1,645
Views
31
CrossRef citations to date
0
Altmetric
Review

Current perspectives in assessing humoral immunity after measles vaccination

, , , &
Pages 75-87 | Received 04 Oct 2018, Accepted 10 Dec 2018, Published online: 26 Dec 2018

References

  • World Health Organization. Progress towards regional measles elimination – worldwide, 2000–2016. Releve epidemiologique hebdomadaire. 2017 Oct 27;92(43):649–659.
  • Dabbagh A, Patel MK, Dumolard L, et al. Progress toward regional measles elimination - worldwide, 2000–2016. MMWR. 2017 Oct 27;66(42):1148–1153.
  • Centers for Disease Control and Prevention. Measles Cases and Outbreaks.[cited 2018 Sep 30] Available from: http://www.cdc.gov/measles/cases-outbreaks.html
  • Hall V, Banerjee E, Kenyon C, et al. Measles outbreak - Minnesota April-May 2017. MMWR. 2017 Jul 14;66(27):713–717.
  • World Health Organization. Europe observes a 4-fold increase in measles cases in 2017 compared to previous year.[cited 2018 Sept 30]. Available from: http://www.euro.who.int/en/media-centre/sections/press-releases/2018/europe-observes-a-4-fold-increase-in-measles-cases-in-2017-compared-to-previous-year
  • Williams S European measles cases quadrupled in 2017. The Scientist. 2018 Available from: https://www.the-scientist.com/the-nutshell/european-measles-cases-quadrupled-in-2017-30261
  • Mina MJ, Metcalf CJ, de Swart RL, et al. Long-term measles-induced immunomodulation increases overall childhood infectious disease mortality. Science. 2015 May 8;348(6235):694–699.
  • de Vries RD, McQuaid S, van AG, et al. Measles immune suppression: lessons from the macaque model. PLoS Pathog. 2012;8(8):E1002885.
  • de Vries RD, de Swart RL. Measles immune suppression: functional impairment or numbers game? PLoS Pathog. 2014 Dec;10(12):E1004482.
  • Laksono BM, Grosserichter-Wagener C, de Vries RD, et al. In vitro measles virus infection of human lymphocyte subsets demonstrates high susceptibility and permissiveness of both naive and memory B cells. J Virol. 2018 Apr 15;92:8.
  • Laksono BM, de Vries RD, McQuaid S, et al. Measles virus host invasion and pathogenesis. Viruses. 2016 Jul 28;8:8.
  • Wendorf KA, Winter K, Zipprich J, et al. Subacute sclerosing panencephalitis: the devastating measles complication that might be more common than previously estimated. Clinl Infect Dis. 2017 Jul 15;65(2):226–232.
  • Muller CP, Kremer JR, Best JM, et al. Reducing global disease burden of measles and rubella: report of the WHO Steering Committee on research related to measles and rubella vaccines and vaccination, 2005. Vaccine. 2007;25(1):1–9.
  • Patel MK, Gacic-Dobo M, Strebel PM, et al. Progress toward regional measles elimination - worldwide, 2000–2015. MMWR. 2016 Nov 11;65(44):1228–1233.
  • Meissner HC, Strebel PM, Orenstein WA. Measles vaccines and the potential for worldwide eradication of measles. Pediatrics. 2004;114(4):1065–1069.
  • Elliman D, Sengupta N. Measles. Curr Opin Infect Dis. 2005;18(3):229–234.
  • Haralambieva IH, Ovsyannikova IG, Pankratz VS, et al. The genetic basis for interindividual immune response variation to measles vaccine: new understanding and new vaccine approaches. Expert Rev Vaccines. 2013 Jan;12(1):57–70.
  • Uzicanin A, Zimmerman L. Field effectiveness of live attenuated measles-containing vaccines: a review of published literature. J Infect Dis. 2011;204(Suppl 1):S133–S148.
  • Boulianne N, De Serres G, Ratnam S, et al. Measles, mumps, and rubella antibodies in children 5–6 years after immunization: effect of vaccine type and age at vaccination. Vaccine. 1995;13:1611–1616.
  • Christenson B, Bttiger M. Measles antibody: comparison of long-term vaccination titres, early vaccination titres and naturally acquired immunity to and booster effects on the measles virus. Vaccine. 1994;12:129–133.
  • Mitchell LA, Tingle AJ, Décarie D, et al. Serologic responses to measles, mumps, and rubella (MMR) vaccine in healthy infants: failure to respond to measles and mumps components may influence decisions on timing of the second dose of MMR. Can J Public Health. 1998;89(5):325–328.
  • Moss W. Measles in vaccinated individuals and the future of measles elimination. Clin Infect Dis. 2018;67:1320–1321.
  • Moss WJ. Measles control and the prospect of eradication. Curr Top Microbiol Immunol. 2009;330:173–189.
  • Measles Fact Sheet No. 286. Available from: http://www.who.int/en/news-room/fact-sheets/detail/measles. 2018.
  • Parker Fiebelkorn A, Redd SB, Gallagher K, et al. Measles in the United States during the postelimination era. J Infect Dis. 2010;202(10):1520–1528.
  • Ga P, Rm J, Am T, et al. Measles re-immunization in children seronegative after initial immunization. JAMA. 1997;277:1156–1158.
  • Seward JF, Orenstein WA. A rare event: A measles outbreak in a population with high 2-dose measles vaccine coverage. Clinl Infect Dis. 2012;55(3):403–405.
  • De Serres G, Boulianne N, Defay F, et al. Higher risk of measles when the first dose of a 2-dose schedule of measles vaccine is given at 12–14 months versus 15 months of age. Clin Infect Dis. 2012;55(3):394–402.
  • Ga P, Rm J. Failure to reach the goal of measles elimination. Apparent paradox of measles infections in immunized persons. Arch Internal Med. 1994;154:1815–1820.
  • Paunio M, Peltola H, Valle M, et al. Explosive school-based measles outbreak: intense exposure may have resulted in high risk, even among revaccinees. Am J Epidemiol. 1998;148(11):1103–1110.
  • Mathias RG, Meekison WG, Arcand TA, et al. The role of secondary vaccine failures in measles outbreaks. Am J Public Health. 1989;79:475–478.
  • Sugerman DE, Barskey AE, Delea MG, et al. Measles outbreak in a highly vaccinated population, San Diego, 2008: role of the intentionally undervaccinated. Pediatrics. 2010;125(4):747–755.
  • Pannuti CS, Morello RJ, Moraes JC, et al. Identification of primary and secondary measles vaccine failures by measurement of immunoglobulin G avidity in measles cases during the 1997 Sao Paulo epidemic. Clin Diagn Lab Immunol. 2004;11(1):119–122.
  • Hickman CJ, Hyde TB, Sowers SB, et al. Laboratory characterization of measles virus infection in previously vaccinated and unvaccinated individuals. J Infect Dis. 2011;204(Suppl 1):S549–S558.
  • Glass K, Grenfell BT. Waning immunity and subclinical measles infections in England. Vaccine. 2004;22(29–30):4110–4116.
  • Mossong J, Nokes DJ, Edmunds WJ, et al. Modeling the impact of subclinical measles transmission in vaccinated populations with waning immunity. Am J Epidemiol. 1999;150(11):1238–1249.
  • Ja W, Ga P. Measles and mumps outbreaks in the United States: think globally, vaccinate locally [editorial]. Vaccine. 2014 Aug 20;32(37):4703–4704.
  • Papania MJ, Wallace GS, Rota PA, et al. Elimination of endemic measles, rubella, and congenital rubella syndrome from the Western hemisphere: the US experience [historical article]. JAMA Pediatr. 2014 Feb;168(2):148–155.
  • Centers for Disease Control and Prevention. Notes from the field: measles outbreak associated with a traveler returning from India - North Carolina, april-may 2013. MMWR. 2013 Sep 13;62(36):753.
  • Centers for Disease Control and Prevention. Notes from the field: measles outbreak among members of a religious community - Brooklyn, New York, March-June 2013. MMWR 2013 Sep 13;62(36):752–753.
  • Zipprich J, Hacker JK, Murray EL, et al. Notes from the field: measles - California, January 1-april 18, 2014. MMWR. 2014 Apr 25;63(16):362–363.
  • Centers for Disease Control and Prevention. Notes from the field: measles outbreak–Indiana, June-July 2011. Morbidity and Mortality Weekly Report. 2011;34:1169.
  • Notes from the field: Multiple cases of measles after exposure during air travel–Australia and New Zealand, January. 2011. MMWR. 2011;60(25):851.
  • Notes from the field: measles outbreak–Hennepin county, Minnesota, February–March. 2011. MMWR. 2011;60(13):421.
  • Katz SL, Hinman AR. Summary and conclusions: measles elimination meeting, 16–17 March 2000. J Infect Dis. 2004;189(Suppl 1):S43–S47.
  • Collective Editorial Team. Measles once again endemic in the United Kingdom [News]. Euro Surveill. Jul 3 2008;13(27).
  • Centers for Disease Control and Prevention. Measles - United States, 2011. MMWR. 2012;61:253–257.
  • Rosen JB, Rota JS, Hickman CJ, et al. Outbreak of measles among persons with prior evidence of immunity, New York City, 2011. Clin Infect Dis. 2014 May;58(9):1205–1210. PubMed PMID: 24585562; eng.
  • Haralambieva IH, Kennedy RB, Ovsyannikova IG, et al. Variability in humoral immunity to measles vaccine: new developments. Trends Mol Med. 2015 Dec;21(12):789–801.
  • LE M, Sr P, Pem F, et al. Duration of live measles vaccine-induced immunity. Pediatr Infect Dis J. 1990;9:101–110.
  • Cherry JD, Zahn M. Clinical characteristics of measles in previously vaccinated and unvaccinated patients in California. Clin Infect Dis. 2018;67(9):1315–1319.
  • Clemmons NS, Wallace GS, Patel M, et al. Incidence of measles in the United States, 2001–2015. JAMA. 2017 Oct 3;318(13):1279–1281.
  • Increased transmission and outbreaks of measles–European Region. 2011. MMWR. 2011 Dec 2;60(47):1605–1610.
  • Griffin DE. Measles Vaccine. Viral Immunol. 2018 Mar;31(2):86–95.
  • Sowers SB, Rota JS, Hickman CJ, et al. High concentrations of measles neutralizing antibodies and high-avidity measles IgG accurately identify measles reinfection cases. Clin Vaccine Immunol. 2016 Aug;23(8):707–716.
  • Hahne SJ, Nic Lochlainn LM, van Burgel ND, et al. Measles outbreak among previously immunized healthcare workers, the Netherlands, 2014. J Infect Dis. 2016 Dec 15;214(12):1980–1986.
  • Tatsuo H, Ono N, Tanaka K, et al. SLAM (CDw150) is a cellular receptor for measles virus. Nature. 2000;406(6798):893–897.
  • Noyce RS, Bondre DG, Ha MN, et al. Tumor cell marker PVRL4 (nectin 4) is an epithelial cell receptor for measles virus. PLoS Pathog. 2011;7(8):e1002240.
  • Muhlebach MD, Mateo M, Sinn PL, et al. Adherens junction protein nectin-4 is the epithelial receptor for measles virus. Nature. 2011;480(7378):530–533.
  • Cattaneo R. Four viruses, two bacteria, and one receptor: membrane cofactor protein (CD46) as pathogens’ magnet. J Virol. 2004;78(9):4385–4388.
  • Griffin DE. The immune response in measles: virus control, clearance and protective immunity. Viruses. 2016 Oct 12;8(10):86–95.
  • Zilliox MJ, Parmigiani G, Griffin DE. Gene expression patterns in dendritic cells infected with measles virus compared with other pathogens. Proc Natl Acad Sci USA. 2006;103(9):3363–3368.
  • Haralambieva IH, Ovsyannikova IG, Dhiman N, et al. Differential cellular immune responses to wild-type and attenuated edmonston tag measles virus strains are primarily defined by the viral phosphoprotein gene. J Med Virol. 2010;82(11):1966–1975.
  • Chen RT, Markowitz LE, Albrecht P, et al. Measles antibody: reevaluation of protective titers. J Infect Dis. 1990;162(5):1036–1042.
  • de Swart RL, Yuksel S, Osterhaus AD. Relative contributions of measles virus hemagglutinin- and fusion protein-specific serum antibodies to virus neutralization. J Virol. 2005;79(17):11547–11551.
  • Polack FP, Lee SH, Permar S, et al. Successful DNA immunization against measles: neutralizing antibody against either the hemagglutinin or fusion glycoprotein protects rhesus macaques without evidence of atypical measles. Nat Med. 2000;6(7):776–781.
  • Bouche FB, Ertl OT, Muller CP. Neutralizing B cell response in measles. Viral Immunol. 2002;15(3):451–471.
  • de Vries RD, de Swart RL. Evaluating measles vaccines: can we assess cellular immunity? Exp Rev Vaccines. 2012 Jul;11(7):779–782.
  • Crotty S. A brief history of T cell help to B cells. Nat Rev Immunol. 2015 Mar;15(3):185–189.
  • Haralambieva IH, Ovsyannikova IG, Vierkant RA, et al. Development of a novel efficient fluorescence-based plaque reduction microneutralization assay for measles immunity. Clin Vaccine Immunol. 2008;15(7):1054–1059.
  • Plotkin SA. Correlates of protection induced by vaccination. Clin Vacci ne Immunol. 2010;17(7):1055–1065.
  • Haralambieva IH, Ovsyannikova IG, O’Byrne M, et al. A large observational study to concurrently assess persistence of measles specific B-cell and T-cell immunity in individuals following two doses of MMR vaccine. Vaccine. 2011;29(27):4485–4491.
  • Polack FP, Hoffman SJ, Crujeiras G, et al. A role for nonprotective complement-fixing antibodies with low avidity for measles virus in atypical measles. Nat Med. 2003;9(9):1209–1213.
  • World Health Organization. Correlates of vaccine-induced protection: methods and implications. Geneva, Switzerland 2013. Available from: http://apps.who.int/iris/bitstream/handle/10665/84288/WHO_IVB_13.01_eng.pdf;jsessionid=F944A52651CB43AEC79D14568617D67D?sequence=1
  • Dimech W, Mulders MN. A review of testing used in seroprevalence studies on measles and rubella. Vaccine. 2016 Jul 29;34(35):4119–4122.
  • Dimech W, Mulders MN. A 16-year review of seroprevalence studies on measles and rubella. Vaccine. 2016 Jul 29;34(35):4110–4118.
  • Thompson KM, Odahowski CL. Systematic review of measles and rubella serology studies. Risk Anal. 2016 Jul;36(7):1459–1486.
  • Binnicker MJ, Jespersen DJ, Rollins LO. Evaluation of the bio-rad bioplex measles, mumps, rubella, and varicella-zoster virus IgG multiplex bead immunoassay. Clin Vaccine Immunol. 2011 Sep;18(9):1524–1526.
  • Smits GP, van Gageldonk PG, Schouls LM, et al. Development of a bead-based multiplex immunoassay for simultaneous quantitative detection of IgG serum antibodies against measles, mumps, rubella, and varicella-zoster virus. Clin Vaccine Immunol. 2012 Mar;19(3):396–400.
  • Tischer A, Andrews N, Kafatos G, et al. Standardization of measles, mumps and rubella assays to enable comparisons of seroprevalence data across 21 European countries and Australia. Epidemiol Infect. 2007 Jul;135(5):787–797.
  • Hubschen JM, Bork SM, Brown KE, et al. Challenges of measles and rubella laboratory diagnostic in the era of elimination. Clin Microbiol Infect. 2017 Aug;23(8):511–515.
  • World Health Organization. Expert committee on Biological standardization, October 8–12, 2007: report on the collaborative study to investigate the relationship between the 1st IRP and the 2nd and 3rd international standards for anti-measles serum/plasma in both ELISA and PRNT. Geneva, Switzerland 2007. Available from: http://www.who.int/biologicals/BS07%202076anti-measles.pdf.
  • Haralambieva IH, Simon WL, Kennedy RB, et al. Profiling of measles-specific humoral immunity in individuals following two doses of MMR vaccine using proteome microarrays. Viruses. 2015;7(3):1113–1133.
  • Mercader S, Garcia P, Bellini WJ. Measles virus IgG avidity assay for use in classification of measles vaccine failure in measles elimination settings. Clin Vaccine Immunol. 2012 Nov;19(11):1810–1817.
  • Latner DR, McGrew M, Williams N, et al. Enzyme-linked immunospot assay detection of mumps-specific antibody-secreting B cells as an alternative method of laboratory diagnosis. Clin Vaccine Immunol. 2011;18(1):35–42.
  • Carter MJ, Mitchell RM, Meyer Sauteur PM, et al. The antibody-secreting cell response to infection: kinetics and clinical applications. Front Immunol. 2017;8:630.
  • Kakoulidou M, Ingelman-Sundberg H, Johansson E, et al. Kinetics of antibody and memory B cell responses after MMR immunization in children and young adults. Vaccine. 2013 Jan 11;31(4):711–717.
  • Buisman AM, de Rond CG, Ozturk K, et al. Long-term presence of memory B-cells specific for different vaccine components. Vaccine. 2009 Dec 10;28(1):179–186.
  • Titanji K, De Milito A, Cagigi A, et al. Loss of memory B cells impairs maintenance of long-term serologic memory during HIV-1 infection. Blood. 2006 Sep 1;108(5):1580–1587.
  • Ellebedy AH, Jackson KJ, Kissick HT, et al. Defining antigen-specific plasmablast and memory B cell subsets in human blood after viral infection or vaccination. Nat Immunol. 2016 Oct;17(10):1226–1234.
  • Hammarlund E, Thomas A, Amanna IJ, et al. Plasma cell survival in the absence of B cell memory. Nat Commun. 2017 Nov 24;8(1):1781.
  • Amanna IJ, Carlson NE, Slifka MK. Duration of humoral immunity to common viral and vaccine antigens. N Engl J Med. 2007;357(19):1903–1915.
  • Amanna IJ, Slifka MK. Mechanisms that determine plasma cell lifespan and the duration of humoral immunity. Immunol Rev. 2010 Jul;236:125–138.
  • Fairfax KA, Kallies A, Nutt SL, et al. Plasma cell development: from B-cell subsets to long-term survival niches. Semin Immunol. 2008 Feb;20(1):49–58.
  • Radbruch A, Muehlinghaus G, Luger EO, et al. Competence and competition: the challenge of becoming a long-lived plasma cell. Nat Rev Immunol. 2006 Oct;6(10):741–750.
  • Pensieroso S, Cagigi A, Palma P, et al. Timing of HAART defines the integrity of memory B cells and the longevity of humoral responses in HIV-1 vertically-infected children. Proc Natl Acad Sci USA. 2009 May 12;106(19):7939–7944.
  • Rocca S, Santilli V, Cotugno N, et al. Waning of vaccine-induced immunity to measles in kidney transplanted children. Medicine (Baltimore). 2016 Sep;95(37):e4738.
  • Kotton CN. Immunization after kidney transplantation-what is necessary and what is safe? Nat Rev Nephrol. 2014 Oct;10(10):555–562.
  • Cotugno N, Finocchi A, Cagigi A, et al. Defective B-cell proliferation and maintenance of long-term memory in patients with chronic granulomatous disease. J Allergy Clin Immunol. 2015 Mar;135(3):753–61 e2.
  • World Health Organization. Global measles & rubella strategic plan 2012–2020. Geneva 2012. http://apps.who.int/iris/bitstream/10665/44855/1/9789241503396_eng.pdf.
  • Ovsyannikova IG, Pankratz VS, Vierkant RA, et al. Consistency of HLA associations between two independent measles vaccine cohorts: a replication study. Vaccine. 2012;30(12):2146–2152.
  • Ovsyannikova IG, Schaid DJ, Larrabee BR, et al. A large population-based association study between HLA and KIR genotypes and measles vaccine antibody responses. PLoS One. 2017;12(2):e0171261.
  • Schellens IM, Meiring HD, Hoof I, et al. Measles virus epitope presentation by HLA: novel insights into epitope selection, dominance, and microvariation. Front Immunol. 2015;6:546.
  • Ovsyannikova IG, Johnson KL, Muddiman DC, et al. Identification and characterization of novel, naturally processed measles virus class II HLA-DRB1 peptides. J Virol. 2004;78(1):42–51.
  • Ovsyannikova IG, Johnson KL, Naylor S, et al. Naturally processed measles virus peptide eluted from class II HLA-DRB1*03 recognized by T lymphocytes from human blood. Virology. 2003;312(2):495–506.
  • Ga P, Ig O, Rb K, et al. Vaccinomics and a new paradigm for the development of preventive vaccines against viral infections. Omics. 2011;15(9):625–636.
  • Ovsyannikova IG, Johnson KL, Bergen HR III, et al. Mass spectrometry and peptide-based vaccine development. Clin Pharmacol Ther. 2007;82(6):644–652.
  • Clifford HD, Hayden CM, Khoo SK, et al. Polymorphisms in key innate immune genes and their effects on measles vaccine responses and vaccine failure in children from Mozambique. Vaccine. 2012 Sep 21;30(43):6180–6185.
  • Ovsyannikova IG, Haralambieva IH, Vierkant RA, et al. The role of polymorphisms in toll-like receptors and their associated intracellular signaling genes in measles vaccine immunity. Hum Genet. 2011;130(4):547–561.
  • Kennedy RB, Ovsyannikova IG, Haralambieva IH, et al. Multigenic control of measles vaccine immunity mediated by polymorphisms in measles receptor, innate pathway, and cytokine genes. Vaccine. 2012;30(12):2159–2167.
  • Ga P, Ig O, Rm J, et al. Heterogeneity in vaccine immune response: the role of immunogenetics and the emerging field of vaccinomics. Clin Pharmacol Ther. 2007;82(6):653–664.
  • Ovsyannikova IG, Haralambieva IH, Vierkant RA, et al. Effects of vitamin A and D receptor gene polymorphisms/haplotypes on immune responses to measles vaccine. Pharmacogenet Genomics. 2012;22(1):20–31.
  • Ovsyannikova IG, Haralambieva IH, Vierkant RA, et al. The association of CD46, SLAM, and CD209 cellular receptor gene SNPs with variations in measles vaccine-induced immune responses–a replication study and examination of novel polymorphisms. Hum Hered. 2011;72(3):206–223.
  • Haralambieva IH, Ovsyannikova IG, Umlauf BJ, et al. Genetic polymorphisms in host antiviral genes: associations with humoral and cellular immunity to measles vaccine. Vaccine. 2011;29(48):8988–8997.
  • Haralambieva IH, Ovsyannikova IG, Kennedy RB, et al. Associations between single nucleotide polymorphisms and haplotypes in cytokine and cytokine receptor genes and immunity to measles vaccination. Vaccine. 2011;29(45):7883–7895. PubMed Central PMCID: PMCPMCID: PMC3191314.
  • Yoo KH, Agarwal K, Butterfield M, et al. Assessment of humoral and cell-mediated immune response to measles-mumps-rubella vaccine viruses among patients with asthma. Allergy Asthma Proc. 2010;31(6):499–506.
  • Ga P, Ig O, Rm J. Application of pharmacogenomics to vaccines. Pharmacogenomics. 2009;10(5):837–852.
  • Dhiman N, Ovsyannikova IG, Vierkant RA, et al. Associations between SNPs in toll-like receptors and related intracellular signaling molecules and immune responses to measles vaccine: preliminary results. Vaccine. 2008;26(14):1731–1736.
  • Ga P, Ig O, Rm J. Genetics and immune response to vaccines. In: Ra K, Jm M, Avs H, editors. Genetic susceptibility to infectious diseases. New York: Oxford University Press; 2008. p. 1–447.
  • Clifford HD, Yerkovich ST, Khoo SK, et al. TLR3 and RIG-I gene variants: associations with functional effects on receptor expression and responses to measles virus and vaccine in vaccinated infants. Hum Immunol. 2012;73(6):677–685.
  • Clifford HD, Hayden CM, Khoo SK, et al. CD46 measles virus receptor polymorphisms influence receptor protein expression and primary measles vaccine responses in naive Australian children. Clin Vaccine Immunol. 2012;19(5):704–710.
  • Clifford HD, Yerkovich ST, Khoo SK, et al. Toll-like receptor 7 and 8 polymorphisms: associations with functional effects and cellular and antibody responses to measles virus and vaccine. Immunogenetics. 2012;64(3):219–228.
  • Clifford HD, Richmond P, Khoo SK, et al. SLAM and DC-SIGN measles receptor polymorphisms and their impact on antibody and cytokine responses to measles vaccine. Vaccine. 2011;29(33):5407–5413.
  • Newport MJ. The genetic regulation of infant immune responses to vaccination. Front Immunol. 2015;6:18.
  • Ovsyannikova IG, Haralambieva IH, Vierkant RA, et al. Associations between polymorphisms in the antiviral TRIM genes and measles vaccine immunity. Hum Immunol. 2013 Jun;74(6):768–774.
  • Haralambieva IH, Ovsyannikova IG, Kennedy RB, et al. Genome-wide associations of CD46 and IFI44L genetic variants with neutralizing antibody response to measles vaccine. Hum Genet. 2017;136(4):421–435.
  • Schaid DJ, Haralambieva IH, Larrabee BR, et al. Heritability of vaccine-induced measles neutralizing antibody titers. Vaccine. 2017;35(10):1390–1394.
  • Marchini J, Donnelly P, Cardon LR. Genome-wide strategies for detecting multiple loci that influence complex diseases. Nat Genet. 2005;37(4):413–417.
  • Feenstra B, Pasternak B, Geller F, et al. Common variants associated with general and MMR vaccine-related febrile seizures. Nat Genet. 2014 Dec;46(12):1274–1282.
  • Pasternak B, Feenstra B, Melbye M, et al. Improving vaccine safety through a better understanding of vaccine adverse events. Clin Infect Dis. 2015 May 15;60(10):1586–1587.
  • Hammer C, Begemann M, McLaren PJ, et al. Amino acid variation in HLA class II proteins is a major determinant of humoral response to common viruses. Am J Hum Genet. 2015 Nov 5;97(5):738–743.
  • Voigt EA, Ovsyannikova IG, Haralambieva IH, et al. Genetically defined race, but not sex, is associated with higher humoral and cellular immune responses to measles vaccination. Vaccine. 2016 Aug 30;34(41):4913–4919.
  • Moss WJ. Measles. Lancet. 2017 Dec 2;390(10111):2490–2502.
  • Hull J, Campino S, Rowlands K, et al. Identification of common genetic variation that modulates alternative splicing. PLoS Genet. 2007 Jun;3(6):e99.
  • Zhao K, Lu ZX, Park JW, et al. GLiMMPS: robust statistical model for regulatory variation of alternative splicing using RNA-seq data. Genome Biol. 2013;14(7):R74.
  • Buchholz CJ, Gerlier D, Hu A, et al. Selective expression of a subset of measles virus receptor-competent CD46 isoforms in human brain. Virology. 1996;217(1):349–355.
  • Iwata K, Seya T, Ueda S, et al. Modulation of complement regulatory function and measles virus receptor function by the serine-threonine-rich domains of membrane cofactor protein (CD46). Biochem J. 1994 Nov 15;304(Pt 1):169–175.
  • Buchholz CJ, Schneider U, Devaux P, et al. Cell entry by measles virus: long hybrid receptors uncouple binding from membrane fusion. Virology. 1996;70:3716–3723.
  • Ni Choileain S, Astier AL. CD46 processing: a means of expression. Immunobiol. 2012 Feb;217(2):169–175.
  • Ni Choileain S, Hay J, Thomas J, et al. TCR-stimulated changes in cell surface CD46 expression generate type 1 regulatory T cells. Sci Signal. 2017 Oct 24;10:502.
  • Flajnik MF, Kasahara M. Origin and evolution of the adaptive immune system: genetic events and selective pressures. Nat Rev Genet. 2010 Jan;11(1):47–59.
  • Oberg AL, McKinney BA, Schaid DJ, et al. Lessons learned in the analysis of high-dimensional data in vaccinomics. Vaccine. 2015;S0264–410X:574–575.
  • Hardy J, Singleton A. Genomewide association studies and human disease. N Engl J Med. 2009 Apr 23;360(17):1759–1768.
  • Wellcome Trust Case Control C. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 2007;447(7145):661–678.
  • Yang J, Benyamin B, McEvoy BP, et al. Common SNPs explain a large proportion of the heritability for human height. Nat Genet. 2010 Jul;42(7):565–569.
  • Willer CJ, Schmidt EM, Sengupta S, et al. Discovery and refinement of loci associated with lipid levels. Nat Genet. 2013 Nov;45(11):1274–1283.
  • Nikpay M, Goel A, Won HH, et al. A comprehensive 1,000 Genomes-based genome-wide association meta-analysis of coronary artery disease. Nat Genet. 2015 Oct;47(10):1121–1130.
  • Al Olama AA, Kote-Jarai Z, Berndt SI, et al. A meta-analysis of 87,040 individuals identifies 23 new susceptibility loci for prostate cancer. Nat Genet. 2014 Oct;46(10):1103–1109.
  • Fuchsberger C, Flannick J, Teslovich TM, et al. The genetic architecture of type 2 diabetes. Nature. 2016 Aug 4;536(7614):41–47.
  • Biological insights from. 108 schizophrenia-associated genetic loci. Nature. 2014 Jul 24;511(7510):421–427.
  • MacArthur J, Bowler E, Cerezo M, et al. The new NHGRI-EBI Catalog of published genome-wide association studies (GWAS Catalog). Nucleic Acids Res. 2017 Jan 4;45(D1):D896–D901.
  • McCarthy S, Das S, Kretzschmar W, et al. A reference panel of 64,976 haplotypes for genotype imputation. Nat Genet. 2016 Oct;48(10):1279–1283.
  • Pruim RJ, Welch RP, Sanna S, et al. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics. 2010 Sep 15;26(18):2336–2337.
  • Manolio TA. Genomewide association studies and assessment of the risk of disease. N Engl J Med. 2010 Jul 8;363(2):166–176.
  • Pe’er I, Yelensky R, Altshuler D, et al. Estimation of the multiple testing burden for genomewide association studies of nearly all common variants. Genet Epidemiol. 2008 May;32(4):381–385.
  • MacArthur DG, Manolio TA, Dimmock DP, et al. Guidelines for investigating causality of sequence variants in human disease. Nature. 2014 Apr 24;508(7497):469–476.
  • van de Bunt M, Cortes A, Brown MA, et al. Evaluating the performance of fine-mapping strategies at common variant GWAS loci. PLoS Genet. 2015;11(9):e1005535.
  • Ding K, Ij K. Methods for the selection of tagging SNPs: a comparison of tagging efficiency and performance. Eur J Hum Genet. 2007 Feb;15(2):228–236.
  • Stram DO. Tag SNP selection for association studies. Genet Epidemiol. 2004 Dec;27(4):365–374.
  • Marchini J, Howie B. Genotype imputation for genome-wide association studies. Nat Rev Genet. 2010 Jul;11(7):499–511.
  • Li Y, Willer C, Sanna S, et al. Genotype imputation [10.1146/annurev.genom.9.081307.164242 doi]. Am J Hum Genet. 2009;10:387–406.
  • Schaid DJ, Chen W, Larson NB. From genome-wide associations to candidate causal variants by statistical fine-mapping. Nat Rev Genet. 2018 Aug;19(8):491–504.
  • Sl S, Jc B. Strategies for fine-mapping complex traits. Hum Mol Genet. 2015 Oct 15;24(R1):R111–R119.
  • Rhee SY, Wood V, Dolinski K, et al. Use and misuse of the gene ontology annotations. Nat Rev Genet. 2008 Jul;9(7):509–515.
  • Harrow J, Frankish A, Gonzalez JM, et al. GENCODE: the reference human genome annotation for the ENCODE project. Genome Res. 2012 Sep;22(9):1760–1774.
  • The ENCODE. (Encyclopedia of DNA elements) Project. Science. 2004 Oct 22;306(5696):636–640.
  • Andersson R, Gebhard C, Miguel-Escalada I, et al. An atlas of active enhancers across human cell types and tissues. Nature. 2014 Mar 27;507(7493):455–461.
  • Kundaje A, Meuleman W, Ernst J, et al. Integrative analysis of 111 reference human epigenome. Nature. 2015 Feb 19;518(7539):317–330.
  • Maurano MT, Humbert R, Rynes E, et al. Systematic localization of common disease-associated variation in regulatory DNA. Science. 2012 Sep 7;337(6099):1190–1195.
  • Millstein J, Zhang B, Zhu J, et al. Disentangling molecular relationships with a causal inference test. BMC Genet. 2009 May;27(10):23.
  • Zhu Z, Zhang F, Hu H, et al. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat Genet. 2016 May;48(5):481–487.
  • Giambartolomei C, Vukcevic D, Schadt EE, et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 2014 May;10(5):e1004383.
  • Hormozdiari F, van de Bunt M, Segre AV, et al. Colocalization of GWAS and eQTL signals detects target genes. Am J Hum Genet. 2016 Dec 1;99(6):1245–1260.
  • Zilliox MJ, Moss WJ, Griffin DE. Gene expression changes in peripheral blood mononuclear cells during measles virus infection. Clin Vaccine Immunol. 2007;14(7):918–923.
  • Dhiman N, Ovsyannikova IG, Oberg AL, et al. Immune activation at effector and gene expression levels after measles vaccination in healthy individuals: a pilot study. Hum Immunol. 2005;66:1125–1136.
  • Haralambieva IH, Zimmermann MT, Ovsyannikova IG, et al. Whole transcriptome profiling identifies CD93 and other plasma cell survival factor genes associated with measles-specific antibody response after vaccination. PLos ONE. 2016;11(8):e0160970.
  • Haralambieva IH, Kennedy RB, Simon WL, et al. Differential miRNA expression in B cells is associated with inter-individual differences in humoral immune response to measles vaccination. PLos ONE. 2018;13(1):e0191812.
  • Querec TD, Akondy RS, Lee EK, et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat Immunol. 2009;10(1):116–125.
  • Nakaya HI, Wrammert J, Lee EK, et al. Systems biology of seasonal influenza vaccination in humans. Nat Immunol. 2011;12(8):786–795.
  • Kazmin D, Nakaya HI, Lee EK, et al. Systems analysis of protective immune responses to RTS,S malaria vaccination in humans. Proc Natl Acad Sci USA. 2017 Feb 28;114(9):2425–2430.
  • Papalexi E, Satija R. Single-cell RNA sequencing to explore immune cell heterogeneity. Nat Rev Immunol. 2018 Jan;18(1):35–45.
  • Friedensohn S, Khan TA, Reddy ST. Advanced methodologies in high-throughput sequencing of immune repertoires. Trends Biotechnol. 2017 Mar;35(3):203–214.
  • Hu Y, An Q, Sheu K, et al. Single cell multi-omics technology: methodology and application. Front Cell Dev Biol. 2018;6:28.
  • Stoeckius M, Hafemeister C, Stephenson W, et al. Simultaneous epitope and transcriptome measurement in single cells. Nat Methods. 2017 Sep;14(9):865–868.
  • Peterson VM, Zhang KX, Kumar N, et al. Multiplexed quantification of proteins and transcripts in single cells. Nat Biotechnol. 2017 Oct;35(10):936–939.
  • Hou Y, Guo H, Cao C, et al. Single-cell triple omics sequencing reveals genetic, epigenetic, and transcriptomic heterogeneity in hepatocellular carcinomas. Cell Res. 2016 Mar;26(3):304–319.
  • Hu Y, Huang K, An Q, et al. Simultaneous profiling of transcriptome and DNA methylome from a single cell. Genome Biol. 2016 5;May(17):88.
  • Frantz PN, Teeravechyan S, Tangy F. Measles-derived vaccines to prevent emerging viral diseases. Microbes Infect. 2018 Oct - Nov;20(9–10):493–500.
  • Griffin DE. Current progress in pulmonary delivery of measles vaccine. Exp Rev Vaccines. 2014 Jun;13(6):751–759.
  • Griffin DE, Pan CH. Measles: old vaccines, new vaccines. Curr Topics Microbiol Immunol. 2009;330:191–212.
  • de Swart RL, de Vries RD, Rennick LJ, et al. Needle-free delivery of measles virus vaccine to the lower respiratory tract of non-human primates elicits optimal immunity and protection. NPJ Vaccines. 2017;2:22.
  • de Vries RD, Stittelaar KJ, Osterhaus AD, et al. Measles vaccination: new strategies and formulations. Expert Rev Vaccines. 2008;7(8):1215–1223.
  • Joyce JC, Carroll TD, Collins ML, et al. A microneedle patch for measles and rubella vaccination is immunogenic and protective in infant rhesus macaques. J Infect Dis. 2018 Jun 5;218(1):124–132.
  • Edens C, Collins ML, Goodson JL, et al. A microneedle patch containing measles vaccine is immunogenic in non-human primates. Vaccine. 2015 Mar 12;S0264–410X(15):285–286.
  • Putz MM, Muller CP. The rationale of a peptide-conjugate vaccine against measles. Vaccine. 2003 Jan 30;21(7–8):663–666.
  • Gala RP, Popescu C, Knipp GT, et al. Physicochemical and preclinical evaluation of a novel buccal measles vaccine. AAPS Pharm Sci Tech. 2017 Feb;18(2):283–292.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.