Acessibilidade / Reportar erro

Twenty Years of Neonatal Screening for Sickle Cell Disease in Brazil: The Challenges of a Continental Country with High Genetic Heterogeneity

Abstract

Sickle cell disease (SCD) is the most common inherited hematological disease worldwide. The benefits of diagnosis and early intervention have led to the wide dissemination of public health programs worldwide. Through neonatal screening programs, it is possible to reduce morbidity and mortality in the first 5 years of life. The prophylactic use of penicillin, the anti-pneumococcal vaccine and other intensive care, increase the survival and quality of life of people with SCD. The aim of this study is to present the 20-year history of screening for hemoglobinopathies in Brazil and its challenges. From 2001 to 2019, an average of 2,400,000 children were screened per year nationwide, with the coverage being of 82,16%. The screening of 54,9% of newborns is collected up to their 5th day of life. The prevalence of SCD was 1:2,263 newborns; therefore, it was the second most-common disease detected by the program of Brazil, being only after hypothyroidism (1/2,175 live births). The healthcare system should provide the necessary infrastructure to confirm the diagnosis of newborns and to provide appropriate counseling and treatment. The early diagnosis and treatment, as well as the follow-up with a multidisciplinary team, are fundamental to the survival rate and the quality of life of patients.

Keywords
Newborn screening; Sickle Cell Disorder; Brazil; National Program

Introduction

Hemoglobins are genetically determined, therefore changes to their genes, a disorder that can lead to the production of proteins with characteristics different from the ones expected and can result in the formation of a pathological hemoglobin, in several cases, it offers pathophysiological consequences to its carrier. Hemoglobinopathies are recognized as one of the most common genetic pathologies around the world. Amongst them, the sickle cell disorder (SCD) and the β thalassemia have a greater impact on morbidity and mortality, affecting millions of individuals worldwide.[11. da Fonseca SF, Amorim T, Purificação A, Gonçalves M, Boa-Sorte N. Hemoglobin A2 values in sickle cell disease patients quantified by high performance liquid chromatography and the influence of alpha thalassemia. Rev Bras Hematol Hemoter. 2015;37(5):296-301. doi:10.1016/j.bjhh.2015.05.005
https://doi.org/10.1016/j.bjhh.2015.05.0...
]

In the scientific world, the SCD has been discovered more than 100 years ago; however, it was mainly in the past decades that the world has advanced towards its prognosis, certainly in virtue of the results provided by Neonatal Screening Programs, its diagnostic benefits and early prophylaxis, which can reduce morbidity and mortality. This early diagnosis has provided a greater knowledge on the early complications of SCD, leading to the creation of prophylactic measures, such as the use of penicillin, the administration of pneumococcal vaccines and other precautions, including the use of hydroxyurea.[22. Field JJ, Nathan DG. Advances in sickle cell therapies in the hydroxyurea era. Mol Med. 2014;20(Suppl 1):S37-S42. doi:10.2119/molmed.2014.00187
https://doi.org/10.2119/molmed.2014.0018...
] These actions significantly increase survival rates and improve the quality of life of patients with SCD, reducing sequelae and complications, such as painful crises and splenic sequestration, thus resulting in a better quality of life.[33. Odame I. Perspective: we need a global solution. Nature. 2014;515(7526):S10. doi:10.1038/515S10a
https://doi.org/10.1038/515S10a...
] In neonatal screening, in addition to the early diagnosis, it is also possible to detect heterozygous newborns and, in these cases, to offer tests for couples, by identifying which couples may be at risk of having children with SCD. According to Pan-European Consensus Conference there is an orientation that the carrier status (all mutations that might cause SCD) should be reported and couselling offered to carriers, but the identification of carriers is still debated in several European countries.[44. Lobitz S, Telfer P, Cela E, et al. Newborn screening for sickle cell disease in Europe: recommendations from a Pan-European Consensus Conference. Br J Haematol. 2018;183(4):648-660. doi:10.1111/bjh.15600.
https://doi.org/10.1111/bjh.15600...
]

Hemoglobinopathies, unlike most other inborn errors of metabolism, are human genetic mutations; that is, they are genetic alterations highly frequent in the global population. Since the population is broadly miscegenated and the severity of complications of SCD can be prevented from adequate actions if they are implemented early in life (penicillin prophylaxis, vaccinations, spleen palpation, parents’ education, etc), all national programs must be universalized, instead of oriented to a specific portion of its citizens. Consequently, any program that offers screening for these anomalies on a national scale must have the necessary infrastructure, not only to confirm the laboratory diagnosis of newborns, but also to provide a complete treatment for patients and a specialized genetic counseling for newborns and families.

Arrival and allocation of immigrants in Brazil

The Brazilian population is characterized by its vast genetic heterogeneity, caused by the diversity of racial groups forming it and by the different degrees of miscegenation in all regions of the country.[55. Callegari-Jacques SM, Grattapaglia D, Salzano FM, Salamoni SP, Crossetti SG, Ferreira ME, Hutz MH. Historical genetics: spatio temporal analysis of the formation of the Brazilian population. Am J Hum Biol. 2003;15(6):824-834. doi:10.1002/ajhb.10217
https://doi.org/10.1002/ajhb.10217...
]

The mutation in the beta globin gene first appeared in Africa, having then spread to other parts of the world, including Brazil. The arrival of Africans in Brazilian territory began almost 400 years old. Up to its discovery in 1500, Brazil was inhabited by Amerindians, encompassing a population of around two million people. From 1,500 onwards, the immigration of Portuguese people began and, between the 16th and 19th centuries, the arrival of African people took place. Therefore, the hemoglobin S (Hb S), responsible for SCD, was introduced by the slave trade, which enslaved people from Africa and brought them to work in Brazil, first in sugarcane plantations, on the Northeast, and later in the extraction of precious metals, in Minas Gerais. This continued miscegenatory activity is believed to have facilitated the spread of variant hemoglobins in different regions of Brazil. There is a strong association between the clinical presentation of the SCD and the different regions of origin of the slaves. Slaves came from different regions of Africa, featuring important association between different haplotypes and clinical features of the disease. HBB*S haplotypes have been studied in different Brazilian populations, as tools to clarify different population origins. The CAR haplotype (Central Africa) was the most frequent one, followed by the BEM (Benin) haplotype, demonstrating the main regions that slaves came from to Brazil.[66. Lindenau JD, Wagner SC, Castro SM, Hutz MH. The effects of old and recent migration waves in the distribution of HBB*S globin gene haplotypes. Genet Mol Biol. 2016; 39(4): 515-523. doi:10.1590/1678-4685-gmb-2016-0032.
https://doi.org/10.1590/1678-4685-gmb-20...
]

In addition to Portugueses and Africans, other immigration flows occurred between the 19th and 20th centuries, primarily of individuals from Italy, Germany and Spain.[77. Salzano FM, Bortolini MC. The evolution and genetics of Latin American populations. Cambridge: Cambridge University Press; 2002.] They arrived mainly in the State of São Paulo and in the Southern region of Brazil.[88. Salzano FM, Freire-Maia N. Problems in human biology. A study of Brazilian populations. Detroit: Wayne State University Press; 1970. doi:10.1002/ajhb.10217.
https://doi.org/10.1002/ajhb.10217...
] Currently, the proportion of races varies according to the region of the country. For instance, the percentage of the African component is lower in the South (11%), whilest the highest values are found in the Northeast and Southeast (18%-20%).[55. Callegari-Jacques SM, Grattapaglia D, Salzano FM, Salamoni SP, Crossetti SG, Ferreira ME, Hutz MH. Historical genetics: spatio temporal analysis of the formation of the Brazilian population. Am J Hum Biol. 2003;15(6):824-834. doi:10.1002/ajhb.10217
https://doi.org/10.1002/ajhb.10217...
] Brazilian studies have demonstrated great regional differences regarding the prevalence of HbS and HbC, which range from 0.2% (Southern region) to 9.8% (Northern region).[99. Wagner SC, de Castro SM, Gonzalez TP, et al. Neonatal screening for hemoglobinopathies: results of a public health system in South Brazil. Genet Test Mol Biomarkers. 2010;14(4):565-569. doi:10.1089/gtmb.2010.0008
https://doi.org/10.1089/gtmb.2010.0008...
] This portays hemoglobinopathies as a public health problem, even though with distinct incidences in the different states. The aim of this study is to present the 20-year history of screening for hemoglobinopathies in Brazil and its challenges.

Methods and Results

Incorporation of hemoglobinopathies in Brazilian neonatal screening programs

Due to the extensive arrival of the African population, resulted from the slave trade in the 16th-18th centuries, the World Health Organization recommends the implementation of programs for hemoglobinopathies prevention and control in Latin America.[1010. WHO. Meeting on Advances in Diagnosis Treatment and Prevention of Hereditary Diseases. Report of a WHO Meeting on Advances in Diagnosis, Treatment and Prevention of Hereditary Diseases. Geneva: WHO; 1989. https://apps.who.int/iris/handle/10665/58835
https://apps.who.int/iris/handle/10665/5...
]

The greatest challenge for developing preventive programs is that they require support from official health agencies, training of the personnel for the diagnosis, and genetic and clinical counseling for patients. Currently, only four countries in Latin America have universal screening programs for hemoglobinopathies; they are Brazil, Costa Rica, Panama and Uruguay.[1111. Silva-Pinto AC, Alencar de Queiroz MC, Antoniazzo PJ, Arruda M, Pimentel dos Santos H. The Neonatal Screening Program in Brazil, Focus on Sickle Cell Disease (SCD). Int J Neonatal Screen. 2019; 5(1):11. doi:10.3390/ijns5010011
https://doi.org/10.3390/ijns5010011...
-1414. Audicio P, Segobia B, Queijo C, Queiruga G. Newborn Screening Pilot Program of Hemoglobinopathies: firstresults in Uruguay. Acta Bioquím Clín Latinoam. 2017;51(2):243-248. https://www.redalyc.org/pdf/535/53552508010.pdf
https://www.redalyc.org/pdf/535/53552508...
] Other countries are conducting pilot studies, in an attempt to demonstrate the importance of implementing national programs.[1515. Knight-Madden J, Lee K, Elana G, Elenga N, Marcheco-Teruel B, Keshi N, Hardy-Dessources MD. Newborn Screening for Sickle Cell Disease in the Caribbean: An Update of the Present Situation and of the Disease Prevalence. Int J Neonatal Screen. 2019;5(1):5. doi:10.3390/ijns5010005
https://doi.org/10.3390/ijns5010005...
]

Nowadays, aproximately 10 million people have abnormal hemoglobins in Brazil, and around three thousand people are annually born with the homozygous form.[1616. Backes CE, Mallmann FG, DassiT, Bazzo ML, Santos-Silva MC. Triagem neonatal como um problema de saúde pública. Rev Bras Hemat Hemoter. 2005;27:43-47. doi:10.1590/S1516-84842005000100011
https://doi.org/10.1590/S1516-8484200500...
]. Therefore, a universal coverage is important because the distribution of sickle cell diseases is also uneven across Brazilian territory. Brazil, with its gigantic territorial area and its geographic, cultural and economic differences, represents a special challenge for any national public health policy, especially neonatal screening. The first neonatal screening program for hemoglobinopathies was developed in the 1990s, in the State of São Paulo.[1717. Brandelise S, Pinheiro V, Gabetta CS, Hambleton I, Serjeant B, Serjeant G. Newborn screening for sickle cell disease in Brazil: the Campinas experience. Clin Lab Haematol. 2004;26(1):15-19. doi:10.1111/j.0141-9854.2003.00576.x
https://doi.org/10.1111/j.0141-9854.2003...
] Independent actions, without any coordination or standardization, were incorporating screening for hemoglobinopathies as a mandatory test in all live births in a particular region of the country. In 1998, the State of Minas Gerais was the first to introduce a Universal Neonatal Screening Program for SCD, inside the already existing Program for phenylketonuria and congenital hypothyroidism.[1111. Silva-Pinto AC, Alencar de Queiroz MC, Antoniazzo PJ, Arruda M, Pimentel dos Santos H. The Neonatal Screening Program in Brazil, Focus on Sickle Cell Disease (SCD). Int J Neonatal Screen. 2019; 5(1):11. doi:10.3390/ijns5010011
https://doi.org/10.3390/ijns5010011...
]

The Brazilian National Neonatal Screening Program (NNSP) was implemented as an official program of the federal government in 2001.The Program establishes the mandatory diagnosis of abnormalities in the newborn metabolism, including screening for hemoglobinopathies.[1616. Backes CE, Mallmann FG, DassiT, Bazzo ML, Santos-Silva MC. Triagem neonatal como um problema de saúde pública. Rev Bras Hemat Hemoter. 2005;27:43-47. doi:10.1590/S1516-84842005000100011
https://doi.org/10.1590/S1516-8484200500...
,1818. Brasil. Ministério da Saúde. Secretaria de Atenção a Saúde. Departamento de Atenção Especializada e Temática. Triagem neonatal biológica: manual técnico. Brasília: Ministério da Saúde; 2016.] The NNSP has the role of detecting, confirming, monitoring and treating suspected cases of six important diseases: phenylketonuria, congenital hypothyroidism, SCD and other hemoglobinopathies, cystic fibrosis, biotinidase deficiency and congenital adrenal hyperplasia. The NNSP was the first federal action to create a universal public health program for the early diagnosis and treatment of selected congenital diseases. Within the scope of the Unified Health System (SUS), the NNSP must be articulated between the Ministry of Health, State and Municipal Health Departments. Its implementation took place gradually in each state, mainly due to regional differences and to the installed capacity to incorporate, from the detection of the disease, confirmatory exams, evaluation by a multidisciplinary team and treatment in high complexity specialized centers.[1919. Ministério da Saúde. Portaria GM/MS nº 822, de 6 de junho de 2001. Institui, no âmbito do Sistema Único de Saúde, o Programa Nacional de Triagem Neonatal/PNTN. Diário Oficial da União. 2001.] As of 2013, all 26 Brazilian states have been qualified to screen and treat hemoglobinopathies; up to 2018, the country had 30 Reference Services distributed along its states, according to report from the Ministry of Health, and 24,300 collection units distributed in all regions.

The Figure 1 presents studies with the different prevalence of SCD and sickle cell traits in newborns from different regions of Brazil.

Figure 1.
Presents studies with the different prevalence of SCD and sickle cell traits in newborns from different regions of Brazil. aRS (Rio Grande do Sul - Cardoso et al., 201720. Cardoso CS, Macedo JL, Diedrich VR, Magalhães CMB, Castro SM. Triagem neonatal de hemoglobinopatias no estado do Rio Grande do Sul no período de 2004 a 2014. Bol Cient Pediatr. 2017;6(3):77-84. https://www.sprs.com.br/sprs2013/bancoimg/171229113516bcped_06_03_a02.pdf
https://www.sprs.com.br/sprs2013/bancoim...
[2020. Cardoso CS, Macedo JL, Diedrich VR, Magalhães CMB, Castro SM. Triagem neonatal de hemoglobinopatias no estado do Rio Grande do Sul no período de 2004 a 2014. Bol Cient Pediatr. 2017;6(3):77-84. https://www.sprs.com.br/sprs2013/bancoimg/171229113516bcped_06_03_a02.pdf
https://www.sprs.com.br/sprs2013/bancoim...
]); bSC (Santa Catarina - Eller et al., 201621. Eller R, Silva DB. Evaluation of a neonatal screening program for sickle-cell disease. J Pediatr (Rio J). 2016;92:409-413. doi:10.1016/j.jped.2015.10.002
https://doi.org/10.1016/j.jped.2015.10.0...
[2121. Eller R, Silva DB. Evaluation of a neonatal screening program for sickle-cell disease. J Pediatr (Rio J). 2016;92:409-413. doi:10.1016/j.jped.2015.10.002
https://doi.org/10.1016/j.jped.2015.10.0...
]); cPA (Paraná - Watanabe et al., 200822. Watanabe AM, Pianovski MA, Zanis Neto J, et al. Prevalence of hemoglobin S in the State of Paraná, Brazil, basedon neonatal screening. Cad Saude Publica. 2008;24(5):993-1000. doi:10.1590/S0102-311X2008000500006
https://doi.org/10.1590/S0102-311X200800...
[2222. Watanabe AM, Pianovski MA, Zanis Neto J, et al. Prevalence of hemoglobin S in the State of Paraná, Brazil, basedon neonatal screening. Cad Saude Publica. 2008;24(5):993-1000. doi:10.1590/S0102-311X2008000500006
https://doi.org/10.1590/S0102-311X200800...
]); dSP (São Paulo - Hadachi et al., 200923. Hadachi S, Iskandar M. Triagem neonatal de hemoglobinopatias - Serviço de referência em triagem neonatal (SRTN) Apae São Paulo. Rev Bras Hematol Hemoter. 2009;31(4):54. [2323. Hadachi S, Iskandar M. Triagem neonatal de hemoglobinopatias - Serviço de referência em triagem neonatal (SRTN) Apae São Paulo. Rev Bras Hematol Hemoter. 2009;31(4):54.]); eRJ (Rio de Janeiro - Lobo et al., 201450. Lobo CL, Ballas SK, Domingos AC, et al. Newborn screening program for hemoglobinopathies in Rio de Janeiro, Brazil. Ped Blood Canc. 2014;61(1):34-39. doi:10.1002/pbc.24711.
https://doi.org/10.1002/pbc.24711...
[2424. Lobo CLC, Ballas SK, Bonini Domingos AC, Moura PG, Nascimento EM, Cardoso GP, Carvalho SMF. Newborn Screening Program for Hemoglobinopathies in Rio de Janeiro. Brazil Pediatr Blood Cancer. 2014;61:34-39. doi:10.1002/pbc.24711
https://doi.org/10.1002/pbc.24711...
]); fDF (Distrito Federal - Diniz et al., 200926. Diniz D, Guedes C, Barbosa L, et al. Prevalence of sickle cell trait and sickle cell anemia among newborns in the Federal District, Brazil, 2004 to 2006. Cad Saude Publica . 2009;25:188-194. doi:10.1590/s0102-311x2009000100020
https://doi.org/10.1590/s0102-311x200900...
[2525. Paixão MC, Cunha Ferraz MH, Januario JN, et al. Reliability of iselectro focusing for the detection of Hb S, Hb C and Hb D in a pioneering population-based program of newborn screening in Brazil. Hemoglobin. 2001;25:297-303. doi:10.1081/hem-100105222
https://doi.org/10.1081/hem-100105222...
]); gMG (Minas Gerais - Paixão et al., 200125. Paixão MC, Cunha Ferraz MH, Januario JN, et al. Reliability of iselectro focusing for the detection of Hb S, Hb C and Hb D in a pioneering population-based program of newborn screening in Brazil. Hemoglobin. 2001;25:297-303. doi:10.1081/hem-100105222
https://doi.org/10.1081/hem-100105222...
[1515. Knight-Madden J, Lee K, Elana G, Elenga N, Marcheco-Teruel B, Keshi N, Hardy-Dessources MD. Newborn Screening for Sickle Cell Disease in the Caribbean: An Update of the Present Situation and of the Disease Prevalence. Int J Neonatal Screen. 2019;5(1):5. doi:10.3390/ijns5010005
https://doi.org/10.3390/ijns5010005...
, 2626. Diniz D, Guedes C, Barbosa L, et al. Prevalence of sickle cell trait and sickle cell anemia among newborns in the Federal District, Brazil, 2004 to 2006. Cad Saude Publica . 2009;25:188-194. doi:10.1590/s0102-311x2009000100020
https://doi.org/10.1590/s0102-311x200900...
]); h,iBA (Bahia - Adorno et al., 200527. Adorno EV, Couto FD, Moura Neto JP, Menezes JF, Rêgo M, Reis MG, et al. Hemoglobinopathies in newborns from Salvador, Bahia, Northeast Brazil. Cad Saude Publica . 2005;21(1):292-298. doi:10.1590/s0102-311x2005000100032
https://doi.org/10.1590/s0102-311x200500...
[2727. Adorno EV, Couto FD, Moura Neto JP, Menezes JF, Rêgo M, Reis MG, et al. Hemoglobinopathies in newborns from Salvador, Bahia, Northeast Brazil. Cad Saude Publica . 2005;21(1):292-298. doi:10.1590/s0102-311x2005000100032
https://doi.org/10.1590/s0102-311x200500...
]; Amorim et al., 201028. Amorim T, Pimentel H, Fontes MIMM, Purificação A, Lessa P, Boa-Sorte N. Avaliação do programa de Triagem Neonatal na Bahia entre 2007 e 2009 - As lições da Doença Falciforme. Gaz Med Bahia. 2010;80(3):10-13. http://www.gmbahia.ufba.br/index.php/gmbahia/article/view/1103
http://www.gmbahia.ufba.br/index.php/gmb...
[2828. Amorim T, Pimentel H, Fontes MIMM, Purificação A, Lessa P, Boa-Sorte N. Avaliação do programa de Triagem Neonatal na Bahia entre 2007 e 2009 - As lições da Doença Falciforme. Gaz Med Bahia. 2010;80(3):10-13. http://www.gmbahia.ufba.br/index.php/gmbahia/article/view/1103
http://www.gmbahia.ufba.br/index.php/gmb...
]); jAL (Alagoas - Lipinski-Figueiredo et al., 200929. Lipinski-Figueiredo E, Estelita S, et al. Dados preliminares da fase II do programa de triagem neonatal do estado de Alagoas. Rev Bras Hemat Hemoter. 2009;31(4):62 [2929. Lipinski-Figueiredo E, Estelita S, et al. Dados preliminares da fase II do programa de triagem neonatal do estado de Alagoas. Rev Bras Hemat Hemoter. 2009;31(4):62]); kRN (Rio Grande do Norte - de Araujo et al., 200430. de Araujo MC, Serafim ES, de Castro WA Jr., et al. Prevalenceof abnormal hemoglobins in newborns in Natal, Rio Grande do Norte, Brazil. Cad Saude Publica . 2004; 20:123-128. doi:10.1590/S0102-311X2004000100027
https://doi.org/10.1590/S0102-311X200400...
[3030. de Araujo MC, Serafim ES, de Castro WA Jr., et al. Prevalenceof abnormal hemoglobins in newborns in Natal, Rio Grande do Norte, Brazil. Cad Saude Publica . 2004; 20:123-128. doi:10.1590/S0102-311X2004000100027
https://doi.org/10.1590/S0102-311X200400...
])

In a historic series of 2014-2016, an average of 2.425.392 children were screened per year nationwide, with the coverage being of 82,16% newborns.[3131. Brasil. Ministério da Saúde. Secretaria de Atenção Especializada à Saúde. Departamento de Atenção Especializada e Temática. Caderno de informação: triagem neonatal. Dados 2014-2016. Brasília: Ministério da Saúde ; 2021. ] Although the Brazilian Program aims at universal, integral and equitable access, there is a variation in coverage rates found throughout the national territory and is associated in differents health care network capacity, to variations in the population’s access to it and also the existence of a private access health network that does not report its data, leaving a gap in the population data of the Ministry of Health.

The collection takes place at the primary care collection points, free of charge through the NNSP, organized in each state. In some states, this collection is carried out at maternity wards, depending on the guidelines of local managers. The guidance for the ideal collection period is from the 3rd to the 5th day, according to the greater sensitivity of the available technologies inherent to diseases within the scope of the national program. The screening of 54,9% of newborns is collected up to their 5th day of life. Collections in non-ideal periods can be considered as neglect, cultural issues or difficulties in access.[3131. Brasil. Ministério da Saúde. Secretaria de Atenção Especializada à Saúde. Departamento de Atenção Especializada e Temática. Caderno de informação: triagem neonatal. Dados 2014-2016. Brasília: Ministério da Saúde ; 2021. ]

The birth prevalence of SCD was 1: 2,263 newborns (0.44 per 1,000 live births); therefore, it was the second most-common disease detected by the NNSP program within the Federative Republic of Brazil, being only after hypothyroidism (1/2,175 live births).[3232. Brasil. Ministério da Saúde. Relatório Anual de Dados do PNTN. Brasília: Ministério da Saúde ; 2018.]

The median indicator of the newborn’s age on the date of the first consultation marks the time that all stages of the screening were performed, culminating in the first consultation of the newborn’s specialty diagnosed with any of the diseases in the program. According to the ministry of health, the median age of the first consultation for SCD was 47 days (18 - 172 days). [3131. Brasil. Ministério da Saúde. Secretaria de Atenção Especializada à Saúde. Departamento de Atenção Especializada e Temática. Caderno de informação: triagem neonatal. Dados 2014-2016. Brasília: Ministério da Saúde ; 2021. ] After diagnostic confirmation, the child initiated penicillin in the first month of life, immunobiological approaches, and then referral to the regional Specialized Care Service. In most states, the reference center is the Blood Center, which is normaly integrated with the local health network to meet other health needs, such as the use of hydroxyurea, transcraneal Doppler and other procedures.

Methods Used in NBS for Hemoglobinopathies

Around the world more than 1,500 molecular defects have been reported in different globin genes. (http://globin.cse.psu.edu/globin/hbvar). The variability of the hemoglobin profiles found in Brazilian studies reflects the heterogeneity of the country’s population.[99. Wagner SC, de Castro SM, Gonzalez TP, et al. Neonatal screening for hemoglobinopathies: results of a public health system in South Brazil. Genet Test Mol Biomarkers. 2010;14(4):565-569. doi:10.1089/gtmb.2010.0008
https://doi.org/10.1089/gtmb.2010.0008...
]

The correct diagnosis of rare variant hemoglobins prevents the application of wrong procedures and therapies, also providing materials for studying structural, functional and anthropological aspects. For the identification of hemoglobin fractions with pathophysiological potential in the neonatal stage, it is also important to highlight another characteristic of human hemoglobins: their gene regulation, which is specific to the development period and in which we find typical proteins of the embryonic, fetal and adult periods.[3333. Sundd P, Gladwin MT, Novelli EM. Pathophysiology of Sickle Cell Disease. Annu Ver Pathol. 2019; 24(14):263-292. doi:10.1146/annurev-pathmechdis-012418-012838
https://doi.org/10.1146/annurev-pathmech...
] Thus, in neonatal screening tests aimed at the early laboratory diagnosis of hemoglobinopathies, we must consider some elements: the stage in which samples are collected (at birth or during the first week after), because the percentage values of adult hemoglobins are still not stabilized, therefore expressing the profile of the fetal period; the amount of variant hemoglobin, which has low percentages in most cases; and the method for collecting samples: on filter paper, the sample oxidizes more easily due to the contact with air; besides, fractions of more fragile or unstable hemoglobins can be lost, degraded. Thus, diagnostic methods for this purpose go beyond what is expected for protein separation, once they need to be accurate for the correct identification of fractions in less quantities.[3434. Dormandy E, Gulliford M, Bryan S, et al. Effectiveness of earlier antenatal screening for sickle cell disease and thalassaemia in primary care: cluster randomised trial. BMJ. 2010;5(341):c5132. doi:10.1136/bmj.c5132
https://doi.org/10.1136/bmj.c5132...
]

Based on the biochemical principle of protein separation, with the goal of identifying fractions with a low percentage and often under oxidation conditions, methodologies have been developed and improved, both nation and worldwide. However, the comparison between them is always present, drawing attention to the characteristics of hemoglobins, the type of collection and the age of the person.[3535. Greene DN, Pyle AL, Chang JS, Hoke C, Lorey T. Comparison of Sebia Capillarys Flex capillary electrophoresis with the BioRad Variant II high pressure liquid chromatography in thee valuation of hemoglobinopathies. Clin Chim Acta. 2012;16(413):1232-1238. doi:10.1016/j.cca.2012.03.027
https://doi.org/10.1016/j.cca.2012.03.02...
] High-performance chromatography (HPLC), capillary electrophoresis (CE), isoelectric focusing (IEF) and Mass spectrometry (MS/MS) are appropriate methods for NBS for SCD as the first methods.

In Brazil, initially, and still in some centers, IEF is used for screening. This method is perfectly accurate because it separates hemoglobin fractions by their isoelectric point, a characteristic of each protein. However, the manual demand, the delay in obtaining results and the number of samples for each collectionled to the implementation of innovations, leaving the system more active and automated. Nonetheless, a few hours were still necessary for its processing and for the diagnostic completion. With the HPLC on automated equipment, the use of specific diagnostic kits to the types of collection and the period of sample collection, it was possible to streamline the entire process. Moreover, the automation in the perforation and identification of samples allowed the reduction of errors in their exchange and an interfacing with the whole laboratory, speeding even more the finalization of tests and delivery of results. The systems were improved as the needs for screening were presented worldwide and, nowadays, specific systems for the neonatal diagnosis of hemoglobinopathies are guaranteed to be used in laboratories around the world.[3636. Khera R, Singh T, Khuana N, Gupta N, Dubey AP. HPLC in characterization of hemoglobin profile in thalassemia syndromes and hemoglobinopathies: a clinico hematological correlation. Indian J Hematol Blood Transfus. 2015;31(1):110-115. doi:10.1007/s12288-014-0409-x
https://doi.org/10.1007/s12288-014-0409-...
]

Due to the often need of confirming the profile found in a methodology by another method, CE was created to mainly assist in the identification of the associated hemoglobins variant forms. Since it is automated, fast and accurate, it has been widely used in laboratories for both the screening and confirmation of the phenotypes obtained.[3737. Degandt S, Coens R, Cauwelier B, Devos H, Langlois M, Emmerechts J. Evaluation of four hemoglobin separation analyzers for hemoglobinopathy diagnosis. J Clin Lab Anal. 2018;32(1):e22224. doi:10.1002/jcla.22224
https://doi.org/10.1002/jcla.22224...
]

Mass spectrometry employs a specific methodology and requires specific training for its execution and interpretation, but it allows a safe correlation of the phenotype/genotype. This methodology is not yet a reality in most services worldwide, even in Brazil. CE and MS / MS are not routinely used in Brazilian laboratories.

However, analyses of the mutations were necessary to carry out genetic counseling for family cases and diagnostic clarification for interactions of thalassemia forms and hemoglobin variants. The confirmation of genotypic profiles was based on the principle of gene amplification by PCR and mutations search by RFLP, specific allele, and sometimes sequencing of genes to identify less frequent mutations.[3838. Hu S, Zhan W, Wang J, et al. Establishment and application of a novel method based on single nucleotide polymorphism analysis for detecting β-globin gene cluster deletions. Sci Rep. 2020;26(1):18298. doi:10.1038/s41598-020-75507-6
https://doi.org/10.1038/s41598-020-75507...
] Knowing the population profile assists in the search for these mutants, and in the cases of miscegenated populations, such as the Brazilian, the molecular profile has been frequently requested. Thus, it is important to have specialized laboratories to support the elucidation of clinically relevant rarer hemoglobin profiles.[3939. Cintron-Garcia J, Guddati AK. Effect of immigration on mortality trends in sickle cell patients. Am J Blood Res. 2020;10(5):172-178. https://www.ncbi.nlm.nih.gov/pmc/articles/pmc7675126/
https://www.ncbi.nlm.nih.gov/pmc/article...
] The high cost of molecular studies does not allow all services to have access to this technology in the day-to-day care of patients with SCD in Brazil.

One of the major challenges of implementing population studies is the cost for the implementation of national programs. Although not a reality in Brazil yet, recent studies using rapid point-of-care (POC) have demonstrated excellent specificity and sensitivity to detect SCD during neonatal screening, shortening health care access for children positive for SCD. The cost for national SCD NBS programs may decrease significantly if efficient, accurate and inexpensive point-of-care (POC) devices become available; a number of these POC testing devices have recently been developed.[4040. McGann PT, Hoppe C. The pressing need for point-of-care diagnostics for sickle cell disease: A review of current and future technologies. Blood Cells Mol Dis. 2017;67: 104-113. doi:10.1016/j.bcmd.2017.08.010
https://doi.org/10.1016/j.bcmd.2017.08.0...
]

The challenges of neonatal screening for hemoglobinopathies in different services and evaluation of the Brazilian National Program

The target disease of a NBS programme for haemoglobinopathies in Brazil is SCD, including all genotypes as in other consensus worldwide.[44. Lobitz S, Telfer P, Cela E, et al. Newborn screening for sickle cell disease in Europe: recommendations from a Pan-European Consensus Conference. Br J Haematol. 2018;183(4):648-660. doi:10.1111/bjh.15600.
https://doi.org/10.1111/bjh.15600...
] And one of the main goals of the NNSP is to ensure that 100% of the country’s live births have access to the tests.[4141. Kato G, Piel F, Reid C, et al. Sickle cell disease. Nat Rev Dis Primers. 2018;4:18010. doi:10.1038/nrdp.2018.10
https://doi.org/10.1038/nrdp.2018.10...
]

In Brazil, the coverage is highly variable.The deficit in the collection coverage is associated to the lack of priority by municipal managers, to the lack of information about the diseases screened in the program, to the exams being carried out in a supplementary network, to the cultural traits not considered relevant for neonatal screening, to the lack of structure and supplies, and to the difficulty in accessing health centers and transporting samples.

According to the great genetic, cultural and economic diversity in the different regions of Brazil, we can cite the profile of the NNSP in two geographically distinct states as an example:

Screening for Hemoglobinopathies in the State of Bahia

In Brazil, SCD is considered a public health problem and the state with the highest occurrence is Bahia, with 1:650 live births and incidence of 1:17 live births HbS mutation.[2727. Adorno EV, Couto FD, Moura Neto JP, Menezes JF, Rêgo M, Reis MG, et al. Hemoglobinopathies in newborns from Salvador, Bahia, Northeast Brazil. Cad Saude Publica . 2005;21(1):292-298. doi:10.1590/s0102-311x2005000100032
https://doi.org/10.1590/s0102-311x200500...
]

The State of Bahia, located in the northeast region of Brazil, has a population of 14,930 million inhabitants, a birth rate of 18.8 live births per 1,000 inhabitants and a Human Development Index (HDI) of 0.660. According to IBGE data (PNAD continues), 81.1% of the population claims to haveblack or brown skin.[4242. IBGE. Projeções da população: Brasil e unidades da federação - revisão 2018. 2nd ed. Rio de Janeiro: IBGE; 2018. https://biblioteca.ibge.gov.br/index.php/biblioteca- catalogo?view=detalhes&id=2101597
https://biblioteca.ibge.gov.br/index.php...
] This data reflects the Afro-descendants miscegenation of the Bahia population.

Neonatal screening for sickle cell disease and other hemoglobinopathies began in July 2000, by means of a contract signed with the State Health Department. In 2001, the APAE-Salvador Neonatal Screening Reference Service was accredited by the Ministry of Health, which was followingthe requirements of Ministerial Decree No. 822/GM, dated June 6, 2001.[1919. Ministério da Saúde. Portaria GM/MS nº 822, de 6 de junho de 2001. Institui, no âmbito do Sistema Único de Saúde, o Programa Nacional de Triagem Neonatal/PNTN. Diário Oficial da União. 2001.] At that moment, the screening was expanded both in coverage and in health care of newborns. Since then, we have confirmed the high incidence of SCD in the population of the State, which has the largest Afro-descendant population in the country.

After these 20 years of neonatal screening, more than 3 million and 300 thousand newborns were screened in the State of Bahia, including the screening for sickle cell disease, with an average coverage varying from 85% to 89% in the recent years.

Thus, in the NNSP more than 5,000 newborns have already been diagnosed with some form of SCD, which requires an organization of the service network to meet the demands of these families, including basic and specialized care.

Screening for Hemoglobinopathies in the State of Rio Grande do Sul

The State of Rio Grande do Sul (RS), located in the Souther most region of Brazil, has a population of 11,400 million inhabitants and a birth rate of 11.6 live births per 1,000 inhabitants, with an HDI of 0.746. Regarding race, 80% of the population declares to have white skin, which is explained by the great European immigration to the region.

The annual average coverage is of 75.88%.[2020. Cardoso CS, Macedo JL, Diedrich VR, Magalhães CMB, Castro SM. Triagem neonatal de hemoglobinopatias no estado do Rio Grande do Sul no período de 2004 a 2014. Bol Cient Pediatr. 2017;6(3):77-84. https://www.sprs.com.br/sprs2013/bancoimg/171229113516bcped_06_03_a02.pdf
https://www.sprs.com.br/sprs2013/bancoim...
] When comparing it to the data from other Southern states, the coverage percentage in RS is below Santa Catarina’s (90%) [4343. Nunes AK, Wachholz RG, Rover MR, Souza LC. Prevalence of disorders detected by newborn screening in Santa Catarina. Arq Bras Endocrinol Metabol. 2013;57(5):360-367. doi:10.1590/s0004-27302013000500005
https://doi.org/10.1590/s0004-2730201300...
] and Paraná’s (100%) [2222. Watanabe AM, Pianovski MA, Zanis Neto J, et al. Prevalence of hemoglobin S in the State of Paraná, Brazil, basedon neonatal screening. Cad Saude Publica. 2008;24(5):993-1000. doi:10.1590/S0102-311X2008000500006
https://doi.org/10.1590/S0102-311X200800...
] and it is also below the national data. By assing the tests carried out in private network laboratories of the state, one can believe that there is a coverage of 99% of newborns. However, there is no concrete data regarding the number and results of the private network, making it difficult to know the real scope of the NNSP in the State of RS.

In accordance with the diversity amongst the different Southern states of the country, RS presents results for sickle cell disease (1:10,226 or 0.0098%) higher than the ones from the States of Santa Catarina [2121. Eller R, Silva DB. Evaluation of a neonatal screening program for sickle-cell disease. J Pediatr (Rio J). 2016;92:409-413. doi:10.1016/j.jped.2015.10.002
https://doi.org/10.1016/j.jped.2015.10.0...
] and Paraná [2222. Watanabe AM, Pianovski MA, Zanis Neto J, et al. Prevalence of hemoglobin S in the State of Paraná, Brazil, basedon neonatal screening. Cad Saude Publica. 2008;24(5):993-1000. doi:10.1590/S0102-311X2008000500006
https://doi.org/10.1590/S0102-311X200800...
], whose population have a similar ethnic composition, differing from other regions of the country where there was a greater African immigration, such as the Southeast and the Northeast.

Although coverage is an essential parameter, a neonatal screening program should not be evaluated without analyzing the consultations of children with abnormal results. While developing neonatal screening, the healthcare system should provide the necessary infrastructure to confirm the laboratory diagnosis of newborns and to provide appropriate counseling and treatment. Without this, the benefits achieved by the early identification of diseases can be lost.[4444. Núcleo de Ações e Pesquisa em Apoio Diagnóstico - NUPAD. Diagnóstico Situacional do Programa Nacional de Triagem Neonatal nos estados brasileiros: relatório técnico. Belo Horizonte: NUPAD; 2013.] Despite the advances made in the diagnosis and treatment of SCD, the survival rates have not yet managed to surpass the fifth decade of life, with an overall life expectancy estimated at around 30 years. It is known that socio-environmental conditions, personal care, antibiotic prophylaxis, vaccination, access to health services, hydration and adequate nutrition have a strong influence on the clinical evolution of the individual with SCD.[4545. Braga JAP. Medidas gerais no tratamento das doenças falciformes. Rev Bras Hematol Hemoter. 2007;29(3):233-238. doi:10.1590/S1516-84842007000300009
https://doi.org/doi:10.1590/S1516-848420...
] This occurs especially in regions where it is more difficult to have an early diagnosis, access to prophylaxis using penicillin and pneumococcal immunization, as well as treatment with Hydroxyurea.[4646. Mariano PC, Queiroz CAI, Costa SM, Bontempo FM, Ferreira Júnior MA, Ivo ML. Fatores de risco para mortalidade em pacientes com doença falciforme: uma revisão integrativa. Esc Anna Nery. 2020;24(2):e20190194. doi:10.1590/2177-9465-EAN-2019-0194
https://doi.org/10.1590/2177-9465-EAN-20...
] Treatment and management of the disease remain costly, making full access to care available only for the most privileged; otherwise, access is very limited because of increasing pressures on public health services.[4747. Piel FB, Steinberg MH, Rees DC. Sickle Cell Disease. N Engl J Med. 2017;376(16):1561-1573. doi:10.1056/nejmra1510865.
https://doi.org/10.1056/nejmra1510865...
]

In Brazil, one of the challenges of treating and monitoring patients can be justified by the lack of standardization in the issue of results. There are no national standardized procedures to guide the active search process. There are several problems with identifying cards for locating patients, with inadequate records of names and addresses, and with changes of address hindering active search processes. An alternative, as it exists in some countries in Europe (Belgium, Cyprus, Germany, Greece, Spain and UK) would be a national registriers for SCD could enhance monitoring of changing demographics, service delivery, and patients outcomes, and improve patient access to care with standardized data collection and coordinated follow-up.[44. Lobitz S, Telfer P, Cela E, et al. Newborn screening for sickle cell disease in Europe: recommendations from a Pan-European Consensus Conference. Br J Haematol. 2018;183(4):648-660. doi:10.1111/bjh.15600.
https://doi.org/10.1111/bjh.15600...
] In 2006, the Ministry of Health established a Technical Advisory Council for Sickle Cell Disease to help states and municipalities organize their programs of care for people with SCD in Brazil. The implementation of the SCD network in all the states is intended to guarantee decentralized care beginning with diagnosis, with the assistance of a multi-professional and multidisciplinary team, providing health education with a focus on self-management and access to specialized and high complexity care in all states of Brazil. Nonetheless, there are still few published studies on follow up and mortality that characterize the different aspects of this hemoglobinopathy in the Brazilian population and the results differ from region to region according to the different levels of organization of the assistance networks existing in the states.

Financial support, specific research, training programs, early diagnosis and intervention programs has made it possible to know the survival rates and the main causes of death amongst people with SCD. In addition, it allows preventive prophylaxis for patients with sickle cell anemia, such as vaccination and the administration of antibiotics (penicillin) up to the 5th year of age, to prevent from infections, which is its major mortality cause. The results of Minas Gerais showed that neonatal screening for SCD was not sufficient to significantly reduce child mortality. Economic and social development and increase of the knowledge about SCD among healthcare professionals and family are needed to overcome the high mortality rate. Several actions of care for patients with SCD need to be reviewed.[4848. Sabarense AP, Lima GO, Silva LML and Viana MB. Characterization of mortality in children with sickle cell disease diagnosed through the Newborn Screening Program. J Pediatr (Rio J) . 2015;91(3):242-247. doi:10.1016/j.jped.2014.08.006.
https://doi.org/10.1016/j.jped.2014.08.0...
] The studies conducted in the states of Rio de Janeiro and Mato Grosso do Sul reported that hydroxycarbamide therapy decreases mortality among patients with SCD. [4949. Arduini GAO, Rodrigues LP, Marqui ABT. Mortality by sickle cell disease in Brazil. Rev Bras Hematol Hemoter. 2017;39(1):52-56. doi:10.1016/j.bjhh.2016.09.008.
https://doi.org/10.1016/j.bjhh.2016.09.0...
-5050. Lobo CL, Ballas SK, Domingos AC, et al. Newborn screening program for hemoglobinopathies in Rio de Janeiro, Brazil. Ped Blood Canc. 2014;61(1):34-39. doi:10.1002/pbc.24711.
https://doi.org/10.1002/pbc.24711...
]

There is a lack of planning, monitoring of actions and federal funding for specific neonatal screening actions, which would qualify the comprehensive care processes for diagnosed patients, and a lack of relationship and integration to the Health Care Network, for the assembeling of programs for care regionalization. The great movement of populations, and their settlement in countries different from their homeland, is resulting in the emergence of diseases in geographic areas where they historically did not exist.

In the recent years, major advances have been made as to the diagnosis and treatment of these diseases, and more will come. However, in poor countries, such as the ones from Africa and Asia, the number of patients tends to increase. Brazil is also included on the list, because the resources for its public health care system not only are not increasing, but they also seem to be reducing. The country is at risk of going backwards in all the progressit has made until today, from the diagnosis to the treatment stage.

Conclusion

The variability of hemoglobin profiles identified in the different regions of Brazil reflects the heterogeneity of its population. Complications of SCD can be delayed and mitigated by early detection, reinforcing the importance of neonatal screening programs. Consequently, neonatal screening for SCD is being introduced progressively in different regions.

Overthese 20 years of a Brazilian neonatal screening program for hemoglobinopathies, the correct diagnosis of hemoglobin variants prevented the application of inappropriate therapies and procedures. Together with notification, these are essential for the planning of public health policies.

SCD population programs will only have social and ethical impacts if they incorporate their actions: to the early diagnosis, which optimizes the effectiveness of preventive and prophylactic actions in reducing morbidity and mortality, through the monitoring carried out by a multidisciplinary team; to the social care that each person with the disease requires; to the access to genetic information/guidance for family members; and to the access to essential medications. Prophylactic care is the essence of the treatment up to the fifth year of life, period with the highest occurrence of deaths and serious complications.

The early diagnosis and treatment, as well as the follow-up with a multidisciplinary team, are fundamental influences to the survival rate and the quality of life of patients with SCD.

Acknowledgments

The authors would like to thank Gabriela Pina for English review.

References

  • 1. da Fonseca SF, Amorim T, Purificação A, Gonçalves M, Boa-Sorte N. Hemoglobin A2 values in sickle cell disease patients quantified by high performance liquid chromatography and the influence of alpha thalassemia. Rev Bras Hematol Hemoter 2015;37(5):296-301. doi:10.1016/j.bjhh.2015.05.005
    » https://doi.org/10.1016/j.bjhh.2015.05.005
  • 2. Field JJ, Nathan DG. Advances in sickle cell therapies in the hydroxyurea era. Mol Med 2014;20(Suppl 1):S37-S42. doi:10.2119/molmed.2014.00187
    » https://doi.org/10.2119/molmed.2014.00187
  • 3. Odame I. Perspective: we need a global solution. Nature 2014;515(7526):S10. doi:10.1038/515S10a
    » https://doi.org/10.1038/515S10a
  • 4. Lobitz S, Telfer P, Cela E, et al Newborn screening for sickle cell disease in Europe: recommendations from a Pan-European Consensus Conference. Br J Haematol 2018;183(4):648-660. doi:10.1111/bjh.15600.
    » https://doi.org/10.1111/bjh.15600
  • 5. Callegari-Jacques SM, Grattapaglia D, Salzano FM, Salamoni SP, Crossetti SG, Ferreira ME, Hutz MH. Historical genetics: spatio temporal analysis of the formation of the Brazilian population. Am J Hum Biol 2003;15(6):824-834. doi:10.1002/ajhb.10217
    » https://doi.org/10.1002/ajhb.10217
  • 6. Lindenau JD, Wagner SC, Castro SM, Hutz MH. The effects of old and recent migration waves in the distribution of HBB*S globin gene haplotypes. Genet Mol Biol 2016; 39(4): 515-523. doi:10.1590/1678-4685-gmb-2016-0032.
    » https://doi.org/10.1590/1678-4685-gmb-2016-0032
  • 7. Salzano FM, Bortolini MC. The evolution and genetics of Latin American populations Cambridge: Cambridge University Press; 2002.
  • 8. Salzano FM, Freire-Maia N. Problems in human biology. A study of Brazilian populations Detroit: Wayne State University Press; 1970. doi:10.1002/ajhb.10217.
    » https://doi.org/10.1002/ajhb.10217
  • 9. Wagner SC, de Castro SM, Gonzalez TP, et al Neonatal screening for hemoglobinopathies: results of a public health system in South Brazil. Genet Test Mol Biomarkers 2010;14(4):565-569. doi:10.1089/gtmb.2010.0008
    » https://doi.org/10.1089/gtmb.2010.0008
  • 10. WHO. Meeting on Advances in Diagnosis Treatment and Prevention of Hereditary Diseases. Report of a WHO Meeting on Advances in Diagnosis, Treatment and Prevention of Hereditary Diseases. Geneva: WHO; 1989. https://apps.who.int/iris/handle/10665/58835
    » https://apps.who.int/iris/handle/10665/58835
  • 11. Silva-Pinto AC, Alencar de Queiroz MC, Antoniazzo PJ, Arruda M, Pimentel dos Santos H. The Neonatal Screening Program in Brazil, Focus on Sickle Cell Disease (SCD). Int J Neonatal Screen 2019; 5(1):11. doi:10.3390/ijns5010011
    » https://doi.org/10.3390/ijns5010011
  • 12. Abarca G, Navarrete M, Trejos R, de Céspedes C, Saborío M. Abnormal haemoglobins in the new Born human population of Costa Rica. Rev Biol Trop 2008; 56:995-1001. https://www.scielo.sa.cr/pdf/rbt/v56n3/art02v56n3.pdf
    » https://www.scielo.sa.cr/pdf/rbt/v56n3/art02v56n3.pdf
  • 13. Echeverry-Coral SJ, Colmenares-Mejía CC, Yepes-Molina ZX, Martínez-Nieto O, Isaza-Ruget MA. Hemoglobinopathy detection through na institutional neonatal screening program in Colombia. J Bras Patol Med Lab 2016; 52(5):299-306. doi:10.5935/1676-2444.20160050
    » https://doi.org/10.5935/1676-2444.20160050
  • 14. Audicio P, Segobia B, Queijo C, Queiruga G. Newborn Screening Pilot Program of Hemoglobinopathies: firstresults in Uruguay. Acta Bioquím Clín Latinoam 2017;51(2):243-248. https://www.redalyc.org/pdf/535/53552508010.pdf
    » https://www.redalyc.org/pdf/535/53552508010.pdf
  • 15. Knight-Madden J, Lee K, Elana G, Elenga N, Marcheco-Teruel B, Keshi N, Hardy-Dessources MD. Newborn Screening for Sickle Cell Disease in the Caribbean: An Update of the Present Situation and of the Disease Prevalence. Int J Neonatal Screen 2019;5(1):5. doi:10.3390/ijns5010005
    » https://doi.org/10.3390/ijns5010005
  • 16. Backes CE, Mallmann FG, DassiT, Bazzo ML, Santos-Silva MC. Triagem neonatal como um problema de saúde pública. Rev Bras Hemat Hemoter 2005;27:43-47. doi:10.1590/S1516-84842005000100011
    » https://doi.org/10.1590/S1516-84842005000100011
  • 17. Brandelise S, Pinheiro V, Gabetta CS, Hambleton I, Serjeant B, Serjeant G. Newborn screening for sickle cell disease in Brazil: the Campinas experience. Clin Lab Haematol 2004;26(1):15-19. doi:10.1111/j.0141-9854.2003.00576.x
    » https://doi.org/10.1111/j.0141-9854.2003.00576.x
  • 18. Brasil. Ministério da Saúde. Secretaria de Atenção a Saúde. Departamento de Atenção Especializada e Temática. Triagem neonatal biológica: manual técnico Brasília: Ministério da Saúde; 2016.
  • 19. Ministério da Saúde. Portaria GM/MS nº 822, de 6 de junho de 2001. Institui, no âmbito do Sistema Único de Saúde, o Programa Nacional de Triagem Neonatal/PNTN. Diário Oficial da União. 2001.
  • 20. Cardoso CS, Macedo JL, Diedrich VR, Magalhães CMB, Castro SM. Triagem neonatal de hemoglobinopatias no estado do Rio Grande do Sul no período de 2004 a 2014. Bol Cient Pediatr 2017;6(3):77-84. https://www.sprs.com.br/sprs2013/bancoimg/171229113516bcped_06_03_a02.pdf
    » https://www.sprs.com.br/sprs2013/bancoimg/171229113516bcped_06_03_a02.pdf
  • 21. Eller R, Silva DB. Evaluation of a neonatal screening program for sickle-cell disease. J Pediatr (Rio J) 2016;92:409-413. doi:10.1016/j.jped.2015.10.002
    » https://doi.org/10.1016/j.jped.2015.10.002
  • 22. Watanabe AM, Pianovski MA, Zanis Neto J, et al Prevalence of hemoglobin S in the State of Paraná, Brazil, basedon neonatal screening. Cad Saude Publica 2008;24(5):993-1000. doi:10.1590/S0102-311X2008000500006
    » https://doi.org/10.1590/S0102-311X2008000500006
  • 23. Hadachi S, Iskandar M. Triagem neonatal de hemoglobinopatias - Serviço de referência em triagem neonatal (SRTN) Apae São Paulo. Rev Bras Hematol Hemoter 2009;31(4):54.
  • 24. Lobo CLC, Ballas SK, Bonini Domingos AC, Moura PG, Nascimento EM, Cardoso GP, Carvalho SMF. Newborn Screening Program for Hemoglobinopathies in Rio de Janeiro. Brazil Pediatr Blood Cancer 2014;61:34-39. doi:10.1002/pbc.24711
    » https://doi.org/10.1002/pbc.24711
  • 25. Paixão MC, Cunha Ferraz MH, Januario JN, et al Reliability of iselectro focusing for the detection of Hb S, Hb C and Hb D in a pioneering population-based program of newborn screening in Brazil. Hemoglobin 2001;25:297-303. doi:10.1081/hem-100105222
    » https://doi.org/10.1081/hem-100105222
  • 26. Diniz D, Guedes C, Barbosa L, et al Prevalence of sickle cell trait and sickle cell anemia among newborns in the Federal District, Brazil, 2004 to 2006. Cad Saude Publica . 2009;25:188-194. doi:10.1590/s0102-311x2009000100020
    » https://doi.org/10.1590/s0102-311x2009000100020
  • 27. Adorno EV, Couto FD, Moura Neto JP, Menezes JF, Rêgo M, Reis MG, et al Hemoglobinopathies in newborns from Salvador, Bahia, Northeast Brazil. Cad Saude Publica . 2005;21(1):292-298. doi:10.1590/s0102-311x2005000100032
    » https://doi.org/10.1590/s0102-311x2005000100032
  • 28. Amorim T, Pimentel H, Fontes MIMM, Purificação A, Lessa P, Boa-Sorte N. Avaliação do programa de Triagem Neonatal na Bahia entre 2007 e 2009 - As lições da Doença Falciforme. Gaz Med Bahia 2010;80(3):10-13. http://www.gmbahia.ufba.br/index.php/gmbahia/article/view/1103
    » http://www.gmbahia.ufba.br/index.php/gmbahia/article/view/1103
  • 29. Lipinski-Figueiredo E, Estelita S, et al. Dados preliminares da fase II do programa de triagem neonatal do estado de Alagoas. Rev Bras Hemat Hemoter 2009;31(4):62
  • 30. de Araujo MC, Serafim ES, de Castro WA Jr., et al Prevalenceof abnormal hemoglobins in newborns in Natal, Rio Grande do Norte, Brazil. Cad Saude Publica . 2004; 20:123-128. doi:10.1590/S0102-311X2004000100027
    » https://doi.org/10.1590/S0102-311X2004000100027
  • 31. Brasil. Ministério da Saúde. Secretaria de Atenção Especializada à Saúde. Departamento de Atenção Especializada e Temática. Caderno de informação: triagem neonatal. Dados 2014-2016 Brasília: Ministério da Saúde ; 2021.
  • 32. Brasil. Ministério da Saúde. Relatório Anual de Dados do PNTN Brasília: Ministério da Saúde ; 2018.
  • 33. Sundd P, Gladwin MT, Novelli EM. Pathophysiology of Sickle Cell Disease. Annu Ver Pathol 2019; 24(14):263-292. doi:10.1146/annurev-pathmechdis-012418-012838
    » https://doi.org/10.1146/annurev-pathmechdis-012418-012838
  • 34. Dormandy E, Gulliford M, Bryan S, et al Effectiveness of earlier antenatal screening for sickle cell disease and thalassaemia in primary care: cluster randomised trial. BMJ 2010;5(341):c5132. doi:10.1136/bmj.c5132
    » https://doi.org/10.1136/bmj.c5132
  • 35. Greene DN, Pyle AL, Chang JS, Hoke C, Lorey T. Comparison of Sebia Capillarys Flex capillary electrophoresis with the BioRad Variant II high pressure liquid chromatography in thee valuation of hemoglobinopathies. Clin Chim Acta 2012;16(413):1232-1238. doi:10.1016/j.cca.2012.03.027
    » https://doi.org/10.1016/j.cca.2012.03.027
  • 36. Khera R, Singh T, Khuana N, Gupta N, Dubey AP. HPLC in characterization of hemoglobin profile in thalassemia syndromes and hemoglobinopathies: a clinico hematological correlation. Indian J Hematol Blood Transfus 2015;31(1):110-115. doi:10.1007/s12288-014-0409-x
    » https://doi.org/10.1007/s12288-014-0409-
  • 37. Degandt S, Coens R, Cauwelier B, Devos H, Langlois M, Emmerechts J. Evaluation of four hemoglobin separation analyzers for hemoglobinopathy diagnosis. J Clin Lab Anal 2018;32(1):e22224. doi:10.1002/jcla.22224
    » https://doi.org/10.1002/jcla.22224
  • 38. Hu S, Zhan W, Wang J, et al Establishment and application of a novel method based on single nucleotide polymorphism analysis for detecting β-globin gene cluster deletions. Sci Rep 2020;26(1):18298. doi:10.1038/s41598-020-75507-6
    » https://doi.org/10.1038/s41598-020-75507-6
  • 39. Cintron-Garcia J, Guddati AK. Effect of immigration on mortality trends in sickle cell patients. Am J Blood Res 2020;10(5):172-178. https://www.ncbi.nlm.nih.gov/pmc/articles/pmc7675126/
    » https://www.ncbi.nlm.nih.gov/pmc/articles/pmc7675126/
  • 40. McGann PT, Hoppe C. The pressing need for point-of-care diagnostics for sickle cell disease: A review of current and future technologies. Blood Cells Mol Dis 2017;67: 104-113. doi:10.1016/j.bcmd.2017.08.010
    » https://doi.org/10.1016/j.bcmd.2017.08.010
  • 41. Kato G, Piel F, Reid C, et al Sickle cell disease. Nat Rev Dis Primers 2018;4:18010. doi:10.1038/nrdp.2018.10
    » https://doi.org/10.1038/nrdp.2018.10
  • 42. IBGE. Projeções da população: Brasil e unidades da federação - revisão 2018. 2nd ed. Rio de Janeiro: IBGE; 2018. https://biblioteca.ibge.gov.br/index.php/biblioteca- catalogo?view=detalhes&id=2101597
    » https://biblioteca.ibge.gov.br/index.php/biblioteca- catalogo?view=detalhes&id=2101597
  • 43. Nunes AK, Wachholz RG, Rover MR, Souza LC. Prevalence of disorders detected by newborn screening in Santa Catarina. Arq Bras Endocrinol Metabol 2013;57(5):360-367. doi:10.1590/s0004-27302013000500005
    » https://doi.org/10.1590/s0004-27302013000500005
  • 44. Núcleo de Ações e Pesquisa em Apoio Diagnóstico - NUPAD. Diagnóstico Situacional do Programa Nacional de Triagem Neonatal nos estados brasileiros: relatório técnico Belo Horizonte: NUPAD; 2013.
  • 45. Braga JAP. Medidas gerais no tratamento das doenças falciformes. Rev Bras Hematol Hemoter 2007;29(3):233-238. doi:10.1590/S1516-84842007000300009
    » https://doi.org/doi:10.1590/S1516-84842007000300009
  • 46. Mariano PC, Queiroz CAI, Costa SM, Bontempo FM, Ferreira Júnior MA, Ivo ML. Fatores de risco para mortalidade em pacientes com doença falciforme: uma revisão integrativa. Esc Anna Nery 2020;24(2):e20190194. doi:10.1590/2177-9465-EAN-2019-0194
    » https://doi.org/10.1590/2177-9465-EAN-2019-0194
  • 47. Piel FB, Steinberg MH, Rees DC. Sickle Cell Disease. N Engl J Med 2017;376(16):1561-1573. doi:10.1056/nejmra1510865.
    » https://doi.org/10.1056/nejmra1510865
  • 48. Sabarense AP, Lima GO, Silva LML and Viana MB. Characterization of mortality in children with sickle cell disease diagnosed through the Newborn Screening Program. J Pediatr (Rio J) . 2015;91(3):242-247. doi:10.1016/j.jped.2014.08.006.
    » https://doi.org/10.1016/j.jped.2014.08.006
  • 49. Arduini GAO, Rodrigues LP, Marqui ABT. Mortality by sickle cell disease in Brazil. Rev Bras Hematol Hemoter 2017;39(1):52-56. doi:10.1016/j.bjhh.2016.09.008.
    » https://doi.org/10.1016/j.bjhh.2016.09.008
  • 50. Lobo CL, Ballas SK, Domingos AC, et al Newborn screening program for hemoglobinopathies in Rio de Janeiro, Brazil. Ped Blood Canc 2014;61(1):34-39. doi:10.1002/pbc.24711.
    » https://doi.org/10.1002/pbc.24711
  • Funding

    This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Publication Dates

  • Publication in this collection
    14 June 2021
  • Date of issue
    2021

History

  • Received
    15 Jan 2021
  • Reviewed
    11 Apr 2021
  • Accepted
    19 Apr 2021
Latin American Society Inborn Errors and Neonatal Screening (SLEIMPN); Instituto Genética para Todos (IGPT) Rua Ramiro Barcelos, 2350, CEP: 90035-903, Porto Alegre, RS - Brasil, Tel.: 55-51-3359-6338, Fax: 55-51-3359-8010 - Porto Alegre - RS - Brazil
E-mail: rgiugliani@hcpa.edu.br