TRIỆU CHỨNG LÂM SÀNG, XÉT NGHIỆM CHẨN ĐOÁN NHIỄM TRÙNG BÀO THAI Ở TRẺ SƠ SINH
Reproductive Toxicology 21 (2006) 350–382 Review Laboratory assessment and diagnosis of congenital viral infections: Rubella, cytomegalovirus (CMV), varicella-zoster virus (VZV), herpes simplex virus (HSV), parvovirus B19 and human immunodeficiency virus (HIV) Ella Mendelson a,∗ , Yair Aboudy b , Zahava Smetana c , Michal Tepperberg d , Zahava Grossman e a Central Virology Laboratory, Ministry of Health and Faculty of Life Sciences, Bar-Ilan University, Chaim Sheba Medical Center, Tel-Hashomer, 52621, Israel b National Rubella, Measles and Mumps Center, Central Virology, Laboratory, Ministry of Health, Chaim Sheba Medical Center, Tel-Hashomer, Israel c National Herpesvirus Center, Central Virology Laboratory, Ministry of Health, Chaim Sheba Medical Center, Tel-Hashomer, Israel d CMV Reference Laboratory, Central Virology Laboratory, Ministry of Health, Chaim Sheba Medical Center, Tel-Hashomer, Israel e National HIV, EBV and Parvovirus B19 Reference Laboratory, Central Virology Laboratory, Ministry of Health, Chaim Sheba Medical Center, Tel-Hashomer, Israel Received 14 October 2004; received in revised form 30 January 2006; accepted February 2006 Abstract Viral infections during pregnancy may cause fetal or neonatal damage Clinical intervention, which is required for certain viral infections, relies on laboratory tests performed during pregnancy and at the neonatal stage This review describes traditional and advanced laboratory approaches and testing methods used for assessment of the six most significant viral infections during pregnancy: rubella virus (RV), cytomegalovirus (CMV), varicella-zoster virus (VZV), herpes simplex virus (HSV), parvovirus B19 and human immunodeficiency virus (HIV) Interpretation of the laboratory tests results according to studies published in recent years is discussed © 2006 Elsevier Inc All rights reserved Keywords: Laboratory diagnosis; Congenital viral infections; Rubella; Cytomegalovirus (CMV); Varicella-zoster virus (VZV); Herpes simplex virus (HSV); Parvovirus B19; Human immunodeficiency virus (HIV) Contents ∗ General introduction Rubella virus 2.1 Introduction 2.1.1 The pathogen 2.1.2 Immunity and protection 2.1.3 Laboratory assessment of primary rubella infection in pregnancy 2.1.4 Pre- and postnatal laboratory assessment of congenital rubella infection 2.2 Laboratory assays for assessment of rubella infection and immunity 2.2.1 Rubella neutralization test (NT) 2.2.2 Hemagglutination inhibition test (HI) 2.2.3 Rubella specific ELISA IgG 2.2.4 Rubella specific ELISA IgM 2.2.5 Rubella specific IgG-avidity assay 2.2.6 Rubella virus isolation in tissue culture Corresponding author Tel.: +972 530 2421; fax: +972 535 0436 E-mail address: ellamen@sheba.health.gov.il (E Mendelson) 0890-6238/$ – see front matter © 2006 Elsevier Inc All rights reserved doi:10.1016/j.reprotox.2006.02.001 352 352 352 352 352 355 357 358 358 358 358 358 359 359 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 351 2.2.7 Rubella RT-PCR assay 2.3 Summary Cytomegalovirus (CMV) 3.1 Introduction 3.1.1 The pathogen 3.1.2 Laboratory assessment of CMV infection in pregnant women 3.1.3 Prenatal assessment of congenital CMV infection 3.2 Laboratory assays for assessment of CMV infection 3.2.1 CMV IgM assays 3.2.2 CMV IgG assays 3.2.3 CMV IgG avidity assays 3.2.4 CMV neutralization assays 3.2.5 Virus isolation in tissue culture 3.2.6 Detection of CMV by PCR 3.2.7 Quantitative PCR-based assays 3.3 Summary Varicella-zoster virus (VZV) 4.1 Introduction 4.1.1 The pathogen 4.1.2 Assessment of VZV infection in pregnancy 4.1.3 Prenatal and perinatal laboratory assessment of congenital VZV infection 4.2 Laboratory assays for assessment of VZV infection and immunity 4.2.1 VZV IgG assays 4.2.2 VZV IgM assays 4.2.3 Virus detection in clinical specimens 4.2.4 Virus isolation in tissue culture 4.2.5 Direct detection of VZV antigen 4.2.6 Molecular methods for detection of viral DNA 4.3 Summary Herpes simplex virus (HSV) 5.1 Introduction 5.1.1 The pathogen 5.1.2 Laboratory assessment of HSV infection in pregnancy and in neonates 5.2 Laboratory assays for assessment of HSV infection and immune status 5.2.1 HSV IgG assays 5.2.2 HSV type-specific IgG assays 5.2.3 HSV IgM assays 5.2.4 Virus isolation in tissue culture 5.2.5 Direct antigen detection of HSV 5.2.6 Detection of HSV DNA by PCR 5.3 Summary Parvovirus B19 6.1 Introduction 6.1.1 The pathogen 6.1.2 Laboratory assessment of parvovirus B19 infection in pregnancy 6.1.3 Prenatal laboratory assessment of congenital B19 infection 6.2 Laboratory assays for assessment of parvovirus B19 infection 6.2.1 B19 IgM and IgG assays 6.2.2 Detection of viral DNA in maternal and fetal specimens 6.2.3 Quantitative assays for detection of viral DNA 6.3 Summary Human immunodeficiency virus 7.1 Introduction 7.1.1 The pathogen 7.1.2 Importance of laboratory assessment of HIV infection in pregnancy 7.1.3 Prenatal laboratory assessment of HIV infection 7.2 Laboratory assessment of HIV infection 7.2.1 HIV antibody assays 7.2.2 Detection of viral DNA in maternal and newborn specimens 7.3 Summary Acknowledgments References 359 359 360 360 360 360 360 361 361 361 361 362 362 362 362 363 363 363 363 363 364 364 364 364 364 364 364 364 365 365 365 365 365 366 366 366 366 366 366 366 367 367 367 367 367 367 368 368 368 369 369 369 369 369 370 370 370 370 371 372 372 372 352 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 General introduction Viral infections during pregnancy carry a risk for intrauterine transmission which may result in fetal damage The consequences of fetal infection depend on the virus type: for many common viral infections there is no risk for fetal damage, but some viruses are teratogenic while others cause fetal or neonatal diseases ranging in severity from mild and transient symptoms to a fatal disease In cases where infection during pregnancy prompts clinical decisions, laboratory diagnostic tests are an essential part of the clinical assessment process This review describes the six most important viruses for which laboratory assessement during pregnancy is required and experience has been gained over many years Rubella virus and CMV are teratogenic viruses, while VZV, HSV, parvovirus B19 and HIV cause fetal or neonatal transient or chronic disease The ability of viruses to cross the placenta, infect the fetus and cause damage depends, among other factors, on the mother’s immune status against the specific virus In general, primary infections during pregnancy are substantially more damaging than secondary infections or reactivations Laboratory testing of maternal immune status is required to diagnose infection and distinguish between primary and secondary infections Assessment of fetal damage and prognosis requires prenatal laboratory testing primarily in those cases where a clinical decision such as drug treatment, pregnancy termination or intrauterine IgG transfusion must be taken This review describes basic virological facts and explains the laboratory approaches and techniques used for the diagnostic process It aims at familiarizing physicians with the rational behind the laboratory requests for specific and timely specimens and with the interpretation of the tests results including its limitations The laboratory methods used for assessment of viral infections in general are of two categories: serology and virus detection Serology is very sensitive but often cannot conclusively determine the time of infection, which may be critical for risk assessment Traditional serological tests, which measure antibody levels without distinction between IgM and IgG, usually require two samples for determination of seroconversion or a substantial rise in titer The modern tests can distinguish between IgG and IgM and may allow diagnosis in one serum sample However, biological and technical difficulties are common and may cause false positive and false negative results The properties of all serological assays used for each of the viruses will be described in detail in the following chapters Virus detection is used primarily for prenatal diagnosis Invasive procedures must be used to obtain samples representing the fetus such as amniotic fluid (AF), cord blood and chorionic villi (CV) The traditional “gold standard” assay for virus detection used to be virus isolation in tissue culture, but other, more rapid and sensitive methods were developed in recent years Among the new methods are direct antigen detection by specific antibodies and amplification and detection of viral nucleic acids The general characteristics of all laboratory assays described in this article are summarized in Table Since the algorithm for maternal and fetal assessment and the interpretation of tests results vary from one virus to another, we have described the approach to each viral infection in a separate chapter Figs 1–6 depict the most common algorithms used for the laboratory diagnosis of each of the viral infections Rubella virus 2.1 Introduction 2.1.1 The pathogen Rubella is a highly transmissible childhood disease which can cause large outbreaks every few years It is a vaccine preventable disease and in developed countries outbreaks are mostly confined to unvaccinated communities [1] Rubella reinfection following natural infection is very rare Rubella virus (RV) is classified as a member of the togaviridae family and is the only virus of the genus rubivirus [2] Hemagglutinating activity and at least three antibody neutralization domains were assigned to the early proteins E1 and E2 [3–5] At least one weak neutralization domain was identified on E2 [5] The main route of postnatal virus transmission is by direct contact with nasopharyngial secretions [6] Postnatal RV infection is a generally mild and self-limited illness [6–8], but primary RV infections during the first trimester of pregnancy have high teratogenic potential leading to severe consequences, known as congenital rubella syndrome (CRS) which may occur in 80–85% of cases [8,9] It should be emphasized that more than 50% of RV infections in non-immunized persons in the general population (and in pregnant women) are subclinical [6–9] 2.1.2 Immunity and protection Antibody level of 10–15 international units (IU) of IgG per millilitre is considered protective Naturally acquired rubella generally confers lifelong and usually high degree of immunity against the disease for the majority of individuals [10,11] Rubella vaccination induces immunity that confers protection from viraemia in the vast majority of vaccinees, which usually persists for more than 16 years [10–12] A small fraction of the vaccinees fail to respond or develop low levels of detectable antibodies which may decline to undetectable levels within 5–8 years from vaccination [13–17] Several methods are used to determine immunity (Table 1) Neutralization test (NT) and hemagglutination inhibition test (HI) correlate well with protective immunity, but since they are difficult to perform and to standardize, they were replaced by the more rapid, facile and sensitive enzyme-linked immunosorbant assay (ELISA) [5,18] In our experience (unpublished data), there is a clear distinction between antibody levels measured using ELISA, and antibody levels measured using functional assays such as NT and HI Moreover, standardization of anti RV antibody assays using different techniques and a variety of antigens (i.e., whole virus, synthetic peptides, recombinant antigen, etc.) has not been achieved, leading to uncertainties regarding the antibody levels that confer immunity and protection against reinfections and against virus transmission to the fetus [19–26] Most of the reinfection cases (9 out of 18 cases; E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 353 Table Summary and characteristics of the laboratory tests used for assessment of viral infections in pregnancy Laboratory test Serology Neutralization (NT) Hemagglutination inhibition (HI) ELISA IgM ELISA IgG IgG avidity (ELISA) Immunofluorescence (IFA; IFAMA, etc.) Western blot (WB) Virus detection Virus isolation in tissue culture Direct antigen detection Shell-vial assay Molecular assays PCR; RT-PCR Test principles Clinical samples Technical advantages Technical disadvantages limitations Interpretation of positive results Inhibition of virus growth in tissue culture by Aba Prevention of hemagglutination by binding of Ab to viral Agb Detection of virus specific Ab bound to a solid phase by a labled secondary anti-IgM Ab Detection of virus specific Ab bound to a solid phase by a labled secondary anti-IgG Ab Removal of low avidity IgG Ab which results in a reduced signal Maternal blood Corresponds with protection Done only in reference laboratories Maternal blood Accurate and corresponds with protection Laborious, not very sensitive, not Ab class-specific Laborious, not Ab class specific Used only for rubella, done only in specialized labs Neutralizing antibodies are present at a certain titer HI antibodies are present at a certain titer Maternal blood, fetal blood, newborn blood Fast and sensitive, commercialized, automated None False positive and false negative IgM antibodies are present Maternal blood, newborn blood Fast and sensitive, commercialized, automated None None IgG antibodies are present (sometimes with units) Maternal blood Fast and sensitive, commercialized, automated Not many available commercially No interpretation for results outside the inclusion or exclusion criteria Detection of IgG or IgM Ab which binds to a spot of virus infected cells on a slide by a labled secondary Ab Separated viral proteins attached to a nylon membrane react with patient’s serum and detected by labled anti-human Ab Maternal blood, fetal blood, newborn blood Can yield titer; short time Manual, reading is subjective Unsuitable for testing large numbers Low avidity: recent infection; medium avidity: not known; high avidity: probably old infection Antibodies are present at a certain titer Maternal blood infant’s blood Detects antibody specific to a viral protein Laborious Not very sensitive Antibodies specific to certain viral antigens are present Innoculation of specific tissue cultures with clinical samples and watching for CPEc Detection of a viral antigen in cells from a clinical sample by IFA or ELISA Any clinical sample which may contain virus Detects and isolates live virus Very labourious Slow Insensitive, done only in virology labs Live virus is present in the clinical sample For IFA: cells from clinical samples For ELISA: any sample Any clinical sample which may contain virus Fast and simple Not sensitive Not sensitive, low positive predictive value The sample most likely contains live virus Detects live virus; rapid: results within 16–72 h Labourious; requires high skills; uses expensive monoclonal Abs Not highly sensitive; done only in virology labs Live virus is present in the clinical sample Any clinical sample which may contain virus Fast, simple, can be automated; very sensitive Very prone to contaminations False positive by contamination; may detect latent virus Viral nucleic acid is present in the sample, not known if live virus is present Innoculation of specific tissue cultures with clinical samples, then fixation and detection of viral cell-bound antigen by IFA Enzymatic amplification of viral nucleic acid and detection of amplified sequences 354 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 Table (Continued ) Laboratory test Real-time PCR/RT-PCR In situ hybridization In situ PCR a b c Test principles Clinical samples Technical advantages Technical disadvantages limitations Interpretation of positive results Detection of accumulating PCR products by a fluorescent dye or probe in a specialized instrument Detection of viral nucleic acid in smears or tissue sections by labled probes Detection of viral nucleic acid in smears or tissue sections by PCR using labled primers Any clinical sample which may contain virus Very fast, simple not prone to contaminations; can be quantitative Expensive instruments Sometimes too sensitive, interpretation of very low result questionable Viral nucleic acid is present in the sample (at a certain amount), not known if live virus is present Cells or tissue from clinical samples Sensitive and specific Difficult to perform Done only in specializing labs Viral nucleic acid is present in the sample, not known if live virus is present Cells or tissue from clinical samples Sensitive and specific Difficult to perform Doe only in specializing labs Viral nucleic acid is present in the sample, not known if live virus is present Antibody Antigen Cytopathic effect 50%) which were detected during an outbreak in Israel in 1992 occurred in the presence of low neutralizing antibody titers of 1:4 (cut off level), and sharp decline in the reinfection rate correlated with the presence of higher titers of neutralizing antibodies (unpublished data) Reinfection rates following vaccination are considerably higher than following natural infection, ranging between 10% and 20% [19] Many developed countries adopted the infant routine vaccination policy using MMR (mumps measles and rubella) vaccine designed to provide indirect protection of child-bearing age Fig Algorithm for assessment of rubella infection in pregnancy: the algorithm shows a stepwise procedure beginning with testing of the maternal blood for IgM and IgG If the maternal blood is IgM negative the IgG result determines if the woman is seropositive (immune) or seronegative (not immune) If not immune the woman should be retested monthly for seroconversion till the end of the 5th month of pregnancy If the maternal blood is IgM and IgG positive the next step would be an IgG avidity assay on the same blood sample to estimate the time of infection Low avidity index (AI) indicates recent infection while high AI indicates past or recurrent infection Medium AI is inconclusive and the test should be repeated on a second blood sample obtained 2–3 weeks later If the maternal blood is IgM positive and IgG negative, recent primary infection is suspected and the same tests should be repeated on a second blood sample obtained 2–3 weeks later If the results remain the same (IgM+ IgG−), then the IgM result is considered non-specific, indicating that the woman has not been infected (however she is seronegative and should be followed to the end of the 5th month as stated above) If the woman has seroconverted (IgM+ IgG+), recent primary infection is confirmed and prenatal diagnosis should take place if the woman wishes to continue her pregnancy Determination of IgM in cord blood is the preferred method with the highest prognostic value Post natal diagnosis is based on the newborn’s serology (IgM for 6–12 m and IgG beyond age m) and on virus isolation from the newborn’s respiratory secretions E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 355 Fig Algorithm for assessment of CMV infection in pregnancy: the algorithm shows a stepwise procedure which begins with detection of IgM in maternal blood If the maternal blood is IgG positive, an IgG avidity assay on the same blood sample should be performed to estimate the time of infection Low avidity index (AI) indicates recent primary infection and prenatal diagnosis should follow Medium or high AI is mostly inconclusive, especially if the maternal blood was obtained on the second or third tremester Continuation of the assessment is based on either maternal blood or fetal prenatal diagnosis If the first maternal blood was IgM positive but IgG negative, a second blood sample should be obtained 2–3 weeks later If the IgG remains negative then the IgM is considered non-specific If the woman has seroconverted and developed IgG, primary infection is confirmed and prenatal diagnosis should follow For prenatal diagnosis amniotic fluid (AF) should be obtained not earlier than the 21st week of gestation and weeks following seroconversion Fetal infection is assessed by virus isolation using standard tissue culture or shell-vial assay, and/or by PCR detection of CMV DNA Positive result by either one of these tests indicates fetal infection women regardless of vaccination status However, in Israel and in other countries with high vaccination coverage, RV still circulates and may cause reinfections in vaccinated women whose immunity has waned [19,20,23, unpublished data] 2.1.3 Laboratory assessment of primary rubella infection in pregnancy Assessment of primary rubella infection in pregnant women relies primarily on the detection of specific maternal IgM antibodies in combination with either seroconversion or a >4-fold rise in rubella specific IgG antibody titer in paired serum samples (acute/convalescent) as shown in Fig Today, due to the high sensitivity of the ELISA-IgM assays low levels of rubella specific IgM are detected more frequently, leading to an increase in the number of therapeutic abortions and reducing the number of CRS cases However, frequently the low level of IgM detected is not indicative of a recent primary infection for several reasons: (a) IgM reactivity after vaccination or primary rubella infection may sometimes persist for up to several years [27–29]; (b) heterotypic IgM antibody reactivity may occur in patients recently infected with Epstein Barr virus (EBV), cytomegalovirus (CMV), human parvovirus B19 and other pathogens, leading to false positive rubella IgM results [30–35]; (c) false positive rubella specific IgM response may occur in patients with autoimmune diseases such as systemic lupus erythematosus (SLE) or juvenile rheumatoid arthritis, etc., due to the presence of rheumatoid factor (RF) [36,37]; (d) low level of specific rubella IgM may occur in pregnancy due to Fig Algorithm for assessment of VZV infection in pregnancy: two situations are shown: (1) clinical varicella in a pregnant woman (top left) should be assessed by serology (IgM and IgG in maternal blood) and by virus isolation or detection in early dermal lesions If either of those approaches confirms maternal VZV infection (positive virus isolation/detection test and/or maternal seroconversion), then fetal infection can be assessed by virus detection in amniotic fluid using direct antigen detection or PCR (2) Exposure of a pregnant woman to a varicella case (top right) should prompt maternal IgG testing within 96 h from exposure If the mother has no IgG (not immune) she should receive VZIG within 96 h from exposure 356 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 Fig Algorithm for assessment of HSV infection in pregnancy and in neonates: the algorithm shows two complementary approaches to the confirmation of genital HSV infection in pregnant women (1) If genital lesions are present (top right), virus isolation and typing is the preferred diagnostic approach A positive woman should be examined during delivery for genital lesions If normal delivery has taken place, the newborn should be examined for HSV infection symptoms and tested by virus isolation or PCR using swabs taken from skin, eye, nasopharynx and rectum or CSF Detection of IgM in the newborn’s blood also confirms the diagnosis Negative infants should be followed for month (2) Serology (top left) is a stepwise procedure beginning with maternal IgM and IgG testing The interpretation of the results is shown: low positive or negative IgM in the presence of IgG indicates previous infection with the same virus type (reactivation), or recurrent infection with the other virus type Type-specific serology may resolve the issue If the IgG test is negative, in the presence or absence of IgM, a second serum sample should be obtained to observe seroconversion If the IgG remains negative then no infection occurred If the woman seroconverted, type specific serology can identify the infecting virus type Fig Algorithm for assessment of parvovirus B19 infection in pregnancy: the algorithm shows a stepwise procedure beginning with maternal serology following clinical symptoms in the mother or in the fetus or maternal contact with a clinical case Negative IgM and positive IgG indicate past infection, but if the IgG is high recent infection cannot be ruled out In all other cases a second serum sample should be obtained and retested Only in the case of repeated negative results for both IgG and IgM recent infection with B19 can be ruled out In all other cases the fetus should be observed for clinical symptoms and if present tested for B19 infection by nested PCR or rt-PCR performed on amniotic fluid or fetal blood Positive result confirms fetal infection while negative result suggests that the fetus was not infected with B19 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 357 Fig Algorithm for assessment of HIV infection in pregnancy and in newborns to infected mothers: (1) diagnosis of maternal infection (top left) is by the routine protocol (testing first by EIA and confirming by WB or IFA) If the mother is positive she should be treated as described in the text (2) A newborn to an HIV positive mother (top right) should be tested at birth by DNA PCR on a blood sample The results, whether positive or negative, should be confirmed by retesting either immediately (if positive) or 14–60 days later (if negative) If positive the newborn should be treated while if negative testing should be repeated at 3–6 months and again at 6–12 months As a general rule, any PCR positive test in a newborn should be repeated on two different blood samples After 12 months serological assessment can replace the PCR test as the infant has lost its maternal antibodies polyclonal B-cells activation trigerred by other viral infections [33,35,38] False negative results may also occur in samples taken too early during the course of primary infection Thus, the presence or absence of rubella specific IgM in an asymptomatic patient should be interpreted in accordance with other clinical and epidemiological information available and prenatal diagnosis may be required A novel assay developed recently to support maternal diagnosis is the IgG avidity assay (Table 1) which can differentiate between antibodies with high or low avidity (or affinity) to the antigen It is used when the mother has both IgM and IgG in the first serum collected (Fig 1) Following postnatal primary infection with rubella virus, the specific IgG avidity is initially low and matures slowly over weeks and months [39–41] Rubella specific IgG avidity measurement proved to be a useful tool for the differentiation between recent primary rubella (clinical and especially subclinical infection), reinfection, remote rubella infection or persistent IgM reactivity This distinction is critical for the clinical management of the case, since infection prompts a therapeutic abortion, reinfection requires fetal assessment, while remote infection or non-specific IgM reactivity carry no risk to the fetus [39–42] 2.1.4 Pre- and postnatal laboratory assessment of congenital rubella infection Maternal primary infection prompts testing for fetal infection (Fig 1) The preferred laboratory method for prenatal diagnosis is determination of IgM antibodies in fetal blood obtained by cordocentesis [27,43] Other options include virus detection in chorionic villi (CV) samples or amniotic fluid (AF) specimens The laboratory methods used for virus detection are virus isolation in tissue culture or amplification of viral nucleic acids by RT/PCR (Table 1) However, using those methods for detection of rubella virus in AF and CV might be unreliable, particularly in AF samples due to low viral load Studies showed that rubella virus may be present in the placenta but not in the fetus, or it can be present in the fetus but not in the placenta, leading to false negative results [6,43,44] Thus, according to one opinion, detection of rubella virus in AF or CV does not justify the risk of fetal loss following these invasive procedures [45], while according to another opinion, laboratory diagnosis of fetal infection should combine a serological assay (detection of rubella specific IgM) with a molecular method (viral RNA detection) in order to enhance the reliability of the diagnosis [46] A recent study showed 83–95% sensitivity and 100% specifity for detection of RV in AF by RT/PCR [47] Postnatal diagnosis of congenital rubella infection [9,27,36] is based on one or more of the following: a Isolation of rubella virus from the infant’s respiratory secretions b Demonstration of rubella specific IgM (or IgA) antibodies in cord blood or in neonatal serum, which remain detectable for 6–12 months of age c Persistence of anti-rubella IgG antibodies in the infant’s serum beyond 3–6 months of age The principles, advantages and disadvantages of each laboratory test, are described below 358 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 2.2 Laboratory assays for assessment of rubella infection and immunity 2.2.1 Rubella neutralization test (NT) Virus neutralization is defined as the loss of infectivity due to reaction of a virus with specific antibody Neutralization can be used to identify virus isolates or, as in the case of rubella diagnosis, to measure the immune response to the virus [24,36,48] As a functional test, neutralization has proven to be highly sensitive, specific and reliable technique, but it can be performed only in virology laboratories which comprise only a small fraction of the laboratories performing rubella serology Rubella virus produces characteristic damage (cytopathic effect, CPE) in the RK-13 cell line that was found most sensitive and suitable for use in rubella neutralization test Other cells such as Vero and SIRC lines can be used if conditions are carefully controlled [36] Principally, 2-fold dilutions of each test serum are mixed and incubated with 100 infectious units of rubella virus under appropriate conditions Then cell monolayers are inoculated with each mixture and followed for CPE Control sera possessing known high and low neutralizing antibody levels and titrations of the virus are included in each test run The neutralization titer is taken as the reciprocal of the highest serum dilution showing complete inhibition of CPE [25,36] 2.2.2 Hemagglutination inhibition test (HI) Until recently, assessment of rubella immunity and diagnosis of rubella infection has been carried out mainly by the HI test which is based on the ability of rubella virus to agglutinate red blood cells [49] HI test is labor intensive, and is currently performed mainly by reference laboratories HI is the “gold standard” test against which almost all other rubella screening and diagnostic tests are measured During the test, the agglutination is inhibited by binding of specific antibodies to the viral agglutinin Titers are expressed as the highest dilution inhibiting hemagglutination under standardized testing conditions [50–53] The HI antibodies increase rapidly after RV infection since the test detects both, IgG and IgM class-specific antibodies A titer of l:8 is commonly considered negative (cut off level: 1:16) and a titer of ≥1:32 indicates an earlier RV infection or successful vaccination and immunity Seroconversion is interpreted as primary rubella infection, and a 4-fold increase in titer between two serum samples (paired sera) in the same test series, is interpreted as a recent primary rubella infection or reinfection [52] Considerable experience has been accumulated over the years in the interpretation of the clinical significance of HI titers [52–54], and the test results accurately correlate with clinical protection [5,18] Although HI is generally considered as not sensitive enough, in certain situations it is still in use for resolution of diagnostic uncertainties Detection of rubella specific IgM class antibodies by HI test which requires tedious methods for purification of IgM or removal of IgG [55,56], are no longer in use due to the development of a variety of rapid, easy to perform and sensitive methods, of which ELISA is the most vastly used [18,57] 2.2.3 Rubella specific ELISA IgG The ELISA technique was established for detection of an increasing range of antibodies to viral antigens In 1976, Voller et al [58] developed an indirect assay for the detection of antiviral antibodies The technique has been successfully applied for the detection of rubella specific antibodies Almost all commercially available ELISA kits for the detection of rubella specific IgG are of the indirect type, employing rubella antigen attached to a solid phase (microtiter polystyrene plates or plastic beads) The source of the antigen (peptide, recombinant or whole virus antigen) affects the sensitivity and specificity of the assay After washing and removal of unbound antigen, diluted test serum is added and incubated with the immobilized antigen The rubella specific antibodies present in the serum bind to the antigen Then, unbound antibodies are removed by washing and an enzyme conjugated anti-human IgG is added and further incubation is carried out The quantity of the conjugate that binds to each well is proportional to the concentration of the rubella specific antibodies present in the patient’s serum The plates are then washed and substrate is added resulting in color development The enzymatic reaction is stopped after a short incubation period, and optical density (OD) is measured by an ELISA-reader instrument The test principle allows the detection of IgM as well by using an appropriate anti-human IgM conjugate [53,57] In most commercial ELISA IgG assays the results are automatically calculated and expressed quantitatively in international units (IU) When performed manually, the procedure takes approximately h but automation has reduced it to about 30 [57–59] It is important to note that in order to obtain reliable results, determination of a significant change in specific IgG activity in paired serum samples should always be performed in the same test run and in the same test dilution The correlation between the ELISA and HI or NT titers is not always high This may be explained by the fact that the three methods detect antibodies directed to different antigenic determinants [54] Certain individuals fail to develop antibodies directed to protective epitopes such as the neutralizing domains of E1 and E2 due to a defect in their rubella specific immune responses [21] but they develop antibodies directed to antigenic sub-regions of rubella virus proteins ELISA assays utilizing whole virus as antigen may fail to distinguish between these different antibody specificities Thus, seroconversion determined by ELISA based on a whole virus antigen does not necessarily correlate with protection against infection [52] 2.2.4 Rubella specific ELISA IgM Commercially available ELISA kits for the detection of IgM are mainly of two types: a Indirect ELISA: The principle of the assay was described above for rubella IgG except for using enzyme labeled antihuman IgM as a conjugate In this assay, false negative results may occur due to a competition in the assay between specific IgG antibodies with high affinity (interfering IgG) while the specific IgM have lower affinity for the antigen [31,32] In the E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 new generation ELISA assays this is avoided by the addition of an absorbent reagent for the removal of IgG from the test serum False positive results may occur if rheumatoid factor (RF: IgM anti-IgG antibodies) is present along with specific IgG in the test serum Absorption or removal of RF and/or IgG is necessary prior to the assay to avoid such reactions [30–32,60] b IgM capture ELISA: In these assays anti-human IgM antibody is attached to the solid phase for capture of serum IgM Rubella virus antigen conjugated to enzyme-labeled anti-rubella virus antibody is added for detection This type of assay eliminates the need for sample pretreatment prior to the assay [32,61] As for the rubella virus antigens, most assays are based on whole virus extracts, but recent developments led to production of recombinant and synthetic rubella virus proteins [5,62] 2.2.5 Rubella specific IgG-avidity assay This assay is based on the ELISA IgG technique and applies the elution principle in which protein denaturant, mostly urea (but also diethylamine, ammonium thiocyanate, guanidine hydrochloride, etc.) is added after binding of the patient’s serum The denaturant disrupts hydrophobic bonds between antibody and antigen, and thus, low avidity IgG antibodies produced during the early stage of infection are removed This results in a significant reduction in the IgG absorbance level [63] The avidity index (AI) is calculated according to the following formula [57]: absorbance of avidity ELISA absorbance of standard ELISA The AI is a useful measure only when the IgG concentration in the patient’s serum is not below 25 IU [39] Low avidity (usually below 50%) is associated with recent primary rubella infection while reinfection is typically associated with high avidity as a result of the stimulation of memory B cells (immunological memory) [39–41] In infants with CRS the low avidity IgG continues to be produced for much longer than in cases of postnatal primary rubella, where it lasts 4–6 week after exposure [39] This may be used for retrospective assessment of initially undiagnosed CRS cases AI = 100 × 2.2.6 Rubella virus isolation in tissue culture Diagnosis of prenatal or postnatal rubella infections are essentially based on the more reliable and rapid serological techniques However, virus isolation is useful in confirming the diagnosis of CRS (Fig 1) and rubella virus strain characterization required for epidemiological purposes Rubella virus can be isolated using a variety of clinical specimens such as: respiratory secretions (nasopharyngeal swabs), urine, heparinized blood, CSF, cataract material, lens fluid, amniotic fluid, synovial fluid and products of conception (fetal tissues: placenta, liver, skin, etc.) obtained following spontaneous or therapeutic abortion [6,36,44,64] In order to avoid virus inactivation, specimens should be inoculated into cell culture immediately or stored at ◦ C for not more than days, or kept frozen (−70◦ C) for longer periods [36] 359 Rubella virus can be grown in a variety of primary cells and cell lines [36,65], but RK-13 and Vero cell lines are the most sensitive and suitable for routine use In these cell systems rubella virus produces characteristic CPE Since the CPE is not always clear upon primary isolation, at least two successive subpassages are required [66] When CPE is evident the identity of the virus isolates should be confirmed using immunological or other methods [36,65,67] 2.2.7 Rubella RT-PCR assay Reverse transcription followed by PCR amplification (RTPCR) is a rapid, sensitive and specific technique for detection of rubella virus RNA in clinical samples using primers from the envelope glycoprotein E1 open reading frame [45,46,68,69] Coding sequences for a major group of antigenic determinants are located between nucleotides 731 and 854 of the E1 gene of RV strain M33 This region is highly conserved in various wild type strains and is likely to be present in most clinical samples from rubella infected patients Specific oligonucleotide primers located in this region were designed for amplification by RT-PCR [70–72] Following rubella genomic RNA extraction from clinical specimens and RT-PCR amplification, the product is visualized by gel electrophoresis Positive samples show a specific band of the expected size compared to size markers [68,69,72] A nested RT-PCR assay, in which the RT-PCR product from the first amplification reaction is re-amplified by internal primers, was developed and shown to provide a higher level of sensitivity for the detection of rubella virus RNA [72] However, the risk of contamination is markedly increased The detection limit of the RT-PCR assay is approximately two RNA copies Clinical specimens for rubella virus genome detection include: products of conception (POC), CV, lens aspirate/biopsy, AF, fetal blood, pharyngeal swabs and spinal fluid (CSF) or brain biopsy when the central nervous system (CNS) is involved [68,69,73–75] An additional advantage of RT-PCR is that it does not require infectious virus [74] RV is extremely thermo-labile and frequently is inactivated during sample transporation to the laboratory Finally, it should be noted that clinical samples may contain PCR inhibitors (such as heparin and hemoglobin), and the extraction procedure itself may cause enzyme inhibition [72,76,77] This underscores the need and importance for strict internal quality control during each step of the RT-PCR procedure and participation in external quality assessment programs is of a high value 2.3 Summary Rubella infection during pregnancy, although rare in countries with routine vaccination programs, is still a problem requiring careful laboratory assessment The laboratory testing should confirm or rule-out recent rubella infection in pregnant women and identify congenital rubella infections in the fetus or neonate Maternal infection is currently assessed by serological assays, primarily by ELISA IgM and IgG Borderline results for the IgG assay can be further assessed by the HI or NT assays available 368 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 does not always develop IgM it is necessary to detect the virus itself in fetal samples B19 cannot be cultivated and therefore detection of viral DNA is sought The type of specimen and the detection methods are still controversial: fetal blood and AF are the most common specimens obtained and the most highly sensitive molecular methods are employed However, 10–25% of the specimens from asymptomatic fetuses may have B19 DNA [289,290] This drives the need to develop quantitative molecular assays which can determine viral load and possibly associate it with the risk to the fetus Those assays include quantitative PCR-ELISA, in situ hybridization and rt-PCR The latter has advantages which will be discussed below [304–308] 6.2 Laboratory assays for assessment of parvovirus B19 infection 6.2.1 B19 IgM and IgG assays Several commercial tests have been developed on the basis of recombinant and synthetic antigens [309–311] These recombinant antigens frequently lack important conformational epitops reducing the sensitivity and specificity of the assays Moreover, “gold standard” methods like virus NT or HI assays are absent, and it is difficult to assess the sensitivity and specificity of the serological assays The variation in specificity and sensitivity among commercial assays is high, and discordant results are often obtained indicating false positive and false negative responses [314–317] Studies aimed at evaluating the sensitivity and specificity of various such assays were mostly based on comparative evaluation, using samples selected on the basis of clinical diagnosis In a study conducted in Norway [315], five commercial ELISA and IF IgM kits were comparatively evaluated in three groups of patients The calculated specificities for the kits were 70.1–94.8%, but the authors refrained from determining sensitivity because of the absence of suitable reference methods In another study conducted in Sweden [316], four commercial IgM assays were evaluated in comparison to an IgM antibody capture radioimmunoassay as a reference method [318] The calculated sensitivity varied between 90% and 97% and the specificities varied from 88% to 96% In a third study conducted in the USA [314] three ELISA systems utilizing one or more conformational antigens for detection of B19 IgM or IgG in sera of 198 pregnant women were comparatively evaluated Agreement with the consensus results varied from 92.3% to 100% for IgM and from 97.9% to 99.5% for IgG The authors note the high agreement in this study compared to their previous study [312] relating to the fact that the antigens utilized in the two studies were substantially different: the earlier study used a linear VP1 antigen while the latter one used a conformational VP2 antigen A study designed to evaluate the most appropriate assays for IgG detection was conducted in Italy [313] which compared the performance of three commercial assays using different antigens: (a) an ELISA assay using VP1 + VP2 recombinant native conformational antigens, (b) an ELISA assay using VP2 recombinant native conformational epitopes, (c) a Western blot assay using denatured linear antigens Four hundred and fourty six serum samples from blood donors with no IgM were tested Overall, 353 sera were found positive by all methods combined Of those 98.6%, 94.6% and 89% were positive by assays a, b and c, respectively Some sera reacted only with conformational epitops The results of this study underscore the need to use ELISA-IgG assays which include conformational epitopes of both VP1 and VP2 or VP2 alone, and demonstrate the lower sensitivity of the Western blot assay In conclusion, when evaluating recent or past infections in pregnant women it is important to use well-established ELISA assays, and in cases of discrepancy between the test results and the clinical and epidemiological circumstances, confirmation must be sought using other commercial assays as well as supportive molecular assays discussed next 6.2.2 Detection of viral DNA in maternal and fetal specimens Due to the limitations of the serological assays described above and since viremia is long-lasting in B19 infections, molecular assays for detection of B19 viral DNA were developed early The methods most frequently used are DNA hybridization and PCR Molecular hybridization assays employing DNA probes derived from most of the viral genome are labeled with 32 P, biotin or digoxigenin [319–322] and are capable of detecting approximately 104 –105 genome copies/ml [275,323] This assay is sensitive enough to detect viral DNA in serum during peak viremia since the viral load exceeds 1010 genome copies/ml, but low levels of viremia may be missed [275,296,298] The PCR assays, developed later relied on a variety of primer sets derived from different genomic regions and were capable of detecting 102 –105 genome copies/ml [324,325] The nested PCR, the PCR-ELISA and the PCR-hybridization assays increased sensitivity and moved the detection limit down to 1–10 genome copies/ml [291,292,296,297,299,326–328] By using those highly sensitive methods it was revealed that in many cases serology failed to detect maternal or fetal infections, as some seronegative mothers as well as fetuses turned out positive in the DNA detection assays This finding was not surprising in view of the known limitations of the serological assays The ability to detect maternal and fetal infection by the highly sensitive molecular methods allowed correct diagnosis of previousely unresolved cases, particularly cases of intrauterine infection which did not result in fetal hydrops or were completely subclinical [275,276,290,292,296,297,324,327,329] However, as stated above, low levels of B19 DNA in maternal blood can be unrelated to recent infection The use of various techniques without standardization, the epidemiologically variable circumstances (epidemic VS nonepidemic years), and the use of different primer sets and clinical specimens led to a high variability in the sensitivity, specificity and interpretation of the assays results Moreover, new genotypes discovered recently were missed by common primer sets Comparison of IgM and DNA detection by PCR in fetal blood was conducted in a study done on 57 pregnant women and their fetuses who had abnormal ultrasonography [327] Viral DNA was found by PCR in 16 out of 58 fetuses (27%) while IgM was E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 detected only in (12.3%) Two fetuses had false-positive IgM result, not supported by any other findings Other researchers [276] compared maternal IgM to fetal serum or AF PCR results in 56 women at high risk for B19 infection They found positive PCR in 24 IgM-negative/IgG-positive and seronegative (total 50%) out of, in addition to positive PCR in 15 (26%) of IgM-positive women Another group [282] reported detection of viral DNA in fetal serum or AF by a PCR-hybridization assay in 11 out of 80 cases of fetal hydrops (14%) while maternal IgM antibodies were detected only in (3.7%) Finally, a comprehensice prospective study in 18 fetal hydrops cases conducted in Italy [275] examined maternal serum, fetal cord blood and amniotic fluid using nested PCR, dot-blot hybridization and in situ hybridization (ISH) The results showed that the ISH assay in fetal blood cells was 100% sensitive while the other methods missed few to many cases The conclusion drawn from this study is that various assays have complementary roles, and reliable diagnosis can be achieved only by a combination of serological and molecular assays done on maternal and fetal samples as outlined in Fig 6.2.3 Quantitative assays for detection of viral DNA The need to determine the quantity of B19 viral DNA present in clinical samples (viral load) arose because B19 can establish long-lasting persistent infection in immunocompetent individuals It is not clear if low viral loads reflect whole genomes or viable infectious virus particles, or only pieces of viral DNA Studies in seronegative plasma-pool recipients showed that only recipients of plasma containing >107 genome copies/ml became infected or seroconverted [305] B19 DNA can also be detected in solid organ tissues by PCR for years [323,330,331] As noted earlier viral DNA can be detected in fetal tissue or blood in the absence of any detectable congenital abnormalities, and the outcome of detectable fetal infection, including fetal hydrops, varies from spontaneous resolution to still-birth Real-time PCR assays developed recently are equivalent to or more sensitive than nested PCR and PCR-ELISA but are much less prone to molecular contamination and produce a quantitative result which can be standardized and automated [304,306–308,332] In a retrospective study, Knoll et al [332] used real-time PCR to investigate the viral load in paired samples from mothers and their abnormal fetuses, and from mothers with normal fetuses who were exposed to B19 The viral load in the maternal serum ranged from 7.2 × 102 to 2.6 × 103 The authors did not report statistically significant correlation between maternal or fetal viral load and fetal condition However, they noted that they could not exclude a correlation between peak maternal viremia levels and fetal condition since most of the mothers were not aware of their infection until onset of fetal symptoms, and their serum was collected after peak viremia Although the viral load in fetal sera was higher than in AF samples, the rtPCR assay detected all positive cases using either one of these fetal specimens The authors recommend testing AF rather than fetal blood because AF can be drawn earlier, is simpler to obtain and less risky to the fetus Clearly, more prospective studies are necessary on larger groups of patients to learn more about the association between viral load and pregnancy outcome 369 6.3 Summary Parvovirus B19 infection during pregnancy may cause fetal damage resulting in fetal loss Early diagnosis of maternal infection will allow fetal assessment and treatment by intrauterine blood transfusion Unfortunately, mothers often are not aware of their infection until fetal damage is observed Confirmation of B19 infection requires laboratory assessment, which is complicated by the nature of the viral infection and immune response Serology, performed by using ELISA assays rely on recombinant antigens and concordance is low among all commercial assays available In the absence of a “gold standard” assay false positive and false negative results prevail Virus culturing is impossible and virus detection is based on various molecular assays In spite of several studies there is no consensus regarding the most appropriate clinical specimen and method for detection of viral DNA Currently, on practical grounds, it is recommended to use ELISA IgM and IgG assays based on recombinant conformational epitopes of VP1 and VP2 or VP2 alone, and to use AF or fetal serum for detection of fetal infection by the most sensitive molecular methds available (nested PCR or rt-PCR) Since B19 may establish long lasting infection in the absence of symptoms, interpretation of viral DNA detection in maternal blood is difficult Assessment of fetal infection and risk should rely on the clinical situation and other prenatal diagnostic means An algorithm describing the most practical approach to laboratory assessment of B19 infection in pregnancy is shown in Fig Human immunodeficiency virus 7.1 Introduction 7.1.1 The pathogen Human immunodeficiency virus (HIV) is a retrovirus that infects helper T cells of the immune system causing a progressive reduction in their numbers, and eventually acquired immunodeficiency syndrome (AIDS) HIV is a member of the Retroviridae family [333], genus Lentivirus (or “slow” viruses) The course of infection with these viruses is characterized by a long interval between initial infection and the onset of serious symptoms The single-stranded RNA viruses exploit their reverse transcriptase enzyme to synthesize DNA using their RNA as a template The DNA is then incorporated into the genome of infected cells AIDS was first diagnosed in European sailors with African connections [334–336] The first AIDS virus, HIV-1, was initially identified in 1983 [337–341] A second AIDS virus, HIV-2, was discovered in 1986 [342–344] Forty million people were estimated to be infected with HIV at the end of 2004 [345] The highest prevalence is found in Sub-Saharan Africa and it is rising mainly in Asia and some of the former Soviet Union countries like Ukraine and the Russian Federation [345,346] During its spread among humans, group M HIV-1 (one of three groups: M, O and M), has evolved into multiple subtypes that differ from one another by 10–30% along their genomes [347–350] 370 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 HIV is passed on primarily via four routes: unprotected sexual intercourse (both homosexual and heterosexual), sharing of needles by IV drug users, medical procedures using HIV-contaminated blood, tissues or equipment, and motherto-child transmission (MTCT) The likelihood of transmission is increased by factors that may damage mucosal linings of exposed tissues, especially by other sexually transmitted diseases that cause ulcers or inflammation HIV can be transmitted from infected mothers to infants during pregnancy (intrauterine) through transplacental passage of the virus [351], during labor and delivery (intrapartum) through exposure to infected maternal fluids (blood or vaginal secretions) [352–355] and during the post partum period through breastfeeding [356–359] Infants who have a positive virologic test (see below) at or before age 48 h are considered to have early (i.e., intrauterine) infection, whereas infants who have a negative virologic test during the first week of life and subsequent positive tests are considered to have late (i.e., intrapartum) infection [360] In the absence of breast-feeding, intrauterine transmission accounts for 10–35% of infection, and 60–75% of transmission occurs during labor and delivery [361–363] Without intervention, maternal infection leads to about 25–30% of babies being infected [364–369] Use of anti-retroviral therapy (ART) during pregnancy in HIV-infected women is associated with improved obstetric outcome of reduced infection rates of babies to less than 2% [370–373] and little maternal toxicity [374–376] The current US guidelines are to offer all pregnant HIV-1-infected women highly active antiretroviral therapy (HAART) to maximally suppress viral replication, reduce the risk of prenatal transmission, and minimize the risk of development of resistant virus In addition, HIV-infected women are offered an elective caesarean section delivery [355] The results also suggest that special attention should be given to women belonging to previously identified risk groups Because testing has proven very successful in helping to prevent the spread of the disease to babies, a US federal panel has recommended that all pregnant women, not just those considered at high risk, be screened for the AIDS virus [377–381] No prenatal (intrauterine) diagnosis is performed because: (a) the infection can occur during delivery and (b) the testing intervention may increase the chance of virus transmission to the baby Assessing the infection status of the mother is critical [381] and following delivery the new-born should be tested The laboratory testing is essential for the diagnosis 7.1.2 Importance of laboratory assessment of HIV infection in pregnancy Identification of HIV-infected women before or during pregnancy is critical to providing optimal therapy for both infected women and their children and to preventing perinatal transmission (see below) For women with unknown HIV status during active labor, ART can still be effective when given during labor and delivery, followed by treatment of the newborn [382] This expedited intervention requires the use of rapid diagnostic testing during labor or rapid return of results from standard testing Knowledge of maternal HIV infection during the antenatal period enables HIV-infected women to receive appropriate antiretroviral therapy during pregnancy, during labor, and to newborns to reduce the risk for HIV transmission from mother to child [370,383,384] It also allows counseling of infected women about the risks for HIV transmission through breast milk and advising against breast feeding in countries where safe alternatives to breast milk are available [385] Early diagnostic evaluation of HIV-exposed infants permits early initiation of aggressive antiretroviral therapy in infected infants and initiation of prophylaxis against Pneumocystis carinii pneumonia (PCP) in all HIV-exposed infants beginning at age 4–6 weeks in accordance with Public Health Services (PHS) guidelines [386] 7.1.3 Prenatal laboratory assessment of HIV infection Prenatal laboratory assessment of congenital HIV infection in AF or cord blood is not recommended as the invasive procedures increases the risk of transmitting the virus from the maternal to the fetal blood stream 7.2 Laboratory assessment of HIV infection 7.2.1 HIV antibody assays The initial screening for HIV infection in adults and children is done by testing for antibodies Viral load assays are not intended for routine diagnosis but could be used in clinical management of HIV-infected persons in conjunction with clinical signs and symptoms and other laboratory markers of disease progression Detection of HIV-1 p24 antigen (Table 1) is used for routine screening in blood and plasma centers but their routine use for diagnosing HIV infection in individuals has been discouraged because the estimated average time from detection of p24 antigen to detection of HIV antibody by standard enzyme-immuno-assay (EIA) is days, and not all recently infected persons have detectable levels of p24 antigen [387] In the USA several FDA approved tests are available that enable the testing of HIV antibodies in different body fluids such as whole blood, serum, plasma, oral fluid and urine The standard testing algorithm for HIV-1 consists of initial screening with an EIA to detect antibodies to the virus Reactive specimens undergo confirmatory testing with a more specific supplemental test, usually a Western blot assay (WB) or, less commonly, IFA (Table 1) [388] Using both tests increases accuracy of the results while maintaining their sensitivity [389–391] Only specimens that are repeatedly reactive by EIA and positive by IFA or reactive by WB are considered HIV-positive and indicative of HIV infection [389,390,392,393] Incomplete antibody responses that produce negative or indeterminate results on WB tests can occur among persons recently infected with HIV who have low levels of detectable antibodies (i.e., seroconverting), persons who have end-stage HIV disease, and perinatally exposed but uninfected infants who are seroreverting (i.e., losing maternal antibody) Non-specific reactions producing indeterminate results in uninfected persons have occurred more frequently among pregnant women than among other persons [389–391,394] False-positive WB results are rare [395] E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 7.2.2 Detection of viral DNA in maternal and newborn specimens 7.2.2.1 Diagnosis of HIV infection in maternal specimens Pregnant women are screened for the presence of antibodies using the standard testing algorithm for adults (EIA plus WB or IFA) as shown in Fig For women with unknown HIV status during active labor, rapid diagnostic tests are used if rapid return of results from standard testing is not available 7.2.2.2 Diagnosis of HIV infection in newborns Infant HIV testing should be done as soon after birth as possible so appropriate treatment interventions can be implemented quickly [377,384] The standard antibody assays used for older children and adults are not useful for diagnosing children younger than 18 months as the presence of maternal antibodies makes serologic tests uninformative Therefore, a definitive diagnosis of HIV infection in early infancy requires viral diagnostic assays, including HIV-1 p24 antigen assays, nucleic acid amplification (e.g., PCR) or viral culture HIV infection can be definitively diagnosed in most infected infants by age month and in virtually all infected infants by age months HIV infection is diagnosed by at least two positive assays using two separate specimens [378] HIV DNA PCR is the preferred virologic method for diagnosing HIV infection during infancy It has 99% specificity to identify HIV proviral DNA in peripheral blood mononuclear cells (PBMC) obtained from whole blood samples collected in EDTA-containing tubes [396] A meta-analysis of published data from 271 infected children indicated that HIV DNA PCR was sensitive for the diagnosis of HIV infection during the neonatal period Thirty-eight percent of infected children had positive HIV DNA PCR tests by age 48 h, 93% by age 14 days and 96% by age 28 days No substantial change in sensitivity during the first week of life was observed, but sensitivity increased rapidly during the second week Quantitative assays that detect HIV RNA in plasma (see Section 7.2.2.3 below) appear to be as sensitive as HIV DNA PCR for early diagnosis of HIV infection in HIV-exposed infants [397–402] The specificity is comparable between the two tests, but results of HIV RNA load below 104 copies/ml should be interpreted with caution [403] Some clinicians use HIV RNA assay as the confirmatory test for infants testing HIV DNA PCR positive since it provides viral load measurement which guides treatment decisions Available quantitative RNA tests include the Amplicor HIV-1 monitor test 1.5 (Roche Diagnostics), the NASBA EasyQ HIV-1 (BioMerieux), the Quantiplex HIV RNA 3.0 (bDNA) (Bayer) and the LCx HIV RNA quantitative assay (Abbott Laboratories) assays [416–421] However, special attention should be taken where non-B HIV is expected HIV culture for the diagnosis of infection has a sensitivity that is similar to that of HIV DNA PCR [404] However, HIV culture is more complex and expensive to perform than DNA PCR, and definitive results may not be available for 2–4 weeks Both standard and immune-complex-dissociated p24 antigen tests are highly specific for HIV infection and have been used to diagnose infection in children However, the use of p24 371 antigen testing alone is not recommended because of its substantiallty reduced sensitivity and specificity compromising the critical need for timely diagnosis [405] Whether the current, more intensive antiretroviral combination regimens women may receive during pregnancy for treatment of their own HIV infection will affect diagnostic test sensitivity in their infants is unknown Similarly, if more complex regimens are administered to HIV-exposed infants for perinatal prophylaxis, the sensitivity of diagnostic assays will need to be re-examined [401] 7.2.2.3 Different subtypes HIV subtype B is the predominant viral subtype found in the U.S and western Europe Non-subtype B viruses predominate in other parts of the world, such as subtype C in regions of Africa and India and subtype E in much of southeast Asia Currently the available HIV DNA PCR commercial tests are less sensitive for detection for non-subtype B HIV, and false negative HIV DNA PCR assays have been reported in infants infected with non-subtype B HIV [406–410] Caution should be exercised in the interpretation of negative HIV DNA PCR test results in infants born to mothers who may have acquired an HIV non-B subtype Some of the currently available HIV RNA assays have improved sensitivity for detection of non-subtype B HIV infection [411–413], although even these assays may not detect some non-B subtypes, particularly group O HIV strains [414] In cases of infants where non-subtype B perinatal exposure may be suspected and HIV DNA PCR is negative, repeat testing using one of the newer RNA assays shown to be more sensitive for non-subtype B HIV is recommended (for example, the Amplicor HIV-1 monitor test 1.5, Nuclisens HIV-1 qt or Quantiplex HIV RNA 3.0 (bDNA) assays) In children with negative HIV DNA PCR and RNA assays but in whom non-subtype B infection continues to be suspected, the clinician should consult with an expert in pediatric HIV infection and the child should undergo close clinical monitoring and definitive HIV serologic testing at 18 months of age 7.2.2.4 Test algorithm for neonates HIV infection is diagnosed by two virological tests performed on separate blood samples, regardless of age The testing rules are summarized below and shown in Fig 6: (1) Initial testing is recommended by age 48 h As many as 40% of infected infants can be identified at this time Blood samples from the umbilical cord should not be used for diagnostic evaluations, because of concerns regarding potential contamination with maternal blood (2) Repeated diagnostic testing can also be considered at age 14 days in infants with negative tests at birth The diagnostic sensitivity of virological assays increases rapidly by age weeks Early identification of infection would permit discontinuation of neonatal ZDV chemoprophylaxis and a further evaluation of the need for more aggressive drug combination therapy (3) Retest infants with initially negative virological tests at age 1–2 months Using ZDV monotherapy to reduce perinatal 372 (4) (5) (6) (7) (8) (9) E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 transmission did not delay the detection of HIV in infants in PACTG protocol 076 [370,400–402,415] At age 3–6 months retest HIV-exposed children who have had repeatedly negative virological assays at birth and at age 1–2 months HIV infection can be reasonably excluded in non-breast fed infants with two or more negative virologic tests performed at age >1 month, with one of those being performed at age >4 months [386] Two or more negative HIV immunoglobulin G (IgG) antibody tests performed at age >6 months with an interval of at least month between the tests can also be used to reasonably exclude HIV infection in HIV-exposed children with no clinical or virologic laboratory evidence of HIV infection Serology after 12 months is recommended to confirm that maternal HIV antibodies transferred to the infant in utero have disappeared if there has not been previous confirmation of two negative antibody tests If the child is still antibody-positive at 12 months, then testing should be repeated between 15 and 18 months [387] Loss of HIV antibody in a child with previously negative HIV DNA PCR tests definitively confirms that the child is HIV uninfected A positive HIV antibody test at >18 months of age indicates HIV infection [378] 7.3 Summary Identification of HIV-infected women before or during pregnancy is critical to providing optimal therapy for both infected women and their children and to preventing perinatal transmission Pregnant women are screened for the presence of antibodies using the standard testing algorithm for adults (EIA plus WB or IFA) Prenatal laboratory assessment of congenital HIV infection is not recommended as it increases the risk of infecting the fetus, but extensive testing is performed to assess the infection status of the baby following delivery A definitive diagnosis of HIV infection in early infancy requires repeated testing using virological assays, with HIV DNA PCR being the currently preferred method All infected infants can be definitively diagnosed by age months An algorithm describing the laboratory diagnostic assays for HIV infection in pregnant women and neonates is shown in Fig Acknowledgments We are grateful to Galit Zemel for her extensive technical support in organizing the citations and reference list and in preparing the review for submission We also thank Zehava Yosefi for helping in reference retrieval and typing References [1] Hahne SJ, Abbink F, van Binnendijk RS, Ruijs WL, van Steenbergen JE, de Melker HE [Rubella epidemic in the Netherlands 2004/’05: awareness of congenital rubella syndrome required] Ned Tijdschr Geneeskd 2005;149(21):1174–8 [2] Melnick JL Taxonomy of viruses Prog Med Virol 1976;22:211– 21 [3] Oker-Blom C, Kalkkinen N, Kaariainen L, Pettersson RF Rubella virus contains one capsid protein and three envelope glycoproteins, E1, E2a, and E2b J Virol 1983;46:964–73 [4] Vesikari T Immune response in rubella infection Scand J Infect Dis 1972;4:1–42 [5] Mitchell LA, Zhang T, Ho M, Decarie D, Tingle AJ, Zrein M, et al Characterization of rubella virus-specific antibody responses by using a new synthetic peptide-based enzyme-linked immunosorbent assay J Clin Microbiol 1992;30:1841–7 [6] Cooper LZ, Krugman S Clinical manifestations of postnatal and congenital rubella Arch Ophthalmol 1967;77:434–9 [7] Cooper LZ The history and medical consequences of rubella Rev Infect Dis 1985;7(Suppl 1):S2–S10 [8] Coulter C, Wood R, Robson J Rubella infection in pregnancy Commun Dis Intell 1999;23:93–6 [9] Webster WS Teratogen update: congenital rubella Teratology 1998;58:13–23 [10] Horstmann DM, Schluederberg A, Emmons JE, Evans BK, Randolph MF, Andiman WA Persistence of vaccine-induced immune responses to rubella: comparison with natural infection Rev Infect Dis 1985;7(Suppl 1):S80–5 [11] Harcourt GC, Best JM, Banatvala JE Rubella-specific serum and nasopharyngeal antibodies in volunteers with naturally acquired and vaccine-induced immunity after intranasal challenge J Infect Dis 1980;142:145–55 [12] Just M, Just V, Berger R, Burkhardt F, Schilt U Duration of immunity after rubella vaccination: a long-term study in Switzerland Rev Infect Dis 1985;7(Suppl 1):S91–4 [13] Balfour Jr HH, Amren DP Rubella, measles and mumps antibodies following vaccination of children A potential rubella problem Am J Dis Child 1978;132:573–7 [14] Horstmann DM Controlling rubella: problems and perspectives Ann Intern Med 1975;83:412–7 [15] Schiff GM, Young BC, Stefanovic GM, Stamler EF, Knowlton DR, Grundy BJ, et al Challenge with rubella virus after loss of detectable vaccine-induced antibody Rev Infect Dis 1985;7(Suppl 1):S157– 63 [16] Horstmann DM, Liebhaber H, Le Bouvier GL, Rosenberg DA, Halstead SB Rubella: reinfection of vaccinated and naturally immune persons exposed in an epidemic N Engl J Med 1970;283:771–8 [17] Banatvala JE, Best JM Rubella In: Collier L, Timbury M, editors Topley and Wilson’s principles of bacteriology, virology and immunity London: Edward Arnold; 1990 p 501–31 [18] Herrmann KL Available rubella serologic tests Rev Infect Dis 1985;7(Suppl 1):S108–12 [19] Aboudy Y, Fogel A, Barnea B, Mendelson E, Yosef L, Frank T, et al Subclinical rubella reinfection during pregnancy followed by transmission of virus to the fetus J Infect 1997;34:273–6 [20] Aboudy Y, Barnea B, Yosef L, Frank T, Mendelson E Clinical rubella reinfection during pregnancy in a previously vaccinated woman J Infect 2000;41:187–9 [21] Best JM, Banatvala JE, Morgan-Capner P, Miller E Fetal infection after maternal reinfection with rubella: criteria for defining reinfection BMJ 1989;299:773–5 [22] Kleeman KT, Kiefer DJ, Halbert SP Rubella antibodies detected by several commercial immunoassays in hemagglutination inhibitionnegative sera J Clin Microbiol 1983;18:1131–7 [23] Cusi MG, Valensin PE, Cellesi C Possibility of reinfection after immunisation with RA27/3 live attenuated rubella virus Arch Virol 1993;129:337–40 [24] Fogel A, Handsher R, Barnea B Subclinical rubella in pregnancy—occurrence and outcome Isr J Med Sci 1985;21:133–8 [25] Schluederberg A, Horstmann DM, Andiman WA, Randolph MF Neutralizing and hemagglutination-inhibiting antibodies to rubella virus as indicators of protective immunity in vaccinees and naturally immune individuals J Infect Dis 1978;138:877–83 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 [26] Best JM, Harcourt GC, Banatvala JE, Congenital Rubella affecting an infant whose mother had Rubella antibodies before conception BMJ 1981;282:1235–6 [27] Best JM, Banatvala JE Rubella In: Zuckerman AJ, Banatvala JEJRP, editors Principles and practice of clinical virology Chichester: John Wiley; 2000 p 387–418 [28] Fogel A, Barnea BS, Aboudy Y, Mendelson E Rubella in pregnancy in Israel: 15 years of follow-up and remaining problems Isr J Med Sci 1996;32:300–5 [29] Banatvala JE, Best JM, O’Shea S, Dudgeon JA Persistence of rubella antibodies after vaccination: detection after experimental challenge Rev Infect Dis 1985;7(Suppl 1):S86–90 [30] Hudson P, Morgan-Capner P Evaluation of 15 commercial enzyme immunoassays for detection of Rubella specific IgM Clin Diagn Virol 1996;5:21–6 [31] Matter L, Gorgievski-Hrisoho M, Germann D Comparison of four enzyme immunoassays for detection of immunoglobulin M antibodies against rubella virus J Clin Microbiol 1994;32:2134–9 [32] Enders G, Knotek F Detection of IgM antibodies against rubella virus: comparison of two indirect ELISAs and an anti-IgM capture immunoassay J Med Virol 1986;19:377–86 [33] Thomas HI, Morgan-Capner P, Roberts A, Hesketh L Persistent rubella-specific IgM reactivity in the absence of recent primary rubella and rubella reinfection J Med Virol 1992;36:188–92 [34] Kurtz JB, Anderson MJ Cross-reactions in rubella and parvovirus specific IgM tests Lancet 1985;2:1356 [35] Thomas HI, Barrett E, Hesketh LM, Wynne A, Morgan-Capner P Simultaneous IgM reactivity by EIA against more than one virus in measles, parvovirus B19 and rubella infection J Clin Virol 1999;14:107–18 [36] Best JM, O‘Shea S Rubella virus In: Lennette EH, Schmidt NY, editors Diagnostic procedure for viral, Reckettsial and Chlamydial infections Washington, DC: American Public Health association; 1989 p 731–95 [37] Ogra PL, Chiba Y, Ogra SS, Dzierba JL, Herd JK Rubella-virus infection in juvenile rheumatoid arthritis Lancet 1975;1:1157–61 [38] Wunderli W, Zbinden R, Biedermann K Rubella IgM detection in pregnancy Lancet 1990;335:1042–3 [39] Hedman K, Seppala I Recent rubella virus infection indicated by a low avidity of specific IgG J Clin Immunol 1988;8:214–21 [40] Thomas HI, Morgan-Capner P, Rubella-specific IgG1 avidity: a comparison of methods J Virol Methods 1991;31:219–28 [41] Bottiger B, Jensen IP Maturation of rubella IgG avidity over time after acute rubella infection Clin Diagn Virol 1997;8:105–11 [42] Pullen GR, Fitzgerald MG, Hosking CS Antibody avidity determination by ELISA using thiocyanate elution J Immunol Methods 1986;86:83–7 [43] Morgan-Capner P Diagnosing rubella BMJ 1989;299:338–9 [44] Cradock-Watson JE, Miller E, Ridehalgh MK, Terry GM, Ho-Terry L Detection of rubella virus in fetal and placental tissues and in the throats of neonates after serologically confirmed rubella in pregnancy Prenat Diagn 1989;9:91–6 [45] D’Alton ME, DeCherney AH Prenatal diagnosis N Engl J Med 1993;328:114–20 [46] Tang JW, Aarons E, Hesketh LM, Strobel S, Schalasta G, Jauniaux E, et al Prenatal diagnosis of congenital rubella infection in the second trimester of pregnancy Prenat Diagn 2003;23:509–12 [47] Mace M, Cointe D, Six C, et al Diagnostic value of reverse transcription-PCR of amniotic fluid for prenatal diagnosis of congenital rubella infection in pregnant women with confirmed primary rubella infection J Clin Microbiol 2004;42(10):4818–20 [48] Skendzel LP Rubella immunity Defining the level of protective antibody Am J Clin Pathol 1996;106:170–4 [49] Stewart GL, Parkman PD, Hopps HE, Douglas RD, Hamilton JP, Meyer Jr HM Rubella-virus hemagglutination-inhibition test N Engl J Med 1967;276:554–7 [50] Enders G Serologic test combinations for safe detection of rubella infections Rev Infect Dis 1985;7(Suppl 1):S113–22 373 [51] Haukenes G, Blom H False positive rubella virus haemagglutination inhibition reactions: occurrence and disclosure Med Microbiol Immunol (Berl) 1975;161:99–106 [52] Field PR, Gong CM Diagnosis of postnatally acquired rubella by use of three enzyme-linked immunosorbent assays for specific immunoglobulins G and M and single radial hemolysis for specific immunoglobulin G J Clin Microbiol 1984;20:951–8 [53] Vaheri A, Salonen EM Evaluation of solid-phase enzymeimmunoassay procedure in immunity surveys and diagnosis of rubella J Med Virol 1980;5:171–81 [54] Shekarchi IC, Sever JL, Tzan N, Ley A, Ward LC, Madden D Comparison of hemagglutination inhibition test and enzyme-linked immunosorbent assay for determining antibody to rubella virus J Clin Microbiol 1981;13:850–4 [55] Vesikari T, Vaheri A Rubella: a method for rapid diagnosis of a recent infection by demonstration of the IgM antibodies Br Med J 1968;1:221–3 [56] Handsher R, Fogel A Modified staphylococcal absorption method used for detecting rubella-specific immunoglobin M antibodies during a rubella epidemic J Clin Microbiol 1977;5:588–92 [57] Enders G, Knotek F, Pacher U Comparison of various serological methods and diagnostic kits for the detection of acute, recent, and previous rubella infection, vaccination, and congenital infections J Med Virol 1985;16:219–32 [58] Voller A, Bidwell DE, Bartlett A Enzyme immunoassays in diagnostic medicine Theory and practice Bull World Health Organ 1976;53:55–65 [59] Field PR, Ho DW, Cunningham AL Evaluation of rubella immune status by three commercial enzyme-linked immunosorbent assays J Clin Microbiol 1988;26:990–4 [60] Salonen EM, Vaheri A, Suni J, Wager O Rheumatoid factor in acute viral infections: interference with determination of IgM, IgG, and IgA antibodies in an enzyme immunoassay J Infect Dis 1980;142:250–5 [61] Grangeot-Keros L, Pillot J, Daffos F, Forestier F Prenatal and postnatal production of IgM and IgA antibodies to rubella virus studied by antibody capture immunoassay J Infect Dis 1988;158:138–43 [62] Starkey WG, Newcombe J, Corbett KM, Liu KM, Sanders PG, Best JM Use of rubella virus E1 fusion proteins for detection of rubella virus antibodies J Clin Microbiol 1995;33:270–4 [63] Condorelli F, Stivala A, Gallo R, Musumeci E, Scata MC, Strano L, et al Improvement in establishing the period of rubella virus primary infection using a mild protein denaturant J Virol Methods 1997;66:109–12 [64] Thompson KM, Tobin JO Isolation of rubella virus from abortion material Br Med J 1970;1:264–6 [65] O’Neill HJ, Russell JD, Wyatt DE, McCaughey C, Coyle PV Isolation of viruses from clinical specimens in microtitre plates with cells inoculated in suspension J Virol Methods 1996;62:169–78 [66] Forsgren M, Carlstrom G, Strangert K Congenital rubella after maternal reinfection Scand J Infect Dis 1979;11:81–3 [67] O’Shea S, Best JM, Banatvala JE Viremia, virus excretion, and antibody responses after challenge in volunteers with low levels of antibody to rubella virus J Infect Dis 1983;148:639–47 [68] Bosma TJ, Corbett KM, O’Shea S, Banatvala JE, Best JM PCR for detection of rubella virus RNA in clinical samples J Clin Microbiol 1995;33:1075–9 [69] Bosma TJ, Corbett KM, Eckstein MB, O’Shea S, Vijayalakshmi P, Banatvala JE, et al Use of PCR for prenatal and postnatal diagnosis of congenital rubella J Clin Microbiol 1995;33:2881–7 [70] Terry GM, Ho-Terry L, Londesborough P, Rees KR Localization of the rubella E1 epitopes Arch Virol 1988;98:189–97 [71] Frey TK, Marr LD, Hemphill ML, Dominguez G Molecular cloning and sequencing of the region of the rubella virus genome coding for glycoprotein E1 Virology 1986;154:228–32 [72] Revello MG, Baldanti F, Sarasini A, Zavattoni M, Torsellini M, Gerna G Prenatal diagnosis of rubella virus infection by direct detection and semiquantitation of viral RNA in clinical samples by reverse transcription-PCR J Clin Microbiol 1997;35:708–13 374 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 [73] Katow S Rubella virus genome diagnosis during pregnancy and mechanism of congenital rubella Intervirology 1998;41:163–9 [74] Ho-Terry L, Terry GM, Londesborough P Diagnosis of foetal rubella virus infection by polymerase chain reaction J Gen Virol 1990;71(Pt 7):1607–11 [75] Givens KT, Lee DA, Jones T, Ilstrup DM Congenital rubella syndrome: ophthalmic manifestations and associated systemic disorders Br J Ophthalmol 1993;77:358–63 [76] Enders G, Bader U, Lindemann L, Schalasta G, Daiminger A Prenatal diagnosis of congenital cytomegalovirus infection in 189 pregnancies with known outcome Prenat Diagn 2001;21:362–77 [77] Cone RW, Hobson AC, Huang ML Coamplified positive control detects inhibition of polymerase chain reactions J Clin Microbiol 1992;30:3185–9 [78] Naessens A, Casteels A, Decatte L, Foulon W A serologic strategy for detecting neonates at risk for congenital cytomegalovirus infection J Pediatr 2005;146(2):194–7 [79] Daiminger A, Bader U, Enders G Pre- and periconceptional primary cytomegalovirus infection: risk of vertical transmission and congenital disease Obstet Gynecol Surv 2005;60(7):420–2 [80] Rowe WP, Hartley JW, Waterman S, Turner HC, Huebner RJ Cytopathogenic agent resembling human salivary gland virus recovered from tissue cultures of human adenoids Proc Soc Exp Biol Med 1956;92:418–24 [81] Craig JM, Macauley JC, Weller TH, Wirth P Isolation of intranuclear inclusion producing agents from infants with illnesses resembling cytomegalic inclusion disease Proc Soc Exp Biol Med 1957;94:4–12 [82] Smith MG Propagation in tissue cultures of a cytopathogenic virus from human salivary gland virus (SGV) disease Proc Soc Exp Biol Med 1956;92:424–30 [83] Liesnard C, Donner C, Brancart F, Gosselin F, Delforge ML, Rodesch F Prenatal diagnosis of congenital cytomegalovirus infection: prospective study of 237 pregnancies at risk Obstet Gynecol 2000;95:881–8 [84] Stagno S, Pass RF, Dworsky ME, Henderson RE, Moore EG, Walton PD, et al Congenital cytomegalovirus infection: the relative importance of primary and recurrent maternal infection N Engl J Med 1982;306:945–9 [85] Stagno S, Pass RF, Cloud G, Britt WJ, Henderson RE, Walton PD, et al Primary cytomegalovirus infection in pregnancy Incidence, transmission to fetus, and clinical outcome JAMA 1986;256:1904–8 [86] Boppana SB, Pass RF, Britt WJ, Stagno S, Alford CA Symptomatic congenital cytomegalovirus infection: neonatal morbidity and mortality Pediatr Infect Dis J 1992;11:93–9 [87] Raynor BD Cytomegalovirus infection in pregnancy Semin Perinatol 1993;17:394–402 [88] Lazzarotto T, Gabrielli L, Lanari M, et al Congenital cytomegalovirus infection: recent advances in the diagnosis of maternal infection Hum Immunol 2004;65(5):410–5 [89] Boppana SB, Miller J, Britt WJ Transplacentally acquired antiviral antibodies and outcome in congenital human cytomegalovirus infection Viral Immunol 1996;9:211–8 [90] Gaytant MA, Steegers EA, Semmekrot BA, Merkus HM, Galama JM Congenital cytomegalovirus infection: review of the epidemiology and outcome Obstet Gynecol Surv 2002;57:245–56 [91] Lipitz S, Yagel S, Shalev E, Achiron R, Mashiach S, Schiff E Prenatal diagnosis of fetal primary cytomegalovirus infection Obstet Gynecol 1997;89:763–7 [92] Melish ME, Hanshaw JB Congenital cytomegalovirus infection Developmental progress of infants detected by routine screening Am J Dis Child 1973;126:190–4 [93] Reimer CB, Black CM, Phillips DJ, Logan LC, Hunter EF, Pender BJ, et al The specificity of fetal IgM: antibody or anti-antibody? Ann N Y Acad Sci 1975;254:77–93 [94] Stagno S, Pass RF, Reynolds DW, Moore MA, Nahmias AJ, Alford CA Comparative study of diagnostic procedures for congenital cytomegalovirus infection Pediatrics 1980;65:251–7 [95] Revello MG, Baldanti F, Furione M, Sarasini A, Percivalle E, Zavattoni M, et al Polymerase chain reaction for prenatal diagnosis of [96] [97] [98] [99] [100] [101] [102] [103] [104] [105] [106] [107] [108] [109] [110] [111] [112] [113] [114] congenital human cytomegalovirus infection J Med Virol 1995;47: 462–6 Revello MG, Sarasini A, Zavattoni M, Baldanti F, Gerna G Improved prenatal diagnosis of congenital human cytomegalovirus infection by a modified nested polymerase chain reaction J Med Virol 1998;56:99–103 Bodeus M, Hubinont C, Bernard P, Bouckaert A, Thomas K, Goubau P Prenatal diagnosis of human cytomegalovirus by culture and polymerase chain reaction: 98 pregnancies leading to congenital infection Prenat Diagn 1999;19:314–7 Catanzarite V, Dankner WM Prenatal diagnosis of congenital cytomegalovirus infection: false-negative amniocentesis at 20 weeks’ gestation Prenat Diagn 1993;13:1021–5 Gouarin S, Palmer P, Cointe D, Rogez S, Vabret A, Rozenberg F, et al Congenital HCMV infection: a collaborative and comparative study of virus detection in amniotic fluid by culture and by PCR J Clin Virol 2001;21:47–55 Donner C, Liesnard C, Brancart F, Rodesch F Accuracy of amniotic fluid testing before 21 weeks’ gestation in prenatal diagnosis of congenital cytomegalovirus infection Prenat Diagn 1994;14:1055–9 Chou S, Merigan TC Rapid detection and quantitation of human cytomegalovirus in urine through DNA hybridization N Engl J Med 1983;308:921–5 Spector SA, Rua JA, Spector DH, McMillan R Detection of human cytomegalovirus in clinical specimens by DNA–DNA hybridization J Infect Dis 1984;150:121–6 Vonsover A, Gotlieb-Stematsky T, Sayar Y, Bardov L, Manor Y, Siegal B Detection of CMV in urine: comparison between DNA–DNA hybridization, virus isolation, and immunoelectron microscopy J Virol Methods 1987;16:29–37 Demmler GJ, Buffone GJ, Schimbor CM, May RA Detection of cytomegalovirus in urine from newborns by using polymerase chain reaction DNA amplification J Infect Dis 1988;158:1177–84 Lazzarotto T, Guerra B, Spezzacatena P, Varani S, Gabrielli L, Pradelli P, et al Prenatal diagnosis of congenital cytomegalovirus infection J Clin Microbiol 1998;36:3540–4 Levy R, Najioullah F, Thouvenot D, Bosshard S, Aymard M, Lina B Evaluation and comparison of PCR and hybridization methods for rapid detection of cytomegalovirus in clinical samples J Virol Methods 1996;62:103–11 Boppana SB, Fowler KB, Britt WJ, Stagno S, Pass RF Symptomatic congenital cytomegalovirus infection in infants born to mothers with preexisting immunity to cytomegalovirus Pediatrics 1999;104: 55–60 Boppana SB, Rivera LB, Fowler KB, Mach M, Britt WJ Intrauterine transmission of cytomegalovirus to infants of women with preconceptional immunity N Engl J Med 2001;344:1366–71 Kyriazopoulou V, Bondis J, Frantzidou F, Athanasiadis A, Diza E, Simitsopoulou M, et al Prenatal diagnosis of fetal cytomegalovirus infection in seropositive pregnant women Eur J Obstet Gynecol Reprod Biol 1996;69:91–5 Joassin L, Reginster M Elimination of nonspecific cytomegalovirus immunoglobulin M activities in the enzyme-linked immunosorbent assay by using anti-human immunoglobulin G J Clin Microbiol 1986;23:576–81 Deyi YM, Goubau P, Bodeus M, False-positive IgM antibody tests for cytomegalovirus in patients with acute Epstein-Barr virus infection Eur J Clin Microbiol Infect Dis 2000;19:557–60 Moller E, Strom H, Al-Balaghi S Role of polyclonal activation in specific immune responses Relevance for findings of antibody activity in various diseases Scand J Immunol 1980;12:177–82 Re MC, Landini MP IgM to human cytomegalovirus: comparison of two enzyme immunoassays and IgM reactivity to viral polypeptides detected by immunoblotting J Clin Lab Anal 1989;3:169– 73 Kangro HO, Griffiths PD, Huber TJ, Heath RB Specific IgM class antibody production following infection with cytomegalovirus J Med Virol 1982;10:203–12 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 [115] Demmler GJ, Six HR, Hurst SM, Yow MD Enzyme-linked immunosorbent assay for the detection of IgM-class antibodies to cytomegalovirus J Infect Dis 1986;153:1152–5 [116] Lazzarotto T, Maine GT, Dal Monte P, Ripalti A, Landini MP A novel Western blot test containing both viral and recombinant proteins for anticytomegalovirus immunoglobulin M detection J Clin Microbiol 1997;35:393–7 [117] Lazzarotto T, Ripalti A, Bergamini G, Battista MC, Spezzacatena P, Campanini F, et al Development of a new cytomegalovirus (CMV) immunoglobulin M (IgM) immunoblot for detection of CMV-specific IgM J Clin Microbiol 1998;36:3337–41 [118] Maine GT, Lazzarotto T, Chovan LE, Flanders R, Landini MP The DNA-binding protein pUL57 of human cytomegalovirus: comparison of specific immunoglobulin M (IgM) reactivity with IgM reactivity to other major target antigens Clin Diagn Lab Immunol 1996;3:358– 60 [119] Schaefer L, Cesario A, Demmler G, Plotkin S, Stagno S, Elrod C, et al Evaluation of Abbott CMV-M enzyme immunoassay for detection of cytomegalovirus immunoglobulin M antibody J Clin Microbiol 1988;26:2041–3 [120] Zerbini M, Musiani M, Gentilomi G, La Placa M Detection of specific immunoglobulin M antibodies to cytomegalovirus by using monoclonal antibody to immunoglobulin M in an indirect immunofluorescence assay J Clin Microbiol 1986;24:166–8 [121] van Zanten J, Harmsen MC, van der Giessen M, van der Bij W, Prop J, de Leij L, et al Humoral immune response against human cytomegalovirus (HCMV)-specific proteins after HCMV infection in lung transplantation as detected with recombinant and naturally occurring proteins Clin Diagn Lab Immunol 1995;2:214–8 [122] Greijer AE, van de Crommert JM, Stevens SJ, Middeldorp JM Molecular fine-specificity analysis of antibody responses to human cytomegalovirus and design of novel synthetic-peptide-based serodiagnostic assays J Clin Microbiol 1999;37:179–88 [123] Daiminger A, Bader U, Eggers M, Lazzarotto T, Enders G Evaluation of two novel enzyme immunoassays using recombinant antigens to detect cytomegalovirus-specific immunoglobulin M in sera from pregnant women J Clin Virol 1999;13:161–71 [124] Genser B, Truschnig-Wilders M, Stunzner D, Landini MP, HalwachsBaumann G Evaluation of five commercial enzyme immunoassays for the detection of human cytomegalovirus-specific IgM antibodies in the absence of a commercially available gold standard Clin Chem Lab Med 2001;39:62–70 [125] Maine GT, Stricker R, Schuler M, Spesard J, Brojanac S, Iriarte B, et al Development and clinical evaluation of a recombinant-antigen-based cytomegalovirus immunoglobulin M automated immunoassay using the Abbott AxSYM analyzer J Clin Microbiol 2000;38:1476–81 [126] Weber B, Berger A, Rabenau H Human cytomegalovirus infection: diagnostic potential of recombinant antigens for cytomegalovirus antibody detection J Virol Methods 2001;96:157–70 [127] Lazzarotto T, Varani S, Spezzacatena P, Gabrielli L, Pradelli P, Guerra B, et al IgG avidity and IgM detected by blot as diagnostic tools to identify pregnant women at risk of transmitting cytomegalovirus Viral Immunol 2000;13:137–41 [128] Blackburn NK, Besselaar TG, Schoub BD, O’Connell KF Differentiation of primary cytomegalovirus infection from reactivation using the urea denaturation test for measuring antibody avidity J Med Virol 1991;33:6–9 [129] Fowler KB, Stagno S, Pass RF, Britt WJ, Boll TJ, Alford CA The outcome of congenital cytomegalovirus infection in relation to maternal antibody status N Engl J Med 1992;326:663–7 [130] Sipewa MJ, Goubau P, Bodeus M Evaluation of a cytomegalovirus glycoprotein B recombinant enzyme immunoassay to discriminate between a recent and a past infection J Clin Microbiol 2002;40:3689–93 [131] Lazzarotto T, Spezzacatena P, Varani S, Gabrielli L, Pradelli P, Guerra B, et al Anticytomegalovirus (anti-CMV) immunoglobulin G avidity in identification of pregnant women at risk of transmitting congenital CMV infection Clin Diagn Lab Immunol 1999;6:127–9 375 [132] Lazzarotto T, Varani S, Gabrielli L, Spezzacatena P, Landini MP New advances in the diagnosis of congenital cytomegalovirus infection Intervirology 1999;42:390–7 [133] Lazzarotto T, Galli C, Pulvirenti R, Rescaldani R, Vezzo R, La Gioia A, et al Evaluation of the Abbott AxSYM cytomegalovirus (CMV) immunoglobulin M (IgM) assay in conjunction with other CMV IgM tests and a CMV IgG avidity assay Clin Diagn Lab Immunol 2001;8:196–8 [134] Bodeus M, Goubau P Predictive value of maternal-IgG avidity for congenital human cytomegalovirus infection J Clin Virol 1999;12: 3–8 [135] Bodeus M, Beulne D, Goubau P Ability of three IgG-avidity assays to exclude recent cytomegalovirus infection Eur J Clin Microbiol Infect Dis 2001;20:248–52 [136] Bodeus M, Van Ranst M, Bernard P, Hubinont C, Goubau P Anticytomegalovirus IgG avidity in pregnancy: a 2-year prospective study Fetal Diagn Ther 2002;17:362–6 [137] Grangeot-Keros L, Mayaux MJ, Lebon P, Freymuth F, Eugene G, Stricker R, et al Value of cytomegalovirus (CMV) IgG avidity index for the diagnosis of primary CMV infection in pregnant women J Infect Dis 1997;175:944–6 [138] Prince HE, Leber AL Validation of an in-house assay for cytomegalovirus immunoglobulin G (CMV IgG) avidity and relationship of avidity to CMV IgM levels Clin Diagn Lab Immunol 2002;9:824–7 [139] Ruellan-Eugene G, Barjot P, Campet M, Vabret A, Herlicoviez M, Muller G, et al Evaluation of virological procedures to detect fetal human cytomegalovirus infection: avidity of IgG antibodies, virus detection in amniotic fluid and maternal serum J Med Virol 1996;50:9–15 [140] Schoppel K, Kropff B, Schmidt C, Vornhagen R, Mach M The humoral immune response against human cytomegalovirus is characterized by a delayed synthesis of glycoprotein-specific antibodies J Infect Dis 1997;175:533–44 [141] Baccard-Longere M, Freymuth F, Cointe D, Seigneurin JM, GrangeotKeros L Multicenter evaluation of a rapid and convenient method for determination of cytomegalovirus immunoglobulin G avidity Clin Diagn Lab Immunol 2001;8:429–31 [142] Mace M, Sissoeff L, Rudent A, Grangeot-Keros L A serological testing algorithm for the diagnosis of primary CMV infection in pregnant women Prenat Diagn 2004;24(11):861–3 [143] Daiminger A, Bader U, Enders G An enzyme linked immunoassay using recombinant antigens for differentiation of primary from secondary or past CMV infections in pregnancy J Clin Virol 1998;11:93–102 [144] Eggers M, Metzger C, Enders G Differentiation between acute primary and recurrent human cytomegalovirus infection in pregnancy, using a microneutralization assay J Med Virol 1998;56:351–8 [145] Eggers M, Bader U, Enders G Combination of microneutralization and avidity assays: improved diagnosis of recent primary human cytomegalovirus infection in single serum sample of second trimester pregnancy J Med Virol 2000;60:324–30 [146] Eggers M, Radsak K, Enders G, Reschke M Use of recombinant glycoprotein antigens gB and gH for diagnosis of primary human cytomegalovirus infection during pregnancy J Med Virol 2001;63:135–42 [147] Hodinka RL Human cytomegalovirus In: Murphy PR, Baron EJ, Pfaller MA, Tenover FC, Yolken RH, editors Manual of Clinical microbiology 1999 p 889–991 [148] Gerna G, Baldanti F, Percivalle E, Zavattoni M, Campanini G, Revello MG Early identification of human cytomegalovirus strains by the shell vial assay is prevented by a novel amino acid substitution in UL123 IE1 gene product J Clin Microbiol 2003;41:4494–5 [149] Zipeto D, Sarasini A, Rossi F, Baldanti F, Revello MG, Milanesi G, et al Identification of human cytomegalovirus strain with immediate-early (IE) antigen-specific monoclonal antibody is prevented by point mutation in IE gene J Clin Microbiol 1994;32: 1402–5 376 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 [150] Donner C, Liesnard C, Content J, Busine A, Aderca J, Rodesch F Prenatal diagnosis of 52 pregnancies at risk for congenital cytomegalovirus infection Obstet Gynecol 1993;82:481–6 [151] Grose C, Meehan T, Weiner CP Prenatal diagnosis of congenital cytomegalovirus infection by virus isolation after amniocentesis Pediatr Infect Dis J 1992;11:605–7 [152] Hagay ZJ, Biran G, Ornoy A, Reece EA Congenital cytomegalovirus infection: a long-standing problem still seeking a solution Am J Obstet Gynecol 1996;174:241–5 [153] Nicolini U, Kustermann A, Tassis B, Fogliani R, Galimberti A, Percivalle E, et al Prenatal diagnosis of congenital human cytomegalovirus infection Prenat Diagn 1994;14:903–6 [154] Skvorc-Ranko R, Lavoie H, St-Denis P, Villeneuve R, Gagnon M, Chicoine R, et al Intrauterine diagnosis of cytomegalovirus and rubella infections by amniocentesis CMAJ 1991;145:649–54 [155] Weiner CP, Grose C Prenatal diagnosis of congenital cytomegalovirus infection by virus isolation from amniotic fluid Am J Obstet Gynecol 1990;163:1253–5 [156] Avidor B, Efrat G, Weinberg M, Kra-oz Z, Satinger J, MitraniRosenbaum S, et al Insight into the intrinsic sensitivity of the PCR assay used to detect CMV infection in amniotic fluid specimens J Clin Virol 2004;29:260–70 [157] Boeckh M, Huang M, Ferrenberg J, Stevens-Ayers T, Stensland L, Nichols WG, et al Optimization of quantitative detection of cytomegalovirus DNA in plasma by real-time PCR J Clin Microbiol 2004;42:1142–8 [158] Gouarin S, Vabret A, Gault E, Petitjean J, Regeasse A, Hurault de Ligny B, et al Quantitative analysis of HCMV DNA load in whole blood of renal transplant patients using real-time PCR assay J Clin Virol 2004;29:194–201 [159] Piiparinen H, Hockerstedt K, Gronhagen-Riska C, Lautenschlager I Comparison of two quantitative CMV PCR tests, Cobas Amplicor CMV Monitor and TaqMan assay, and pp65-antigenemia assay in the determination of viral loads from peripheral blood of organ transplant patients J Clin Virol 2004;30:258–66 [160] Nedelec O, Bellagra N, Devisme L, Hober D, Wattre P, Dewilde A Congenital human cytomegalovirus infection: value of human cytomegalovirus DNA quantification in amniotic fluid Ann Biol Clin (Paris) 2002;60:201–7 [161] Revello MG, Zavattoni M, Furione M, Baldanti F, Gerna G Quantification of human cytomegalovirus DNA in amniotic fluid of mothers of congenitally infected fetuses J Clin Virol 1999;37:3350–2 [162] Picone O, Costa JM, Leruez-Ville M, Ernault P, Olivi M, Ville Y, et al Cytomegalovirus (CMV) glycoprotein B genotype and CMV DNA load in the amniotic fluid of infected fetuses Prenat Diagn 2004;24(12):1001–6 [163] Gouarin S, Gault E, Vabret A, Cointe D, Rozenberg F, Grangeot-Keros L, et al Real-time PCR quantification of human cytomegalovirus DNA in amniotic fluid samples from mothers with primary infection J Clin Microbiol 2002;40:1767–72 [164] Guerra B, Lazzarotto T, Quarta S, Lanari M, Bovicelli L, Nicolosi A, et al Prenatal diagnosis of symptomatic congenital cytomegalovirus infection Am J Obstet Gynecol 2000;183:476–82 [165] Herrmann B, Larsson VC, Rubin CJ, Sund F, Eriksson BM, Arvidson J, et al Comparison of a duplex quantitative real-time PCR assay and the COBAS Amplicor CMV Monitor test for detection of cytomegalovirus J Clin Microbiol 2004;42:1909–14 [166] Kalpoe JS, Kroes AC, de Jong MD, Schinkel J, de Brouwer CS, Beersma MF, et al Validation of clinical application of cytomegalovirus plasma DNA load measurement and definition of treatment criteria by analysis of correlation to antigen detection J Clin Microbiol 2004;42:1498–504 [167] O’Riordan M, O’Gorman C, Morgan C, Mc Cafferkey M, Henry G, McKenna PI Sera prevalence of varicella zoster virus in pregnant women in Dublin Ir J Med Sci 2000;169:288 [168] Leikin E, Figueroa R, Bertkau A, Lysikiewicz A, Visintainer P, Tejani N Seronegativity to varicella-zoster virus in a tertiary care obstetric population Obstet Gynecol 1997;90:511–3 [169] Gershon AA, Raker R, Steinberg S, Topf-Olstein B, Drusin LM Antibody to varicella-zoster virus in parturient women and their offspring during the first year of life Pediatrics 1976;58:692–6 [170] Linder N, Waintraub I, Smetana Z, Barzilai A, Lubin D, Mendelson E, et al Placental transfer and decay of varicella-zoster virus antibodies in preterm infants J Pediatr 2000;137:85–9 [171] Enders G, Miller E, Cradock-Watson J, Bolley I, Ridehalgh M Consequences of varicella and herpes zoster in pregnancy: prospective study of 1739 cases Lancet 1994;343:1548–51 [172] Chapman SJ Varicella in pregnancy Semin Perinatol 1998;22:339–46 [173] Pastuszak AL, Levy M, Schick B, Zuber C, Feldkamp M, Gladstone J, et al Outcome after maternal varicella infection in the first 20 weeks of pregnancy N Engl J Med 1994;330:901–5 [174] Paryani SG, Arvin AM Intrauterine infection with varicella-zoster virus after maternal varicella N Engl J Med 1986;314:1542–6 [175] Dufour P, de Bievre P, Vinatier D, Tordjeman N, Da Lage B, Vanhove J, et al Varicella and pregnancy Eur J Obstet Gynecol Reprod Biol 1996;66:119–23 [176] Nikkels AF, Delbecque K, Pierard GE, Wienkotter B, Schalasta G, Enders M Distribution of varicella-zoster virus DNA and gene products in tissues of a first-trimester varicella-infected fetus J Infect Dis 2005;191(4):540–5 [177] Sauerbrei A, Pawlak J, Luger C, Wutzler P Intracerebral varicellazoster virus reactivation in congenital varicella syndrome Dev Med Child Neurol 2003;45(12):837–40 [178] Enders G Varicella-zoster virus infection in pregnancy Prog Med Virol 1984;29:166–96 [179] Prober CG, Gershon AA, Grose C, McCracken Jr GH, Nelson JD Consensus: varicella-zoster infections in pregnancy and the perinatal period Pediatr Infect Dis J 1990;9:865–9 [180] Grose C Varicella infection during pregnancy Herpes 1999;6:33–7 [181] Stein O, Smetana Z, Dulitzki M, Stevenson DK, Mashiach S, Seidman DS, et al Zoster antibody status in pregnant women exposed to varricella Isr J Obst Gynecol 1996;7:106–10 [182] Linder N, Ferber A, Kopilov U, Smetana Z, Barzilai A, Mendelson E, et al Reported exposure to chickenpox: a predictor of positive anti-varicella-zoster antibodies in parturient women Fetal Diagn Ther 2001;16:423–6 [183] Mouly F, Mirlesse V, Meritet JF, Rozenberg F, Poissonier MH, Lebon P, et al Prenatal diagnosis of fetal varicella-zoster virus infection with polymerase chain reaction of amniotic fluid in 107 cases Am J Obstet Gynecol 1997;177:894–8 [184] Steinberg SP, Gershon AA Measurement of antibodies to varicellazoster virus by using a latex agglutination test J Clin Microbiol 1991;29:1527–9 [185] Landry ML, Ferguson D Comparison of latex agglutination test with enzyme-linked immunosorbent assay for detection of antibody to varicella-zoster virus J Clin Microbiol 1993;31:3031–3 [186] Leventon-Kriss S, Yoffe R, Rannon L, Modan M Seroepidemiologic aspects of varicella zoster virus infections in an Israeli Jewish population Isr J Med Sci 1978;14:766–70 [187] Haikin H, Leventon-Kriss S, Sarov I Antibody to varicella-zoster virus-induced membrane antigen: immunoperoxidase assay with airdried target cells J Infect Dis 1979;140:601–4 [188] Shehab Z, Brunell PA Enzyme-linked immunosorbent assay for susceptibility to varicella J Infect Dis 1983;148:472–6 [189] Waner LJ, Weller TH In: Emmons NJSRW, editor Varicella zoster virus diagnostic procedures for viral, Rickettsial and Chlamydial infection ApHA American public Health Association; 1989 p 379–406 [190] Qureshi F, Jacques SM Maternal varicella during pregnancy: correlation of maternal history and fetal outcome with placental histopathology Hum Pathol 1996;27:191–5 [191] Puchhammer-Stockl E, Kunz C, Wagner G, Enders G Detection of varicella zoster virus (VZV) DNA in fetal tissue by polymerase chain reaction J Perinat Med 1994;22:65–9 [192] Mehraein Y, Rehder H, Draeger HG, Froster-Iskenius UG, Schwinger E, Holzgreve W [Diagnosis of fetal virus infections by in situ hybridization] Geburtshilfe Frauenheilkd 1991;51:984–9 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 [193] Gottardi H, Rabensteiner A, Delucca A, Lobbiani A, Nocco A, Colucci G [Detection of varicella virus with a DNA probe in fetal blood and amniotic fluid] Geburtshilfe Frauenheilkd 1991;51:63–4 [194] Schmidt NJ, Gallo D, Devlin V, Woodie JD, Emmons RW Direct immunofluorescence staining for detection of herpes simplex and varicella-zoster virus antigens in vesicular lesions and certain tissue specimens J Clin Microbiol 1980;12:651–5 [195] Ziegler T Detection of varicella-zoster viral antigens in clinical specimens by solid-phase enzyme immunoassay J Infect Dis 1984;150:149–54 [196] Brinker JP, Doern GV Comparison of MRC-5 and A-549 cells in conventional culture tubes and shell vial assays for the detection of varicella-zoster virus Diagn Microbiol Infect Dis 1993;17:75–7 [197] Gershon AA, LaRussa P, Steinberg SP Varicella-zoster virus In: Murray RR, Barron EJ, Pfaller MA, Tenover FC, Yolken RH, editors Manual of Clinical Microbiology 7th ed Washington, DC: American Society for Microbiology; 1999 p 900–11 [198] Arvin AM In: Fields B, editor Varicella-zoster virus virology New York: Raven Press; 1995 p 2547–86 [199] Levin MJ, Leventhal S, Masters HA Factors influencing quantitative isolation of varicella-zoster virus J Clin Microbiol 1984;19:880–3 [200] Arvin AM Varicella-zoster virus Clin Microbiol Rev 1996;9:361–81 [201] Rawlinson WD, Dwyer DE, Gibbons VL, Cunningham AL Rapid diagnosis of varicella-zoster virus infection with a monoclonal antibody based direct immunofluorescence technique J Virol Methods 1989;23:13–8 [202] Sadick NS, Swenson PD, Kaufman RL, Kaplan MH Comparison of detection of varicella-zoster virus by the Tzanck smear, direct immunofluorescence with a monoclonal antibody, and virus isolation J Am Acad Dermatol 1987;17:64–9 [203] Perez JL, Garcia A, Niubo J, Salva J, Podzamczer D, Martin R Comparison of techniques and evaluation of three commercial monoclonal antibodies for laboratory diagnosis of varicella-zoster virus in mucocutaneous specimens J Clin Microbiol 1994;32:1610–3 [204] Nahass GT, Goldstein BA, Zhu WY, Serfling U, Penneys NS, Leonardi CL Comparison of Tzanck smear, viral culture, and DNA diagnostic methods in detection of herpes simplex and varicella-zoster infection JAMA 1992;268:2541–4 [205] Kido S, Ozaki T, Asada H, Higashi K, Kondo K, Hayakawa Y, et al Detection of varicella-zoster virus (VZV) DNA in clinical samples from patients with VZV by the polymerase chain reaction J Clin Microbiol 1991;29:76–9 [206] Weidmann M, Meyer-Konig U, Hufert FT Rapid detection of herpes simplex virus and varicella-zoster virus infections by real-time PCR J Clin Microbiol 2003;41:1565–8 [207] Cuthbertson G, Grose C Biotinylated and radioactive DNA probes for detection of varicella-zoster virus genome in infected human cells Mol Cell Probes 1988;2:197–207 [208] Vonsover A, Leventon-Kriss S, Langer A, Smetana Z, Zaizov R, Potaznick D, et al Detection of varicella-zoster virus in lymphocytes by DNA hybridization J Med Virol 1987;21:57–66 [209] Stranska R, Schuurman R, de Vos M, van Loon AM Routine use of a highly automated and internally controlled real-time PCR assay for the diagnosis of herpes simplex and varicella-zoster virus infections J Clin Virol 2004;30:39–44 [210] Stocher M, Holzl G, Stekel H, Berg J Automated detection of five human herpes virus DNAs by a set of LightCycler PCRs complemented with a single multiple internal control J Clin Virol 2004;29:171–8 [211] Whitley RJ Neonatal herpes simplex virus infections Clin Perinatol 1988;15:903–16 [212] Lafferty WE, Downey L, Celum C, Wald A Herpes simplex virus type as a cause of genital herpes: impact on surveillance and prevention J Infect Dis 2000;181:1454–7 [213] Nahmias AJ, Keyserling HL, Lee FK Herpes simplex virus and In: Evans AS, editor Viral infections of humans: epidemiology and control 3rd ed 1989 p 393–417 [214] Corey L, Spear PG Infections with herpes simplex viruses (2) N Engl J Med 1986;314:749–57 377 [215] Nilson A, Myrmel H Changing trends in genital herpes simplex virus infection Obstet Gynecol 2000;79:693–6 [216] Hutto C, Arvin A, Jacobs R, Steele R, Stagno S, Lyrene R, et al Intrauterine herpes simplex virus infections J Pediatr 1987;110:97– 101 [217] Stagno S, Whitley RJ Herpes virus infections in the neonate and children In: K.K.H., editor Sexully transmitted diseases New York: McGraw Hill; 1990 p 872–3 [218] Brown ZA, Benedetti J, Selke S, Ashley R, Watts DH, Corey L Asymptomatic maternal shedding of herpes simplex virus at the onset of labor: relationship to preterm labor Obstet Gynecol 1996;87: 483–8 [219] Brown ZA, Benedetti J, Ashley R, Burchett S, Selke S, Berry S, et al Neonatal herpes simplex virus infection in relation to asymptomatic maternal infection at the time of labor N Engl J Med 1991;324:1247–52 [220] Malkin JE, Beumont MG Herpes simplex virus infection in pregnancy Herpes 1999;6(2):47–50 [221] Gibbs RS, Dinsmoor MJ, Newton ER, Ramamurthy RS A randomized trial of intrapartum versus immediate postpartum treatment of women with intra-amniotic infection Obstet Gynecol 1988;72:823–8 [222] Blanchier H, Huraux JM, Huraux-Rendu C, Sainte-Croix le Baleur A Genital herpes and pregnancy—preventive measures Eur J Obstet Gynecol Reprod Biol 1994;53:33–8 [223] Brown ZA, Selke S, Zeh J, Kopelman J, Maslow A, Ashley RL, et al The acquisition of herpes simplex virus during pregnancy N Engl J Med 1997;337:509–15 [224] ACOG criteria set Cesarean delivery Number 35, July 1998 Committee on quality assessment American College of Obstetricians and Gynecologists Int J Gynaecol Obstet 1998;63:206–7 [225] Amann ST, Fagnant RJ, Chartrand SA, Monif GR Herpes simplex infection associated with short-term use of a fetal scalp electrode A case report J Reprod Med 1992;37:372–4 [226] Kimura H, Futamura M, Kito H, Ando T, Goto M, Kuzushima K, et al Detection of viral DNA in neonatal herpes simplex virus infections: frequent and prolonged presence in serum and cerebrospinal fluid J Infect Dis 1991;164:289–93 [227] Malm G, Forsgren M Neonatal herpes simplex virus infections: HSV DNA in cerebrospinal fluid and serum Arch Dis Child Fetal Neonatal Ed 1999;81:F24–9 [228] Lakeman FD, Whitley RJ Diagnosis of herpes simplex encephalitis: application of polymerase chain reaction to cerebrospinal fluid from brain-biopsied patients and correlation with disease National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group J Infect Dis 1995;171:857–63 [229] Kimberlin DW, Lakeman FD, Arvin AM, Prober CG, Corey L, Powell DA, et al Application of the polymerase chain reaction to the diagnosis and management of neonatal herpes simplex virus disease National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group J Infect Dis 1996;174:1162–7 [230] Ashley LR Type specific antibodies to HSV1 and Review of methology Herpes 1998;5:33–8 [231] Cleary KL, Pare E, Stamilio D, Macones GA Type-specific screening for asymptomatic herpes infection in pregnancy: a decision analysis BJOG 2005;112(6):731–6 [232] Baker D, Brown Z, Hollier LM, et al Cost-effectiveness of herpes simplex virus type serologic testing and antiviral therapy in pregnancy Am J Obstet Gynecol 2004;191(6):2074–84 [233] Twagira M, Hadzic N, Smith M, et al Disseminated neonatal herpes simplex virus (HSV) type infection diagnosed by HSV DNA detection in blood and successfully managed by liver transplantation Eur J Pediatr 2004;163(3):166–9 [234] Leventon-Kriss S, Rannon L, Smetana Z, Yoffe R Increase in laboratory-confermed cases of herpes genitalis and neonatal herpes infection in Israel Isr J Med Sci 1983;19:946–9 [235] Kimberlin DW, Whitley RJ Antiviral resistance: mechanisms, clinical significance, and future implications J Antimicrob Chemother 1996;37:403–21 378 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 [236] Stamos JK, Rowley AH Timely diagnosis of congenital infections Pediatr Clin North Am 1994;41:1017–33 [237] Ashley LR, Corey L Herpes simplex viruses In: Schmidt N, Whitely R, Emmons, editors Diagnostic procedures for viral, Rickettial and Chlamydial infection ApHA American Public Health Association; 1989 p 265–319 [238] Rawls WE, Iwamato K, Adam E, Melnick JL Measurement of antibodies to herpesvirus type and type in human sera J Immunol 1970;104:599–606 [239] Strick L, Wald A Type-specific testing for herpes simplex virus Expert Rev Mol Diagn 2004;4(4):443–53 [240] Ashley RL, Wald A Genital herpes: review of the epidemic and potential use of type-specific serology Clin Microbiol Rev 1999;12:1–8 [241] Ashley RL Performance and use of HSV type-specific serology test kits Herpes 2002;9:38–45 [242] Olofsson S, Lundstrom M, Marsden H, Jeansson S, Vahlne A Characterization of a herpes simplex virus type 2-specified glycoprotein with affinity for N-acetylgalactosamine-specific lectins and its identification as g92K or gG J Gen Virol 1986;67(Pt 4):737–44 [243] Proffit MR, Schindler SA Rapid detection of HSV with enzyme-linked virus inducible system employing a genetically modified cell line Clin Diagn Virol 1995;4:175–82 [244] Kimberlin DW Advances in the treatment of neonatal herpes simplex infections Rev Med Virol 2001;11:157–63 [245] Espy MJ, Uhl JR, Mitchell PS, Thorvilson JN, Svien KA, Wold AD, et al Diagnosis of herpes simplex virus infections in the clinical laboratory by LightCycler PCR J Clin Microbiol 2000;38:795–9 [246] Ozawa K, Ayub J, Hao YS, Kurtzman G, Shimada T, Young N Novel transcription map for the B19 (human) pathogenic parvovirus J Virol 1987;61:2395–406 [247] van Elsacker-Niele AM, Kross AC Human parvovirus B19: relevance in internal medicine Neth J Med 1999;54(6):221–30 [248] Anderson MJ, Jones SE, Fisher-Hoch SP, Lewis E, Hall SM, Bartlett CL, et al Human parvovirus, the cause of erythema infectiosum (fifth disease)? Lancet 1983;1:1378 [249] Anderson MJ, Pattison JR The human parvovirus Brief review Arch Virol 1984;82:137–48 [250] Anderson MJ, Lewis E, Kidd IM, Hall SM, Cohen BJ An outbreak of erythema infectiosum associated with human parvovirus infection J Hyg (Lond) 1984;93:85–93 [251] Anderson MJ, Higgins PG, Davis LR, Willman JS, Jones SE, Kidd IM, et al Experimental parvoviral infection in humans J Infect Dis 1985;152:257–65 [252] Reid DM, Reid TM, Brown T, Rennie JA, Eastmond CJ Human parvovirus-associated arthritis: a clinical and laboratory description Lancet 1985;1:422–5 [253] White DG, Woolf AD, Mortimer PP, Cohen BJ, Blake DR, Bacon PA Human parvovirus arthropathy Lancet 1985;1:419–21 [254] Pattison JR, Jones SE, Hodgson J, Davis LR, White JM, Stroud CE, et al Parvovirus infections and hypoplastic crisis in sickle-cell anaemia Lancet 1981;1:664–5 [255] Zerbini M, Musiani M, Venturoli S, Gallinella G, Gibellini D, Gentilomi G, et al Different syndromes associated with B19 parvovirus viraemia in paediatric patients: report of four cases Eur J Pediatr 1992;151:815–7 [256] Heegaard ED, Brown KE Human parvovirus B19 Clin Microbiol Rev 2002;15:485–505 [257] Naides SJ Rheumatic manifestations of parvovirus B19 infection Rheum Dis Clin North Am 1998;24:375–401 [258] Van Horn DK, Mortimer PP, Young N, Hanson GR Human parvovirusassociated red cell aplasia in the absence of underlying hemolytic anemia Am J Pediatr Hematol Oncol 1986;8:235–9 [259] Kurtzman GJ, Ozawa K, Cohen B, Hanson G, Oseas R, Young NS Chronic bone marrow failure due to persistent B19 parvovirus infection N Engl J Med 1987;317:287–94 [260] Zerbini M Human parvoviruses In: Murray PR, Baron EJ, Pfallar MA, Tenover FC, Yolken RH, editors Manual of clinical microbiol Washington, DC: American Society for Microbiology; 1999 p 1089–98 [261] Takahashi T, Ozawa K, Takahashi K, Okuno Y, Muto Y, Takaku FSS DNA replication of Parvovirus B19 in a human erythrooid leukemia cell-line (TK-1) in vitro Arch Virol 1993;131:201–8 [262] Ozawa K, Kurtzman G, Young N Replication of the B19 parvovirus in human bone marrow cell cultures Science 1986;233:883–6 [263] Anand A, Gray ES, Brown T, Clewley JP, Cohen BJ Human parvovirus infection in pregnancy and hydrops fetalis N Engl J Med 1987;316:183–6 [264] Morey AL, Keeling JW, Porter HJ, Fleming KA Clinical and histopathological features of parvovirus B19 infection in the human fetus Br J Obstet Gynaecol 1992;99:566–74 [265] Brown T, Anand A, Ritchie LD, Clewley JP, Reid TM Intrauterine parvovirus infection associated with hydrops fetalis Lancet 1984;2:1033–4 [266] Knott PD, Welply GA, Anderson MJ Serologically proved intrauterine infection with parvovirus Br Med J (Clin Res Ed) 1984;289:1660 [267] Mortimer PP, Cohen BJ, Buckley MM, Cradock-Watson JE, Ridehalgh MK, Burkhardt F, et al Human parvovirus and the fetus Lancet 1985;2:1012 [268] Cohen BJ, Buckley MM The prevalence of antibody to human parvovirus B19 in England and Wales J Med Microbiol 1988;25:151–3 [269] Harger JH, Adler SP, Koch WC, Harger GF Prospective evaluation of 618 pregnant women exposed to parvovirus B19: risks and symptoms Obstet Gynecol 1998;91:413–20 [270] Rodis JF Parvovirus infection Clin Obstet Gynecol 1999;42:107–20, quiz 74–5 [271] Gratacos E, Torres PJ, Vidal J, Antolin E, Costa J, Jimenez de Anta MT, et al The incidence of human parvovirus B19 infection during pregnancy and its impact on perinatal outcome J Infect Dis 1995;171:1360–3 [272] Hemauer A, Gigler A, Searle K, Beckenlehner K, Raab U, Broliden K, et al Seroprevalence of parvovirus B19 NS1-specific IgG in B19infected and uninfected individuals and in infected pregnant women J Med Virol 2000;60:48–55 [273] Enders M, Weidner A, Zoellner I, Searle K, Enders G Fetal morbidity and mortality after acute human parvovirus B19 infection in pregnancy: prospective evaluation of 1018 cases Prenat Diagn 2004;24(7): 513–8 [274] Fairley CK, Smoleniec JS, Caul OE, Miller E Observational study of effect of intrauterine transfusions on outcome of fetal hydrops after parvovirus B19 infection Lancet 1995;346:1335–7 [275] Zerbini M, Musiani M, Gentilomi G, Venturoli S, Gallinella G, Morandi R Comparative evaluation of virological and serological methods in prenatal diagnosis of parvovirus B19 fetal hydrops J Clin Microbiol 1996;34:603–8 [276] Torok TJ, Wang QY, Gary Jr GW, Yang CF, Finch TM, Anderson LJ Prenatal diagnosis of intrauterine infection with parvovirus B19 by the polymerase chain reaction technique Clin Infect Dis 1992;14:149– 55 [277] Peters MT, Nicolaides KH Cordocentesis for the diagnosis and treatment of human fetal parvovirus infection Obstet Gynecol 1990;75:501–4 [278] Forestier F, Tissot JD, Vial Y, Daffos F, Hohlfeld P Haematological parameters of parvovirus B19 infection in 13 fetuses with hydrops foetalis Br J Haematol 1999;104:925–7 [279] Kaliasam C, Brennand J, Cameron AD Congenital parvoviral B19 infection: experience of a recent epidemic Fetal Diagn Ther 2001;16:18–22 [280] Miyagawa E, Yoshida T, Takahashi H, Yamaguchi K, Nagano T, Kiriyama Y, et al Infection of the erythroid cell line KU812Ep6 with human parvovirus B19 and its application to titration of B19 infectivity J Virol Methods 1999;83:45–54 [281] Shimomura S, Komatsu N, Frickhofen N, Anderson S, Kajigaya S, Young NS First continuous propagation of B19 parvovirus in a cell line Blood 1992;79:18–24 [282] Soderlund M, Brown CS, Spaan WJ, Hedman L, Hedman K Epitope type-specific IgG responses to capsid proteins VP1 and VP2 of human parvovirus B19 J Infect Dis 1995;172:1431–6 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 [283] Dorsch S, Kaufmann B, Schaible U, Prohaska E, Wolf H, Modrow S The VP1-unique region of parvovirus B19: amino acid variability and antigenic stability J Gen Virol 2001;82:191–9 [284] Kerr S, O’Keeffe G, Kilty C, Doyle S Undenatured parvovirus B19 antigens are essential for the accurate detection of parvovirus B19 IgG J Med Virol 1999;57:179–85 [285] Manaresi E, Zuffi E, Gallinella G, Gentilomi G, Zerbini M, Musiani M Differential IgM response to conformational and linear epitopes of parvovirus B19 VP1 and VP2 structural proteins J Med Virol 2001;64:67–73 [286] Corcoran A, Mahon BP, Doyle S B cell memory is directed toward conformational epitopes of parvovirus B19 capsid proteins and the unique region of VP1 J Infect Dis 2004;189:1873–80 [287] Heegaard ED, Rasksen CJ, Christensen J Detection of parvovirus B19 NS1-specific antibodies by ELISA and Western blotting employing recombinant NS1 protein as antigen J Med Virol 2002;67:375–83 [288] Manaresi E, Gallinella G, Zerbini M, Venturoli S, Gentilomi G, Musiani M IgG immune response to B19 parvovirus VP1 and VP2 linear epitopes by immunoblot assay J Med Virol 1999;57:174–8 [289] Nunoue T, Kusuhara K, Hara T Human fetal infection with parvovirus B19: maternal infection time in gestation, viral persistence and fetal prognosis Pediatr Infect Dis J 2002;21:1133–6 [290] Koch WC, Harger JH, Barnstein B, Adler SP Serologic and virologic evidence for frequent intrauterine transmission of human parvovirus B19 with a primary maternal infection during pregnancy Pediatr Infect Dis J 1998;17:489–94 [291] Wattre P, Dewilde A, Subtil D, Andreoletti L, Thirion V A clinical and epidemiological study of human parvovirus B19 infection in fetal hydrops using PCR Southern blot hybridization and chemiluminescence detection J Med Virol 1998;54:140–4 [292] Musiani M, Azzi A, Zerbini M, Gibellini D, Venturoli S, Zakrzewska K, et al Nested polymerase chain reaction assay for the detection of B19 parvovirus DNA in human immunodeficiency virus patients J Med Virol 1993;40:157–60 [293] Hamilton H, Gomos J, Berns KI, Falck-Pedersen E Adenoassociated virus site-specific integration and AAVS1 disruption J Virol 2004;78:7874–82 [294] Musiani M, Zerbini M, Gentilomi G, Rodorigo G, De Rosa V, Gibellini D, et al Persistent B19 parvovirus infections in haemophilic HIV-1 infected patients J Med Virol 1995;46:103–8 [295] Hendrie PC, Hirata RK, Russell DW Chromosomal integration and homologous gene targeting by replication-incompetent vectors based on the autonomous parvovirus minute virus of mice J Virol 2003;77:13136–45 [296] Gallinella G, Zuffi E, Gentilomi G, Manaresi E, Venturoli S, Bonvicini F, et al Relevance of B19 markers in serum samples for a diagnosis of parvovirus B19-correlated diseases J Med Virol 2003;71:135–9 [297] Cassinotti P, Weitz M, Siegl G Human parvovirus B19 infections: routine diagnosis by a new nested polymerase chain reaction assay J Med Virol 1993;40:228–34 [298] Musiani M, Zerbini M, Gentilomi G, Plazzi M, Gallinella G, Venturoli S Parvovirus B19 clearance from peripheral blood after acute infection J Infect Dis 1995;172:1360–3 [299] Patou G, Pillay D, Myint S, Pattison J Characterization of a nested polymerase chain reaction assay for detection of parvovirus B19 J Clin Microbiol 1993;31:540–6 [300] Searle K, Guilliard C, Wallat S, Schalasta G, Enders G Acute parvovirus B19 infection in pregnant women—an analysis of serial samples by serological and semi-quantitative PCR techniques Infection 1998;26:139–43 [301] Azzi A, Zakrzewska K, Bertoni E, Guidi S, Salvadori M Persistent parvovirus B19 infections with different clinical outcomes in renal transplant recipients: diagnostic relevance of polymerase chain reaction (PCR) and of quantification of B19 DNA in sera Clin Microbiol Infect 1996;2:105–8 [302] Jordan J, Tiangco B, Kiss J, Koch W Human parvovirus B19: prevalence of viral DNA in volunteer blood donors and clinical outcomes of transfusion recipients Vox Sang 1998;75:97–102 379 [303] McOmish F, Yap PL, Jordan A, Hart H, Cohen BJ, Simmonds P Detection of parvovirus B19 in donated blood: a model system for screening by polymerase chain reaction J Clin Microbiol 1993;31:323–8 [304] Braham S, Gandhi J, Beard S, Cohen B Evaluation of the Roche LightCycler parvovirus B19 quantification kit for the diagnosis of parvovirus B19 infections J Clin Virol 2004;31:5–10 [305] Brown KE Detection and quantitation of parvovirus B19 J Clin Virol 2004;31:1–4 [306] Aberham C, Pendl C, Gross P, Zerlauth G, Gessner M A quantitative, internally controlled real-time PCR assay for the detection of parvovirus B19 DNA J Virol Methods 2001;92:183–91 [307] Manaresi E, Gallinella G, Zuffi E, Bonvicini F, Zerbini M, Musiani M Diagnosis and quantitative evaluation of parvovirus B19 infections by real-time PCR in the clinical laboratory J Med Virol 2002;67:275–81 [308] Schorling S, Schalasta G, Enders G, Zauke M Quantification of parvovirus B19 DNA using COBAS AmpliPrep automated sample preparation and LightCycler real-time PCR J Mol Diagn 2004;6:37–41 [309] Brown CS, van Bussel MJ, Wassenaar AL, van Elsacker-Niele AM, Weiland HT, Salimans MM An immunofluorescence assay for the detection of parvovirus B19 IgG and IgM antibodies based on recombinant viral antigen J Virol Methods 1990;29:53–62 [310] Brown CS, Salimans MM, Noteborn MH, Weiland HT Antigenic parvovirus B19 coat proteins VP1 and VP2 produced in large quantities in a baculovirus expression system Virus Res 1990;15:197–211 [311] Fridell E, Trojnar J, Wahren B A new peptide for human parvovirus B19 antibody detection Scand J Infect Dis 1989;21:597–603 [312] Jordan JA Comparison of a baculovirus-based VP2 enzyme immunoassay (EIA) to an Escherichia coli-based VP1 EIA for detection of human parvovirus B19 immunoglobulin M and immunoglobulin G in sera of pregnant women J Clin Microbiol 2000;38:1472–5 [313] Manaresi E, Gallinella G, Venturoli S, Zerbini M, Musiani M Detection of parvovirus B19 IgG: choice of antigens and serological tests J Clin Virol 2004;29:51–3 [314] Butchko AR, Jordan JA Comparison of three commercially available serologic assays used to detect human parvovirus B19-specific immunoglobulin M (IgM) and IgG antibodies in sera of pregnant women J Clin Microbiol 2004;42:3191–5 [315] Bruu AL, Nordbo SA Evaluation of five commercial tests for detection of immunoglobulin M antibodies to human parvovirus B19 J Clin Microbiol 1995;33:1363–5 [316] Tolfvenstam T, Ruden U, Broliden K Evaluation of serological assays for identification of parvovirus B19 immunoglobulin M Clin Diagn Lab Immunol 1996;3:147–50 [317] Patou G, Ayliffe U Evaluation of commercial enzyme linked immunosorbent assay for detection of B19 parvovirus IgM and IgG J Clin Pathol 1991;44:831–4 [318] Cohen BJ, Mortimer PP, Pereira MS Diagnostic assays with monoclonal antibodies for the human serum parvovirus-like virus (SPLV) J Hyg (Lond) 1983;91:113–30 [319] Anderson MJ, Jones SE, Minson AC Diagnosis of human parvovirus infection by dot-blot hybridization using cloned viral DNA J Med Virol 1985;15:163–72 [320] Mori J, Field AM, Clewley JP, Cohen BJ Dot blot hybridization assay of B19 virus DNA in clinical specimens J Clin Microbiol 1989;27:459–64 [321] Zerbini M, Musiani M, Venturoli S, Gallinella G, Gibellini D, Gentilomi G, et al Rapid screening for B19 parvovirus DNA in clinical specimens with a digoxigenin-labeled DNA hybridization probe J Clin Microbiol 1990;28:2496–9 [322] Zerbini M, Gentilomi G, Cricca M, Manaresi E, Bonvicini F, Musiani M A system to enhance the sensitivity of digoxigenin-labelled probe: detection of B19 DNA in serum samples J Virol Methods 2001;93:137–44 [323] Wong S, Young NS, Brown KE Prevalence of parvovirus B19 in liver tissue: no association with fulminant hepatitis or hepatitis-associated aplastic anemia J Infect Dis 2003;187:1581–6 [324] Sevall JS, Ritenhous J, Peter JB Laboratory diagnosis of parvovirus B19 infection J Clin Lab Anal 1992;6:171–5 380 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 [325] Kovacs BW, Carlson DE, Shahbahrami B, Platt LD Prenatal diagnosis of human parvovirus B19 in nonimmune hydrops fetalis by polymerase chain reaction Am J Obstet Gynecol 1992;167:461–6 [326] Carriere C, Boulanger P, Delsert C Rapid and sensitive method for the detection of B19 virus DNA using the polymerase chain reaction with nested primers J Virol Methods 1993;44:221–34 [327] Dieck D, Schild RL, Hansmann M, Eis-Hubinger AM Prenatal diagnosis of congenital parvovirus B19 infection: value of serological and PCR techniques in maternal and fetal serum Prenat Diagn 1999;19:1119–23 [328] Schwartz TF, Jager G, Holzgreve W, Roggendorf M Diagnosis of human parvovirus B19 by polymerase chain reaction scan J Infect Dis 1992;24:691–6 [329] Eis-Hubinger AM, Dieck D, Schild R, Hansmann M, Schneweis KE Parvovirus B19 infection in pregnancy Intervirology 1998;41:178–84 [330] Cassinotti P, Burtonboy G, Fopp M, Siegl G Evidence for persistence of human parvovirus B19 DNA in bone marrow J Med Virol 1997;53:229–32 [331] Soderlund M, von Essen R, Haapasaari J, Kiistala U, Kiviluoto O, Hedman K Persistence of parvovirus B19 DNA in synovial membranes of young patients with and without chronic arthropathy Lancet 1997;349:1063–5 [332] Knoll A, Louwen F, Kochanowski B, Plentz A, Stussel J, Beckenlehner K, et al Parvovirus B19 infection in pregnancy: quantitative viral DNA analysis using a kinetic fluorescence detection system (TaqMan PCR) J Med Virol 2002;67:259–66 [333] Varmus H Retroviruses Science 1988;240(4858):1427–35 [334] Zhu T, Korber BT, Nahmias AJ, Hooper E, Sharp PM, Ho DD An African HIV-1 sequence from 1959 and implications for the origin of the epidemic Nature 1998;391(6667):594–7 [335] Corbitt G, Bailey AS, Williams G HIV infection in Manchester Lancet 1990;336(8706):51 [336] Williams G, Stretton TB, Leonard JC AIDS in 1959? Lancet 1983;2(8359):1136 [337] Barre-Sinoussi F, Chermann JC, Rey F, Nugeyre MT, Chamaret S, Gruest J, et al Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS) Science 1983;220(4599):868–71 [338] Popovic M, Sarngadharan MG, Read E, Gallo RC Detection, isolation, and continuous production of cytopathic retroviruses (HTLV-III) from patients with AIDS and pre-AIDS Science 1984;224(4648):497–500 [339] Wain-Hobson S, Alizon M, Montagnier L Relationship of AIDS to other retroviruses Nature 1985;313(6005):743 [340] Wain-Hobson S, Sonigo P, Danos O, Cole S, Alizon M Nucleotide sequence of the AIDS virus, LAV Cell 1985;40(1):9–17 [341] Shaw GM, Hahn BH, Arya SK, Groopman JE, Gallo RC, Wong-Staal F Molecular characterization of human T-cell leukemia (lymphotropic) virus type III in the acquired immune deficiency syndrome Science 1984;226(4679):1165–71 [342] Clavel F HIV-2, the West African AIDS virus AIDS 1987;1(3):135–40 [343] Clavel F, Guetard D, Brun-Vezinet F, Chamaret S, Rey MA, SantosFerreira MO, et al Isolation of a new human retrovirus from West African patients with AIDS Science 1986;233(4761):343–6 [344] Horsburgh Jr CR, Holmberg SD The global distribution of human immunodeficiency virus type (HIV-2) infection Transfusion 1988;28(2):192–5 [345] UNAIDS 2004 Report on the global AIDS epidemic Geneva: UnAIDS; 2004 [346] Hahn BH, Shaw GM, De Cock KM, Sharp PM AIDS as a zoonosis: scientific and public health implications Science 2000;287(5453):607–14 [347] Korber B, Muldoon M, Theiler J, Gao F, Gupta R, Lapedes A, et al Timing the ancestor of the HIV-1 pandemic strains Science 2000;288(5472):1789–96 [348] Robertson DL, Anderson JL, Bradac JA, Carr JK, Foley B, Funkhouser RK, et al HIV-1 nomenclature proposal: a reference guide to HIV-1 classification In: Kuiken CL, Foley B, Hahn BH, Marx P, McCutchan [349] [350] [351] [352] [353] [354] [355] [356] [357] [358] [359] [360] [361] [362] [363] [364] [365] [366] [367] FE, Mellors J, Mullins JI, Wolanski S, Korber B, editors Human retroviruses and AIDS: a compilation and analysis of nucleic and amino acid sequences Los Alamos, NM: Los Alamos National Laboratory; 2000 p 492–505 Robertson DL, Anderson JP, Bradac JA, Carr JK, Foley B, Funkhouser RK, et al HIV-1 nomenclature proposal Science 2000;288(5463):55–6 Coffin JM HIV population dynamics in vivo: implications for genetic variation, pathogenesis, and therapy Science 1995;267(5197):483–9 Lewis SH, Reynolds-Kohler C, Fox HE, Nelson JA HIV-1 in trophoblastic and villous Hofbauer cells, and haematological precursors in eight-week fetuses Lancet 1990;335(8689):565–8 Rogers MF, Ou CY, Rayfield M, Thomas PA, Schoenbaum EE, Abrams E, et al Use of the polymerase chain reaction for early detection of the proviral sequences of human immunodeficiency virus in infants born to seropositive mothers New York City Collaborative Study of Maternal HIV Transmission and Montefiore Medical Center HIV Perinatal Transmission Study Group N Engl J Med 1989;320(25):1649– 54 Burgard M, Mayaux MJ, Blanche S, Ferroni A, Guihard-Moscato ML, Allemon MC, et al The use of viral culture and p24 antigen testing to diagnose human immunodeficiency virus infection in neonates The HIV Infection in Newborns French Collaborative Study Group N Engl J Med 1992;327(17):1192–7 Goedert JJ, Duliege AM, Amos CI, Felton S, Biggar RJ High risk of HIV-1 infection for first-born twins The international registry of HIV-exposed twins Lancet 1991;338(8781):1471–5 Villari P, Spino C, Chalmers TC, Lau J, Sacks HS Cesarean section to reduce perinatal transmission of human immunodeficiency virus A metaanalysis Online J Curr Clin Trials 1993 Doc No 74:[5107 words; 46 paragraphs] Oxtoby MJ Human immunodeficiency virus and other viruses in human milk: placing the issues in broader perspective Pediatr Infect Dis J 1988;7(12):825–35 Hira SK, Mangrola UG, Mwale C, Chintu C, Tembo G, Brady WE, et al Apparent vertical transmission of human immunodeficiency virus type by breast-feeding in Zambia J Pediatr 1990;117(3):421–4 Van de Perre P, Hitimana DG, Simonon A, Dabis F, Msellati P, Karita P, et al Postnatal transmission of HIV-1 associated with breast abscess Lancet 1992;339(8807):1490–1 Van de Perre P, Simonon A, Msellati P, Hitimana DG, Vaira D, Bazubagira A, et al Postnatal transmission of human immunodeficiency virus type from mother to infant A prospective cohort study in Kigali, Rwanda N Engl J Med 1991;325(9):593–8 Bryson YJ, Luzuriaga K, Sullivan JL, Wara DW Proposed definitions for in utero versus intrapartum transmission of HIV-1 N Engl J Med 1992;327(17):1246–7 Fowler MG, Simonds RJ, Roongpisuthipong A Update on perinatal HIV transmission Pediatr Clin North Am 2000;47(1):21–38 Mohlala BK, Tucker TJ, Besser MJ, Williamson C, Yeats J, Smit L, et al Investigation of HIV in amniotic fluid from HIV-infected pregnant women at full term J Infect Dis 2005;192(3):488–91 Magder LS, Mofenson L, Paul ME, Zorrilla CD, Blattner WA, Tuomala RE, et al Risk factors for in utero and intrapartum transmission of HIV J Acquir Immune Defic Syndr 2005;38(1):87–95 Newell ML, Dunn DT, Peckham CS, Semprini AE, Pardi G Vertical transmission of HIV-1: maternal immune status and obstetric factors The European Collaborative Study AIDS 1996;10(14):1675–81 Pitt J, Brambilla D, Reichelderfer P, Landay A, McIntosh K, Burns D, et al Maternal immunologic and virologic risk factors for infant human immunodeficiency virus type infection: findings from the Women and Infants Transmission Study J Infect Dis 1997;175(3):567–75 Orloff SL, Simonds RJ, Steketee RW, St Louis ME Determinants of perinatal HIV-1 transmission Clin Obstet Gynecol 1996;39(2):386–95 Dabis F, Msellati P, Dunn D, Lepage P, Newell ML, Peckham C, et al Estimating the rate of mother-to-child transmission of HIV Report of a workshop on methodological issues Ghent (Belgium), 17–20 February 1992 The Working Group on Mother-to-Child Transmission of HIV AIDS 1993;7(8):1139–48 E Mendelson et al / Reproductive Toxicology 21 (2006) 350–382 [368] De Cock KM, Fowler MG, Mercier E, de Vincenzi I, Saba J, Hoff E, et al Prevention of mother-to-child HIV transmission in resourcepoor countries: translating research into policy and practice JAMA 2000;283(9):1175–82 [369] Best JM Laboratory diagnosis of intrauterine and perinatal virus infections Clin Diagn Virol 1996;5(2–3):121–9 [370] Connor EM, Sperling RS, Gelber R, Kiselev P, Scott G, O’Sullivan MJ, et al Reduction of maternal-infant transmission of human immunodeficiency virus type with zidovudine treatment Pediatric AIDS Clinical Trials Group Protocol 076 Study Group N Engl J Med 1994;331(18):1173–80 [371] Simonds RJ, Steketee R, Nesheim S, Matheson P, Palumbo P, Alger L, et al Impact of zidovudine use on risk and risk factors for perinatal transmission of HIV Perinatal AIDS Collaborative Transmission Studies AIDS 1998;12(3):301–8 [372] Shaffer N, Chuachoowong R, Mock PA, Bhadrakom C, Siriwasin W, Young NL, et al Short-course zidovudine for perinatal HIV1 transmission in Bangkok Thailand: a randomised controlled trial Bangkok Collaborative Perinatal HIV Transmission Study Group Lancet 1999;353(9155):773–80 [373] Garcia PM, Kalish LA, Pitt J, Minkoff H, Quinn TC, Burchett SK, et al Maternal levels of plasma human immunodeficiency virus type RNA and the risk of perinatal transmission Women and Infants Transmission Study Group N Engl J Med 1999;341(6):394– 402 [374] Tuomala RE, Watts DH, Li D, Vajaranant M, Pitt J, Hammill H, et al Improved obstetric outcomes and few maternal toxicities are associated with antiretroviral therapy, including highly active antiretroviral therapy during pregnancy J Acquir Immune Defic Syndr 2005;38(4):449–73 [375] Mofenson LM, Lambert JS, Stiehm ER, Bethel J, Meyer 3rd WA, Whitehouse J, et al Risk factors for perinatal transmission of human immunodeficiency virus type in women treated with zidovudine Pediatric AIDS Clinical Trials Group Study 185 Team N Engl J Med 1999;341(6):385–93 [376] Ioannidis JP, Abrams EJ, Ammann A, Bulterys M, Goedert JJ, Gray L, et al Perinatal transmission of human immunodeficiency virus type by pregnant women with RNA virus loads 1 month, with one of those being performed at age >4 months [386] Two or more negative HIV immunoglobulin G (IgG) antibody tests performed at age >6 months... blotting (WB) for HSV is the most accurate method for type-specific serology [241] This method is the serology gold standard and is performed only by few laboratories WB is expensive to perform and