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 32 Change from baseline in growth.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 33 Quality of life: general health perception.
Figures and Tables -
Analysis 1.33

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 33 Quality of life: general health perception.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 34 Quality of life: pain recall.
Figures and Tables -
Analysis 1.34

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 34 Quality of life: pain recall.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 35 Quality of life: social function.
Figures and Tables -
Analysis 1.35

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 35 Quality of life: social function.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 36 Changes in 'Ladder of Life'.
Figures and Tables -
Analysis 1.36

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 36 Changes in 'Ladder of Life'.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 37 Proportion of participants with signs of organ damage.
Figures and Tables -
Analysis 1.37

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 37 Proportion of participants with signs of organ damage.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 38 Signs of organ damage ‐ change from baseline in DTPA GFR.
Figures and Tables -
Analysis 1.38

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 38 Signs of organ damage ‐ change from baseline in DTPA GFR.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 39 Signs of organ damage ‐ change from baseline in Howell‐Jolley body (per 106 red blood cells).
Figures and Tables -
Analysis 1.39

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 39 Signs of organ damage ‐ change from baseline in Howell‐Jolley body (per 106 red blood cells).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 40 Signs of organ damage ‐ change from baseline in pitted cells (%).
Figures and Tables -
Analysis 1.40

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 40 Signs of organ damage ‐ change from baseline in pitted cells (%).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 41 Signs of organ damage ‐ change from baseline in spleen: liver ratio of counts.
Figures and Tables -
Analysis 1.41

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 41 Signs of organ damage ‐ change from baseline in spleen: liver ratio of counts.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 42 Signs of organ damage ‐ change from baseline in spleen volume (cm3).
Figures and Tables -
Analysis 1.42

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 42 Signs of organ damage ‐ change from baseline in spleen volume (cm3).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 43 Signs of organ damage ‐ change from baseline in creatinine (mg/L).
Figures and Tables -
Analysis 1.43

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 43 Signs of organ damage ‐ change from baseline in creatinine (mg/L).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 44 Signs of organ damage ‐ change from baseline in Schwartz GFR.
Figures and Tables -
Analysis 1.44

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 44 Signs of organ damage ‐ change from baseline in Schwartz GFR.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 45 Signs of organ damage ‐ change from baseline in cystatin C.
Figures and Tables -
Analysis 1.45

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 45 Signs of organ damage ‐ change from baseline in cystatin C.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 46 Signs of organ damage ‐ change from baseline in urine osmolality.
Figures and Tables -
Analysis 1.46

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 46 Signs of organ damage ‐ change from baseline in urine osmolality.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 47 Signs of organ damage ‐ change from baseline in urine pH.
Figures and Tables -
Analysis 1.47

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 47 Signs of organ damage ‐ change from baseline in urine pH.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 48 Signs of organ damage ‐ change from baseline in urine‐specific gravity.
Figures and Tables -
Analysis 1.48

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 48 Signs of organ damage ‐ change from baseline in urine‐specific gravity.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 49 Signs of organ damage ‐ change from baseline in total kidney volume.
Figures and Tables -
Analysis 1.49

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 49 Signs of organ damage ‐ change from baseline in total kidney volume.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 50 Signs of organ damage ‐ change from baseline in TCD ultrasound velocity (time‐averaged mean maximum velocity).
Figures and Tables -
Analysis 1.50

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 50 Signs of organ damage ‐ change from baseline in TCD ultrasound velocity (time‐averaged mean maximum velocity).

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 51 Signs of organ damage ‐ change from baseline in CNS measures.
Figures and Tables -
Analysis 1.51

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 51 Signs of organ damage ‐ change from baseline in CNS measures.

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 52 Proportion of participants experiencing adverse events and toxicity.
Figures and Tables -
Analysis 1.52

Comparison 1 Hydroxyurea versus placebo for participants with sickle cell disease, Outcome 52 Proportion of participants experiencing adverse events and toxicity.

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 1 Proportion experiencing pain.
Figures and Tables -
Analysis 2.1

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 1 Proportion experiencing pain.

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 2 Proportion experiencing life‐threatening events during study.
Figures and Tables -
Analysis 2.2

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 2 Proportion experiencing life‐threatening events during study.

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 3 Deaths during the study.
Figures and Tables -
Analysis 2.3

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 3 Deaths during the study.

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 4 Change from baseline in fetal haemoglobin (HbF %).
Figures and Tables -
Analysis 2.4

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 4 Change from baseline in fetal haemoglobin (HbF %).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 5 Change from baseline in absolute neutrophil count (x10⁹/L).
Figures and Tables -
Analysis 2.5

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 5 Change from baseline in absolute neutrophil count (x10⁹/L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 6 Change from baseline in mean corpuscular volume (fL).
Figures and Tables -
Analysis 2.6

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 6 Change from baseline in mean corpuscular volume (fL).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 7 Change from baseline in sickle haemoglobin (%).
Figures and Tables -
Analysis 2.7

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 7 Change from baseline in sickle haemoglobin (%).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 8 Change from baseline in haemoglobin (g/L).
Figures and Tables -
Analysis 2.8

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 8 Change from baseline in haemoglobin (g/L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 9 Change from baseline in absolute reticulocyte count (10⁹ / L).
Figures and Tables -
Analysis 2.9

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 9 Change from baseline in absolute reticulocyte count (10⁹ / L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 10 Change from baseline in white blood count (10⁹ / L).
Figures and Tables -
Analysis 2.10

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 10 Change from baseline in white blood count (10⁹ / L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 11 Change from baseline in platelets (10⁹ / L).
Figures and Tables -
Analysis 2.11

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 11 Change from baseline in platelets (10⁹ / L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 12 Change from baseline in total bilirubin (mg/L).
Figures and Tables -
Analysis 2.12

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 12 Change from baseline in total bilirubin (mg/L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 13 Change from baseline in liver iron concentration.
Figures and Tables -
Analysis 2.13

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 13 Change from baseline in liver iron concentration.

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 14 Change from baseline in serum ferritin (ng/mL).
Figures and Tables -
Analysis 2.14

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 14 Change from baseline in serum ferritin (ng/mL).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 15 Change from baseline in lactate dehydrogenase (U/L).
Figures and Tables -
Analysis 2.15

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 15 Change from baseline in lactate dehydrogenase (U/L).

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 16 Signs of organ damage ‐ CNS measures at the end of the study.
Figures and Tables -
Analysis 2.16

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 16 Signs of organ damage ‐ CNS measures at the end of the study.

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 17 Proportion of participants experiencing non‐neurological adverse events and toxicity.
Figures and Tables -
Analysis 2.17

Comparison 2 Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke, Outcome 17 Proportion of participants experiencing non‐neurological adverse events and toxicity.

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 1 Proportion experiencing life‐threatening events during the study.
Figures and Tables -
Analysis 3.1

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 1 Proportion experiencing life‐threatening events during the study.

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 2 Signs of organ damage ‐ proportion of participants with a change in CNS measures.
Figures and Tables -
Analysis 3.2

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 2 Signs of organ damage ‐ proportion of participants with a change in CNS measures.

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 3 Signs of organ damage ‐ change from baseline in CNS measures.
Figures and Tables -
Analysis 3.3

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 3 Signs of organ damage ‐ change from baseline in CNS measures.

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 4 Adverse events and toxicity.
Figures and Tables -
Analysis 3.4

Comparison 3 Hydroxyurea compared to observation for participants with SCD and risk of stroke, Outcome 4 Adverse events and toxicity.

Summary of findings for the main comparison. Summary of findings ‐ Hydroxyurea compared with placebo for sickle cell disease

Hydroxyurea compared with placebo for sickle cell disease

Patient or population: adults and children with sickle cell disease

Settings: outpatients

Intervention: hydroxyurea

Comparison: placebo

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Placebo

Hydroxyurea

Pain alteration1

Follow‐up: 6 ‐ 24 months

See comment

See comment

NA

577

(4 studies)2

⊕⊕⊕⊝
moderate5

All studies showed a significant advantage to hydroxyurea compared to placebo (different measures of pain alteration presented)1.

Life‐threatening illness

Follow‐up: 6 ‐ 24 months

See comment

See comment

NA

552

(3 studies)

⊕⊕⊕⊝
moderate5

Significantly fewer occurrences of ACS (2 studies) and transfusions (3 studies) on hydroxyurea compared to placebo. No significant differences in terms of stroke, hepatic or splenic sequestration (two studies).

Death during the study (all deaths)

Follow‐up: 6 ‐ 24 months

26 per 1000

10 per 1000

(0 to 51 per 1000)

RR 0.39 (0.08 to 1.96)

577

(4 studies)2

⊕⊕⊕⊝
moderate5

There was also no significant difference between groups in terms of deaths related to SCD.

Measures of HbF (%)

Follow‐up: 6 ‐ 24 months

See comment

See comment

NA

577

(4 studies)2

⊕⊕⊕⊝
moderate5

There was a significant increase in HbF(%) in the hydroxyurea group compared to the placebo group in all studies (different measures presented)3.

Measures of ANC

Follow‐up: 6 ‐ 24 months

See comment

See comment

NA

517

(3 studies)2

⊕⊕⊕⊝
moderate5

There was a significant decrease in ANC in the hydroxyurea group compared to the placebo group in all studies (different measures presented)3.

Quality of life: 'Health Status Survey' the 'Profile of Mood States' and the 'Ladder of Life'

Follow‐up: 24 months

See comment

See comment

NA

up to 277

(1 study)

⊕⊕⊝⊝

low5,6

No significant difference in terms of any domain of any scale except for pain recall at 18 months (MD 0.70, 95% CI 0.11 to 1.29, P = 0.02)4.

Adverse events or toxicity: differences in rates of specific adverse events

Follow‐up: 6 ‐ 24 months

See comment

See comment

NA

577

(4 studies)2

⊕⊕⊝⊝

low5,7

Significantly fewer events of dactylitis and gastroenteritis on hydroxyurea compared to placebo.

No significant differences between groups in terms of all other events.

The basis for the assumed risk is the event rate in the control group unless otherwise stated in the comments and footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
ACS: acute chest syndrome; ANC: absolute neutrophil counts; CI: confidence interval; HbF: fetal haemoglobin; MD: mean difference;NA: not applicable; RR: risk ratio; SCD: sickle cell disease.

GRADE Working Group grades of evidence
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

  1. Pain alteration measured by mean annual pain crisis rate, time to initiation of treatment to first, second or third crisis, number of vaso‐occlusive crises, proportion of participants experiencing pain, proportion of hospitalisation for painful episodes.

  2. One study of 25 participants is of a cross‐over design (Belgian Study 1996). Participants are counted only once in this total.

  3. Different measures presented ‐ change from baseline or post intervention measures ‐ therefore, data from all studies could not be pooled.

  4. Within the study (MSH 1995, reported in Ballas 2006), to allow for multiple statistical testing of the quality of life domains, a P value < 0.01 was considered significant. Therefore this result not interpreted as significant in the study publication.

  5. Downgraded once due to applicability: only individuals with HbSS or HbSβº‐thalassemia genotypes were included therefore results are not applicable to individuals with HbSC genotype.

  6. Downgraded once due to imprecision/uncertainty: caution is encouraged regarding the interpretation of these results as not all participants contributed data to all quality of life domains and the study publication defines statistical significance differently to this review.

  7. Downgraded once due to imprecision/uncertainty: caution is encouraged regarding the interpretation of these results due to the number of separate outcomes considered in analysis and the increased probability of a statistical type I error.

Figures and Tables -
Summary of findings for the main comparison. Summary of findings ‐ Hydroxyurea compared with placebo for sickle cell disease
Summary of findings 2. Summary of findings ‐ Hydroxyurea and phlebotomy compared to transfusion and chelation for people with sickle cell disease and an increased risk of stroke

Hydroxyurea and phlebotomy compared to transfusion and chelation for people with sickle cell disease and an increased risk of stroke

Patient or population: adults and children with sickle cell disease and an increased risk of stroke

Settings: outpatients

Intervention: hydroxyurea and phlebotomy

Comparison: transfusion and chelation

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Transfusion and chelation

Hydroxyurea and phlebotomy

Pain alteration: proportion experiencing serious VOC or sickle‐related pain events

Follow‐up: 24 to 30 months

213 per 1000

718 per 1000

(339 to 1000 per 1000)

RR 3.37 (95% CI 1.59 to 7.11)

254

(2 studies)

⊕⊕⊝⊝

low4,5

No significant difference between treatment groups in terms of all pain events (serious and non‐serious) in 1 study (RR 1.03, 95% CI 0.81 to 1.30).

Life‐threatening illness

Follow‐up: 24 ‐ 30 months

See comment

See comment

NA

254

(2 studies)

⊕⊕⊕⊝
moderate4

No significant difference between groups in life‐threatening neurological events, hepatobiliary disease and splenic sequestration; significantly more ACS and infections and infestations in the hydroxyurea and phlebotomy compared to the transfusion and chelation group.

Death during the study (all deaths)

Follow‐up: 24 ‐ 30 months

1 death occurred in the transfusion and chelation group of 1 study1.

1 death occurred in the hydroxyurea and phlebotomy group of 1 study1.

RR 0.99 (95% CI 0.06 to 15.42)

254

(2 studies)

⊕⊕⊝⊝

low4,5

Measures of HbF (%)

Follow‐up: 24 to 30 months

See comment

See comment

NA

254

(2 studies)

⊕⊕⊕⊝
moderate4

There was a significant increase in HbF(%) in the hydroxyurea and phlebotomy group compared to the transfusion and chelation group for both studies (different measures presented)2.

Measures of ANC

Follow‐up: 24 to 30 months

See comment

See comment

NA

254

(2 studies)

⊕⊕⊕⊝
moderate4

There was a significant decrease in ANC in the hydroxyurea and phlebotomy group compared to the transfusion and chelation group for both studies (different measures presented)2.

Quality of life

Outcome not reported3

NA

Adverse events or toxicity: differences in rates of specific adverse events

See comment

See comment

NA

254

(2 studies)

⊕⊕⊝⊝

low4,6

There was a statistically significant difference in terms of immune disorders (more in transfusion and chelation group), reticulocytopenia, neutropenia and anaemia (more in hydroxyurea and phlebotomy group) in 1 study and the rate of adverse events was balanced across groups in the other study.

The basis for the assumed risk is the event rate in the control group unless otherwise stated in the comments and footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
ACS: acute chest syndrome; ANC: absolute neutrophil counts; CI: confidence interval; HbF: fetal haemoglobin; NA: not applicable; RR: risk ratio; VOC: vaso‐occlusive crisis.

GRADE Working Group grades of evidence
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

1. Absolute data presented for number of deaths as the confidence interval of the relative effect very large due to the small number of events.
2. Different measures presented ‐ mean or median change from baseline ‐ therefore data from all studies could not be pooled.
3. Quality of life data was collected in TWiTCH 2016; to date, primary results of this study have been published but not quality of life data. When available, quality of life data will be included in an update of this review.
4. Downgraded once due to applicability: only children with HbSS or HbSβº‐thalassemia were included therefore results are not applicable to adults or individuals with HbSC genotype.
5. Downgraded once due to imprecision: small number of events and large CI around the relative effect.
6. Downgraded once due to imprecision/uncertainty: caution is encouraged regarding the interpretation of these results due to the number of separate outcomes considered in analysis and the increased probability of a statistical type I error.

Figures and Tables -
Summary of findings 2. Summary of findings ‐ Hydroxyurea and phlebotomy compared to transfusion and chelation for people with sickle cell disease and an increased risk of stroke
Summary of findings 3. Summary of findings ‐ Hydroxyurea compared with observation for people with sickle cell disease and an increased risk of stroke

Hydroxyurea compared with observation for people with sickle cell disease and an increased risk of stroke

Patient or population: adults and children with sickle cell disease and an increased risk of stroke

Settings: outpatients

Intervention: hydroxyurea

Comparison: observation

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

Observation

Hydroxyurea

Pain alteration

Follow‐up: NA

Outcome not reported

NA

Life‐threatening illness

Follow‐up: 15 months

See comment

See comment

NA

22

(1 study)

⊕⊝⊝⊝
very low3,4

No significant differences between groups in terms of ACS, blood transfusions required or acute splenic sequestration.

Death during the study

Follow‐up: 15 months

No deaths occurred

No deaths occurred

NA

22

(1 study)

⊕⊝⊝⊝
very low3,4

Measures of HbF

Follow‐up: 15 months

See comment

See comment

NA

22

(1 study)

⊕⊝⊝⊝
very low3,4

There was a significant increase in HbF in the hydroxyurea group compared to the observation group1.

Measures of ANC

Follow‐up: 15 months

See comment

See comment

NA

22

(1 study)

⊕⊝⊝⊝
very low3,4

There was a significant decrease in ANC in the hydroxyurea group compared to the observation group1.

Quality of Life

Follow‐up: NA

Outcome not reported2

NA

Adverse events or toxicity: differences in rates of specific adverse events

Follow‐up: 15 months

See comment

See comment

NA

22

(1 study)

⊕⊝⊝⊝
very low3,4

No significant differences between groups in terms of transient neutropenia, reticulocytopenia, parasite infestation, headache and dizziness.

The basis for the assumed risk is the event rate in the control group unless otherwise stated in the comments and footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
ACS: acute chest syndrome; ANC: absolute neutrophil counts; CI: confidence interval; HbF: fetal haemoglobin; NA: not applicable; RR: risk ratio.

GRADE Working Group grades of evidence
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

1. Median values reported so data cannot be entered into analysis.
2. Outcome was not collected or presented due to early termination of study.
3. Downgraded twice due to serious imprecision: study terminated early with only 22 of target 100 participants recruited. Small number of participants included in final analyses which are likely to be underpowered.
4. Downgraded once due to applicability: only children with HbSS or HbSβº‐thalassemia were included therefore results are not applicable to adults or individuals with HbSC genotype.

Figures and Tables -
Summary of findings 3. Summary of findings ‐ Hydroxyurea compared with observation for people with sickle cell disease and an increased risk of stroke
Summary of findings 4. Summary of findings ‐ Hydroxyurea compared with no hydroxyurea for people with sickle cell disease

Hydroxyurea compared with no hydroxyurea for sickle cell disease

Patient or population: adults and children with sickle cell disease

Settings: outpatients

Intervention: hydroxyurea

Comparison: no hydroxyurea

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

No hydroxyurea

Hydroxyurea

Pain alteration

Follow‐up: NA

Outcome not reported

NA

Life‐threatening illness

Follow‐up: NA

Outcome not reported

NA

Death during the study

Follow‐up: 11 months

No deaths occurred

No deaths occurred

NA

up to 44

(1 study)1

⊕⊝⊝⊝
very low2,3,4

Measures of HbF

Follow‐up: 24 weeks

See comment

See comment

NA

up to 44

(1 study)1

⊕⊝⊝⊝
very low2,3,4

There was a significant increase in HbF in the hydroxyurea group compared to the no hydroxyurea group1.

Measures of ANC

Follow‐up: 24 weeks

See comment

See comment

NA

up to 44

(1 study)1

⊕⊝⊝⊝
very low2,3,4

There was no significant difference in ANC between treatment groups1.

Quality of life

Follow‐up: NA

Outcome not reported

NA

Adverse events or toxicity

Follow‐up: 11 months

See comment

See comment

NA

up to 44

(1 study)1

⊕⊝⊝⊝
very low2,3,4

Vaso‐occlusive pain crises, headache / migraine, upper respiratory infection, skin rash diarrhoea and abdominal pain were the most common adverse events during the trial and these events were evenly distributed across treatment groups (not separated by group).

The basis for the assumed risk is the event rate in the control group unless otherwise stated in the comments and footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
ANC: absolute neutrophil counts; CI: confidence interval; HbF: fetal haemoglobin; NA: not applicable; RR: risk ratio.

GRADE Working Group grades of evidence.
High quality: further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: we are very uncertain about the estimate.

1. Due to the factorial design of the study (22 participants randomised to a treatment arm including hydroxyurea and 22 randomised to a treatment arm without hydroxyurea), results are not entered into analysis. All results of this trial are considered exploratory (CHAMPS 2011).
2. Downgraded once due to indirectness: factorial design of the study makes comparison of hydroxyurea to no hydroxyurea indirect.
3. Downgraded once due to imprecision and risk of bias: study was terminated early with only 44 out of 188 participants recruited and outcome data is presented for only those who completed each follow‐up time.
4. Downgraded once due to applicability: participants with HbSC were included therefore results are not applicable to individuals with HbSS or HbSβº‐thalassemia genotypes.

Figures and Tables -
Summary of findings 4. Summary of findings ‐ Hydroxyurea compared with no hydroxyurea for people with sickle cell disease
Table 1. Clinical events and markers of response in Jain 2012

Hydroxyurea

Placebo

P value

Baseline

18 months

Baseline

18 months

Clinical events (number of events per participant per year)

VOC

12.13 (8.56)

0.60 (1.37)

11.46 (3.01)

10.2 (3.24)

< 0.001

Blood transfusions

2.43 (0.69)

0.13 (0.43)

2.13 (0.98)

1.98 (0.82)

< 0.001

Hospitalisations

10.13 (6.56)

0.70 (1.28)

9.56 (2.91)

9.59 (2.94)

< 0.001

Haematological parameters

Hb (g/dL)

8.1 (0.68)

9.29 (0.55)

8.21 (0.68)

7.90 (0.58)

< 0.001

HbF(%)

19.8 (0.9)

24 (5.9)

19.21 (6.37)

18.92 (5.77)

< 0.001

Reticulocytes (x10⁵ /mm³)

1.83 (0.96)

1.15 (0.1)

1.73 (0.49)

1.81 (0.67)

< 0.001

Leucocytes (x10³ /mm³)

7.36 (6.03)

6.54 (5.54)

7.26 (4.91)

7.38 (2.85)

< 0.001

Platelets (x10³ /mm³)

1.78 (0.26)

2.01 (0.18)

1.91 (0.21)

2.06 (0.26)

0.28

RBC (x10⁶ /mm³)

2.89 (0.57)

1.98 (0.22)

1.84 (0.47)

3.11 (0.20)

0.05

Total bilirubin (mg/dL)

2.32 (1.42)

1.10 (0.42)

2.27 (1.28)

2.71 (0.93)

< 0.001

Values are mean (standard deviation) P values are calculated using independent t‐test.

Hb: haemoglobin
HbF: fetal haemoglobin
RBC: red blood count
VOC: vaso‐occlusive crises
WBC: white blood count

Figures and Tables -
Table 1. Clinical events and markers of response in Jain 2012
Table 2. Laboratory measurements from MSH 1995

Baseline

Hyrdoxyurea

Placebo

P value

2 years

Baseline

2 years

2 years

WBC (10⁹/L)

12.6 (3.4)

9.9 (3.1)

12.3 (3.2)

12.2 (2.8)

0.0001

Neutrophils (10⁹/L)

6.9 (2.4)

4.9 (2.0)

6.7 (2.3)

6.4 (2.0)

0.0001

Platelets (10⁹/L)

468 (147)

399 (124)

457 (130)

423 (122)

0.12

Hb (g/dL)

8.5 (1.4)

9.1 (1.5)

8.5 (1.2)

8.5 (1.3)

0.0009

PCV (%)

24.9 (4.4)

27 (5)

25.2 (4.0)

25.1 (4.2)

0.0007

MCV (fl)

94 (9)

103 (14)

93 (9)

93 (9)

0.0001

Reticulocytes (10⁹/L)

327 (98)

231 (100)

325 (94)

300 (99)

0.0001

HbF (%)

5 (3.5)

8.6 (6.8)

5.2 (3.4)

4.7 (3.3)

0.0001

F cells (%)

33 (17)

48 (23)

33 (17)

35 (18)

0.0001

F reticulocytes

15 (8)

17 (9)

15 (8)

15 (7)

0.0036

Dense cells (%)

14 (6)

11 (6)

14 (7)

13 (7)

0.004

Creatinine (mg/dL)

0.9 (0.3)

1.0 (0.5)

0.9 (0.2)

1.0 (0.5)

0.64

Total bilirubin (mg/dL)

3.7 (2.4)

2.9 (2.5)

3.7 (2.5)

4.2 (4.6)

0.004

Direct bilirubin (mg/dL)

0.5 (0.3)

0.4 (0.3)

0.5 (0.4)

0.7 (2.2)

0.08

Aspartate aminotransferase

44 (23)

39 (20)

41 (21)

43 (27)

0.16

Alkaline phosphatase

120 (59)

117 (48)

119 (67)

119 (71)

0.71

Values are mean (standard deviation) P values are calculated using independent t‐test.

Hb: haemoglobin
HbF: fetal haemoglobin
MCV: mean corpuscular volume
PCV: packed cell volume
WBC: white blood count

Figures and Tables -
Table 2. Laboratory measurements from MSH 1995
Table 3. Laboratory evaluations from the SWiTCH trial

Outcome

Hydroxyurea and

phlebotomy group (n = 67)

Transfusions and

chelation group (n = 66)

P value

HbF (%)

17.9 (92 to 22.9)

‐0.2 (‐0.8 to 0.4)

< 0.001

ANC (x10⁹/L)

‐3.3 (‐5.1 to ‐1.4)

0.8 (‐1.3 to 2.4)

< 0.001

Hb (g /dL)

0.0 (‐0.7 to 0.7)

0.0 (‐0.5 to 0.6)

0.898

HbA (%)

‐50.9 (‐66.8 to ‐33.7)

0.0 (‐12.7 to 6.7)

< 0.001

HbS (%)

35.0 (21.7 to 46.2)

0.3 (‐7.5 to 12.3)

< 0.001

MCV (fL)

19.5 (7.5 to 28.5)

0.1 (‐2.0 to 2.5)

< 0.001

WBC (x10⁹/L)

‐5.4 (‐8.1 to ‐2.2)

0.2 (‐2.0 to 2.3)

< 0.001

ARC (x10⁹/L)

‐149.1 (‐231.0 to ‐19.0)

‐11.8 (‐88.2 to 93.2)

< 0.001

Platelets (x10⁹/L)

‐83.0 (‐171.0 to ‐8.0)

‐28.0 (‐70.0 to 18.0)

0.0022

Total bilirubin (mg/dL)

‐1.1 (‐1.9 to ‐0.6)

0.4 (‐0.3 to 1.2)

< 0.001

LIC (mg Fe/g)

‐1.2 (‐2.8 to 7.2)

‐2.2 (‐5.5 to 4.9)

0.48888

Serum ferritin (ng/mL)

‐966.0 (‐1629.0 to 49.0)

1159.5 (‐662.0 to 2724.0)

< 0.001

LDH (U/L)

‐67.0 (‐143.0 to 7.0)

‐8.5 (‐74.0 to 74.0)

0.0015

ANC: absolute neutrophil count
ARC: absolute reticulocyte count
Hb: haemoglobin
HbA: adult haemoglobin
HbF: fetal haemoglobin
HbS: sickle haemoglobin
LDH: lactate dehydrogenase
LIC: liver iron concentration
MCV: mean corpuscular volume
WBC: white blood count

Values are median change from baseline and interquartile range. P values are calculated using Wilcoxon rank sum test.

Figures and Tables -
Table 3. Laboratory evaluations from the SWiTCH trial
Table 4. Laboratory evaluations from the SCATE trial

Outcome

Hydroxyurea (n = 11)

Observation (n = 11)

P value

Hb (g/dL)

1.6

‐0.5

< 0.0001

MCV (fL)

8.7

1

0.0001

ARC ( x10⁹/L)

22.7

‐33.2

0.76

WBC ( x10⁹/L)

‐4.6

1.3

0.07

ANC ( x10⁹/L)

‐2.2

1.4

0.05

Platelets ( x10⁹/L)

‐76

‐35

0.56

HbF (%)

8.9

0.3

0.002

Weight (kg)

2.5

1.8

0.51

Height (cm)

6.8

3.8

0.22

ANC: absolute neutrophil count
ARC: absolute reticulocyte count
Hb: haemoglobin
HbF: fetal haemoglobin
MCV: mean corpuscular volume
WBC: white blood count.

Values are median change from baseline and P values are calculated using Wilcoxon rank sum test.

Figures and Tables -
Table 4. Laboratory evaluations from the SCATE trial
Table 5. Pregnancies in the MSH Study

Pregnancy

Hydroxyurea

Placebo

Patients

Normal full‐term delivery

1

2

Elective termination

2

1

Partners of patients

Normal full‐term delivery

2

0

Spontaneous abortion

1

0

Still pregnant

0

1

TOTAL

6

4

Figures and Tables -
Table 5. Pregnancies in the MSH Study
Comparison 1. Hydroxyurea versus placebo for participants with sickle cell disease

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain crises Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

1.1 Mean annual crisis rate at 2 years (all crises)

1

Mean Difference (IV, Fixed, 95% CI)

‐2.80 [‐4.74, ‐0.86]

1.2 Mean annual crisis rate at 2 years (all crises requiring hospitalisation)

1

Mean Difference (IV, Fixed, 95% CI)

‐1.50 [‐2.58, ‐0.42]

1.3 Number of vaso‐occlusive crises after 18 months of treatment

1

Mean Difference (IV, Fixed, 95% CI)

‐9.6 [‐10.86, ‐8.34]

2 Proportion experiencing pain Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

3 Proportion experiencing life threatening events during study Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 Acute chest syndrome

2

492

Risk Ratio (M‐H, Fixed, 95% CI)

0.43 [0.29, 0.63]

3.2 Hepatic sequestration

1

299

Risk Ratio (M‐H, Fixed, 95% CI)

0.32 [0.03, 3.06]

3.3 Stroke

2

492

Risk Ratio (M‐H, Fixed, 95% CI)

0.54 [0.12, 2.53]

3.4 Patients transfused

2

492

Risk Ratio (M‐H, Fixed, 95% CI)

0.66 [0.52, 0.82]

3.5 Splenic sequestration

1

193

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.36, 2.23]

4 Number of life‐threatening events during study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

4.1 Blood transfusions

1

Mean Difference (IV, Fixed, 95% CI)

‐1.85 [‐2.18, ‐1.52]

4.2 Hospitalisations

1

Mean Difference (IV, Fixed, 95% CI)

‐8.89 [‐10.04, ‐7.74]

4.3 Duration of hospitalisation (days)

1

Mean Difference (IV, Fixed, 95% CI)

‐2.00 [‐4.87, ‐3.13]

5 Deaths during the study Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

5.1 All deaths

3

552

Risk Ratio (M‐H, Fixed, 95% CI)

0.39 [0.08, 1.96]

5.2 Deaths related to SCD

3

552

Risk Ratio (M‐H, Fixed, 95% CI)

0.48 [0.09, 2.60]

6 Change from baseline in fetal haemoglobin (HbF %) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

7 Fetal haemoglobin (HbF %) after treatment Show forest plot

2

359

Mean Difference (IV, Fixed, 95% CI)

4.07 [2.95, 5.18]

8 Change from baseline in absolute neutrophil count (x10³ per μL) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

9 Neutrophil response (10⁹/L) after treatment Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

9.1 Neutrophils (x10 9/l) at 10 weeks

1

Mean Difference (IV, Fixed, 95% CI)

‐1.90 [‐2.51, ‐1.29]

9.2 Neutrophils (x10 9/l) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐1.50 [‐2.01, ‐0.99]

10 Change from baseline in haemoglobin (g/L) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

11 Change from baseline in m corpuscular volume (fL) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

12 Change from baseline in white blood cells (x10³ per μL) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

13 Change from baseline in absolute reticulocyte count (x10³ per μL) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

14 Change from baseline in reticulocytes (%) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

15 Change from baseline in total bilirubin (mg/L) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

16 Change from baseline in platelet count (x10³ per μL) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

17 Haemoglobin (g/dL) Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

17.1 At 10 weeks

1

299

Mean Difference (IV, Fixed, 95% CI)

0.5 [0.19, 0.81]

17.2 At the end of the study

2

359

Mean Difference (IV, Fixed, 95% CI)

1.04 [0.82, 1.25]

18 Mean corpuscular volume (fL) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

18.1 At 10 weeks

1

Mean Difference (IV, Fixed, 95% CI)

12.30 [9.69, 14.91]

18.2 At 2 years

1

Mean Difference (IV, Fixed, 95% CI)

10.0 [7.34, 12.66]

19 Total bilirubin (mg/L) Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Subtotals only

19.1 At the end of the study

2

359

Mean Difference (IV, Fixed, 95% CI)

‐1.56 [‐1.90, ‐1.23]

20 Reticulocytes Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

20.1 Reticulocytes (10⁵/mm³) at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

‐0.66 [‐0.90, ‐0.42]

20.2 Reticulocytes (10⁹/L) at 10 weeks

1

Mean Difference (IV, Fixed, 95% CI)

‐130.0 [‐152.17, ‐107.83]

20.3 Reticulocytes (10⁹/L) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐69.0 [‐91.56, ‐46.44]

21 Platelet count Show forest plot

2

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

21.1 Platelet count (10³/mm³) at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

‐0.05 [‐0.16, 0.06]

21.2 Platelet count (x10⁹/L) at 10 weeks

1

Mean Difference (IV, Fixed, 95% CI)

‐35.0 [‐75.19, 5.19]

21.3 Platelet count (x10⁹/L) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐24.0 [‐51.88, 3.88]

22 Packed cell volume Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

22.1 Packed cell volume (%) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

1.90 [0.85, 2.95]

23 F reticulocytes Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

23.1 F reticulocytes at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

2.0 [0.18, 3.82]

24 F cells Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

24.1 F cells (%) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

13.0 [8.33, 17.67]

25 Red blood count Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

25.1 Red blood count (10⁶/mm³) at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

‐1.13 [‐1.24, ‐1.02]

26 White blood cells Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

26.1 White blood cells (109/L) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐2.30 [‐2.97, ‐1.63]

27 Dense cells Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

27.1 Dense cells (%) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐2.0 [‐3.48, ‐0.52]

28 Leucocytes Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

28.1 Leucocytes (10³/mm³) at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

‐0.84 [‐3.07, 1.39]

29 Creatinine Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

29.1 Creatinine (mg/dL) at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [‐0.11, 0.11]

30 Aspartate aminotransferase Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

30.1 Aspartate aminotransferase at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐4.0 [‐9.40, 1.40]

31 Alkaline phosphatase Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

31.1 Alkaline phosphatase at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

‐2.0 [‐15.78, 11.78]

32 Change from baseline in growth Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

32.1 Height (cm)

1

Mean Difference (Fixed, 95% CI)

‐0.2 [1.00, 0.60]

32.2 Weight (kg)

1

Mean Difference (Fixed, 95% CI)

0.10 [‐0.20, 0.40]

32.3 Head circumference (cm)

1

Mean Difference (Fixed, 95% CI)

‐0.2 [‐0.60, 0.20]

33 Quality of life: general health perception Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

33.1 General health perception at 6 months

1

Mean Difference (IV, Fixed, 95% CI)

0.30 [‐0.54, 1.14]

33.2 General health perception at 1 year

1

Mean Difference (IV, Fixed, 95% CI)

0.60 [‐0.18, 1.38]

33.3 General health perception at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

0.9 [0.08, 1.72]

33.4 General health perception at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

0.4 [‐0.51, 1.31]

34 Quality of life: pain recall Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

34.1 Pain recall at 6 months

1

Mean Difference (IV, Fixed, 95% CI)

0.40 [‐0.13, 0.93]

34.2 Pain recall at 1 year

1

Mean Difference (IV, Fixed, 95% CI)

0.40 [‐0.18, 0.98]

34.3 Pain recall at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

0.70 [0.11, 1.29]

34.4 Pain recall at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

0.3 [‐0.30, 0.90]

35 Quality of life: social function Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

35.1 Social function at 6 months

1

Mean Difference (IV, Fixed, 95% CI)

0.4 [‐0.15, 0.95]

35.2 Social function at 1 year

1

Mean Difference (IV, Fixed, 95% CI)

0.2 [‐0.36, 0.76]

35.3 Social function at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

0.40 [‐0.21, 1.01]

35.4 Social function at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

0.3 [‐0.31, 0.91]

36 Changes in 'Ladder of Life' Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

36.1 Changes in 'Ladder of Life' at 6 months

1

Mean Difference (IV, Fixed, 95% CI)

0.0 [‐0.52, 0.52]

36.2 Changes in 'Ladder of Life' at 1 year

1

Mean Difference (IV, Fixed, 95% CI)

0.4 [‐0.15, 0.95]

36.3 Changes in 'Ladder of Life' at 18 months

1

Mean Difference (IV, Fixed, 95% CI)

0.30 [‐0.22, 0.82]

36.4 Changes in 'Ladder of Life' at 2 years

1

Mean Difference (IV, Fixed, 95% CI)

0.3 [‐0.23, 0.83]

37 Proportion of participants with signs of organ damage Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

37.1 New leg ulcers

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.85 [0.44, 1.64]

37.2 Aseptic necrosis (humerus or femur)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.39, 2.37]

37.3 Decreased spleen function at exit (compared to baseline)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.72 [0.44, 1.16]

38 Signs of organ damage ‐ change from baseline in DTPA GFR Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

39 Signs of organ damage ‐ change from baseline in Howell‐Jolley body (per 106 red blood cells) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

40 Signs of organ damage ‐ change from baseline in pitted cells (%) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

41 Signs of organ damage ‐ change from baseline in spleen: liver ratio of counts Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

42 Signs of organ damage ‐ change from baseline in spleen volume (cm3) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

43 Signs of organ damage ‐ change from baseline in creatinine (mg/L) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

44 Signs of organ damage ‐ change from baseline in Schwartz GFR Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

44.1 Schwartz glomerular filtration rate (mL/min per 1.73m2)

1

Mean Difference (Fixed, 95% CI)

‐8.0 [‐35.00, 21.00]

45 Signs of organ damage ‐ change from baseline in cystatin C Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

46 Signs of organ damage ‐ change from baseline in urine osmolality Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

47 Signs of organ damage ‐ change from baseline in urine pH Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

48 Signs of organ damage ‐ change from baseline in urine‐specific gravity Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

49 Signs of organ damage ‐ change from baseline in total kidney volume Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

50 Signs of organ damage ‐ change from baseline in TCD ultrasound velocity (time‐averaged mean maximum velocity) Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

51 Signs of organ damage ‐ change from baseline in CNS measures Show forest plot

1

Mean Difference (Fixed, 95% CI)

Totals not selected

51.1 Bayley Mental Development Index

1

Mean Difference (Fixed, 95% CI)

3.0 [0.00, 8.00]

51.2 Bayley motor performance development index

1

Mean Difference (Fixed, 95% CI)

2.0 [‐1.00, 7.00]

52 Proportion of participants experiencing adverse events and toxicity Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

52.1 hair loss at 1 or 2 visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.25 [0.69, 2.26]

52.2 hair loss at 3 or more visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.61 [0.31, 1.21]

52.3 skin rash at 1 or 2 visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.62, 1.51]

52.4 skin rash at 3 or more visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.58, 1.60]

52.5 fever at 1 or 2 visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.55, 1.69]

52.6 fever at 3 or more visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.68, 1.17]

52.7 Gastroinestinal disturbance at 1 or 2 visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.83 [0.50, 1.36]

52.8 Gastrointestinal disturbance at 3 or more visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.74, 1.31]

52.9 Other abnormalities at 1 or 2 visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.60 [0.33, 1.11]

52.10 Other abnormalities at 3 or more visits

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.83, 1.40]

52.11 Hospitalisation (for any reason)

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.83 [0.72, 0.96]

52.12 Dactylitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.34 [0.20, 0.58]

52.13 Priapism

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.52 [0.26, 8.87]

52.14 Sepsis or bacteraemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.40 [0.08, 2.03]

52.15 Splenomegaly

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.61, 1.32]

52.16 Absolute Neutrophil Count < 500

1

Risk Ratio (M‐H, Fixed, 95% CI)

2.53 [0.50, 12.71]

52.17 Absolute Neutrophil 500 ‐ 1250

1

Risk Ratio (M‐H, Fixed, 95% CI)

2.53 [1.58, 4.03]

52.18 Thrombocytopaenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.59 [0.64, 3.92]

52.19 Alanine transaminase > 150 U/L

1

Risk Ratio (M‐H, Fixed, 95% CI)

2.02 [0.19, 21.92]

52.20 Severe anaemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.51 [0.05, 5.48]

52.21 Bilirubin

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.34 [0.01, 8.17]

52.22 Creatinine

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

52.23 Skin and subcutaneous disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.91 [0.75, 1.10]

52.24 Splenic sequestration

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.36, 2.23]

52.25 Gastroenteritis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.44 [0.28, 0.71]

52.26 Nausea

1

Risk Ratio (M‐H, Fixed, 95% CI)

5.0 [0.25, 99.95]

52.27 Skin Rash

1

Risk Ratio (M‐H, Fixed, 95% CI)

7.0 [0.38, 129.93]

Figures and Tables -
Comparison 1. Hydroxyurea versus placebo for participants with sickle cell disease
Comparison 2. Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion experiencing pain Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

1.1 Vaso‐occlusive or sickle cell‐related pain (all)

1

133

Risk Ratio (M‐H, Fixed, 95% CI)

1.03 [0.81, 1.30]

1.2 Vaso‐occlusive or sickle cell‐related pain (serious)

2

254

Risk Ratio (M‐H, Fixed, 95% CI)

3.37 [1.59, 7.11]

2 Proportion experiencing life‐threatening events during study Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

2.1 Stroke (secondary)

1

133

Risk Ratio (M‐H, Fixed, 95% CI)

14.78 [0.86, 253.66]

2.2 Transient ischaemic attack (primary)

1

121

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.21, 4.84]

2.3 Transient ischaemic attack (secondary)

1

133

Risk Ratio (M‐H, Fixed, 95% CI)

0.66 [0.25, 1.74]

2.4 Other neurological event (primary)

1

121

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.07, 15.88]

2.5 Acute chest syndrome

2

254

Risk Ratio (M‐H, Fixed, 95% CI)

2.84 [1.25, 6.42]

2.6 Infections and Infestations

2

254

Risk Ratio (M‐H, Fixed, 95% CI)

3.65 [1.05, 12.76]

2.7 Splenic sequestration or splenectomy

2

254

Risk Ratio (M‐H, Fixed, 95% CI)

0.34 [0.01, 8.16]

2.8 Hepatobiliary disease

1

121

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.07, 15.88]

2.9 Total with serious adverse events

2

254

Risk Ratio (M‐H, Fixed, 95% CI)

1.93 [1.17, 3.20]

2.10 Total with sickle cell related, non‐neurological adverse events

1

133

Risk Ratio (M‐H, Fixed, 95% CI)

3.10 [1.42, 6.75]

3 Deaths during the study Show forest plot

2

254

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.06, 15.42]

4 Change from baseline in fetal haemoglobin (HbF %) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

5 Change from baseline in absolute neutrophil count (x10⁹/L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

6 Change from baseline in mean corpuscular volume (fL) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

7 Change from baseline in sickle haemoglobin (%) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

8 Change from baseline in haemoglobin (g/L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

9 Change from baseline in absolute reticulocyte count (10⁹ / L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

10 Change from baseline in white blood count (10⁹ / L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

11 Change from baseline in platelets (10⁹ / L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

12 Change from baseline in total bilirubin (mg/L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

13 Change from baseline in liver iron concentration Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

14 Change from baseline in serum ferritin (ng/mL) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

15 Change from baseline in lactate dehydrogenase (U/L) Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

16 Signs of organ damage ‐ CNS measures at the end of the study Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

16.1 Final TCD ultrasound velocity

1

Mean Difference (IV, Fixed, 95% CI)

‐5.0 [‐9.16, ‐0.84]

17 Proportion of participants experiencing non‐neurological adverse events and toxicity Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

17.1 Infections and infestations

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.24 [0.89, 1.72]

17.2 Gastrointestinal disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.16 [0.74, 1.80]

17.3 Fever

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.66, 1.75]

17.4 Musculoskelatal disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.14 [0.59, 2.20]

17.5 Immune system disorders

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.12 [0.02, 0.96]

17.6 Cholelithiasis

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.74 [0.17, 3.17]

17.7 Cholecystitis

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.97 [0.18, 21.21]

17.8 Asthma

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.74 [0.17, 3.17]

17.9 Acute chest syndrome

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.72 [0.53, 5.61]

17.10 Renal or urinary disorder

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.66 [0.11, 3.80]

17.11 Priapism

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.97 [0.18, 21.21]

17.12 Cathether‐related complications

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.48 [0.26, 8.56]

17.13 Cardiac disorder

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.06, 15.42]

17.14 Hyderbilirubianemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.49 [0.24, 1.02]

17.15 Alanine transaminase increase

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.55, 2.01]

17.16 Aspartate transaminase increase

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.38 [0.66, 2.88]

17.17 Serum creatinine increase

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.97 [0.51, 7.55]

17.18 Thrombocytopenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

6.90 [0.87, 54.51]

17.19 Reticuloctopenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

6.90 [2.16, 22.02]

17.20 Neutropenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

9.85 [1.30, 74.80]

17.21 Anaemia

1

Risk Ratio (M‐H, Fixed, 95% CI)

6.57 [2.05, 21.05]

17.22 Sickle cell pain

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.29 [0.92, 1.82]

17.23 Sickle cell‐related events

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.03 [0.81, 1.30]

17.24 All adverse events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.92, 1.05]

Figures and Tables -
Comparison 2. Hydroxyurea and phlebotomy compared to transfusion and chelation for participants with SCD and risk of stroke
Comparison 3. Hydroxyurea compared to observation for participants with SCD and risk of stroke

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion experiencing life‐threatening events during the study Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

1.1 Vaso‐occlusive events

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.4 [0.10, 1.64]

1.2 Acute splenic sequestration

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.07, 14.05]

1.3 Blood transfusions required

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.04, 2.73]

2 Signs of organ damage ‐ proportion of participants with a change in CNS measures Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

2.1 Participants converting from conditional to abnormal TCD ultrasound velocity

1

Risk Ratio (M‐H, Fixed, 95% CI)

0.2 [0.03, 1.45]

3 Signs of organ damage ‐ change from baseline in CNS measures Show forest plot

1

Mean Difference (IV, Fixed, 95% CI)

Totals not selected

3.1 TCD ultrasound velocity (time‐averaged mean maximum velocity)

1

Mean Difference (IV, Fixed, 95% CI)

‐25.7 [‐45.38, ‐6.02]

4 Adverse events and toxicity Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Totals not selected

4.1 Dizziness

1

Risk Ratio (M‐H, Fixed, 95% CI)

3.0 [0.14, 66.53]

4.2 Headaches

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.07, 14.05]

4.3 Transient neutropenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

3.0 [0.14, 66.53]

4.4 Reticulocytopenia

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.07, 14.05]

4.5 Parasite infection

1

Risk Ratio (M‐H, Fixed, 95% CI)

1.0 [0.07, 14.05]

Figures and Tables -
Comparison 3. Hydroxyurea compared to observation for participants with SCD and risk of stroke