Skip to main content
Clinical Microbiology Reviews logoLink to Clinical Microbiology Reviews
. 2019 Dec 11;33(1):e00130-18. doi: 10.1128/CMR.00130-18

Performance of Zika Assays in the Context of Toxoplasma gondii, Parvovirus B19, Rubella Virus, and Cytomegalovirus (TORCH) Diagnostic Assays

Bettie Voordouw a,b,, Barry Rockx a, Thomas Jaenisch c, Pieter Fraaij a, Philippe Mayaud d, Ann Vossen e, Marion Koopmans a,
PMCID: PMC6927310  PMID: 31826871

Infections during pregnancy that may cause congenital abnormalities have been recognized for decades, but their diagnosis is challenging. This was again illustrated with the emergence of Zika virus (ZIKV), highlighting the inherent difficulties in estimating the extent of pre- and postnatal ZIKV complications because of the difficulties in establishing definitive diagnoses.

KEYWORDS: TORCH, Zika virus, congenital infections, diagnostics, fetal infection, maternal infection, molecular methods, serology, virology

SUMMARY

Infections during pregnancy that may cause congenital abnormalities have been recognized for decades, but their diagnosis is challenging. This was again illustrated with the emergence of Zika virus (ZIKV), highlighting the inherent difficulties in estimating the extent of pre- and postnatal ZIKV complications because of the difficulties in establishing definitive diagnoses. We reviewed the epidemiology, infection kinetics, and diagnostic methods used for Toxoplasma gondii, parvovirus B19, rubella virus, and cytomegalovirus (TORCH) infections and compared the results with current knowledge of ZIKV diagnostic assays to provide a basis for the inclusion of ZIKV in the TORCH complex evaluations. Similarities between TORCH pathogens and ZIKV support inclusion of ZIKV as an emerging TORCH infection. Our review evaluates the diagnostic performance of various TORCH diagnostic assays for maternal screening, fetal screening, and neonatal screening. We show that the sensitivity, specificity, and positive and negative predictive value of TORCH complex pathogens are widely variable, stressing the importance of confirmatory testing and the need for novel techniques for earlier and accurate diagnosis of maternal and congenital infections. In this context it is also important to acknowledge different needs and access to care for different geographic and resource settings.

INTRODUCTION

The present Zika virus (ZIKV) epidemic was first noted through an alert from the Brazilian health authorities notifying the World Health Organization (WHO) of an illness characterized by skin rash in the northeastern states and subsequently signaling an almost 20-fold increase in the incidence of microcephaly in newborns coinciding with rapid spread of ZIKV after incursion into the continent (14). Until then, ZIKV infection was generally assumed to be associated with mild and transient disease, estimated to be asymptomatic in approximately 80% of the cases (5). Hence, it is likely that the infection is underdiagnosed or underreported in a setting where disease is endemic (6).

The association of ZIKV infection with congenital neurological disease has since then been the subject of numerous publications. First, establishment of a causal association with neuropathological processes came from a study showing widespread ZIKV infection in the brain of a fetus from a pregnancy that had been terminated due to severe fetal malformation (7). Further evidence for the association came from larger case series, retrospective analysis of notification data from regions with prior outbreaks (8), and replication of the syndrome in animal models (913). Although there is general agreement on such an association, many uncertainties remain with regard to the actual risk of fetal infection during pregnancy (14). A systematic review estimated the prevalence of microcephaly at 2.3% (95% confidence interval [CI], 1.0 to 5.3%) (15) though estimates range widely. Little is known about the risk of complications in relation to timing of maternal infection (first trimester versus later exposures), prior (flavivirus) exposure, or the rates of transplacental transmission, fetal infection, and congenital disease once infection is present (Fig. 1). Two recent reports suggest a decreasing risk over the course of pregnancy (16, 17). A recent European study showed an overall congenital anomaly prevalence of any (nongenetic) cause of 1.5/100 total births, but for microcephaly interpretation was hampered by differences in diagnostic criteria (18).

FIG 1.

FIG 1

Seroprevalence, maternal infection risk, fetal transmission risk, and congenital infection risk of the following selected infections: TOXO, PB19, rubella, CMV, and ZIKV. Seroprevalence at childbearing age is shown. IR/year, maternal annual infection rate; FTR, fetal transmission rate; CIR, congenital infection rate, determined as the number of congenital infections per 1,000 live births (TOXO) or per 1,000 pregnancies (RV, CMV, PB19, and ZIKV).

At present, ZIKV diagnostic algorithms are based on the use of reverse transcriptase PCR (RT-PCR) for virus detection and/or serological determination of pathogen-specific IgM and IgG antibodies, supplemented with virus neutralization assays if available. All of the assays have benefits but also known limitations, challenging interpretation at different stages of pregnancy, particularly in relation to the wide diversity of flavivirus background exposures in the regions where ZIKV circulates. Virus genome detection by RT-PCR is considered confirmatory but has a very short detection window as ZIKV viremia is of short duration. Virus may persist for longer periods in other body fluids, with reported persistence up to 120 days for semen (19). In addition, pregnant women may experience prolonged viremia (20, 21), with reported (transient) presence of ZIKV in fetal blood and amniotic fluid (21). Antibody-based testing is severely hampered by cross-reactivity with antibodies from prior flavivirus exposures. Other infections during pregnancy are associated with congenital and subsequent neonatal disease, sometimes referred to as TORCH infections (Toxoplasma gondii [TOXO], other [e.g., varicella zoster and parvovirus B19 (PB19)], rubella virus [RV], cytomegalovirus [CMV], and herpes simplex virus [HSV] with or without syphilis) (22). Diagnosis of fetal infection and linking fetal infections to clinical outcomes require knowledge of infection kinetics, including timing and differentiation of primary from nonprimary infection (i.e., reactivation or reinfection), of maternal and fetal immune responses in relation to pathology, and of the availability of biomarkers predictive of vertical transmission and presence and/or severity of fetal abnormalities (2325). For instance, TOXO and CMV cause persistent or latent infections, whereas RV, PB19, and ZIKV infection are thought to be primarily self-limiting. Immunocompetent pregnant women with previous infection with TOXO are considered not at risk for congenital abnormalities, whereas for CMV primary as well as subsequent infections are associated with congenital infection and abnormalities, albeit with a lower attributable risk. The recent ZIKV epidemic and its possible association with microcephaly have initiated the discussion to include ZIKV as a novel TORCH pathogen (26).

Although the described maternal infections are important causes of fetal and neonatal morbidity and mortality on a global scale, the overall contribution to fetal and congenital diseases is limited (2729) due to cumulative low and/or limited a priori risks of maternal infection, vertical transmission, and subsequent congenital infection (Fig. 1).

Generally, the risk of congenital disease following maternal infection is linked to primary infection and timing in pregnancy infection, which is pathogen dependent (25, 3037). Nonprimary maternal infections may result in fetal transmission, but only in the case of CMV does this contribute significantly to congenital disease (38).

The low attributive risk of TORCH and ZIKV infections to congenital disease has consequences for diagnostic accuracy and the ability to provide information relevant for clinical decision-making. This is further complicated by the high proportion of asymptomatic maternal infections (6, 35, 3941), challenging timely detection of fetal infection and early neonatal congenital disease, which may remain asymptomatic for years (40, 42). Early diagnosis of fetal disease risk in pregnancy, however, is important particularly when early therapeutic management is available, e.g., for TOXO infection (43). Consequently, diagnostic algorithms should reliably detect maternal infection in a timely fashion, determine (risk of) vertical transmission, and establish or exclude congenital infection. This review assesses diagnostic methods presently used for TORCH infections, their correlation with congenital and/or neonatal disease, and their predictive value in prenatal screening; it will document gaps in methods used; and it will draw implications for diagnostic algorithm development in novel or (re)emerging infections such as Zika virus.

EPIDEMIOLOGY, INFECTION, AND TRANSMISSION RISKS OF SELECTED TORCH PATHOGENS

The risk of infection during pregnancy varies by pathogen and depends upon geographic region, prevalence in the population, and preventive (vaccination) practices.

The seroprevalence of Toxoplasma gondii among women of childbearing age shows a broad range, from <2% in a large Chinese cohort (44) up to 75% in Brazil (45), with a mean estimate of around 40% (4648). Similar, broad ranges in seroprevalence between 30 and 72% are reported for PB19 and CMV (4956) although for CMV seroprevalences up to 100% are also reported (57). RV seroprevalence estimates depend on (differences in) vaccination practices (5860). ZIKV seroprevalence has a geographic distribution, varying from <2% in travelers returning from areas of endemicity or blood donors in settings of nonendemicity (6163) to up to 39% in healthy individuals living in areas of endemicity (64, 65) and increasing to >60% following outbreaks (66, 67). These wide ranges in background seroprevalence affect the likelihood of primary infection during pregnancy, as well as the interpretation of diagnostic assays, and need to be taken into account in the development of diagnostic algorithms.

To assess the clinical impact of exposure to TORCH pathogens and ZIKV during pregnancy, it is important to consider maternal infection risk (MIR), fetal transmission risk (FTR), and congenital infection risk (CIR) for each pathogen (Fig. 1). MIR estimates, defined as the annual infection rate for selected TORCH infections, range from 0.1 to 0.6% for TOXO (31) to 2 to 7% for PB19 and CMV (6873), with an epidemic rise up to 10% in PB19. Reliable data for RV MIR is lacking in an elimination setting, but the annual incidence is estimated at 1.3/100,000 pregnancies in the general population (74). A 6.4% IgG seroconversion rate was reported in women with nonimmune RV titers prior to pregnancy (75). Although efforts have been made to calculate the MIR for ZIKV, reliable data are still lacking due to several factors, including diagnostic limitations (e.g., cross-reactivity) (76, 77) and differences and rapid changes in epidemiology.

Fetal transmission risk (FTR), defined as the proportion of transplacental transmission following (primary) infection during pregnancy, is also pathogen dependent and is linked to the timing of infection during pregnancy. The FTR may increase (TOXO, PB19, and CMV) or decrease (RV) during the pregnancy period, with a variable mean FTR estimated to range between 24 and 80% for these pathogens (25, 33, 42, 78, 79). For ZIKV, the FTR is thought to be highest during the first trimester, but more data are needed (16, 17). Perinatal transmission has also been reported for ZIKV (80).

Despite a high FTR, the congenital infection risk (CIR), defined as the number of congenital infections per 1,000 live births (or number of fetal deaths/hydrops fetalis per 1,000 infected fetuses in PB19), is low, ranging from <1/100,000 pregnancies (RV) up to 0.1/1,000 to 20/1,000 pregnancies for TOXO, PB19, and CMV infection (33, 34, 8189). Overall congenital CMV infection is most prevalent in the developed world (5/1,000 to 20/1,000 live births) (30, 90), followed by infection with TOXO (0.1/1,000 to 5/1,000 live births) (31, 46), and RV (annual incidence of 0.4/100,000) (81). Parvovirus B19 infection is associated with hydrops fetalis (8284) and fetal death, with an estimated annual incidence of <4/1,000 fetuses (85). Recent studies estimated the contribution of symptomatic ZIKV during pregnancy to ZIKV-associated congenital disease at 7% (17) and evidence of acute infection in pregnancy at less than 4% (77). How this translates to the overall contribution of ZIKV to, e.g., congenital microcephaly prevalence depends on the baseline risks, and these are uncertain (18, 91). A retrospective analysis in French Polynesia estimated a risk of microcephaly associated with ZIKV infection at 9.5/1,000 pregnancies, with an overall risk of microcephaly of 0.2/1,000 neonates (8).

The CIR does not necessarily follow the FTR, with the highest CIR in the first trimester for TOXO, RV, and CMV (25, 34, 35, 42) and the highest CIR in the second trimester for PB19 (36, 37). In addition, for ZIKV, the highest risk is reported in the first trimester (8, 17). Overall, CIR is limited to primary infection, except for CMV, where reinfection or reactivation contributes mainly to congenital CMV disease burden (56, 92). Latent or chronic TOXO infection does not exclude transmission but does not result in CIR in immunocompetent pregnant women (93, 94). In general, the low attributive risk of the reported infections to overall prevalence of congenital disease (2729) impacts the performance of diagnostic assays.

This implies that the low a priori attributive risk of TORCH and ZIKV to congenital infections needs to be included in every step excluding or confirming maternal, fetal, and/or congenital infection.

MATERNAL DIAGNOSTIC TESTING

Infection Kinetics

Interpretation of diagnostic testing during pregnancy requires knowledge on infection kinetics. These are defined by many factors, including the presence and duration of symptoms, duration of pathogen presence in different body fluids, loads, timing of development of specific antibodies, background antibody prevalence, and relationship between these parameters. Molecular detection in early symptomatic infection is generally considered proof of acute primary infection (17, 40, 59, 95) although this is not true for each pathogen, i.e., CMV. Primary infections typically show IgM and IgG development, determined with serological assays with or without confirmatory testing (96, 97). Reported antibody kinetics differ between selected pathogens. TOXO IgM seroconversion occurs relatively late, between 15 and 30 days (98, 99), while an early IgM rise is observed for PB19 infection, i.e., toward the end of the first week of infection and coinciding with peak viremia (56), and for infection with RV, whose IgM rises within 5 days after rash onset (100). CMV IgM may become detectable between 0 and 3 weeks (101), with peak IgM observed between 1 and 3 months (102). IgM antibodies against ZIKV show an early rise and can be first detected within the first week after clinical symptoms, while IgG antibodies can be detected within the first 2 weeks (103). PB19 IgM can persist up to 3 months following infection (104). ZIKV IgM can persist beyond 3 months (105). Long-term persistence of rubella IgM is reported following vaccination (106108) due to the natural occurrence of nonspecific IgM (109, 110) and despite attempts to improve assays (111114). Differentiation of acute infection from latent infection or reactivation/reinfection is important in TOXO and CMV infections as IgM/IgG may coincide, thereby making it difficult to diagnose primary infection if the first consultation yields an IgM/IgG-positive test result (115). In this case, confirmatory testing is needed, e.g., by avidity index (AI) or immunoblotting (IB), using presently available assays (47, 116, 117). Development of CMV-specific IgG with a negative sample collected earlier in pregnancy is considered proof of primary CMV infection; however, in the absence of routine screening this is usually not feasible (118).

Molecular Assay Performance and Limitations

Although PCR assay specificity is high in acute primary infection, the window of PCR positivity may be short, as shown for PB19 (95) (Fig. 2). Limited data from ZIKV showed a similar pattern. A recent external quality assessment (EQA) suggested similar more robust specificity but variable sensitivity between participating laboratories (119). Furthermore, most acute (primary) infections in pregnancy are asymptomatic, and the day of infection is unknown, precluding use of this gold standard test. In acute primary PB19 infection in pregnancy, high viral load is associated with an early positive IgM test (39, 56, 120, 121). Long-term, low-load DNA persistence is observed following PB19 infection (104, 122). In one study, the use of endonuclease treatment before molecular testing differentiated naked DNA persistence from true viremia (123). For ZIKV, rapid degradation of RNA was reported in urine samples (124). In acute maternal PB19 infection, the positive predictive value (PPV) of PB19 PCR is high, but at the time of fetal symptoms, the PPV of PB19 DNA detection in maternal blood is generally low as clinical symptoms in the fetus are usually observed when maternal viremia has ceased. This temporal relation has not been established for other primary infections such as TOXO (125, 126), or CMV (127). Viremia in pregnant women is associated with vertical transmission risk and increased CIR, but the relationship between maternal infection, FTR, and CIR is different for the selected pathogens (128134). Currently, there is no obvious predictor for transmission risk. For instance, viral load does not differentiate transmitters from nontransmitters in CMV infection (129, 130, 135). Absence of a relationship with maternal disease severity or viral load was also recently described for congenital ZIKV infection (136). Low-viral-load positives may occasionally not show IgM seroconversion (RV and ZIKV) (137139). Low assay sensitivity was suggested as one of the possible explanations (139). Genotype differences for individual pathogens may impact the sensitivity of assays (140142), which is important in considering the use of assays in different regions.

FIG 2.

FIG 2

Routine maternal diagnostic methods: sensitivity and specificity, positive predictive value, and negative predictive value median point estimate ± 95% CI. For IgM and IgG data, plus and minus signs indicate allocation of “gray zone” results to the seropositive or seronegative results, respectively. NA, not available. References for the data for each virus are as follows: TOXO (n = 28), 48, 99, 113, 134, 143, 150, 153, 160163, 165, 170, 171, 175, 177, 179, 183, 189, 195, 199201, 204, 205, 209, 262, and 302; PB19 (n = 14), 29, 36, 53, 79, 84, 86, 121, 146, 176, 197, 211, 224, 303, and 304; RV (n = 9), 59, 106, 108, 137, 149, 151, 186, 204, and 303; CMV (n = 24), 23, 53, 92, 101, 112, 128, 145, 148, 155, 156, 159, 166, 169, 173, 178, 190, 196, 204, 206, 250, 265, 281, 303, and 305; ZIKV (n = 15), 184, 289, and 306318.

Serological Assays and Performance

Primary diagnostic assays.

Generally, TORCH immunoassays report relatively high specificity for IgM-positive and IgG-positive samples or for IgG in IgM-negative samples (34, 108, 113, 143), but as the a priori likelihood of a maternal infection with TORCH pathogens is generally low, even a relatively low false positivity rate translates to a low PPV for all pathogens (including ZIKV) except PB19, stressing the need for confirmatory testing (144147) (Fig. 2). Comparative studies of assays reporting relative performance data tend to overestimate sensitivity and specificity (108, 113, 143, 148). In view of the above, a positive IgM test result always requires confirmation with other assays (36, 101, 113, 149) and follow-up samples. More specific (recombinant) peptide-specific IgM assays may provide solutions, but their performance also needs to be fully evaluated (121, 150).

Confirmatory testing.

The use of confirmatory testing with avidity index measurements (AI), immunoblotting (IB), and virus neutralization testing (VNT) is not consistent between pathogens and also shows variable performance levels (Fig. 2). Testing for AI is common practice for TOXO and CMV diagnostics but not for PB19 (121) or RV (151). The rationale for avidity testing is that the avidity of antibodies increases with time, and a high AI correlates with infection in the more distant past (152158). The sensitivity of the AI as a confirmatory test depends upon the initial screening platform used, as was shown in one study. A negative initial IgM screening is unlikely to be confirmed (145). IgM positivity combined with a low AI increases the sensitivity and PPV of the combined assays in diagnosing recent infection (159162). In contrast, a low AI with positive IgG has a relatively high negative predictive value (NPV) (163, 164). A high AI plus positive IgG usually confirms past infection; however, for TOXO infection, AI maturation may never occur (165). A rapid increase in the AI in CMV infection was associated with the false exclusion of recent infection (166), with a higher FTR (167) and CIR (168). Therefore, exclusion of acute infection based on (high) AI requires a predefined time window (101, 114, 169171), and the size of the window varies depending on pathogen, thresholds, and platforms (172). Different antigens, including recombinant antigens as targets for antibody responses, may improve AI assay performance (170).

Antibody test results may also be confirmed by IB (101, 173) or different complementary assays (174). Epitope-specific IgG IB used in TOXO and PB19 infection could confirm IgM/IgG measurements (175, 176). This is particularly relevant in equivocal outcomes (177) or in (false) negative results with high viral load (146) and may help timing of the infection by correlating IB with virus neutralization in CMV infection (178) or with the AI in TOXO infection (179).

An interesting application is the use of IB in TOXO infection for discrimination of maternal and neonatal antibody responses by comparing patterns of antibody binding to different proteins or peptides in the blood from mother and neonate (180). Limitations of IB include lack of standardization with variable concordance between assays, particularly in acute infection (181, 182), and different diagnostic accuracies of band patterns in the blots (175). The predictive value of IB depends upon the target, and IgM blots often have poorer predictive value than IgG blots (173, 183). Virus neutralization data are primarily available for CMV (96) but are also commonly used in confirmation of ZIKV infection (184), with generally high assay performance.

Limitations of Serodiagnostic Assays

Major limitations for all of the diagnostic methods described include interassay variability (98, 185), use of different cutoffs, differences in classifications of positives (185, 186), low agreement between AI index assays (187), and variability between platforms (114, 165, 171, 172, 188191). Other serious concerns are the so-called “gray zone” classification, i.e., the area between the negative outcome and the positive outcome of a test (98, 114, 153, 163, 164, 166, 186, 187, 192, 193), which differs considerably between assays (194), and the lack of standardization of cutoff values for the same assay (183). How gray-zone results are interpreted has a profound impact on the sensitivity or specificity of a given assay. This is indicated in Fig. 2, where, for example, the category Tox+ denotes the assignment of the gray zone to the seropositive group and Tox– indicates assignment to the seronegative group (148, 195, 196). This variability stresses the need for common standards for assay development and validation (197201), use of standard curves (202), and/or (international) standardization, as shown for RV (186, 203). Even when general standards for defining seropositivity are applied, different assays show different performance characteristics which are impacted by the assignment of equivocal results (i.e., gray zone) to the positive or negative outcome (108, 204, 205). Since this mainly affects sensitivity, it increases the NPV, particularly when prevalence decreases (92, 168, 206208).

FETAL INFECTION DIAGNOSTIC TESTING

In case of suspected fetal infection, molecular detection of virus DNA or RNA in amniotic fluid (AF) or cord blood (CB) is the primary diagnostic option in most cases except in TOXO infection. Limited serological data on AF include IgM determination (209, 210). IgG determination is not informative as IgG is usually of maternal origin. IgM and/or IgA determination in AF or fetal blood (FB) has low diagnostic value (211215), whereas cell culture isolation (virological confirmation) is more specific, for example, in CMV infection (215). Loads in FB or AF may be 100-fold to 1,000-fold higher than those in maternal blood (129, 157, 216), particularly in symptomatic fetuses (217219), as shown in PB19, CMV, and TOXO infection. Although (viral) load in primary infection may be high in the fetus (211), its presence is not necessarily associated with symptomatic infection (220223). These discrepancies possibly reflect different windows of infection detection. But overall, PCR on AF or FB has good specificity and NPV in the fetus (Fig. 3) (210, 224229). In the presence of maternal viremia, sensitivity has been shown to increase for PB19 (216) and for TOXO with a shorter interval to AF or FB sampling (224, 226) or use of multicopy genes (230). Assay performance may be different between AF and FB, with a reported concordance between 73% and 99% (210, 213). Although (transient) ZIKV was reported in AF and FB in fetuses of women with proven infection during pregnancy (21), there are no quantitative data on FB/AF in ZIKV available at this point (231).

FIG 3.

FIG 3

Routine fetal diagnostics methods: sensitivity and specificity, positive predictive value, and negative predictive value median point estimate ± 95% CI. References for the data for each virus are as follows: TOXO (n = 13), 33, 43, 126, 154, 164, 209, 212, 226228, 230, 319, and 320; PB19 (n = 5), 79, 213, 220, 222, and 223; RV (n = 1), 320; CMV (n = 10), 89, 129, 169, 210, 222224, 229, 277, and 320.

POSTPARTUM DIAGNOSTIC TESTING

Postpartum sequelae of fetal infections have been observed for TOXO, RV, CMV, and ZIKV. Although the literature is not consistent on this issue, fetal anemia following PB19 infection may result in severe postpartum sequelae (232). In general, timely postpartum diagnosis is hampered by the low sensitivity of IgM testing (33, 36, 40, 205, 228, 233235), the presence of maternal antibodies, and the high proportion of asymptomatic CMV-infected or TOXO-infected newborns (31, 128, 224, 228, 236). Consequently, ascertainment of congenital disease typically requires longer-term follow-up, posing challenges to the differentiation from postpartum infection (40, 237, 238).

IgM positivity in cord blood or peripheral blood in newborns at <24 h confirms prenatal infection with RV and CMV when supported by viral load testing (233, 239). IgM assay performance is better when testing is done more selectively in symptomatic neonates, as reported for TOXO (218). Contamination with maternal blood should be excluded within the first 10 days postpartum if the first sample was taken from CB, as reported for TOXO (35).

The majority of IgG detected at birth will be of maternal origin (31, 240) but may be neonatal (241). Generally, maternal IgG is assumed to persist for less than 6 months (242, 243). Therefore, in TOXO infection, persistence of the IgG levels in the infant at 12 months is used to confirm or exclude congenital toxoplasmosis. Comparison of IgG immunoblot patterns in mother and child is used to reduce the period of the uncertainty about IgG origin (205, 244). The use of immunoblotting or other multiantigen assays early postpartum (245) has thus been shown to provide an opportunity to differentiate congenital from noncongenital infection by comparing maternal and neonatal antibody binding patterns (246, 247).

The feasibility of using differences in the AI for this purpose has also been studied in different settings, e.g., in infection with RV, TOXO, and CMV (245, 248251). In RV infection, slow IgG AI maturation in neonates, in combination with presence of IgM, correlated with congenital infection (251, 252). In TOXO infection, identical neonatal and maternal AI excluded congenital toxoplasmosis (35).

Molecular testing of neonatal blood or urine has generally good specificity (239, 252) (Fig. 4) with higher viral load and longer RNA/DNA persistence in symptomatic babies, particularly in urine or throat samples for selected pathogens within a selected period after birth (239, 253255) (Fig. 4). Viral load has been used to differentiate congenital from postpartum infection when early samples are available, but it is not clear if these findings can be generalized (256).

FIG 4.

FIG 4

Routine neonatal screening methods: sensitivity and specificity, positive predictive value, and negative predictive value median point estimate ± 95% CI. References for the data for each virus are as follows: TOXO (n = 9), 174, 201, 212, 228, 234, 245, 247, 249, and 321; PB19 (n = 2), 36 and 85; RV (n = 2), 233 and 251; CMV (n = 11), 102, 115, 128, 158, 159, 215, 238, 250, 253, 277, and 322.

EFFORTS AT IMPROVING DIAGNOSTIC ACCURACY

There have been many efforts to improve diagnostic accuracy; however, this has not yet resulted in significant improvements. These efforts include development of recombinant (multi-)proteins and peptide-specific tests using different techniques (e.g., immunoproteomics) (257) in (multiplex) assays to improve sensitivity and specificity (143, 258260), distinction between primary and postprimary infection (261263), timing (264), and differentiation of transmitters from nontransmitters (265). For example, recombinant proteins in novel avidity assays reported a PPV of >85% (266) and were better suited for IgG detection in TOXO infection (267) or could serve as proxies for functional antibody measurements, for example, virus neutralization in RV infection (268). Multiplex assays are used for simultaneous detection of different antibodies in TORCH infections (34, 108, 113, 143), which is important for differential diagnostic approaches. However, assays still have the performance limitations of the standard assays described. Microarray-based assays have been developed to improve simultaneous testing of antibodies of different pathogens, including (extended) TORCH (269). Use of dry blood spots in multiplex serological assays allows the use of small volumes and may result in shorter diagnostic delays (270272). Additional use of novel platforms, such as plasmonic gold chip multiplex immunoassay platforms in TOXO infection, may yield potentially better assay performance (273275). Cell-mediated immunity (CMI) assay data (gamma interferon release assay [IGRA] or enzyme-linked immunosorbent spot [ELISpot] assay) particularly come from CMV, but these assays are not routinely used. A higher CD4+/CD8 proliferative T-cell response was associated with primary infection (276280), improving assay sensitivity for low IgG avidity (281), but here also variability in assay performance was reported in different settings, e.g., in primary infection and transmitters (282). Since CMI in the neonate is never of maternal origin, it is hypothesized that it might aid in differentiating maternal from fetal ZIKV infection. Rapid point-of-care testing, such as immunochromatography (98), loop-mediated isothermal amplification (LAMP) (141, 283), or digital microfluidic (DMF) diagnostic platforms (284), studied for different pathogens, may further reduce the time to first positive test and increase sensitivity and/or decentralized availability of assays in resource-limited settings. Such developments are also reported for ZIKV (285288). Alternative, novel methods in AF samples include comparisons of metabolic profiles (metabolomics) of transmitter versus nontransmitter infections (289, 290), cytokine profiles (291), or peptidome prognostic classifiers (229) to differentiate infected from noninfected fetuses or to distinguish symptomatic from asymptomatic infections postpartum. Such developments are particularly important as they may provide early (prenatal) information on the risk of overt clinical congenital disease postpartum. Other nonpathogen-related methods are those comparing differential gene or protein expression levels between fetal cells and maternal cells (292). In analogy with previously developed tests for noninfectious prenatal screening (293, 294), genes associated with neurodevelopment were studied as biomarkers in cell-free RNA transcripts in AF samples (295). Paper-based, cell-free detection of RNA was recently evaluated for rapid point-of-care testing of ZIKV (296).

CONCLUSIONS

Our review of approaches to diagnose acute maternal infection, determine vertical transmission risk, and establish presence or absence of congenital infection has shown similarities but also large variations in approaches between pathogens, risking underexploration of methods for optimal diagnostic algorithms. Present diagnosis of TORCH and ZIKV infections is primarily based on serological testing with a focus on IgM and/or IgG detection, for which a variety of commercial assays is available. These assays show variable levels of performance and may not differentiate between primary and nonprimary infections (115) or persistence and/or may be limited by cross-reactivity (108, 112). A positive serological test thus always requires confirmatory testing, including IgG avidity index determinations, immunoblotting, virus neutralization, and molecular testing (43, 212, 224).

The use of different assays and the lack of (international) standardization hamper the interpretation of, and agreement between, different studies (297), despite availability of (WHO) recommended antigens, primers, and probes (298). Efforts to improve detection of primary infection and determination of its time of occurrence during pregnancy have not yet resulted in reliable biomarkers for fetal or congenital disease risk (299). Even if protocols and/or algorithms are in place, variabilities between assays interfere with unambiguous and timely decision-making (300). Predefined evidence-based approaches with standardized diagnostic assays and algorithms are clearly needed to improve adequate and timely diagnosis of (primary) maternal infections and subsequent postpartum congenital disease (297). This is of particular relevance in settings of low endemicity where suboptimal diagnostic performance may increase the risk of false-positive outcomes. Lessons to draw from this review for novel challenges such as ZIKV are to directly combine methods (52), increase epitope specificity (e.g., avidity, immunoblotting, and virus neutralization), and implement paired mother-fetus and/or mother-child testing, as was recently reported for ZIKV neutralizing antibodies (301). Differences in background exposure to ZIKV and other flaviviruses will have an (age-dependent) effect on cross-reactivity and on interpretation of protein-driven assays, such as IB or microarray analysis. In these instances, CMI might be explored as an alternative method to differentiate maternal from congenital ZIKV infection. Standardization of (validated) reference methods is critical in order to compare different methods and might need reference centers to confirm acute infection. As discussed in this review, diagnostics for the TORCH infections, including ZIKV infection, remain complex although techniques continue to develop. Systematic explorations of the published methods across pathogens is important to improve diagnostic algorithms. In the meantime, it is essential to raise awareness among medical microbiologists and treating physicians about the limitations of the presently applied tests and algorithms and the need to improve protocols for diagnostic testing and clinical decision-making. Given the observed disconnect between the different pathogen specialist fields, we conclude that there is a clear case to be made for an integrated TORCH-ZIKV diagnostic approach.

ACKNOWLEDGMENTS

Special thanks to the ZIKAlliance consortium for discussions and for their expertise.

Biographies

graphic file with name CMR.00130-18-f0005.gif

Bettie Voordouw, M.D., Ph.D., M.P.H., was trained at Leiden University, the Netherlands. She has worked since 2016 as a medical microbiologist with a focus on clinical virology, vaccinology, and public health microbiology at the National Institute of Public Health and Environment, Bilthoven. There, she is also a project leader for reference laboratories. Furthermore, since 2017 she has been a senior scientist at the Department of Viroscience, Erasmus Medical Centre Rotterdam, with particular interest in emerging (viral) infections and optimizing diagnostic algorithms. She has previous long-term experience in the (international) regulatory field, public health agencies, academia, and scientific committees, leading assessment teams and being responsible for and/or participating in scientific advisory bodies and for clinical, treatment, and vaccine guidelines. Since 2017 she has been the Dutch alternate National Microbiology Focal Point to the European Center for Disease Prevention and Control (ECDC). Since 2019 she has been a member of the board of the National Working Group of Clinical Virology of the Dutch Society of Medical Microbiology.

graphic file with name CMR.00130-18-f0006.gif

Barry Rockx, Ph.D., is Head of the Exotic Viruses working group in the Department of Viroscience, Erasmus Medical Centre, Rotterdam. His main research lines involve studies on the tropism, pathogenesis, and host responses of emerging zoonotic viruses causing hemorrhagic, respiratory, and neurological diseases, including orthohantaviruses and arboviruses in a variety of in vitro and in vivo models. He has authored over 50 peer-reviewed scientific publications and several book chapters and has supervised several postdoctoral fellows and students. He has coordinated several NIH-funded projects and contracts and is currently task leader of projects funded by Horizon 2020 of the European Union and by the Netherlands Organization for Health Research and Development (ZonMw). Fields of expertise include tropism, pathogenesis, and the host responses following emerging virus infection.

graphic file with name CMR.00130-18-f0007.gif

Thomas Jaenisch, M.D. Ph.D., is clinical researcher and infectious disease epidemiologist at Heidelberg University Hospital (UKHD). In his research, he focuses on emerging arboviruses of global importance, notably dengue viruses (DENV) and Zika virus (ZIKV). Until recently, he was also responsible for the parasitology reference laboratory at UKHD. Dr. Jaenisch has coordinated in direct sequence two multicenter observational dengue consortia and is now the work package leader in the ZIKAlliance consortium (funded by the European Commission [EC] Horizon 2020 program), where he coordinates the multicenter cohorts in pregnant women and children in Latin America and the Caribbean. He is also responsible for harmonization and data sharing between different Zika consortia, aiming at a pooled analysis of cohort data between the three EC-funded Zika consortia and, in the context of a WHO-moderated effort, with other Zika maternal cohort studies globally.

graphic file with name CMR.00130-18-f0008.gif

Pieter Fraaij is a pediatric infectious disease and immunology consultant and head of the subdepartment of Pediatric Infectious Diseases, Immunology, and Rheumatology at the Sophia Children’s Hospital of the Erasmus University Medical Centre in Rotterdam, The Netherlands. In addition, he is employed at the Department of Viroscience at the same institution as a clinical scientist. This unique combination of professional environments has allowed him to set up and implement translational research studies, initiating and coordinating interactions between the scientific and clinical departments. He obtained his Ph.D. at the Erasmus University Rotterdam in clinical, virological, and immunological aspects of highly active antiretroviral therapy in HIV-1-infected children. His current research focuses on the impact of emerging and reemerging viruses and the effects of childhood growth and development on host-virus interactions.

graphic file with name CMR.00130-18-f0009.gif

Philippe Mayaud, M.D., M.Sc., is a Professor of Infectious Diseases and Reproductive Health at the London School of Hygiene and Tropical Medicine (Faculty of Infectious and Tropical Diseases) and the Head of the HIV Epidemiology and Intervention Research Programme at the Medical Research Unit/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Research Unit in Uganda, which he joined in 2019. His research focuses on epidemiological, clinical, and intervention aspects of HIV/AIDS and sexually transmitted infections, particularly in sub-Saharan Africa. In addition, he has conducted research projects on arbovirus/Zika epidemiology research in Brazil where he held the position of Visiting Professor at the University of Sao Paulo (2015 to 2018).

graphic file with name CMR.00130-18-f0010.gif

Ann Vossen, M.D., Ph.D., is Associate Professor of Clinical Virology at the Department of Medical Microbiology, Leiden University Medical Center. Her research focuses on congenital cytomegalovirus infection, including several large cohort studies on disease burden, virus-host interaction, and antiviral treatment. She is actively involved in the European Congenital CMV Initiative. Since 2018, she has been President of the Dutch Society of Medical Microbiology.

graphic file with name CMR.00130-18-f0011.gif

Marion Koopmans, D.V.M., Ph.D., is head of the Department of Viroscience and director of the WHO collaborating center for arboviruses and viral hemorrhagic fevers, Erasmus Medical Centre, Rotterdam, The Netherlands. Her research focuses on the global population-level impact of rapidly spreading zoonotic and vector-borne virus infections, with an emphasis on improving preparedness and outbreak response through advanced laboratory studies. She is scientific coordinator of COMPARE, a large Horizon 2020-funded project (20 MEuro) exploring the potential uses of next-generation sequencing techniques for outbreak detection and tracking (www.compare-europe.eu), coprincipal investigator in the FP7-funded PREPARE project (www.prepare-europe.eu) aimed at building a pan-European operational network for rapid and large-scale European clinical research in response to infectious disease outbreaks with epidemic potential, and coprincipal investigator in ZIKALLIANCE. She has coauthored >500 papers that have been cited >20,000 times.

REFERENCES

  • 1.Kindhauser MK, Allen T, Frank V, Santhana R, Dye C. 2016. Zika: the origin and spread of a mosquito-borne virus. Bull World Health Organ 94:675–686. doi: 10.2471/BLT.16.171082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kleber de Oliveira W, Cortez-Escalante J, De Oliveira WT, do Carmo GM, Henriques CM, Coelho GE, Araújo de França GV. 2016. Increase in reported prevalence of microcephaly in infants born to women living in areas with confirmed Zika virus transmission during the first trimester of pregnancy, Brazil, 2015. MMWR Morb Mortal Wkly Rep 65:242–247. doi: 10.15585/mmwr.mm6509e2. [DOI] [PubMed] [Google Scholar]
  • 3.FrançA GVA, Schuler-Faccini L, Oliveira WK, Henriques CMP, Carmo EH, Pedi VD, Nunes ML, Castro MC, Serruya S, Silveira MF, Barros FC, Victora CG. 2016. Congenital Zika virus syndrome in Brazil: a case series of the first 1501 livebirths with complete investigation. Lancet 359:891–897. doi: 10.1016/S0140-6736(16)30902-3. [DOI] [PubMed] [Google Scholar]
  • 4.Baud D, Gubler DJ, Schaub B, Lanteri MC, Musso D. 2017. An update on Zika virus infection. Lancet 390:2099–2109. doi: 10.1016/S0140-6736(17)31450-2. [DOI] [PubMed] [Google Scholar]
  • 5.Duffy MR, Chen TH, Hancock WT, Powers AM, Kool JL, Lanciotti RS, Pretrick M, Marfel M, Holzbauer S, Dubray C, Guillaumot L, Griggs A, Bel M, Lambert AJ, Laven J, Kosoy O, Panella A, Biggerstaff BJ, Fischer M, Hayes EB. 2009. Zika virus outbreak on Yap Island, Federated States of Micronesia. N Engl J Med 360:2536–2543. doi: 10.1056/NEJMoa0805715. [DOI] [PubMed] [Google Scholar]
  • 6.Gourinat A-C, O'Connor O, Calvez E, Goarant C, Dupont-Rouzeyrol M. 2015. Detection of Zika virus in urine. Emerg Infect Dis 21:84–86. doi: 10.3201/eid2101.140894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Mlakar J, Korva M, Tul N, Popović M, Poljšak-Prijatelj M, Mraz J, Kolenc M, Resman Rus K, Vesnaver Vipotnik T, Fabjan Vodušek V, Vizjak A, Pižem J, Petrovec M, Avšič Županc T. 2016. Zika virus associated with microcephaly. N Engl J Med 374:951–958. doi: 10.1056/NEJMoa1600651. [DOI] [PubMed] [Google Scholar]
  • 8.Cauchemez S, Besnard M, Bompard P, Dub T, Guillemette-Artur P, Eyrolle-Guignot D, Salje H, Van Kerkhove MD, Abadie V, Garel C, Fontanet A, Mallet HP. 2016. Association between Zika virus and microcephaly in French Polynesia, 2013–15: a retrospective study. Lancet 387:2125–2132. doi: 10.1016/S0140-6736(16)00651-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Russo FB, Jungmann P, Beltrão-Braga PC. 2017. Zika infection and the development of neurological defects. Cell Microbiol 19:e12744. doi: 10.1111/cmi.12744. [DOI] [PubMed] [Google Scholar]
  • 10.Kim K, Shresta S. 2016. Neuroteratogenic viruses and lessons for Zika virus models. Trends Microbiol 24:622–636. doi: 10.1016/j.tim.2016.06.002. [DOI] [PubMed] [Google Scholar]
  • 11.Da Silva SR, Gao S-J. 2016. Zika virus: an update on epidemiology, pathology, molecular biology and animal model. J Med Virol 88:1291–1296. doi: 10.1002/jmv.24563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Van den Pol AN, Mao G, Yang Y, Ornaghi S, Davis JN. 2017. Zika virus targeting in the developing brain. J Neurosci 37:2161–2175. doi: 10.1523/JNEUROSCI.3124-16.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Koide F, Goebel S, Snyder B, Walters KB, Gast A, Hagelin K, Kalkeri R, Rayner J. 2016. Development of a Zika virus infection model in cynomolgus macaques. Front Microbiol 7:2028. doi: 10.3389/fmicb.2016.02028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Panchaud A, Stojanov M, Ammerdorffer A, Vouga M, Baud D. 2016. Emerging role of Zika virus in adverse fetal and neonatal outcomes. Clin Microbiol Rev 29:659–694. doi: 10.1128/CMR.00014-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Coelho AVC, Crovella S. 2017. Microcephaly prevalence in infants born to Zika virus-infected women: a systematic review and meta-analysis. Int J Mol Sci 18:E1714. doi: 10.3390/ijms18081714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Shapiro-Mendoza CK, Rice ME, Galang RR, Fulton AC, VanMaldeghem K, Prado MV, Ellis E, Anesi MS, Simeone RM, Petersen EE, Ellington SR, Jones AM, Williams T, Reagan-Steiner S, Perez-Padilla J, Deseda CC, Beron A, Tufa AJ, Rosinger A, Roth NM, Green C, Martin S, Lopez CD, deWilde L, Goodwin M, Pagano HP, Mai CT, Gould C, Zaki S, Ferrer LN, Davis MS, Lathrop E, Polen K, Cragan JD, Reynolds M, Newsome KB, Huertas MM, Bhatangar J, Quiñones AM, Nahabedian JF, Adams L, Sharp TM, Hancock WT, Rasmussen SA, Moore CA, Jamieson DJ, Munoz-Jordan JL, Garstang H, Kambui A, Masao C, Honein MA, Meaney-Delman D, Zika Pregnancy and Infant Registries Working Group. 2017. Pregnancy outcomes after maternal Zika virus infection during pregnancy—U.S. Territories, January 1, 2016–April 25, 2017. MMWR Morb Mortal Wkly Rep 66:615–621. doi: 10.15585/mmwr.mm6623e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hoen B, Schaub B, Funk AL, Ardillon V, Boullard M, Cabié A, Callier C, Carles G, Cassadou S, Césaire R, Douine M, Herrmann-Storck C, Kadhel P, Laouénan C, Madec Y, Monthieux A, Nacher M, Najioullah F, Rousset D, Ryan C, Schepers K, Stegmann-Planchard S, Tressières B, Voluménie J-L, Yassinguezo S, Janky E, Fontanet A. 2018. Pregnancy outcomes after ZIKV infection in French Territories in the Americas. N Engl J Med 378:985–994. doi: 10.1056/NEJMoa1709481. [DOI] [PubMed] [Google Scholar]
  • 18.Morris JK, Springett AL, Greenlees R, Loane M, Addor M-C, Arriola L, Barisic I, Bergman JEH, Csaky-Szunyogh M, Dias C, Draper ES, Garne E, Gatt M, Khoshnood B, Klungsoyr K, Lynch C, McDonnell R, Nelen V, Neville AJ, O'Mahony M, Pierini A, Queisser-Luft A, Randrianaivo H, Rankin J, Rissmann A, Kurinczuk J, Tucker D, Verellen-Dumoulin C, Wellesley D, Dolk H. 2018. Trends in congenital anomalies in Europe from 1980 to 2012. PLoS One 13:e0194986. doi: 10.1371/journal.pone.0194986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Paz-Bailey G, Rosenberg ES, Doyle K, Munoz-Jordan J, Santiago GA, Klein L, Perez-Padilla J, Medina FA, Waterman SH, Gubern CG, Alvarado LI, Sharp TM. 2018. Persistence of Zika virus in body fluids—final report. N Engl J Med 379:1234–1243. doi: 10.1056/NEJMoa1613108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Meaney-Delman D, Oduyebo T, Polen KN, White JL, Bingham AM, Slavinski SA, Heberlein-Larson L, St George K, Rakeman JL, Hills S, Olson CK, Adamski A, Culver Barlow L, Lee EH, Likos AM, Munoz JL, Petersen EE, Dufort EM, Dean AB, Cortese MM, Santiago GA, Bhatnagar J, Powers AM, Zaki S, Petersen LR, Jamieson DJ, Honein MA. 2016. Prolonged detection of Zika virus RNA in pregnant women. Obstet Gynecol 128:724–730. doi: 10.1097/AOG.0000000000001625. [DOI] [PubMed] [Google Scholar]
  • 21.Schaub B, Vouga M, Najioullah F, Gueneret M, Monthieux A, Harte C, Muller F, Jolivet E, Adenet C, Dreux S, Leparc-Goffart I, Cesaire R, Volumenie J-L, Baud D. 2017. Analysis of blood from Zika virus-infected fetuses: a prospective case series. Lancet Infect Dis 17:520–527. doi: 10.1016/S1473-3099(17)30102-0. [DOI] [PubMed] [Google Scholar]
  • 22.Nahmias AJ, Walls KW, Stewart JA, Herrmann KL, Flynt WJ Jr. 1971. The ToRCH complex-perinatal infections associated with toxoplasma and rubella, cytomegol- and herpes simplex viruses. Pediatr Res 5:405–406. doi: 10.1203/00006450-197108000-00144. [DOI] [Google Scholar]
  • 23.Munro SC, Hall B, Whybin LR, Leader L, Robertson P, Maine GT, Rawlinson WD. 2005. Diagnosis of and screening for cytomegalovirus infection in pregnant women. J Clin Microbiol 43:4713–4718. doi: 10.1128/JCM.43.9.4713-4718.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Forner G, Saldan A, Mengoli C, Gussetti N, Palù G, Abate D. 2016. Cytomegalovirus (CMV) enzyme-linked immunosorbent spot assay but not CMV QuantiFERON assay is a novel biomarker to determine risk of congenital CMV infection in pregnant women. J Clin Microbiol 54:2149–2154. doi: 10.1128/JCM.00561-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Silasi M, Cardenas I, Racicot K, Kwon J-Y, Aldo P, Mor G. 2015. Viral infections during pregnancy. Am J Reprod Immunol 73:199–213. doi: 10.1111/aji.12355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Coyne CB, Lazear HM. 2016. Zika virus—reigniting the TORCH. Nat Rev Microbiol 14:707–715. doi: 10.1038/nrmicro.2016.125. [DOI] [PubMed] [Google Scholar]
  • 27.Pasquini L, Seravalli V, Sisti G, Battaglini C, Nepi F, Pelagalli R, Di Tommaso M. 2016. Prevalence of a positive TORCH and parvovirus B19 screening in pregnancies complicated by polyhydramnios. Prenat Diagn 36:290–293. doi: 10.1002/pd.4769. [DOI] [PubMed] [Google Scholar]
  • 28.Naresh A, Simhan H. 2012. Absence of viruses in amniotic fluid of women with PPROM: a case series. J Reprod Immunol 96:79–83. doi: 10.1016/j.jri.2012.08.003. [DOI] [PubMed] [Google Scholar]
  • 29.Brkic S, Bogavac MA, Simin N, Hrnjakovic-Cvetkovic I, Milosevic V, Maric D. 2011. Unusual high rate of asymptomatic maternal parvovirus B19 infection associated with severe fetal outcome. J Matern Fetal Neonatal Med 24:647–649. doi: 10.3109/14767058.2010.511330. [DOI] [PubMed] [Google Scholar]
  • 30.Vossen A. 2014. Virale infecties in de zwangerschap met risico voor het kind. Stand van zaken. Ned Tijdschr Geneeskd 158:A7418. [PubMed] [Google Scholar]
  • 31.Bollani L, Strocchio L, Stronati M. 2013. Congenital toxoplasmosis. Early Hum Dev 89(Suppl 4):S70–S72. doi: 10.1016/S0378-3782(13)70107-5. [DOI] [Google Scholar]
  • 32.Lamont RF, Sobel JD, Vaisbuch E, Kusanovic JP, Mazaki-Tovi S, Kim SK, Uldbjerg N, Romero R. 2011. Parvovirus B19 infection in human pregnancy. BJOG 118:175–186. doi: 10.1111/j.1471-0528.2010.02749.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bessières MH, Berrebi A, Cassaing S, Fillaux J, Cambus JP, Berry A, Assouline C, Ayoubi JM, Magnaval JF. 2009. Diagnosis of congenital toxoplasmosis: prenatal and neonatal evaluation of methods used in Toulouse University Hospital and incidence of congenital toxoplasmosis. Mem Inst Oswaldo Cruz 104:389–392. doi: 10.1590/s0074-02762009000200038. [DOI] [PubMed] [Google Scholar]
  • 34.Cordier AG, Vauloup-Fellous C, Grangeot-Keros L, Pinet C, Benachi A, Ayoubi JM, Picone O. 2012. Pitfalls in the diagnosis of congenital rubella syndrome in the first trimester of pregnancy. Prenat Diagn 32:496–497. doi: 10.1002/pd.2943. [DOI] [PubMed] [Google Scholar]
  • 35.Sensini A. 2006. Toxoplasma gondii infection in pregnancy- opportunities and pitfalls of serological diagnosis. Clin Microbiol Infect 12:504–512. doi: 10.1111/j.1469-0691.2006.01444.x. [DOI] [PubMed] [Google Scholar]
  • 36.Bonvicini F, Puccetti C, Salfi NC, Guerra B, Gallinella G, Rizzo N, Zerbini M. 2011. Gestational and fetal outcomes in B19 maternal infection: a problem of diagnosis. J Clin Microbiol 49:3514–3518. doi: 10.1128/JCM.00854-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Žegarac Ž, Duić Ž, Borovečki A. 2013. Recent parvovirus B19 infection in late pregnancy. Int J Gynaecol Obstet 122:262–263. doi: 10.1016/j.ijgo.2013.04.006. [DOI] [PubMed] [Google Scholar]
  • 38.de Vries JJ, van Zwet EW, Dekker FW, Kroes AC, Verkerk PH, Vossen AC. 2013. The apparent paradox of maternal seropositivity as a risk factor for congenital cytomegalovirus infection: a population-based prediction model. Rev Med Virol 23:241–249. doi: 10.1002/rmv.1744. [DOI] [PubMed] [Google Scholar]
  • 39.Crane J, Mundle W, Boucoiran I, Maternal Fetal Medicine Committee. 2014. Parvovirus B19 infection in pregnancy. J Obstet Gynaecol Can 36:1107–1116. doi: 10.1016/S1701-2163(15)30390-X. [DOI] [PubMed] [Google Scholar]
  • 40.Saldan A, Forner G, Mengoli C, Gussetti N, Palù G, Abate D. 2017. Testing for cytomegalovirus in pregnancy. J Clin Microbiol 55:693–702. doi: 10.1128/JCM.01868-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Vauloup-Fellous C, Grangeot-Keros L. 2007. Humoral immune response after primary rubella virus infection and after vaccination. Clin Vaccine Immunol 14:644–647. doi: 10.1128/CVI.00032-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Weisblum Y, Panet A, Haimov-Kochman R, Wolf DG. 2014. Models of vertical cytomegalovirus (CMV) transmission and pathogenesis. Semin Immunopathol 36:615–625. doi: 10.1007/s00281-014-0449-1. [DOI] [PubMed] [Google Scholar]
  • 43.de Oliveira Azevedo CT, do Brasil PE, Guida L, Lopes Moreira ME. 2016. Performance of polymerase chain reaction analysis of the amniotic fluid of pregnant women for diagnosis of congenital toxoplasmosis: a systematic review and meta-analysis. PLoS One 11:e0149938. doi: 10.1371/journal.pone.0149938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Li Z, Yan C, Liu P, Yan R, Feng Z. 2009. Prevalence of serum antibodies to TORCH among women before pregnancy or in the early period of pregnancy in Beijing. Clin Chim Acta 403:212–215. doi: 10.1016/j.cca.2009.03.027. [DOI] [PubMed] [Google Scholar]
  • 45.Ribeiro AC, Mutis MS, Fernandes O. 2008. Association of the presence of residual anti-Toxoplasma gondii IgM in pregnant women and their respective family groups in Miracema, Northwest Rio de Janeiro, Brazil. Mem Inst Oswaldo Cruz 103:591–594. doi: 10.1590/s0074-02762008000600013. [DOI] [PubMed] [Google Scholar]
  • 46.Munoz-Zanzi CA, Fry P, Lesina B, Hill D. 2010. Toxoplasma gondii oocyst-specific antibodies and source of infection. Emerg Infect Dis 16:1591–1593. doi: 10.3201/eid1610.091674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Villena I, Ancelle T, Delmas C, Garcia P, Brezin AP, Thulliez P, Wallon M, King L, Goulet V, Toxosurv Network and National Reference Centre for Toxoplasmosis. 2010. Congenital toxoplasmosis in France in 2007: first results from a national surveillance system. Euro Surveill 15:19600. doi: 10.2807/ese.15.25.19600-en. [DOI] [PubMed] [Google Scholar]
  • 48.Kamal AM, Ahmed AK, Abdellatif MZ, Tawfik M, Hassan EE. 2015. Seropositivity of Toxoplasmosis in pregnant women by ELISA at Minia University Hospital, Egypt. Korean J Parasitol 53:605–610. doi: 10.3347/kjp.2015.53.5.605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Candotti D, Etiz N, Parsyan A, Allain JP. 2004. Identification and characterization of persistent human erythrovirus infection in blood donor samples. J Virol 78:12169–12178. doi: 10.1128/JVI.78.22.12169-12178.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Emiasegen SE, Nimzing L, Adoga MP, Ohagenyi AY, Lekan R. 2011. Parvovirus B19 antibodies and correlates of infection in pregnant women attending an antenatal clinic in central Nigeria. Mem Inst Oswaldo Cruz 106:227–231. doi: 10.1590/s0074-02762011000200018. [DOI] [PubMed] [Google Scholar]
  • 51.Lamarre V, Gilbert NL, Rousseau C, Gyorkos TW, Fraser WD. 2016. Seroconversion for cytomegalovirus infection in a cohort of pregnant women in Québec, 2010–2013. Epidemiol Infect 144:1701–1709. doi: 10.1017/S0950268815003167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zajkowska A, Garkowski A, Czupryna P, Moniuszko A, Król ME, Szamatowicz J, Pancewicz S. 2015. Seroprevalence of parvovirus B19 antibodies among young pregnant women or planning pregnancy, tested for toxoplasmosis. Przegl Epidemiol 69:479–482. [PubMed] [Google Scholar]
  • 53.Zhou Y, Bian G, Zhou Q, Gao Z, Liao P, Liu Y, He M. 2015. Detection of cytomegalovirus, human parvovirus B19, and herpes simplex virus-1/2 in women with first-trimester spontaneous abortions. J Med Virol 87:1749–1753. doi: 10.1002/jmv.24218. [DOI] [PubMed] [Google Scholar]
  • 54.Barlinn R, Vainio K, Samdal HH, Nordbø SA, Nøkleby H, Dudman SG. 2014. Susceptibility to cytomegalovirus, parvovirus B19 and age-dependent differences in levels of rubella antibodies among pregnant women. J Med Virol 86:820–826. doi: 10.1002/jmv.23757. [DOI] [PubMed] [Google Scholar]
  • 55.Karacan M, Batukan M, Cebi Z, Berberoglugil M, Levent S, Kır M, Baksu A, Ozel E, Camlıbel T. 2014. Screening cytomegalovirus, rubella and toxoplasma infections in pregnant women with unknown pre-pregnancy serological status. Arch Gynecol Obstet 290:1115–1120. doi: 10.1007/s00404-014-3340-3. [DOI] [PubMed] [Google Scholar]
  • 56.de Jong EP, Walther FJ, Kroes AC, Oepkes D. 2011. Parvovirus B19 infection in pregnancy: new insights and management. Prenat Diagn 5:419–425. doi: 10.1002/pd.2714. [DOI] [PubMed] [Google Scholar]
  • 57.Cannon MJ, Schmid DS, Hyde TB. 2010. Review of cytomegalovirus seroprevalence and demographic characteristics associated with infection. Rev Med Virol 20:202–213. doi: 10.1002/rmv.655. [DOI] [PubMed] [Google Scholar]
  • 58.Vynnycky E, Adams EJ, Cutts FT, Reef SE, Navar AM, Simons E, Yoshida LM, Brown DW, Jackson C, Strebel PM, Dabbagh AJ. 2016. Using seroprevalence and immunisation coverage data to estimate the global burden of congenital rubella syndrome, 1996–2010: a systematic review. PLoS One 11:e0149160. doi: 10.1371/journal.pone.0149160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Zanga J, Mbanzulu MK, Kabasele A-F, Ngatu NR, Wumba DR. 2017. Rubella seroprevalence and real-time PCR detection of RUBV among Congolese pregnant women. BMC Infect Dis 17:250. doi: 10.1186/s12879-017-2352-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mirambo MM, Majigo M, Aboud S, Groß U, Mshana SE. 2015. Serological markers of rubella infection in Africa in the pre vaccination era: a systematic review. BMC Res Notes 8:716. doi: 10.1186/s13104-015-1711-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Loconsole D, Metallo A, De Robertis AL, Morea A, Quarto M, Chironna M. 2018. Seroprevalence of dengue virus, West Nile virus, chikungunya virus, and Zika virus in international travelers attending a travel and migration center in 2015–2017, Southern Italy. Vector Borne Zoonotic Dis 18:331–334. doi: 10.1089/vbz.2017.2260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Seruyange E, Gahutu JB, Muvunyi CM, Katare S, Ndahindwa V, Sibomana H, Nyamusore J, Rutagarama F, Hannoun C, Norder H, Bergström T. 2018. Seroprevalence of Zika virus and rubella virus IgG among blood donors in Rwanda and in Sweden. J Med Virol 90:1290–1296. doi: 10.1002/jmv.25198. [DOI] [PubMed] [Google Scholar]
  • 63.Borena W, Hofer T, Stiasny K, Aberle SW, Gaber M, von Laer D, Schennach H. 2017. No molecular or serological evidence of Zikavirus infection among healthy blood donors living in or travelling to regions where Aedes albopictus circulates. PLoS One 12:e0178175. doi: 10.1371/journal.pone.0178175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Saba Villarroel PM, Nurtop E, Pastorino B, Roca Y, Drexler JF, Gallian P, Jaenisch T, Leparc-Goffart I, Priet S, Ninove L, de Lamballerie X. 2018. Zika virus epidemiology in Bolivia: a seroprevalence study in volunteer blood donors. PLoS Negl Trop Dis 12:e0006239. doi: 10.1371/journal.pntd.0006239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Gake B, Vernet MA, Leparc-Goffart I, Drexler JF, Gould EA, Gallian P, de Lamballerie X. 2017. Low seroprevalence of Zika virus in Cameroonian blood donors. Braz J Infect Dis 21:481–483. doi: 10.1016/j.bjid.2017.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Aubry M, Teissier A, Huart M, Merceron S, Vanhomwegen J, Roche C, Vial A, Teururai S, Sicard S, Paulous S, Desprès P, Manuguerra JC, Mallet HP, Musso D, Deparis X, Cao-Lormeau VM. 2017. Zika virus seroprevalence, French Polynesia, 2014–2015. Emerg Infect Dis 23:669–672. doi: 10.3201/eid2304.161549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Netto EM, Moreira-Soto A, Pedroso C, Höser C, Funk S, Kucharski AJ, Rockstroh A, Kümmerer BM, Sampaio GS, Luz E, Vaz SN, Dias JP, Bastos FA, Cabral K, Kistemann T, Ulbert S, de Lamballerie X, Jaenisch T, Brady OJ, Drosten C, Sarno M, Brites C, Drexler JF. 2017. High Zika virus seroprevalence in Salvador, Northeastern Brazil limits the potential for further outbreaks. mBio 8:e01390-17. doi: 10.1128/mBio.01390-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Hyde TB, Schmid DS, Cannon MJ. 2010. Cytomegalovirus seroconversion rates and risk factors: implications for congenital CMV. Rev Med Virol 20:311–326. doi: 10.1002/rmv.659. [DOI] [PubMed] [Google Scholar]
  • 69.Revello MG, Tibaldi C, Masuelli G, Frisina V, Sacchi A, Furione M, Arossa A, Spinillo A, Klersy C, Ceccarelli M, Gerna G, Todros T, CCPE Study Group. 2015. Prevention of primary cytomegalovirus infection in pregnancy. EBioMedicine 2:1205–1210. doi: 10.1016/j.ebiom.2015.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lazzarotto T, Guerra B, Gabrielli L, Lanari M, Landini MP. 2011. Update on the prevention, diagnosis and management of cytomegalovirus infection during pregnancy. Clin Microbiol Infect 17:1285–1293. doi: 10.1111/j.1469-0691.2011.03564.x. [DOI] [PubMed] [Google Scholar]
  • 71.Adler SP. 2011. Screening for cytomegalovirus during pregnancy. Infect Dis Obstet Gynecol 2011:942937. doi: 10.1155/2011/942937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Valeur-Jensen AK, Pedersen CB, Westergaard T, Jensen IP, Lebech M, Andersen PK, Aaby P, Pedersen BN, Melbye M. 1999. Risk factors for parvovirus B19 infection in pregnancy. JAMA 281:1099–1105. doi: 10.1001/jama.281.12.1099. [DOI] [PubMed] [Google Scholar]
  • 73.Mossong J, Hens N, Friederichs V, Davidkin I, Broman M, Litwinska B, Siennicka J, Trzcinska A, van Damme P, Beutels P, Vyse A, Shkedy Z, Aerts M, Massari M, Gabutti G. 2008. Parvovirus B19 infection in five European countries: seroepidemiology, force of infection and maternal risk of infection. Epidemiol Infect 136:1059–1068. doi: 10.1017/S0950268807009661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Bouthry E, Picone O, Hamdi G, Grangeot-Keros L, Ayoubi JM, Vauloup-Fellous C. 2014. Rubella and pregnancy: diagnosis, management and outcomes. Prenat Diagn 34:1246–1253. doi: 10.1002/pd.4467. [DOI] [PubMed] [Google Scholar]
  • 75.Hutton J, Rowan P, Greisinger A, Mouzoon M. 2014. Rubella monitoring in pregnancy as a means for evaluating a possible reemergence of rubella. Am J Obstet Gyn 211:534.e1–e4. doi: 10.1016/j.ajog.2014.05.046. [DOI] [PubMed] [Google Scholar]
  • 76.Champagne C, Salthouse DG, Paul R, Cao-Lormeau V-M, Roche B, Cazelles B. 2016. Structure in the variability of the basic reproductive number (R0) for Zika epidemics in the Pacific islands. Elife 5:e19874. doi: 10.7554/eLife.19874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Shiu C, Starker R, Kwal J, Bartlett M, Crane A, Greissman S, Gunaratne N, Lardy M, Picon M, Rodriguez P, Gonzalez I, Curry CL. 2018. Zika virus testing and outcomes during pregnancy, Florida, USA, 2016. Emerg Infect Dis 24:1–8. doi: 10.3201/eid2401.170979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Chalouhi GE, Benedetti S, Alby C, Benzina N, Ville Y. 2014. Cause of fetal demise in first-trimester parvovirus infection: anemia, placentitis or myocarditis? Ultrasound Obstet Gynecol 44:618–619. doi: 10.1002/uog.13416. [DOI] [PubMed] [Google Scholar]
  • 79.Puccetti C, Contoli M, Bonvicini F, Cervi F, Simonazzi G, Gallinella G, Murano P, Farina A, Guerra B, Zerbini M, Rizzo N. 2012. Parvovirus B19 in pregnancy: possible consequences of vertical transmission. Prenat Diagn 32:897–902. doi: 10.1002/pd.3930. [DOI] [PubMed] [Google Scholar]
  • 80.Citil Dogan A, Wayne S, Bauer S, Ogunyemi D, Kulkharni SK, Maulik D, Carpenter CF, Bahado-Singh RO. 2017. The Zika virus and pregnancy: evidence, management and prevention. J Matern Fetal Neonatal Med 30:386–396. doi: 10.3109/14767058.2016.1174210. [DOI] [PubMed] [Google Scholar]
  • 81.Giambi C, Montaño-Remacha C, Celentano LP, Derrough T, National Focal Points for Rubella. 2015. Surveillance of congenital rubella and rubella infections in pregnancy in EU/EEA countries, 2012: current status and future perspective to monitor elimination. Vaccine 33:4929–4937. doi: 10.1016/j.vaccine.2015.07.041. [DOI] [PubMed] [Google Scholar]
  • 82.Landolsi H, Yacoubi MT, Bouslama L, Lahmar A, Trabelsi A, Hmissa S, Aouni M, Korbi S. 2009. Detection of the human parvovirus B19 in nonimmune hydrops fetalis using immunohistochemistry and nested-PCR in formalin-fixed and paraffin-embedded placenta and fetal tissues. Pathol Biol (Paris) 57:e1–e7. doi: 10.1016/j.patbio.2007.11.001. [DOI] [PubMed] [Google Scholar]
  • 83.Beigi RH, Wiesenfeld HC, Landers DV, Simhan HN. 2008. High rate of severe fetal outcomes associated with maternal parvovirus b19 infection in pregnancy. Infect Dis Obstet Gynecol 2008:524601. doi: 10.1155/2008/524601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Enders M, Weidner A, Zoellner I, Searle K, Enders G. 2004. Fetal morbidity and mortality after acute human parvovirus B19 infection in pregnancy: prospective evaluation of 1018 cases. Prenat Diagn 24:513–518. doi: 10.1002/pd.940. [DOI] [PubMed] [Google Scholar]
  • 85.Lassen J, Jensen AK, Bager P, Pedersen CB, Panum I, Nørgaard-Pedersen B, Aaby P, Wohlfahrt J, Melbye M. 2012. Parvovirus B19 infection in the first trimester of pregnancy and risk of fetal loss: a population-based case-control study. Am J Epidemiol 176:803–807. doi: 10.1093/aje/kws177. [DOI] [PubMed] [Google Scholar]
  • 86.Sarfraz AA, Samuelsen SO, Bruu AL, Jenum PA, Eskild A. 2009. Maternal human parvovirus B19 infection and the risk of fetal death and low birthweight: a case-control study within 35,940 pregnant women. BJOG 116:1492–1498. doi: 10.1111/j.1471-0528.2009.02211.x. [DOI] [PubMed] [Google Scholar]
  • 87.Miyakawa M, Yoshino H, Yoshida LM, Vynnycky E, Motomura H, Tho Le H, Thiem VD, Ariyoshi K, Anh DD, Moriuchi H. 2014. Seroprevalence of rubella in the cord blood of pregnant women and congenital rubella incidence in Nha Trang, Vietnam. Vaccine 32:1192–1198. doi: 10.1016/j.vaccine.2013.08.076. [DOI] [PubMed] [Google Scholar]
  • 88.Li JM, Zhang HF, Zhang XQ, Huang GL, Huang HZ, Yu WW. 2015. Genetic mechanism associated with congenital cytomegalovirus infection and analysis of effects of the infection on pregnancy outcome. Genet Mol Res 14:13247–13257. doi: 10.4238/2015.October.26.21. [DOI] [PubMed] [Google Scholar]
  • 89.Lipitz S, Yinon Y, Malinger G, Yagel S, Levit L, Hoffman C, Rantzer R, Weisz B. 2013. Risk of cytomegalovirus-associated sequelae in relation to time of infection and findings on prenatal imaging. Ultrasound Obstet Gynecol 41:508–514. doi: 10.1002/uog.12377. [DOI] [PubMed] [Google Scholar]
  • 90.Schleiss MR. 2013. Cytomegalovirus in the neonate: immune correlates of infection and protection. Review article. Clin Dev Immunol 2013:501801. doi: 10.1155/2013/501801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Ticconi C, Pietropolli A, Rezza G. 2016. Zika virus infection and pregnancy: what we do and do not know. Path Global Health 110:262–284. doi: 10.1080/20477724.2016.1234804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Tanimura K, Tairaku S, Morioka I, Ozaki K, Nagamata S, Morizane M, Deguchi M, Ebina Y, Minematsu T, Yamada H. 2017. Universal screening with use of immunoglobulin G avidity for congenital cytomegalovirus infection. Clin Infect Dis 65:1652–1658. doi: 10.1093/cid/cix621. [DOI] [PubMed] [Google Scholar]
  • 93.Braakenburg AM, Crespi CM, Holland GN, Wu S, Yu F, Rothova A. 2014. Recurrence rates of ocular toxoplasmosis during pregnancy. Am J Ophthalmol 157:767–773. doi: 10.1016/j.ajo.2014.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kaňková S, Sulc J, Křivohlavá R, Kuběna A, Flegr J. 2012. Slower postnatal motor development in infants of mothers with latent toxoplasmosis during the first 18 months of life. Early Hum Dev 88:879–884. doi: 10.1016/j.earlhumdev.2012.07.001. [DOI] [PubMed] [Google Scholar]
  • 95.Heegaard ED, Brown KE. 2002. Human parvovirus B19. Clin Microbiol Rev 15:485–505. doi: 10.1128/cmr.15.3.485-505.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Fornara C, Furione M, Lilleri D, Cane I, Revello MG, Zavattoni M, Gerna G. 2015. Primary human cytomegalovirus infections: kinetics of ELISA-IgG and neutralizing antibody in pauci/asymptomatic pregnant women vs symptomatic non-pregnant subjects. J Clin Virol 64:45–51. doi: 10.1016/j.jcv.2015.01.004. [DOI] [PubMed] [Google Scholar]
  • 97.Maine GT, Stricker R, Stricker R. 2012. Kinetics of CMV seroconversion in a Swiss pregnant women population. Diagn Microbiol Infect Dis 73:275–277. doi: 10.1016/j.diagmicrobio.2012.03.016. [DOI] [PubMed] [Google Scholar]
  • 98.Armengol C, Cassaing S, Roques-Malecaze C, Chauvin P, Iriart X, Berry A, Fillaux J. 2017. Time before anti-Toxoplasma IgG seroconversion detection by 7 commercial assays in French pregnant women. Diagn Microbiol Infect Dis 87:103–107. doi: 10.1016/j.diagmicrobio.2016.10.020. [DOI] [PubMed] [Google Scholar]
  • 99.Mahmoudi S, Mamishi S, Suo X, Keshavarz H. 2017. Early detection of Toxoplasma gondii infection by using a interferon gamma release assay: a review. Exp Parasitol 172:39–43. doi: 10.1016/j.exppara.2016.12.008. [DOI] [PubMed] [Google Scholar]
  • 100.Tipples GA. 2011. Rubella diagnostic issues in Canada. J Infect Dis 204(Suppl 2):S659–S663. doi: 10.1093/infdis/jir430. [DOI] [PubMed] [Google Scholar]
  • 101.Enders G, Daiminger A, Bäder U, Exler S, Schimpf Y, Enders M. 2013. The value of CMV IgG avidity and immunoblot for timing the onset of primary CMV infection in pregnancy. J Clin Virol 56:102–107. doi: 10.1016/j.jcv.2012.09.015. [DOI] [PubMed] [Google Scholar]
  • 102.Revello MG, Gerna G. 2002. Diagnosis and management of human cytomegalovirus infection in the mother, fetus and newborn infant. Clin Microbiol Rev 15:680–715. doi: 10.1128/cmr.15.4.680-715.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Pasquier C, Joguet G, Mengelle C, Chapuy-Regaud S, Pavili L, Prisant N, Izopet J, Bujan L, Mansuy JM. 2018. Kinetics of anti-ZIKV antibodies after Zika infection using two commercial enzyme-linked immunoassays. Diagn Microbiol Infect Dis 90:26–30. doi: 10.1016/j.diagmicrobio.2017.09.001. [DOI] [PubMed] [Google Scholar]
  • 104.Lindblom A, Isa A, Norbeck O, Wolf S, Johansson B, Broliden K, Tolfvenstam T. 2005. Slow clearance of human parvovirus B19 viremia following acute infection. Clin Infect Dis 4:1201–1203. doi: 10.1086/444503. [DOI] [PubMed] [Google Scholar]
  • 105.Oduyebo T, Polen KD, Walke HT, Reagan-Steiner S, Lathrop E, Rabe IB, Kuhnert-Tallman WL, Martin SW, Walker AT, Gregory CJ, Ades EW, Carroll DS, Rivera M, Perez-Padilla J, Gould C, Nemhauser JB, Ben Beard C, Harcourt JL, Viens L, Johansson M, Ellington SR, Petersen E, Smith LA, Reichard J, Munoz-Jordan J, Beach MJ, Rose DA, Barzilay E, Noonan-Smith M, Jamieson DJ, Zaki SR, Petersen LR, Honein MA, Meaney-Delman D. 2017. Update: interim guidance for health care providers caring for pregnant women with possible Zika virus exposure—United States (including U.S. territories), July 2017. MMWR Morb Mortal Wkly Rep 66:781–793. doi: 10.15585/mmwr.mm6629e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Khorrami SM, Mokhtari-Azad T, Yavarian J, Nasab GS, Naseri M, Jandaghi NZ. 2015. The etiology of rubella IgM positivity in patients with rubella-like illness in Iran from 2011 to 2013. J Med Virol 87:1846–1852. doi: 10.1002/jmv.24250. [DOI] [PubMed] [Google Scholar]
  • 107.Wandinger KP, Saschenbrecker S, Steinhagen K, Scheper T, Meyer W, Bartelt U, Enders G. 2011. Diagnosis of recent primary Rubella virus infections: significance of glycoprotein-based IgM serology, IgG avidity and immunoblot analysis. J Virol Methods 174:85–93. doi: 10.1016/j.jviromet.2011.04.001. [DOI] [PubMed] [Google Scholar]
  • 108.van Helden J, Grangeot-Keros L, Vauloup-Fellous C, Vleminckx R, Masset F, Revello MG. 2014. Evaluation of fully automated assays for the detection of rubella IgM and IgG antibodies by the Elecsys immunoassay system. J Virol Methods 199:108. doi: 10.1016/j.jviromet.2014.01.009. [DOI] [PubMed] [Google Scholar]
  • 109.Gussetti N, D'Elia R, Mottola A, Rigoli E. 1990. Natural immunoglobulin M antibodies against Toxoplasma gondii during pregnancy. Am J Obstet Gyn 162:1359–1360. doi: 10.1016/0002-9378(90)90083-J. [DOI] [PubMed] [Google Scholar]
  • 110.Konishi E. 1987. A pregnant woman with a high level of naturally occurring immunoglobulin M antibodies to Toxoplasma gondii. Am J Obstet Gyn 157:832–833. doi: 10.1016/S0002-9378(87)80066-2. [DOI] [PubMed] [Google Scholar]
  • 111.Prince HE, Lapé-Nixon M. 2014. Role of cytomegalovirus (CMV) IgG avidity testing in diagnosing primary CMV infection during pregnancy. Clin Vaccine Immunol 21:1377–1384. doi: 10.1128/CVI.00487-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Revello MG, Vauloup-Fellous C, Grangeot-Keros L, van Helden J, Dickstein Y, Lipkin I, Mühlbacher A, Lazzarotto T. 2012. Clinical evaluation of new automated cytomegalovirus IgM and IgG assays for the Elecsys analyzer platform. Eur J Clin Microbiol Infect Dis 31:3331–3339. doi: 10.1007/s10096-012-1700-0. [DOI] [PubMed] [Google Scholar]
  • 113.Gay-Andrieu F, Fricker-Hidalgo H, Sickinger E, Espern A, Brenier-Pinchart MP, Braun HB, Pelloux H. 2009. Comparative evaluation of the ARCHITECT Toxo IgG, IgM, and IgG avidity assays for anti-Toxoplasma antibodies detection in pregnant women sera. Diagn Microbiol Infect Dis 65:279–287. doi: 10.1016/j.diagmicrobio.2009.07.013. [DOI] [PubMed] [Google Scholar]
  • 114.Murat J-B, L'Ollivier C, Fricker Hidalgo H, Franck J, Pelloux H, Piarroux R. 2012. Evaluation of the new Elecsys Toxo IgG avidity assay for toxoplasmosis and new insights into the interpretation of avidity results. Clin Vaccine Immunol 19:1838–1843. doi: 10.1128/CVI.00333-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Dogan K, Kafkasli A, Kaya C, Cengiz H. 2013. Seroprevalence and seroconversion rates of cytomegalovirus pp65 antigen and cord blood screening of pregnant women in Malatya, Turkey. Eurasian J Med 45:88–91. doi: 10.5152/eajm.2013.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Nissapatorn V, Suwanrath C, Sawangjaroen N, Ling LY, Chandeying V. 2011. Toxoplasmosis-serological evidence and associated risk factors among pregnant women in southern Thailand. Am J Trop Med Hyg 85:243–247. doi: 10.4269/ajtmh.2011.10-0633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.El Sanousi SM, Osman ZA, Mohmed AB, Al Awfi MS. 2016. Cytomegalovirus infection in a cohort of pregnant women. Am J Infect Control 44:e41–e3. doi: 10.1016/j.ajic.2015.11.002. [DOI] [PubMed] [Google Scholar]
  • 118.Leruez-Ville M, Ville Y. 2017. Fetal cytomegalovirus infection. Best Pract Res Clin Obstet Gynaecol 38:97–107. doi: 10.1016/j.bpobgyn.2016.10.005. [DOI] [PubMed] [Google Scholar]
  • 119.Charrel R, Mögling R, Pas S, Papa A, Baronti C, Koopmans M, Zeller H, Leparc-Goffart I, Reusken CB. 2017. Variable sensitivity in molecular detection of Zika virus in European expert laboratories: external quality assessment, November 2016. J Clin Microbiol 55:3219–3226. doi: 10.1128/JCM.00987-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Enders M, Weidner A, Rosenthal T, Baisch C, Hedman L, Söderlund-Venermo M, Hedman K. 2008. Improved diagnosis of gestational parvovirus B19 infection at the time of nonimmune fetal hydrops. J Infect Dis 197:58–62. doi: 10.1086/524302. [DOI] [PubMed] [Google Scholar]
  • 121.Weseslindtner L, Aberle JH, Hedman L, Hedman K. 2017. The chemokine CXCL-10 is a marker of infection stage in individuals with DNAemia due to parvovirus B19. J Infect Dis 215:214–220. doi: 10.1093/infdis/jiw509. [DOI] [PubMed] [Google Scholar]
  • 122.Baylis SA, Shah N, Minor PD. 2004. Evaluation of different assays for the detection of parvovirus B19 DNA in human plasma. J Virol Methods 121:7–16. doi: 10.1016/j.jviromet.2004.05.011. [DOI] [PubMed] [Google Scholar]
  • 123.-de Backer MWA, Russcher A, Kroes ACM, Koppelman MHGM, Lanfermeijer M, Zaaijer HL. 2016. Detection of parvovirus B19 in blood: viruses or DNA remnants? J Clin Virol 84:19–23. doi: 10.1016/j.jcv.2016.09.004. [DOI] [PubMed] [Google Scholar]
  • 124.Tan SK, Sahoo MK, Milligan S, Taylor N, Pinsky BA. 2017. Stability of Zika virus in urine: specimen processing considerations and implications for the detection of RNA targets in urine. J Virol Methods 248:66–70. doi: 10.1016/j.jviromet.2017.04.018. [DOI] [PubMed] [Google Scholar]
  • 125.Bin Dajem SM, Almushait MA. 2012. Detection of Toxoplasma gondii DNA by PCR in blood samples collected from pregnant Saudi women from the Aseer region, Saudi Arabia. Ann Saudi Med 32:507–512. doi: 10.5144/0256-4947.2012.14.7.1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Turcekova L, Spisak F, Dubinsky P, Ostro A. 2012. Molecular diagnosis of Toxoplasma gondii in pregnant women. Bratisl Lek Listy 113:307–310. doi: 10.4149/BLL_2012_071. [DOI] [PubMed] [Google Scholar]
  • 127.Revello MG, Furione M, Rognoni V, Arossa A, Gerna G. 2014. Cytomegalovirus DNAemia in pregnant women. J Clin Virol 61:590–592. doi: 10.1016/j.jcv.2014.10.002. [DOI] [PubMed] [Google Scholar]
  • 128.Simonazzi G, Cervi F, Zavatta A, Pellizzoni L, Guerra B, Mastroroberto M, Morselli-Labate AM, Gabrielli L, Rizzo N, Lazzarotto T. 2017. Congenital cytomegalovirus infection. Prognostic value of maternal DNAemia at amniocentesis. Clin Infect Dis 64:207–210. doi: 10.1093/cid/ciw700. [DOI] [PubMed] [Google Scholar]
  • 129.Zavattoni M, Furione M, Lanzarini P, Arossa A, Rustico M, Tassis B, Piralla A, Baldanti F. 2016. Monitoring of human cytomegalovirus DNAemia during primary infection in transmitter and non-transmitter mothers. J Clin Virol 82:89–93. doi: 10.1016/j.jcv.2016.07.005. [DOI] [PubMed] [Google Scholar]
  • 130.Eldar-Yedidia Y, Bar-Meir M, Hillel M, Abitbol G, Broide E, Falk R, Assous M, Schlesinger Y. 2016. Low interferon relative response to cytomegalovirus is associated with low likelihood of intrauterine transmission of the virus. PLoS One 11:e0147883. doi: 10.1371/journal.pone.0147883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Ding ZY, Xu F, Chen DZ, Meng XN, Xu TS, Lu MD, Zhuge HX. 2015. A multifactorial analysis of the pregnancy outcomes in cytomegalovirus-infected women. Gynecol Obstet Invest 80:106–112. doi: 10.1159/000370333. [DOI] [PubMed] [Google Scholar]
  • 132.Wujcicka W, Wilczyński J, Nowakowska D. 2014. Do the placental barrier, parasite genotype and Toll-like receptor polymorphisms contribute to the course of primary infection with various Toxoplasma gondii genotypes in pregnant women? Eur J Clin Microbiol Infect Dis 33:703–709. doi: 10.1007/s10096-013-2017-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Curti SP, Figueiredo CA, de Oliveira MI, Andrade JQ, Zugaib M, Frugis Yu AL, Oliveira DB, Durigon EL. 2013. Molecular epidemiology of rubella viruses involved in congenital rubella infections in São Paulo, Brazil, between 1996 and 2009. J Med Virol 85:2034–2041. doi: 10.1002/jmv.23675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Ferreira IMR, Vidal JE, de Mattos C. d C B, de Mattos LC, Qu D, Su C, Pereira-Chioccola VL. 2011. Toxoplasma gondii isolates: multilocus RFLP-PCR genotyping from human patients in Sao Paulo State, Brazil identified distinct genotypes. Exp Parasitol 129:190–195. doi: 10.1016/j.exppara.2011.06.002. [DOI] [PubMed] [Google Scholar]
  • 135.Lilleri D, Zelini P, Fornara C, Comolli G, Revello MG, Gerna G. 2009. Human cytomegalovirus-specific CD4+ and CD8+ T cell responses in primary infection of the immunocompetent and the immunocompromised host. Clin Immunol 131:395–403. doi: 10.1016/j.clim.2009.02.002. [DOI] [PubMed] [Google Scholar]
  • 136.Halai UA, Nielsen-Saines K, Moreira ML, de Sequeira PC, Junior J, de Araujo Zin A, Cherry J, Gabaglia CR, Gaw SL, Adachi K, Tsui I, Pilotto JH, Nogueira RR, de Filippis A, Brasil P. 2017. Maternal Zika virus disease severity, virus load, prior dengue antibodies, and their relationship to birth outcomes. Clin Infect Dis 65:877–883. doi: 10.1093/cid/cix472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Zhu Z, Xu W, Abernathy ES, Chen MH, Zheng Q, Wang T, Zhang Z, Li C, Wang C, He W, Zhou S, Icenogle J. 2007. Comparison of four methods using throat swabs to confirm rubella virus infection. J Clin Microbiol 45:2847–2852. doi: 10.1128/JCM.00289-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Okamoto K, Fujii K, Komase K. 2010. Development of a novel TaqMan real-time PCR assay for detecting rubella virus RNA. J Virol Methods 168:267–271. doi: 10.1016/j.jviromet.2010.05.016. [DOI] [PubMed] [Google Scholar]
  • 139.Lustig Y, Cotar AI, Ceianu CS, Castilletti C, Zelena H, Burdino E, van Tienen C, Avsic T, Aarons E, Reusken C. 2018. Lack of Zika virus antibody response in confirmed patients in non-endemic countries. J Clin Virol 99–100:31–34. doi: 10.1016/j.jcv.2017.12.007. [DOI] [PubMed] [Google Scholar]
  • 140.Toppinen M, Norja P, Aaltonen L-M, Wessberg S, Hedman L, Söderlund-Venermo M, Hedman K. 2015. A new quantitative PCR for human parvovirus B19 genotypes. J Virol Methods 218:40–45. doi: 10.1016/j.jviromet.2015.03.006. [DOI] [PubMed] [Google Scholar]
  • 141.Abo H, Okamoto K, Anraku M, Otsuki N, Sakata M, Icenogle J, Zheng Q, Kurata T, Kase T, Komase K, Takeda M, Mori Y. 2014. Development of an improved RT-LAMP assay for detection of currently circulating rubella viruses. J Virol Methods 207:73–77. doi: 10.1016/j.jviromet.2014.06.013. [DOI] [PubMed] [Google Scholar]
  • 142.Tran DN, Pham NT, Tran TT, Khamrin P, Thongprachum A, Komase K, Hayakawa S, Mizuguchi M, Ushijima H. 2012. Phylogenetic analysis of rubella viruses in Vietnam during 2009–2010. J Med Virol 84:705–710. doi: 10.1002/jmv.23199. [DOI] [PubMed] [Google Scholar]
  • 143.Rodrigues JP, Andrade HF. Jr, 2015. Efficient duplex solid-phase fluorescent assay (dFISA) for the simultaneous detection of specific anti-T. gondii IgG and IgM due to refined conjugates. J Immunol Methods 420:11–17. doi: 10.1016/j.jim.2015.03.007. [DOI] [PubMed] [Google Scholar]
  • 144.De Carolis S, Santucci S, Botta A, Salvi S, Degennaro VA, Garufi C, Garofalo S, Ferrazzani S, Scambia G. 2012. The relationship between TORCH complex false positivity and obstetric outcome in patients with antiphospholipid syndrome. Lupus 21:773–775. doi: 10.1177/0961203312447866. [DOI] [PubMed] [Google Scholar]
  • 145.Yoshida M, Matsuda H, Yoshinaga Y, Asai K, Kawashima A, Sei K, Furuya K. 2013. Can measurement of maternal anti-cytomegalovirus immunoglobulin-M antibody levels be used to screen for cytomegalovirus infection in embryos and fetuses? J Obstet Gynaecol Res 39:166–169. doi: 10.1111/j.1447-0756.2012.01900.x. [DOI] [PubMed] [Google Scholar]
  • 146.Bredl S, Plentz A, Wenzel JJ, Pfister H, Möst J, Modrow S. 2011. False-negative serology in patients with acute parvovirus B19 infection. J Clin Virol 51:115–120. doi: 10.1016/j.jcv.2011.03.012. [DOI] [PubMed] [Google Scholar]
  • 147.Kadkhoda K, Gretchen A, Racano A. 2017. Evaluation of a commercially available Zika virus IgM ELISA: specificity in focus. Diagn Microbiol Infect Dis 88:233–235. doi: 10.1016/j.diagmicrobio.2017.04.002. [DOI] [PubMed] [Google Scholar]
  • 148.Bal TA, Armstrong G, Han XY. 2012. Evaluation of the IMMULITE 2000 CMV IgM assay. Herpesviridae 3:2. doi: 10.1186/2042-4280-3-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Medici MC, Martinelli M, Albonetti V, Chezzi C, Dettori G. 2008. Evaluation of rubella virus immunoglobulin G (IgG) and IgM assays with the new Vidia instrument. J Clin Microbiol 46:1847–1849. doi: 10.1128/JCM.00468-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Golkar M, Azadmanesh K, Khalili G, Khoshkholgh-Sima B, Babaie J, Mercier C, Brenier-Pinchart MP, Fricker-Hidalgo H, Pelloux H, Cesbron-Delauw MF. 2008. Serodiagnosis of recently acquired Toxoplasma gondii infection in pregnant women using enzyme-linked immunosorbent assays with a recombinant dense granule GRA6 protein. Diagn Microbiol Infect Dis 61:31–39. doi: 10.1016/j.diagmicrobio.2007.09.003. [DOI] [PubMed] [Google Scholar]
  • 151.Hamkar R, Jalilvand S, Abdolbaghi MH, Jelyani KN, Esteghamati A, Hagh-Goo A, Mohktari-Azad T, Nategh R. 2009. Distinguishing between primary infection and reinfection with rubella vaccine virus by IgG avidity assay in pregnant women. East Mediterr Health J 15:94–103. doi: 10.26719/2009.15.1.94. [DOI] [PubMed] [Google Scholar]
  • 152.Lutsenko MT, Andrievskaya IA. 2014. Activity of histidine in peripheral blood erythrocytes of pregnant women during exacerbation of cytomegalovirus infection. Bull Exp Biol Med 157:765–768. doi: 10.1007/s10517-014-2662-2. [DOI] [PubMed] [Google Scholar]
  • 153.Alver O, Göral G, Ercan İ. 2014. Investigation of serological results of patients with suspected toxoplasmosis admitted to the ELISA laboratory of Uludağ University Hospital between 2002–2008. Turkiye Parazitol Derg 38:141–146. doi: 10.5152/tpd.2014.3350. [DOI] [PubMed] [Google Scholar]
  • 154.Yamada H, Nishikawa A, Yamamoto T, Mizue Y, Yamada T, Morizane M, Tairaku S, Nishihira J. 2011. Prospective study of congenital toxoplasmosis screening with use of IgG avidity and multiplex nested PCR methods. J Clin Microbiol 49:2552–2556. doi: 10.1128/JCM.02092-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Kamel N, Metwally L, Gomaa N, Sayed Ahmed WA, Lotfi M, Younis S. 2014. Primary cytomegalovirus infection in pregnant Egyptian women confirmed by cytomegalovirus IgG avidity testing. Med Princ Pract 23:29–33. doi: 10.1159/000354758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Seo S, Cho Y, Park J. 2009. Serologic screening of pregnant Korean women for primary human cytomegalovirus infection using IgG avidity test. Korean J Lab Med 29:557–562. doi: 10.3343/kjlm.2009.29.6.557. [DOI] [PubMed] [Google Scholar]
  • 157.Berger A, Reitter A, Harter PN, Buxmann H, Allwinn R, Louwen F, Doerr HW. 2011. Problems and challenges in the diagnosis of vertical infection with human cytomegalovirus (CMV): lessons from two accidental cases. J Clin Virol 51:285–288. doi: 10.1016/j.jcv.2011.05.007. [DOI] [PubMed] [Google Scholar]
  • 158.Nozawa N, Fang-Hoover J, Tabata T, Maidji E, Pereira L. 2009. Cytomegalovirus-specific, high-avidity IgG with neutralizing activity in maternal circulation enriched in the fetal bloodstream. J Clin Virol 46(Suppl 4):S58–S63. doi: 10.1016/j.jcv.2009.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Kaneko M, Ohhashi M, Minematsu T, Muraoka J, Kusumoto K, Sameshima H. 2017. Maternal immunoglobulin G avidity as a diagnostic tool to identify pregnant women at risk of congenital cytomegalovirus infection. J Infect Chemother 23:173–176. doi: 10.1016/j.jiac.2016.12.001. [DOI] [PubMed] [Google Scholar]
  • 160.Hashoosh DA, Majeed IA. 2014. Comparison of two assays in the diagnosis of toxoplasmosis: immunological and molecular. East Mediterr Health J 20:46–50. doi: 10.26719/2014.20.1.46. [DOI] [PubMed] [Google Scholar]
  • 161.De Paschale M, Agrappi C, Belvisi L, Cagnin D, Cerulli T, Clerici P, Mirri P, Manco MT, Cavallari S, Viganò EF. 2008. Revision of the positive predictive value of IgM anti-Toxoplasma antibodies as an index of recent infection. New Microbiol 31:105–111. [PubMed] [Google Scholar]
  • 162.Trotta M, Borchi B, Zammarchi L, Sterrantino G, Brogi M, Kiros ST, Lorini C, Bonaccorsi G, Colao MG, Bartoloni A. 2016. Immunoglobulin M indirect fluorescent antibody test for the diagnosis of acute toxoplasmosis during pregnancy in the avidity era. A 14-year experience. J Obstet Gynaecol Res 42:1699–1703. doi: 10.1111/jog.13144. [DOI] [PubMed] [Google Scholar]
  • 163.Emelia O, Rahana AR, Mohamad Firdaus A, Cheng HS, Nursyairah MS, Fatinah AS, Azmawati MN, Siti NA, Aisah MY. 2014. IgG avidity assay: a tool for excluding acute toxoplasmosis in prolonged IgM titer sera from pregnant women. Trop Biomed 31:633–640. [PubMed] [Google Scholar]
  • 164.Findal G, Stray-Pedersen B, Holter EK, Berge T, Jenum PA. 2015. Persistent low Toxoplasma IgG avidity is common in pregnancy: experience from antenatal testing in Norway. PLoS One 10:e0145519. doi: 10.1371/journal.pone.0145519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Bobic B, Klun I, Vujanic M, Nikolic A, Ivovic V, Zivkovic T, Djurkovic-Djakovic O. 2009. Comparative evaluation of three commercial Toxoplasma-specific IgG antibody avidity tests and significance in different clinical settings. J Med Microbiol 58:358–364. doi: 10.1099/jmm.0.006668-0. [DOI] [PubMed] [Google Scholar]
  • 166.Sellier Y, Guilleminot T, Ville Y, Leruez-Ville M. 2015. Comparison of the LIAISON CMV IgG Avidity II and the VIDAS CMV IgG Avidity II assays for the diagnosis of primary infection in pregnant women. J Clin Virol 72:46–48. doi: 10.1016/j.jcv.2015.08.018. [DOI] [PubMed] [Google Scholar]
  • 167.Furione M, Rognoni V, Sarasini A, Zavattoni M, Lilleri D, Gerna G, Revello MG. 2013. Slow increase in IgG avidity correlates with prevention of human cytomegalovirus transmission to the fetus. J Med Virol 85:1960–1967. doi: 10.1002/jmv.23691. [DOI] [PubMed] [Google Scholar]
  • 168.Ebina Y, Minematsu T, Morioka I, Deguchi M, Tairaku S, Tanimura K, Sonoyama A, Nagamata S, Morizane M, Yamada H. 2015. Rapid increase in the serum cytomegalovirus IgG avidity index in women with a congenitally infected fetus. J Clin Virol 66:44–47. doi: 10.1016/j.jcv.2015.03.002. [DOI] [PubMed] [Google Scholar]
  • 169.Lazzarotto T, Guerra B, Lanari M, Gabrielli L, Landini MP. 2008. New advances in the diagnosis of congenital cytomegalovirus infection. J Clin Virol 41:192–197. doi: 10.1016/j.jcv.2007.10.015. [DOI] [PubMed] [Google Scholar]
  • 170.Elyasi H, Babaie J, Fricker-Hidalgo H, Brenier-Pinchart MP, Zare M, Sadeghiani G, Assmar M, Pelloux H, Golkar M. 2010. Use of dense granule antigen GRA6 in an immunoglobulin G avidity test to exclude acute Toxoplasma gondii infection during pregnancy. Clin Vaccine Immunol 17:1349–1355. doi: 10.1128/CVI.00199-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Smets A, Fauchier T, Michel G, Marty P, Pomares C. 2016. Comparison of Toxoplasma gondii IgG avidity Architect and Vidas assays with the estimated date of infection in pregnant women. Parasite 23:45. doi: 10.1051/parasite/2016056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Vauloup-Fellous C, Lazzarotto T, Revello MG, Grangeot-Keros L. 2014. Clinical evaluation of the Roche Elecsys CMV IgG Avidity assay. Eur J Clin Microbiol Infect Dis 33:1365–1369. doi: 10.1007/s10096-014-2080-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Rajasekariah H, Scott G, Robertson PW, Rawlinson WD. 2013. Improving diagnosis of primary cytomegalovirus infection in pregnant women using immunoblots. J Med Virol 85:315–319. doi: 10.1002/jmv.23471. [DOI] [PubMed] [Google Scholar]
  • 174.Flori P, Bellete B, Crampe C, Maudry A, Patural H, Chauleur C, Hafid J, Raberin H, Tran Manh Sung R. 2008. A technique for dating toxoplasmosis in pregnancy and comparison with the Vidas anti-toxoplasma IgG avidity test. Clin Microbiol Infect 14:242–249. doi: 10.1111/j.1469-0691.2007.01905.x. [DOI] [PubMed] [Google Scholar]
  • 175.Khammari I, Saghrouni F, Lakhal S, Bouratbine A, Ben Said M, Boukadida J. 2014. A new IgG immunoblot kit for diagnosis of toxoplasmosis in pregnant women. Korean J Parasitol 52:493–499. doi: 10.3347/kjp.2014.52.5.493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Palmer P, Pallier C, Leruez-Ville M, Deplanche M, Morinet F. 1996. Antibody response to human parvovirus B19 in patients with primary infection by immunoblot assay with recombinant proteins. Clin Diagn Lab Immunol 3:236–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Khammari I, Saghrouni F, Yaacoub A, Gaied Meksi S, Ach H, Garma L, Fathallah A, Ben Saïd M. 2013. IgG western blot for confirmatory diagnosis of equivocal cases of toxoplasmosis by EIA-IgG and fluorescent antibody test. Korean J Parasitol 51:485–488. doi: 10.3347/kjp.2013.51.4.485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Eggers M, Radsak K, Enders G, Reschke M. 2001. Use of recombinant glycoprotein antigens gB and gH for diagnosis of primary human cytomegalovirus infection during pregnancy. J Med Virol 63:135–142. doi:. [DOI] [PubMed] [Google Scholar]
  • 179.Sroka J, Wójcik-Fatla A, Zając V, Sawczyn A, Cisak E, Karamon J, Dutkiewicz J, Bojar I. 2016. Comparison of the efficiency of two commercial kits—ELFA and Western blot in estimating the phase of Toxoplasma gondii infection in pregnant women. Ann Agric Environ Med 23:570–575. doi: 10.5604/12321966.1226848. [DOI] [PubMed] [Google Scholar]
  • 180.L'Ollivier C, Wallon M, Faucher B, Piarroux R, Peyron F, Franck J. 2012. Comparison of mother and child antibodies that target high-molecular-mass Toxoplasma gondii antigens by immunoblotting improves neonatal diagnosis of congenital toxoplasmosis. Clin Vaccine Immunol 19:1326–1328. doi: 10.1128/CVI.00060-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Vauloup-Fellous C, Berth M, Heskia F, Dugua JM, Grangeot-Keros L. 2013. Re-evaluation of the VIDAS cytomegalovirus (CMV) IgG avidity assay: determination of new cut-off values based on the study of kinetics of CMV-IgG maturation. J Clin Virol 56:118–123. doi: 10.1016/j.jcv.2012.10.017. [DOI] [PubMed] [Google Scholar]
  • 182.Villard O, Breit L, Cimon B, Franck J, Fricker-Hidalgo H, Godineau N, Houze S, Paris L, Pelloux H, Villena I, Candolfi E, French National Reference Center for Toxoplasmosis Network. 2013. Comparison of four commercially available avidity tests for Toxoplasma gondii-specific IgG antibodies. Clin Vaccine Immunol 20:197–204. doi: 10.1128/CVI.00356-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Jost C, Touafek F, Fekkar A, Courtin R, Ribeiro M, Mazier D, Paris L. 2011. Utility of immunoblotting for early diagnosis of toxoplasmosis seroconversion in pregnant women. Clin Vaccine Immunol 18:1908–1912. doi: 10.1128/CVI.05303-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Shan C, Xie X, Ren P, Loeffelholz MJ, Yang Y, Furuya A, Dupuis AP, Kramer LD, Wong SJ, Shi PY. 2017. A rapid Zika diagnostic assay to measure neutralizing antibodies in patients. EBioMed 17:157–162. doi: 10.1016/j.ebiom.2017.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Revello MG, Genini E, Gorini G, Klersy C, Piralla A, Gerna G. 2010. Comparative evaluation of eight commercial human cytomegalovirus IgG avidity assays. J Clin Virol 48:255–259. doi: 10.1016/j.jcv.2010.05.004. [DOI] [PubMed] [Google Scholar]
  • 186.Bouthry E, Furione M, Huzly D, Ogee-Nwankwo A, Hao L, Adebayo A, Icenogle J, Sarasini A, Revello MG, Grangeot-Keros L, Vauloup-Fellous C. 2016. Assessing immunity to rubella virus: a plea for standardization of IgG (immuno)assays. J Clin Microbiol 54:1720–1725. doi: 10.1128/JCM.00383-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Lumley S, Patel M, Griffiths PD. 2014. The combination of specific IgM antibodies and IgG antibodies of low avidity does not always indicate primary infection with cytomegalovirus. J Med Virol 86:834–837. doi: 10.1002/jmv.23863. [DOI] [PubMed] [Google Scholar]
  • 188.Sickinger E, Gay-Andrieu F, Jonas G, Schultess J, Stieler M, Smith D, Hausmann M, Stricker R, Stricker R, Dhein J, Braun HB. 2008. Performance characteristics of the new ARCHITECT Toxo IgG and Toxo IgG avidity assays. Diagn Microbiol Infect Dis 62:235–244. doi: 10.1016/j.diagmicrobio.2008.07.005. [DOI] [PubMed] [Google Scholar]
  • 189.Sickinger E, Braun HB, Praast G, Stieler M, Gundlach C, Birkenbach C, Prostko J, Palafox MA, Frias E, Hsu S, Matias M, Pucci D, Hausmann M, Sagel U, Smith D. 2009. Evaluation of the Abbott ARCHITECT Toxo IgM assay. Diagn Microbiol Infect Dis 64:275–282. doi: 10.1016/j.diagmicrobio.2009.03.016. [DOI] [PubMed] [Google Scholar]
  • 190.Lagrou K, Bodeus M, Van Ranst M, Goubau P. 2009. Evaluation of the new architect cytomegalovirus immunoglobulin M (IgM), IgG, and IgG avidity assays. J Clin Microbiol 47:1695–1699. doi: 10.1128/JCM.02172-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Lachaud L, Calas O, Picot MC, Albaba S, Bourgeois N, Pratlong F. 2009. Value of 2 IgG avidity commercial tests used alone or in association to date toxoplasmosis contamination. Diagn Microbiol Infect Dis 64:267–274. doi: 10.1016/j.diagmicrobio.2009.02.014. [DOI] [PubMed] [Google Scholar]
  • 192.Rahbar N, Vali Zadeh S, Ghorbani R, Kheradmand P. 2015. Prevalence of parvovirus B19 specific antibody in pregnant women with spontaneous abortion. Acta Med Iran 53:168–172. [PubMed] [Google Scholar]
  • 193.Fricker-Hidalgo H, Cimon B, Chemla C, Darde ML, Delhaes L, L'ollivier C, Godineau N, Houze S, Paris L, Quinio D, Robert-Gangneux F, Villard O, Villena I, Candolfi E, Pelloux H. 2013. Toxoplasma seroconversion with negative or transient immunoglobulin M in pregnant women: myth or reality? A French multicenter retrospective study. J Clin Microbiol 51:2103–2111. doi: 10.1128/JCM.00169-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Carlier P, Harika N, Bailly R, Vranken G. 2010. Laboratory evaluation of the new Access cytomegalovirus immunoglobulin IgM and IgG assays. J Clin Virol 49:192–197. doi: 10.1016/j.jcv.2010.07.024. [DOI] [PubMed] [Google Scholar]
  • 195.Maudry A, Chene G, Chatelain R, Patural H, Bellete B, Tisseur B, Hafid J, Raberin H, Beretta S, Sung RT, Belot G, Flori P. 2009. Bicentric evaluation of six anti-toxoplasma immunoglobulin G (IgG) automated immunoassays and comparison to the Toxo II IgG Western blot. Clin Vaccine Immunol 16:1322–1326. doi: 10.1128/CVI.00128-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Gentile M, Galli C, Pagnotti P, Di Marco P, Tzantzoglou S, Bellomi F, Ferreri ML, Selvaggi C, Antonelli G. 2009. Measurement of the sensitivity of different commercial assays in the diagnosis of CMV infection in pregnancy. Eur J Clin Microbiol Infect Dis 28:977–981. doi: 10.1007/s10096-009-0738-0. [DOI] [PubMed] [Google Scholar]
  • 197.Enders M, Helbig S, Hunjet A, Pfister H, Reichhuber C, Motz M. 2007. Comparative evaluation of two commercial enzyme immunoassays for serodiagnosis of human parvovirus B19 infection. J Virol Methods 146:409–413. doi: 10.1016/j.jviromet.2007.08.008. [DOI] [PubMed] [Google Scholar]
  • 198.Ghoneim NH, Shalaby SI, Hassanain NA, Zeedan GS, Soliman YA, Abdalhamed AM. 2010. Comparative study between serological and molecular methods for diagnosis of toxoplasmosis in women and small ruminants in Egypt. Foodborne Pathog Dis 7:17–22. doi: 10.1089/fpd.2008.0223. [DOI] [PubMed] [Google Scholar]
  • 199.Kasper DC, Prusa AR, Hayde M, Gerstl N, Pollak A, Herkner KR, Reiter-Reisacher R. 2009. Evaluation of the Vitros ECiQ immunodiagnostic system for detection of anti-Toxoplasma immunoglobulin G and immunoglobulin M antibodies for confirmatory testing for acute Toxoplasma gondii infection in pregnant women. J Clin Microbiol 47:164–167. doi: 10.1128/JCM.01435-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Prusa AR, Hayde M, Unterasinger L, Pollak A, Herkner KR, Kasper DC. 2010. Evaluation of the Roche Elecsys Toxo IgG and IgM electrochemiluminescence immunoassay for the detection of gestational Toxoplasma infection. Diagn Microbiol Infect Dis 68:352–357. doi: 10.1016/j.diagmicrobio.2010.07.011. [DOI] [PubMed] [Google Scholar]
  • 201.Franck J, Garin YJ, Dumon H. 2008. LDBio-Toxo II immunoglobulin G Western blot confirmatory test for anti-toxoplasma antibody detection. J Clin Microbiol 46:2334–2338. doi: 10.1128/JCM.00182-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Stowe RP, Ruiz RJ, Fagundes CP, Stowe RH, Chen M, Glaser R. 2014. An ELISA method to compute endpoint titers to Epstein-Barr virus and cytomegalovirus: application to population-based studies. J Immunol Methods 408:64–69. doi: 10.1016/j.jim.2014.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Dimech W, Grangeot-Keros L, Vauloup-Fellous C. 2016. Standardization of assays that detect anti-rubella virus IgG antibodies. Clin Microbiol Rev 29:163. doi: 10.1128/CMR.00045-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Centonze AR, Tonolli E, Fontana R. 2013. Performance characteristics of current-generation Immulite 2000 TORCH Assays. Clin Vaccine Immunol 20:122–126. doi: 10.1128/CVI.00106-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Murat JB, Dard C, Fricker Hidalgo H, Dardé ML, Brenier-Pinchart MP, Pelloux H. 2013. Comparison of the Vidas system and two recent fully automated assays for diagnosis and follow-up of toxoplasmosis in pregnant women and newborns. Clin Vaccine Immunol 20:1203–1212. doi: 10.1128/CVI.00089-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Paschale de M, Agrappi M, Manco MT, Clerici P. 2010. Positive predictive value of anti-HCMV IgM as an index of primary infection. J Virol Methods 168:121–125. doi: 10.1016/j.jviromet.2010.05.001. [DOI] [PubMed] [Google Scholar]
  • 207.Drew RJ, Stapleton P, Abu H, Healy E, Ferguson W, De Gascun C, O'Gorman J, Eogan M. 2015. Pregnancy outcomes of mothers with detectable CMV-specific IgM antibodies: a three-year review in a large Irish tertiary referral maternity hospital. Infect Dis Obstet Gynecol 2015:218080. doi: 10.1155/2015/218080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Toriyabe K, Morikawa F, Minematsu T, Ikejiri M, Suga S, Ikeda T. 2017. Anti-cytomegalovirus immunoglobulin M titer for congenital infection in first trimester pregnancy with primary infection: a multicenter prospective cohort study. J Perinatol 37:1272–1277. doi: 10.1038/jp.2017.133. [DOI] [PubMed] [Google Scholar]
  • 209.Tanimura K, Nishikawa A, Tairaku S, Shinozaki N, Deguchi M, Morizane M, Ebina Y, Morioka I, Yamada H. 2015. The IgG avidity value for the prediction of Toxoplasma gondii infection in the amniotic fluid. J Infect Chemother 21:668–671. doi: 10.1016/j.jiac.2015.05.013. [DOI] [PubMed] [Google Scholar]
  • 210.Enders M, Daiminger A, Exler S, Ertan K, Enders G, Bald R. 2017. Prenatal diagnosis of congenital cytomegalovirus infection in 115 cases- a 5 years’ single center experience. Prenat Diagn 37:389–398. doi: 10.1002/pd.5025. [DOI] [PubMed] [Google Scholar]
  • 211.Weiffenbach J, Bald R, Gloning KP, Minderer S, Gärtner BC, Weidner A, Hanke M, Enders M. 2012. Serological and virological analysis of maternal and fetal blood samples in prenatal human parvovirus b19 infection. J Infect Dis 205:782–788. doi: 10.1093/infdis/jir855. [DOI] [PubMed] [Google Scholar]
  • 212.Stajner T, Bobic B, Klun I, Nikolic A, Srbljanovic J, Uzelac A, Rajnpreht I, Djurkovic-Djakovic O. 2016. Prenatal and early postnatal diagnosis of congenital toxoplasmosis in a setting with no systematic screening in pregnancy. Medicine (Baltimore) 95:e2979. doi: 10.1097/MD.0000000000002979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Bonvicini F, Manaresi E, Gallinella G, Gentilomi GA, Musiani M, Zerbini M. 2009. Diagnosis of fetal parvovirus B19 infection: value of virological assays in fetal specimens. BJOG 116:813–817. doi: 10.1111/j.1471-0528.2009.02109.x. [DOI] [PubMed] [Google Scholar]
  • 214.Van Le S, Le DH, Hoang HT, Hoang H, Nguyen NT, Chu HH. 2015. Characterization of rubella virus genotypes among pregnant women in northern Vietnam, 2011–2013. J Med Virol 87:338–343. doi: 10.1002/jmv.24049. [DOI] [PubMed] [Google Scholar]
  • 215.Fabbri E, Revello MG, Furione M, Zavattoni M, Lilleri D, Tassis B, Quarenghi A, Rustico M, Nicolini U, Ferrazzi E, Gerna G. 2011. Prognostic markers of symptomatic congenital human cytomegalovirus infection in fetal blood. BJOG 118:448–456. doi: 10.1111/j.1471-0528.2010.02822.x. [DOI] [PubMed] [Google Scholar]
  • 216.Ishikawa A, Yoto Y, Asakura H, Tsutsumi H. 2015. Quantitative analysis of human parvovirus B19 DNA in maternal and fetal serum, and amniotic fluid during an early stage of pregnancy. J Med Virol 87:683–685. doi: 10.1002/jmv.24105. [DOI] [PubMed] [Google Scholar]
  • 217.Chen CP, Su YN, Chern SR, Wang TY, Tsai FJ, Lin HH, Wu PC, Wang W. 2010. Detection and comparison of cytomegalovirus DNA levels in amniotic fluid and fetal ascites in a second-trimester fetus with massive ascites, hyperechogenic bowel, ventriculomegaly and intrauterine growth restriction. Taiwan J Obstet Gyn 49:206–210. doi: 10.1016/S1028-4559(10)60044-7. [DOI] [PubMed] [Google Scholar]
  • 218.Yamamoto L, Targa LS, Sumita LM, Shimokawa PT, Rodrigues JC, Kanunfre KA, Okay TS. 2017. Association of parasite load levels in amniotic fluid with clinical outcome in congenital toxoplasmosis. Obstet Gynecol 130:335–345. doi: 10.1097/AOG.0000000000002131. [DOI] [PubMed] [Google Scholar]
  • 219.Hsu ST, Chen YT, Huang YF, Yeh TT, Chen WC, Ho ES, Chou MM. 2007. Prenatal diagnosis and perinatal management of maternal-fetal congenital parvovirus B19 infection. Taiwan J Obstet Gyn 46:417–422. doi: 10.1016/S1028-4559(08)60014-5. [DOI] [PubMed] [Google Scholar]
  • 220.Yoshida M, Matsuda H, Yoshinaga Y, Asai K, Kawashima A, Sei K, Horii M, Nakanishi A, Soyama H, Furuya K. 2013. Analysis about the influence on the fetus infected with parvovirus B19 using amniotic erythropoietin and troponin-T. Arch Gynecol Obstet 288:521–525. doi: 10.1007/s00404-013-2815-y. [DOI] [PubMed] [Google Scholar]
  • 221.Schiesser M, Sergi C, Enders M, Maul H, Schnitzler P. 2009. Discordant outcomes in a case of parvovirus b19 transmission into both dichorionic twins. Twin Res Hum Genet 12:175–179. doi: 10.1375/twin.12.2.175. [DOI] [PubMed] [Google Scholar]
  • 222.Adams LL, Gungor S, Turan S, Kopelman JN, Harman CR, Baschat AA. 2012. When are amniotic fluid viral PCR studies indicated in prenatal diagnosis? Prenat Diagn 32:88–93. doi: 10.1002/pd.3835. [DOI] [PubMed] [Google Scholar]
  • 223.Gervasi MT, Romero R, Bracalente G, Chaiworapongsa T, Erez O, Dong Z, Hassan SS, Yeo L, Yoon BH, Mor G, Barzon L, Franchin E, Militello V, Palù G. 2012. Viral invasion of the amniotic cavity (VIAC) in the midtrimester of pregnancy. J Matern Fetal Neonatal Med 25:2002–2013. doi: 10.3109/14767058.2012.683899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Zavattoni M, Paolucci S, Sarasini A, Tassis B, Rustico M, Quarenghi A, Piralla A, Baldanti F. 2016. Diagnostic and prognostic value of molecular and serological investigation of human parvovirus B19 infection during pregnancy. New Microbiol 39:181–185. [PubMed] [Google Scholar]
  • 225.Goegebuer T, Van Meensel B, Beuselinck K, Cossey V, Van Ranst M, Hanssens M, Lagrou K. 2009. Clinical predictive value of real-time PCR quantification of human cytomegalovirus DNA in amniotic fluid samples. J Clin Microbiol 47:660–665. doi: 10.1128/JCM.01576-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Prusa AR, Kasper DC, Pollak A, Olischar M, Gleiss A, Hayde M. 2015. Amniocentesis for the detection of congenital toxoplasmosis: results from the nationwide Austrian prenatal screening program. Clin Microbiol Infect 21:191.e1–e8. doi: 10.1016/j.cmi.2014.09.018. [DOI] [PubMed] [Google Scholar]
  • 227.Teixeira LE, Kanunfre KA, Shimokawa PT, Targa LS, Rodrigues JC, Domingues W, Yamamoto L, Okay TS. 2013. The performance of four molecular methods for the laboratory diagnosis of congenital toxoplasmosis in amniotic fluid samples. Rev Soc Bras Med Trop 46:584–588. doi: 10.1590/0037-8682-0095-2013. [DOI] [PubMed] [Google Scholar]
  • 228.Gratzl R, Hayde M, Kohlhauser C, Hermon M, Burda G, Strobl W, Pollak A. 1998. Follow-up of infants with congenital toxoplasmosis detected by PCR analysis of amniotic fluid. Eur J Clin Microbiol Infect Dis 17:853–858. doi: 10.1007/s100960050206. [DOI] [PubMed] [Google Scholar]
  • 229.Desveaux C, Klein J, Leruez-Ville M, Ramirez-Torres A, Lacroix C, Breuil B, Froment C, Bascands JL, Schanstra JP, Ville Y. 2016. Identification of symptomatic fetuses infected with cytomegalovirus using amniotic fluid peptide biomarkers. PLoS Pathog 12:e1005395. doi: 10.1371/journal.ppat.1005395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Costa JM, Alanio A, Moukoury S, Clairet V, Debruyne M, Poveda JD, Bretagne S. 2013. Direct genotyping of Toxoplasma gondii from amniotic fluids based on B1 gene polymorphism using minisequencing analysis. BMC Infect Dis 13:552. doi: 10.1186/1471-2334-13-552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Benjamin I, Fernandez G, Figueira JV, Parpacen L, Urbina MT, Medina R. 2017. Zika virus detected in amniotic fluid and umbilical cord blood in an in vitro fertilization-conceived pregnancy in Venezuela. Fertil Steril 107:1319–1322. doi: 10.1016/j.fertnstert.2017.02.112. [DOI] [PubMed] [Google Scholar]
  • 232.Ornoy A, Ergaz Z. 2017. Parvovirus B19 infection during pregnancy and risks to the fetus. Birth Defects Res 109:311–323. doi: 10.1002/bdra.23588. [DOI] [PubMed] [Google Scholar]
  • 233.Durski KN, Tituli C, Ogaoga D, Musto J, Joshua C, Dofai A, Leydon J, Nilles E. 2016. An outbreak investigation of congenital rubella syndrome in Solomon Islands, 2013. Western Pac Surveill Response J 7:10–13. doi: 10.5365/WPSAR.2015.6.4.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Olariu TR, Remington JS, Montoya JG. 2014. Polymerase chain reaction in cerebrospinal fluid for the diagnosis of congenital toxoplasmosis. Pediatr Infect Dis J 33:566–570. doi: 10.1097/INF.0000000000000256. [DOI] [PubMed] [Google Scholar]
  • 235.Revello MG, Zavattoni M, Baldanti F, Sarasini A, Paolucci S, Gerna G. 1999. Diagnostic and prognostic value of human cytomegalovirus load and IgM antibody in blood of congenitally infected newborns. J Clin Virol 14:57–66. doi: 10.1016/S1386-6532(99)00016-5. [DOI] [PubMed] [Google Scholar]
  • 236.Wilson CB, Remington JS, Stagno S, Reynolds DW. 1980. Development of adverse sequelae in children born with subclinical congenital Toxoplasma infection. Pediatrics 66:767–774. [PubMed] [Google Scholar]
  • 237.Nijman J, de Vries LS, Koopman-Esseboom C, Uiterwaal CS, van Loon AM, Verboon-Maciolek MA. 2012. Postnatally acquired cytomegalovirus infection in preterm infants: a prospective study on risk factors and cranial ultrasound findings. Arch Dis Child Fetal Neonatal ed 97:F259–63. doi: 10.1136/archdischild-2011-300405. [DOI] [PubMed] [Google Scholar]
  • 238.Forner G, Abate D, Mengoli C, Palù G, Gussetti N. 2015. High cytomegalovirus (CMV) DNAemia predicts CMV sequelae in asymptomatic congenitally infected newborns born to women with primary infection during pregnancy. J Infect Dis 212:67–71. doi: 10.1093/infdis/jiu627. [DOI] [PubMed] [Google Scholar]
  • 239.Samedi VM, Skappak C, Jantzie L, Trevenen C, Kamaluddeen M, Ekwalanga P, Al Awad EH. 2016. Comparison of presentation, course, and outcome of congenital and acquired cytomegalovirus infection in twins. AJP Rep 6:e1–e5. doi: 10.1055/s-0035-1563387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Pejcic I, Rankovic Janevski M, Knezevic A, Jevtovic D, Stanojevic M. 2016. Rubella immune status of neonates-a window towards seroprevalence among childbearing women. BMC Public Health 16:838. doi: 10.1186/s12889-016-3514-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Armstrong L, Isaacs D, Evans N. 2004. Severe neonatal toxoplasmosis after third trimester maternal infection. Pediatr Infect Dis J 23:968–969. doi: 10.1097/01.inf.0000137573.08226.55. [DOI] [PubMed] [Google Scholar]
  • 242.Goeyvaerts N, Hens N, Aerts M, Beutels P. 2011. Model structure analysis to estimate basic immunological processes and maternal risk for parvovirus B19. Biostatistics 12:283–302. doi: 10.1093/biostatistics/kxq059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.van den Berg JP, Westerbeek EA, Smits GP, van der Klis FR, Berbers GA, van Elburg RM. 2014. Lower transplacental antibody transport for measles, mumps, rubella and varicella zoster in very preterm infants. PLoS One 9:e94714. doi: 10.1371/journal.pone.0094714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Murat J-B, Hidalgo HF, Brenier-Pinchart M-P, Pelloux H. 2013. Human toxoplasmosis: which biological diagnostic tests are best suited to which clinical situations? Expert Rev Anti Infect Ther 11:943–956. doi: 10.1586/14787210.2013.825441. [DOI] [PubMed] [Google Scholar]
  • 245.Kaneko M, Sameshima H, Minematsu T, Kusumoto K, Yamauchi A, Ikenoue T. 2013. Maternal IgG avidity, IgM and ultrasound abnormalities: combined method to detect congenital cytomegalovirus infection with sequelae. J Perinatol 33:831–835. doi: 10.1038/jp.2013.87. [DOI] [PubMed] [Google Scholar]
  • 246.Marangoni A, Capretti MG, De Angelis M, Nardini P, Compri M, Foschi C, Orlandi A, Marsico C, Righetti F, Faldella G, Cevenini R. 2014. Evaluation of a new protocol for retrospective diagnosis of congenital toxoplasmosis by use of Guthrie cards. J Clin Microbiol 52:2963–2970. doi: 10.1128/JCM.00106-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Tridapalli E, Capretti MG, Farneti G, Marangoni A, Cevenini R, Faldella G. 2008. Congenital toxoplasmosis: the importance of the western blot method to avoid unnecessary therapy in potentially infected newborns. Acta Paediatr 97:1298–1300. doi: 10.1111/j.1651-2227.2008.00905.x. [DOI] [PubMed] [Google Scholar]
  • 248.Fonseca ZC, Rodrigues I, Melo N, Avelar JB, Castro AM, Avelino MM. 2017. IgG avidity test in congenital toxoplasmosis diagnoses in newborns. Pathogens 6:26. doi: 10.3390/pathogens6020026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Chumpitazi BF, Boussaid A, Pelloux H, Racinet C, Bost M, Goullier-Fleuret A. 1995. Diagnosis of congenital toxoplasmosis by immunoblotting and relationship with other methods. J Clin Microbiol 33:1479–1485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Marchant E, Bishop L, Flaxman D, Jagodzinski J, Nanjundappa M, Muniyappa P, Cordery R. 2016. A case of congenital rubella syndrome and infection in South-East London in 2015: prevention, diagnosis, and the public health response. Br J Gen Pract 66:635–636. doi: 10.3399/bjgp16X688321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Thomas HI, Morgan-Capner P, Cradock-Watson JE, Enders G, Best JM, O'Shea S. 1993. Slow maturation of IgG1 avidity and persistence of specific IgM in congenital rubella: implications for diagnosis and immunopathology. J Med Virol 41:196–200. doi: 10.1002/jmv.1890410305. [DOI] [PubMed] [Google Scholar]
  • 252.Ligozzi M, Poggi M, Saletti M, Gibellini D. 2016. Development of a real-time quantitative polymerase chain reaction assay for the detection of congenital human cytomegalovirus infection in urine samples. Mol Cell Probes 30:50–52. doi: 10.1016/j.mcp.2015.11.006. [DOI] [PubMed] [Google Scholar]
  • 253.Sugishita Y, Akiba T, Sumitomo M, Hayata N, Hasegawa M, Tsunoda T, Okazaki T, Murauchi K, Hayashi Y, Kai A, Seki N, Kayebeta A, Iwashita Y, Kurita M, Tahara N. 2016. Shedding of rubella virus among infants with congenital rubella syndrome born in Tokyo, Japan, 2013–2014. Jpn J Infect Dis 69:418–423. doi: 10.7883/yoken.JJID.2015.316. [DOI] [PubMed] [Google Scholar]
  • 254.Cannon MJ, Hyde TB, Schmid DS. 2011. Review of cytomegalovirus shedding in bodily fluids and relevance to congenital cytomegalovirus infection. Rev Med Virol 21:240–255. doi: 10.1002/rmv.695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Jin L, Thomas B. 2007. Application of molecular and serological assays to case based investigations of rubella and congenital rubella syndrome. J Med Virol 79:1017–1024. doi: 10.1002/jmv.20847. [DOI] [PubMed] [Google Scholar]
  • 256.Nijman J, Mandemaker FS, Verboon-Maciolek MA, Aitken SC, van Loon AM, de Vries LS, Schuurman R. 2014. Genotype distribution, viral load and clinical characteristics of infants with postnatal or congenital cytomegalovirus infection. PLoS One 9:e108018. doi: 10.1371/journal.pone.0108018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Sun XM, Ji YS, Elashram SA, Lu ZM, Liu XY, Suo X, Chen QJ, Wang H. 2012. Identification of antigenic proteins of Toxoplasma gondii RH strain recognized by human immunoglobulin G using immunoproteomics. J Proteomics 77:423–432. doi: 10.1016/j.jprot.2012.09.018. [DOI] [PubMed] [Google Scholar]
  • 258.Baschirotto PT, Krieger MA, Foti L. 2017. Preliminary multiplex microarray IgG immunoassay for the diagnosis of toxoplasmosis and rubella. Mem Inst Oswaldo Cruz 112:428–436. doi: 10.1590/0074-02760160509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Wang Y, Hedman L, Perdomo MF, Elfaitouri A, Bölin-Wiener A, Kumar A, Lappalainen M, Söderlund-Venermo M, Blomberg J, Hedman K. 2016. Microsphere-based antibody assays for human parvovirus B19V, CMV and T. gondii. BMC Infect Dis 16:8. doi: 10.1186/s12879-015-1194-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Kotresha D, Noordin R. 2010. Recombinant proteins in the diagnosis of toxoplasmosis. APMIS 118:529–542. doi: 10.1111/j.1600-0463.2010.02629.x. [DOI] [PubMed] [Google Scholar]
  • 261.Dai JF, Jiang M, Qu LL, Sun L, Wang YY, Gong LL, Gong RJ, Si J. 2013. Toxoplasma gondii: enzyme-linked immunosorbent assay based on a recombinant multi-epitope peptide for distinguishing recent from past infection in human. Exp Parasitol 133:95–100. doi: 10.1016/j.exppara.2012.10.016. [DOI] [PubMed] [Google Scholar]
  • 262.Liu L, Liu T, Yu L, Cai Y, Zhang A, Xu X, Luo Q, Hu Y, Song W, Lun Z, Lu F, Wang Y, Shen J. 2012. rROP2(186–533): a novel peptide antigen for detection of IgM antibodies against Toxoplasma gondii. Foodborne Pathog Dis 9:7–12. doi: 10.1089/fpd.2011.0942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Costa JG, Peretti LE, García VS, Peverengo L, González VD, Gugliotta LM, Dalla Fontana ML, Lagier CM, Marcipar IS. 2017. P35 and P22 Toxoplasma gondii antigens abbreviate regions to diagnose acquired toxoplasmosis during pregnancy: toward single-sample assays. Clin Chem Lab Med 55:595–604. doi: 10.1515/cclm-2016-0331. [DOI] [PubMed] [Google Scholar]
  • 264.Lilleri D, Gerna G, Furione M, Zavattoni M, Spinillo A. 2016. Neutralizing and ELISA IgG antibodies to human cytomegalovirus glycoprotein complexes may help date the onset of primary infection in pregnancy. J Clin Virol 81:16–24. doi: 10.1016/j.jcv.2016.05.007. [DOI] [PubMed] [Google Scholar]
  • 265.Lilleri D, Kabanova A, Revello MG, Percivalle E, Sarasini A, Genini E, Sallusto F, Lanzavecchia A, Corti D, Gerna G. 2013. Fetal human cytomegalovirus transmission correlates with delayed maternal antibodies to gH/gL/pUL128-130-131 complex during primary infection. PLoS One 8:e59863. doi: 10.1371/journal.pone.0059863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Teh AY, Amerizadeh A, Osman S, Yunus MH, Noordin R. 2016. Identification, production and assessment of two Toxoplasma gondii recombinant proteins for use in a Toxoplasma IgG avidity assay. Pathog Glob Health 110:277–286. doi: 10.1080/20477724.2016.1238186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.de Souza Macre M, Pires M, Meireles LR, Angel SO, de Andrade HF Jr. 2009. Serology using rROP2 antigen in the diagnostic of toxoplasmosis in pregnant women. Rev Inst Med Trop Sao Paulo 51:283–288. doi: 10.1590/s0036-46652009000500009. [DOI] [PubMed] [Google Scholar]
  • 268.Petrova EK, Dmitrieva AA, Trifonova EA, Nikitin NA, Karpova OV. 2016. The key role of rubella virus glycoproteins in the formation of immune response, and perspectives on their use in the development of new recombinant vaccines. Vaccine 34:1006–1011. doi: 10.1016/j.vaccine.2016.01.010. [DOI] [PubMed] [Google Scholar]
  • 269.Ardizzoni A, Capuccini B, Baschieri MC, Orsi CF, Rumpianesi F, Peppoloni S, Cermelli C, Meacci M, Crisanti A, Steensgaard P, Blasi E. 2009. A protein microarray immunoassay for the serological evaluation of the antibody response in vertically transmitted infections. Eur J Clin Microbiol Infect Dis 28:1067–1075. doi: 10.1007/s10096-009-0748-y. [DOI] [PubMed] [Google Scholar]
  • 270.Madi JM, Souza R. d S d, Araújo B. F d, Oliveira Filho P F d, Rombaldi RL, Mitchell C, Lorencetti J, Marcon NO. 2010. Prevalence of toxoplasmosis, HIV, syphilis and rubella in a population of puerperal women using Whatman 903 filter paper. Braz J Infect Dis 14:24–29. doi: 10.1016/S1413-8670(10)70006-4. [DOI] [PubMed] [Google Scholar]
  • 271.Cañedo-Solares I, Ortiz-Alegría LB, Figueroa-Damián R, Bustos-Bahena ML, González-Henkel H, Calderón-Segura E, Luna-Pastén H, Correa D. 2009. Toxoplasmosis in pregnancy: determination of IgM, IgG and avidity in filter paper-embedded blood. J Perinatol 29:668–672. doi: 10.1038/jp.2009.79. [DOI] [PubMed] [Google Scholar]
  • 272.Boa-Sorte N, Purificação A, Amorim T, Assunção L, Reis A, Galvão-Castro B. 2014. Dried blood spot testing for the antenatal screening of HTLV, HIV, syphilis, toxoplasmosis and hepatitis B and C: prevalence, accuracy and operational aspects. Braz J Infect Dis 18:618–624. doi: 10.1016/j.bjid.2014.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Augustine S. 2016. Towards universal screening for toxoplasmosis- rapid, cost-effective, and simultaneous detection of anti-toxoplasma IgG, IgM, and IgA antibodies by use of very small serum volumes. J Clin Microbiol 54:1684–1685. doi: 10.1128/JCM.00913-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Li X, Pomares C, Gonfrier G, Koh B, Zhu S, Gong M, Montoya JG, Dai H. 2016. Multiplexed anti-toxoplasma IgG, IgM, and IgA assay on plasmonic gold chips: towards making mass screening possible with dye test precision. J Clin Microbiol 54:1726–1733. doi: 10.1128/JCM.03371-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Pomares C, Zhang B, Arulkumar S, Gonfrier G, Marty P, Zhao S, Cheng S, Tang M, Dai H, Montoya JG. 2017. Validation of IgG, IgM multiplex plasmonic gold platform in French clinical cohorts for the serodiagnosis and follow-up of Toxoplasma gondii infection. Diagn Microbiol Infect Dis 87:213–218. doi: 10.1016/j.diagmicrobio.2016.09.001. [DOI] [PubMed] [Google Scholar]
  • 276.Fornara C, Cassaniti I, Zavattoni M, Furione M, Adzasehoun KMG, De Silvestri A, Comolli G, Baldanti F. 2017. Human cytomegalovirus-specific memory CD4+ T-cell response and its correlation with virus transmission to the fetus in pregnant women with primary infection. Clin Infect Dis 65:1659–1665. doi: 10.1093/cid/cix622. [DOI] [PubMed] [Google Scholar]
  • 277.Mele F, Fornara C, Jarrossay D, Furione M, Arossa A, Spinillo A, Lanzavecchia A, Gerna G, Sallusto F, Lilleri D. 2017. Phenotype and specificity of T cells in primary human cytomegalovirus infection during pregnancy: IL-7Rpos long-term memory phenotype is associated with protection from vertical transmission. PLoS One 12:e0187731. doi: 10.1371/journal.pone.0187731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Lilleri D, Fornara C, Revello MG, Gerna G. 2008. Human cytomegalovirus-specific memory CD8+ and CD4+ T cell differentiation after primary infection. J Infect Dis 198:536–543. doi: 10.1086/590118. [DOI] [PubMed] [Google Scholar]
  • 279.Fornara C, Lilleri D, Revello MG, Furione M, Zavattoni M, Lenta E, Gerna G. 2011. Kinetics of effector functions and phenotype of virus-specific and γδ T lymphocytes in primary human cytomegalovirus infection during pregnancy. J Clin Immunol 31:1054–1064. doi: 10.1007/s10875-011-9577-8. [DOI] [PubMed] [Google Scholar]
  • 280.Fornara C, Furione M, Arossa A, Gerna G, Lilleri D. 2016. Comparative magnitude and kinetics of human cytomegalovirus-specific CD4+ and CD8+ T-cell responses in pregnant women with primary versus remote infection and in transmitting versus non-transmitting mothers: its utility for dating primary infection in pregnancy. J Med Virol 88:1238–1246. doi: 10.1002/jmv.24449. [DOI] [PubMed] [Google Scholar]
  • 281.Saldan A, Forner G, Mengoli C, Gussetti N, Palù G, Abate D. 2015. Strong cell mediated immune response to human cytomegalovirus is associated with increased risk of fetal infection in primarily infected pregnant women. Clin Infect Dis 61:1228–1234. doi: 10.1093/cid/civ561. [DOI] [PubMed] [Google Scholar]
  • 282.Saldan A, Forner G, Mengoli C, Tinto D, Fallico L, Peracchi M, Gussetti N, Palù G, Abate D. 2016. Comparison of the cytomegalovirus (CMV) enzyme-linked immunosorbent spot and CMV QuantiFERON cell-mediated immune assays in CMV-seropositive and -seronegative pregnant and nonpregnant women. J Clin Microbiol 54:1352–1356. doi: 10.1128/JCM.03128-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Wang X, Li X, Hu S, Qu H, Zhang Y, Ni H, Wang X. 2015. Rapid detection of active human cytomegalovirus infection in pregnancy using loop-mediated isothermal amplification. Mol Med Rep 12:2269–2274. doi: 10.3892/mmr.2015.3572. [DOI] [PubMed] [Google Scholar]
  • 284.Ng AH, Lee M, Choi K, Fischer AT, Robinson JM, Wheeler AR. 2015. Digital microfluidic platform for the detection of rubella infection and immunity: a proof of concept. Clin Chem 61:420–429. doi: 10.1373/clinchem.2014.232181. [DOI] [PubMed] [Google Scholar]
  • 285.Fernández-Carballo BL, McBeth C, McGuiness I, Kalashnikov M, Baum C, Borrós S, Sharon A, Sauer-Budge AF. 2018. Continuous-flow, microfluidic, qRT-PCR system for RNA virus detection. Anal Bioanal Chem 410:33–43. doi: 10.1007/s00216-017-0689-8. [DOI] [PubMed] [Google Scholar]
  • 286.Kaushik A, Tiwari S, Jayant RD, Vashist A, Nikkhah-Moshaie R, El-Hage N, Nair M. 2017. Electrochemical biosensors for early stage Zika diagnostics. Trends Biotechnol 35:308–317. doi: 10.1016/j.tibtech.2016.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Song J, Liu C, Mauk MG, Rankin SC, Lok JB, Greenberg RM, Bau HH. 2017. Two-stage isothermal enzymatic amplification for concurrent multiplex molecular detection. Clin Chem 63:714–722. doi: 10.1373/clinchem.2016.263665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Calvert AE, Biggerstaff BJ, Tanner NA, Lauterbach M, Lanciotti RS. 2017. Rapid colorimetric detection of Zika virus from serum and urine specimens by reverse transcription loop-mediated isothermal amplification (RT-LAMP). PLoS One 12:e0185340. doi: 10.1371/journal.pone.0185340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Barberini L, Noto A, Saba L, Palmas F, Fanos V, Dessì A, Zavattoni M, Fattuoni C, Mussap M. 2016. Multivariate data validation for investigating primary HCMV infection in pregnancy. Data Brief 9:220–230. doi: 10.1016/j.dib.2016.08.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Fattuoni C, Palmas F, Noto A, Barberini L, Mussap M, Grapov D, Dessì A, Casu M, Casanova A, Furione M, Arossa A, Spinillo A, Baldanti F, Fanos V, Zavattoni M. 2016. Primary HCMV infection in pregnancy from classic data towards metabolomics. An exploratory analysis. Clin Chim Acta 460:23–32. doi: 10.1016/j.cca.2016.06.005. [DOI] [PubMed] [Google Scholar]
  • 291.Scott GM, Chow SS, Craig ME, Pang CN, Hall B, Wilkins MR, Jones CA, Lloyd AR, Rawlinson WD. 2012. Cytomegalovirus infection during pregnancy with maternofetal transmission induces a proinflammatory cytokine bias in placenta and amniotic fluid. J Infect Dis 205:1305–1310. doi: 10.1093/infdis/jis186. [DOI] [PubMed] [Google Scholar]
  • 292.Geyer H, Bauer M, Neumann J, Lüdde A, Rennert P, Friedrich N, Claus C, Perelygina L, Mankertz A. 2016. Gene expression profiling of rubella virus infected primary endothelial cells of fetal and adult origin. Virol J 13:21. doi: 10.1186/s12985-016-0475-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Wataganara T, Bianchi DW. 2004. Fetal cell-free nucleic acids in the maternal circulation. Ann N Y Acad Sci 1022:90–99. doi: 10.1196/annals.1318.015. [DOI] [PubMed] [Google Scholar]
  • 294.Sifakis S, Koukou Z, Spandidos DA. 2015. Cell-free fetal DNA and pregnancy-related complications (review). Mol Med Rep 11:2367–2372. doi: 10.3892/mmr.2014.3118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Hui L, Tong S, Uhevaha T, U-Lino K, Hannan NJ. 2016. A comparison of sample collection methods for quantifying cell-free fetal neurodevelopment transcripts in amniotic fluid. BMC Res Notes 9:335. doi: 10.1186/s13104-016-2146-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Pardee K, Green AA, Takahashi MK, Braff D, Lambert G, Lee JW, Ferrante T, Ma D, Donghia N, Fan M, Daringer NM, Bosch I, Dudley DM, O'Connor DH, Gehrke L, Collins JJ. 2016. Rapid, low-cost detection of Zika virus using programmable biomolecular components. Cell 165:1255–1266. doi: 10.1016/j.cell.2016.04.059. [DOI] [PubMed] [Google Scholar]
  • 297.Tomasoni LR, Meroni V, Bonfanti C, Bollani L, Lanzarini P, Frusca T, Castelli F. 2014. Multidisciplinary approach to congenital Toxoplasma infection: an Italian nationwide survey. New Microbiol 37:347–354. [PubMed] [Google Scholar]
  • 298.Messedi E, Fki-Berrajah L, Gargouri S, Chouikha A, Chaari A, Bouaziz M, Jallouli H, Yahia AB, Hammami A, Triki H, Karray-Hakim H. 2014. Clinical epidemiological and molecular aspects of rubella outbreak with high number of neurological cases, Tunisia 2011–2012. J Clin Virol 61:248–254. doi: 10.1016/j.jcv.2014.07.003. [DOI] [PubMed] [Google Scholar]
  • 299.Lilleri D, Gerna G. 2017. Maternal immune correlates of protection from human cytomegalovirus transmission to the fetus after primary infection in pregnancy. Rev Med Virol 27:e1921. doi: 10.1002/rmv.1921. [DOI] [PubMed] [Google Scholar]
  • 300.Frischknecht F, Sell W, Trummer I, Brühwiler H. 2011. Serological testing for infectious diseases in pregnant women: are the guidelines followed? Swiss Med Wkly 140:w13138. doi: 10.4414/smw.2010.13138. [DOI] [PubMed] [Google Scholar]
  • 301.Cordeiro MT, Brito CA, Pena LJ, Castanha PM, Gil LH, Lopes KG, Dhalia R, Meneses JA, Ishigami AC, Mello LM, Alencar LX, Guarines KM, Rodrigues LC, Marques ET. 2016. Results of a Zika virus (ZIKV) immunoglobulin M-specific diagnostic assay are highly correlated with detection of neutralizing anti-ZIKV antibodies in neonates with congenital disease. J Infect Dis 214:1897–1904. doi: 10.1093/infdis/jiw477. [DOI] [PubMed] [Google Scholar]
  • 302.Binnicker MJ, Jespersen DJ, Harring JA. 2010. Multiplex detection of IgM and IgG class antibodies to Toxoplasma gondii, rubella virus, and cytomegalovirus using a novel multiplex flow immunoassay. Clin Vaccine Immunol 17:1734–1738. doi: 10.1128/CVI.00332-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Maple PA, Hedman L, Dhanilall P, Kantola K, Nurmi V, Söderlund-Venermo M, Brown KE, Hedman K. 2014. Identification of past and recent parvovirus B19 infection in immunocompetent individuals by quantitative PCR and enzyme immunoassays: a dual-laboratory study. J Clin Microbiol 52:947–956. doi: 10.1128/JCM.02613-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Beersma MF, Claas ECJ, Sopaheluakan T, Kroes A. 2005. Parvovirus B19 viral loads in relation to VP1 and VP2 antibody responses in diagnostic blood samples. J Clin Virol 34:71–75. doi: 10.1016/j.jcv.2005.04.005. [DOI] [PubMed] [Google Scholar]
  • 305.Revello MG, Furione M, Zavattoni M, Tassis B, Nicolini U, Fabbri E, Gerna G. 2008. Human cytomegalovirus (HCMV) DNAemia in the mother at amniocentesis as a risk factor for iatrogenic HCMV infection of the fetus. J Infect Dis 197:593–596. doi: 10.1086/526499. [DOI] [PubMed] [Google Scholar]
  • 306.Colombo ET, Estofolete CF, Reis AFN, da Silva NS, Aguiar ML, Cabrera EMS, Dos Santos INP, Costa FR, Cruz L, Rombola PL, Terzian ACB, Nogueira ML. 2017. Clinical laboratory and virological data from suspected ZIKV patients in an endemic arbovirus area. J Clin Virol 96:20–25. doi: 10.1016/j.jcv.2017.09.002. [DOI] [PubMed] [Google Scholar]
  • 307.Garza-González E, Mendoza-Olazarán S, Roman-Campos R, Téllez-Marroquín R, Saldívar-Rodríguez D, Soria-López JA, Guzman A, Flores-Treviño S, Camacho-Ortiz A. 2017. Rapid spread of an ongoing outbreak of Zika virus disease in pregnant women in a Mexican hospital. Braz J Infect Dis 21:554–555. doi: 10.1016/j.bjid.2017.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Ren P, Ortiz DA, Terzian A, Colombo TE, Nogueira ML, Vasilakis N, Loeffelholz MJ. 2017. Evaluation of Aptima Zika virus assay. J Clin Microbiol 55:2198–2203. doi: 10.1128/JCM.00603-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Santiago GA, Vázquez J, Courtney S, Matías KY, Andersen LE, Colón C, Butler AE, Roulo R, Bowzard J, Villanueva JM, Muñoz-Jordan JL. 2018. Performance of the Trioplex real-time RT-PCR assay for detection of Zika, dengue, and chikungunya viruses. Nature Comm 9:1391. doi: 10.1038/s41467-018-03772-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Pabbaraju K, Wong S, Gill K, Fonseca K, Tipples GA, Tellier R. 2016. Simultaneous detection of Zika, Chikungunya and Dengue viruses by multiplex real-time PCR assay. J Clin Virol 83:66–71. doi: 10.1016/j.jcv.2016.09.001. [DOI] [PubMed] [Google Scholar]
  • 311.Waggoner JJ, Gresh L, Mohamed-Hadley A, Ballesteros G, Davila MJ, Tellez Y, Sahoo MK, Balmaseda A, Harris E, Pinsky BA. 2016. Single-reaction multiplex reverse transcription PCR for detection of Zika, chikungunya, and dengue viruses. Emerg Infect Dis 22:1295–1297. doi: 10.3201/eid2207.160326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Steinhagen K, Probst C, Radzimski C, Schmidt-Chanasit J, Emmerich P, van Esbroeck M, Schinkel J, Grobusch MP, Goorhuis A, Warnecke JM, Lattwein E, Komorowski L, Deerberg A, Saschenbrecker S, Stöcker W, Schlumberger W. 2016. Serodiagnosis of Zika virus (ZIKV) infections by a novel NS1-based ELISA devoid of cross-reactivity with dengue virus antibodies: a multicohort study of assay performance, 2015 to 2016. Euro Surveill 21:304266. doi: 10.2807/1560-7917.ES.2016.21.50.30426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Bingham AM, Cone M, Mock V, Heberlein-Larson L, Stanek D, Blackmore C, Likos A. 2016. Comparison of test results for Zika virus RNA in urine, serum, and saliva specimens from persons with travel-associated Zika virus disease Florida, 2016. MMWR Morb Mortal Wkly Rep 65:475–478. doi: 10.15585/mmwr.mm6518e2. [DOI] [PubMed] [Google Scholar]
  • 314.Safronetz D, Sloan A, Stein DR, Mendoza E, Barairo N, Ranadheera C, Scharikow L, Holloway K, Robinson A, Traykova-Andonova M, Makowski K, Dimitrova K, Giles E, Hiebert J, Mogk R, Beddome S, Drebot M. 2017. Evaluation of 5 commercially available Zika virus immunoassays. Emerg Infect Dis 23:1577–1580. doi: 10.3201/eid2309.162043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315.L'Huillier AG, Hamid-Allie A, Kristjanson E, Papageorgiou L, Hung S, Wong CF, Stein DR, Olsha R, Goneau LW, Dimitrova K, Drebot M, Safronetz D, Gubbay JB. 2017. Evaluation of Euroimmun anti-Zika virus IgM and IgG enzyme-linked immunosorbent assays for Zika virus serologic testing. J Clin Microbiol 55:2462–2471. doi: 10.1128/JCM.00442-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316.Granger D, Hilgart H, Misner L, Christensen J, Bistodeau S, Palm J, Strain AK, Konstantinovski M, Liu D, Tran A, Theel ES. 2017. Serologic testing for Zika virus: comparison of three Zika virus IgM-screening enzyme-linked immunosorbent assays and initial laboratory experiences. J Clin Microbiol 55:2127–2136. doi: 10.1128/JCM.00580-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317.Lustig Y, Zelena H, Venturi G, Van Esbroeck M, Rothe C, Perret C, Koren R, Katz-Likvornik S, Mendelson E, Schwartz E. 2017. Sensitivity and kinetics of an NS1-based Zika virus enzyme-linked immunosorbent assay in Zika virus-infected travelers from Israel, the Czech Republic, Italy, Belgium, Germany, and Chile. J Clin Microbiol 55:1894–1901. doi: 10.1128/JCM.00346-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.St George K, Sohi IS, Dufort EM, Dean AB, White JL, Limberger R, Sommer JN, Ostrowski S, Wong SJ, Backenson PB, Kuhles D, Blog D, Taylor J, Hutton B, Zucker HA. 2017. Zika virus testing considerations: lessons learned from the first 80 real-time reverse transcription-PCR-positive cases diagnosed in New York State. J Clin Microbiol 55:535–544. doi: 10.1128/JCM.01232-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319.Findal G, Helbig A, Haugen G, Jenum PA, Stray-Pedersen B. 2017. Management of suspected primary Toxoplasma gondii infection in pregnant women in Norway: twenty years of experience of amniocentesis in a low-prevalence population. BMC Pregnancy Childbirth 17:127. doi: 10.1186/s12884-017-1300-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320.Margioula-Siarkou C, Kalogiannidis I, Petousis S, Prapa S, Dagklis T, Mamopoulos A, Prapas N, Rousso D. 2015. Cytomegalovirus, Toxoplasma gondii and rubella vertical transmission rates according to mid-trimester amniocentesis: a retrospective study. Int J Prev Med 6:32. doi: 10.4103/2008-7802.154774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321.Machado AS, Andrade GMQ, Januario JN, Fernandes MD, Carneiro AC, Carneiro M, Carellos EV, Romanelli RM, Vasconcelos-Santos DV, Vitor RW. 2010. IgG and IgM western blot assay for diagnosis of congenital toxoplasmosis. Mem Inst Oswaldo Cruz 105:757–761. doi: 10.1590/s0074-02762010000600005. [DOI] [PubMed] [Google Scholar]
  • 322.Ross SA, Ahmed A, Palmer AL, Michaels MG, Sánchez PJ, Bernstein DI, Tolan RW, Novak Z, Chowdhury N, Fowler KB, Boppana SB, National Institute on Deafness and Other Communication Disorders CHIMES Study. 2014. Detection of congenital cytomegalovirus infection by real-time polymerase chain reaction analysis of saliva or urine specimens. J Infect Dis 210:1415–1418. doi: 10.1093/infdis/jiu263. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical Microbiology Reviews are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES