Abstract

Background

Primary human cytomegalovirus (HCMV) infection during pregnancy is the major cause of congenital viral sequelae. The HCMV-specific T-cell response may have a role in the prevention of virus transmission to the fetus.

Methods

HCMV-specific memory T cells were investigated in the second month after primary infection onset in 44 pregnant women (15 transmitting the infection to the fetus) and 8 pregnant women with remote infection. Peripheral blood mononuclear cells were stimulated for 12 days with peptide pools of HCMV proteins IE-1, IE-2, and pp65, and subsequently restimulated for 24 hours with the same peptide pools in a cultured enzyme-linked immunospot (ELISPOT) assay.

Results

In pregnant women with primary infection, the cultured ELISPOT assay detected a higher T-cell response to pp65 than to IE-1 or IE-2, whereas in remote infection pp65-, IE-1–, and IE-2–specific T cells were detected at comparable levels. During primary infection, the cultured ELISPOT response was mainly mediated by CD4+ T cells, and was lower than in remote infection. Strikingly, the cultured ELISPOT response to pp65 (but not to IE-1 or IE-2) was significantly higher in nontransmitting mothers. To detect other factors potentially associated with nontransmission, different serological parameters were analyzed. Only immunoglobulin G avidity index was higher in nontransmitting mothers, who showed also a lower DNAemia level. These 2 parameters remained associated with congenital infection in multivariate analysis.

Conclusions

Determination of HCMV-specific T cells by cultured ELISPOT, in pregnant women with primary HCMV infection, in association with avidity index and DNAemia may help to assess the risk of HCMV fetal transmission.

(See the Major Article by Tanimura et al on pages 1652-8 and Editorial Commentary by Schleiss on pages 1666-9.)

Human cytomegalovirus (HCMV) is a major cause of congenital viral infection, occurring in 0.5%–2% of pregnancies [1] and in 20% of cases results in sensory and developmental impairment [2]. HCMV transmission only rarely occurs during nonprimary infection [3], whereas primary infection in pregnancy confers a major risk (about 40%) of congenital infection [4]. Thus, a preexisting immune response appears to provide a strong degree of protection from virus transmission to the fetus. As for the humoral response, in one study, women with primary infections who did not transmit the infection to the fetus exhibited earlier development of neutralizing antibodies against the HCMV pentameric complex gH/gL/pUL128L [5]. Also, the early development of an HCMV-specific CD4+ T-cell lymphoproliferative response [6, 7] has been reported in nontransmitting women. In addition, reduced reexpression of CD45RA, on the surface of HCMV-specific effector memory T cells, correlates with HCMV transmission to the fetus [8, 9]. Studies on transplant recipients have also documented the importance of the HCMV-specific CD4+ T-cell response for the control of viral infection [10–13].

In recent years, the simple tools Quantiferon and enzyme-linked immunospot (ELISPOT) assay were largely used to detect antigen-specific T-cell responses. Using Quantiferon, Eldar-Yedidia and colleagues demonstrated an association between high interferon-gamma (IFN-γ) relative response to HCMV and high risk of transmission to the fetus [14]; in addition, a higher response detected by the ELISPOT assay was shown to be associated with an increased risk of congenital infection [15]. According to the size of the peptides used in both studies, the response was most likely sustained by HCMV-specific CD8+ T cells. Both Quantiferon and standard ELISPOT assays quantify T cells capable of prompt IFN-γ production. These cells are thought to be mainly effector or effector-memory T cells [16]. However, the ELISPOT assay can also be used to quantify antigen-specific T cells with proliferative capacity [17]. In this assay, antigen-specific lymphocytes are expanded for 12 days and then restimulated with the same antigens (cultured ELISPOT). These expandable antigen-specific T cells are thought to be central memory T cells [18]. The aim of this study was to investigate the HCMV-specific memory T-cell response with the cultured ELISPOT assay in pregnant women with primary HCMV infection and evaluate its correlation with HCMV transmission to the fetus, in parallel with the antibody response and viral load.

METHODS

Blood Samples

Forty-four pregnant women experiencing primary HCMV infection during the first or second trimester of gestation were enrolled in the study (Table 1). Heparinized blood samples were collected at a median estimated time of 45 (range, 26–66) days after onset of infection. In addition, 8 pregnant women (median age, 32 [range, 25–36] years) with remote infection (>5 years) were enrolled. This retrospective study was performed according to the guidelines of the Institutional Review Board of Fondazione IRCCS Policlinico San Matteo on the use of biological specimens for scientific purposes in keeping with Italian legislation (art.13 D.Lgs 196/2003).

Table 1.

Characteristics of the 44 Primary Human Cytomegalovirus–Infected Pregnant Women

ParametersNontransmitters (n = 29)Transmitters (n = 15)P Value
Age, y, median (range)33 (23–40)35 (27–41)NS
Gestational week at onset of maternal infection, median (range)15 (1–26)18 (4–25)NS
Estimated days after onset of infection, median (range)47 (26–66)43 (29–65)NS
Parameters used for dating infection onset, No. (%) of cases
 Presence of symptoms and/or biochemical signs22 (67)14 (93)NS
 IgG seroconversion19 (65)10 (66)NS
 Low IgG avidity index12 (41)4 (27)NS
 IgM ratio7 (24)3 (20)NS
 Neutralizing antibody seroconversion20 (67)10 (67)NS
Diagnosis of congenital infection, No. (%) of cases
 At amniocentesis13 (45)5 (33)NS
 At birth29 (100)14a (93)NS
Infant outcome at birth, No. (%) of cases
 SymptomaticNA1 (7)NA
 AsymptomaticNA11 (73)NA
 UnknownNA3 (20)NA
ParametersNontransmitters (n = 29)Transmitters (n = 15)P Value
Age, y, median (range)33 (23–40)35 (27–41)NS
Gestational week at onset of maternal infection, median (range)15 (1–26)18 (4–25)NS
Estimated days after onset of infection, median (range)47 (26–66)43 (29–65)NS
Parameters used for dating infection onset, No. (%) of cases
 Presence of symptoms and/or biochemical signs22 (67)14 (93)NS
 IgG seroconversion19 (65)10 (66)NS
 Low IgG avidity index12 (41)4 (27)NS
 IgM ratio7 (24)3 (20)NS
 Neutralizing antibody seroconversion20 (67)10 (67)NS
Diagnosis of congenital infection, No. (%) of cases
 At amniocentesis13 (45)5 (33)NS
 At birth29 (100)14a (93)NS
Infant outcome at birth, No. (%) of cases
 SymptomaticNA1 (7)NA
 AsymptomaticNA11 (73)NA
 UnknownNA3 (20)NA

Abbreviations: IgG, immunoglobulin G; IgM, immunoglobulin M; NA, not applicable; NS, not significant.

aOne termination of pregnancy after amniocentesis.

Table 1.

Characteristics of the 44 Primary Human Cytomegalovirus–Infected Pregnant Women

ParametersNontransmitters (n = 29)Transmitters (n = 15)P Value
Age, y, median (range)33 (23–40)35 (27–41)NS
Gestational week at onset of maternal infection, median (range)15 (1–26)18 (4–25)NS
Estimated days after onset of infection, median (range)47 (26–66)43 (29–65)NS
Parameters used for dating infection onset, No. (%) of cases
 Presence of symptoms and/or biochemical signs22 (67)14 (93)NS
 IgG seroconversion19 (65)10 (66)NS
 Low IgG avidity index12 (41)4 (27)NS
 IgM ratio7 (24)3 (20)NS
 Neutralizing antibody seroconversion20 (67)10 (67)NS
Diagnosis of congenital infection, No. (%) of cases
 At amniocentesis13 (45)5 (33)NS
 At birth29 (100)14a (93)NS
Infant outcome at birth, No. (%) of cases
 SymptomaticNA1 (7)NA
 AsymptomaticNA11 (73)NA
 UnknownNA3 (20)NA
ParametersNontransmitters (n = 29)Transmitters (n = 15)P Value
Age, y, median (range)33 (23–40)35 (27–41)NS
Gestational week at onset of maternal infection, median (range)15 (1–26)18 (4–25)NS
Estimated days after onset of infection, median (range)47 (26–66)43 (29–65)NS
Parameters used for dating infection onset, No. (%) of cases
 Presence of symptoms and/or biochemical signs22 (67)14 (93)NS
 IgG seroconversion19 (65)10 (66)NS
 Low IgG avidity index12 (41)4 (27)NS
 IgM ratio7 (24)3 (20)NS
 Neutralizing antibody seroconversion20 (67)10 (67)NS
Diagnosis of congenital infection, No. (%) of cases
 At amniocentesis13 (45)5 (33)NS
 At birth29 (100)14a (93)NS
Infant outcome at birth, No. (%) of cases
 SymptomaticNA1 (7)NA
 AsymptomaticNA11 (73)NA
 UnknownNA3 (20)NA

Abbreviations: IgG, immunoglobulin G; IgM, immunoglobulin M; NA, not applicable; NS, not significant.

aOne termination of pregnancy after amniocentesis.

Diagnosis and Timing of Primary Infection

Diagnosis of primary HCMV infection was based on 1 or more of the following criteria: HCMV-specific immunoglobulin G (IgG) seroconversion, HCMV-specific immunoglobulin M (IgM) antibody detection, low IgG avidity index (AI), and HCMV DNAemia. HCMV-specific IgG and IgM were determined by ETI-CYTOK-G and ETI-CYTOK-M (DiaSorin). IgM results obtained by the commercial assay were confirmed by an in-house–developed capture enzyme-linked immunosorbent assay (ELISA) [19]. IgG AI was determined by an in-house–developed ELISA test using HCMV nuclear antigen [20]. HCMV DNA was determined by real-time polymerase chain reaction [21, 22]. Estimated timing of primary infection was based on the following criteria: decreasing levels of HCMV-specific IgM antibody, increasing levels of AI, increasing levels of neutralizing antibodies detected on human embryonic lung fibroblasts (HELF) and the presence of clinical symptoms along with laboratory findings [23, 24]. Additional serological assays were performed to analyze the possible correlation between the antibody response to HCMV and virus transmission to the fetus. The neutralization assay was performed also in epithelial (ARPE-19) cells [25] and the IgG antibody titers to gH/gL/pUL128L, gH/gL/gO, and gB were determined by ELISA, as reported [5].

Diagnosis of Congenital Human Cytomegalovirus Infection

Congenital HCMV infection was diagnosed before birth by HCMV isolation and/or viral DNA detection in amniotic fluid or fetal blood [26] or within 2 weeks after birth by viral DNA detection in urine.

Isolation of Peripheral Blood Mononuclear Cells

Peripheral blood mononuclear cells (PBMCs) were isolated by standard density gradient centrifugation using Lymphoprep (Axis-Shield). Isolated PBMCs were cryopreserved in RPMI-1640 (Euroclone) supplemented with 10% dimethyl sulfoxide (Sigma-Aldrich) and 5% human albumin (Grifold Biologicals) and preserved in liquid nitrogen.

Cultured Enzyme-Linked Immunospot Assay

PBMCs were thawed, washed, and resuspended in culture medium (RPMI-1640 medium supplemented with 2 mM l-glutamine, 100 U/mL penicillin, 100 µg/mL streptomycin, and 10% heat-inactivated fetal calf serum, Euroclone). Cells were kept overnight at 37°C in a humidified 5% CO2 atmosphere. PBMCs were then transferred to a 48-well flat-bottom plate (5 × 105 cells/mL per well) and stimulated with (i) peptide pools (15-mer overlapping by 11 aa) encompassing the HCMV proteins immediate early (IE) -1, IE-2, and the phosphoprotein (pp) 65 (JPT Peptide Technologies) at a final concentration of 0.25 µg/mL; (ii) culture medium only; (iii) or phytohemagglutinin (PHA; 5 µg/mL, Sigma-Aldrich). PBMCs were then incubated at 37°C in a humidified 5% CO2 atmosphere for 12 days. On days 4 and 8, half of the supernatant from each well was removed and replaced with fresh culture medium supplemented with 20 IU/mL recombinant human interleukin 2 (Peprotech). After 12 days, cells were harvested, washed 3 times with culture medium, and resuspended at a concentration of 4 × 105 cells/mL before their use in the ELISPOT assay. The human IFN-γ ELISPOT kit (Diaclone) and Multiscreen-IP 96-well plates (Merck Millipore) were used as described elsewhere [17]. Cells were added to the plate in triplicate (4 × 104 cells/well) and restimulated for 24 hours with the corresponding peptide pool (0.25 µg/mL) used for the 12-day expansion or staphylococcal enterotoxin B (SEB, 2 µg/mL, Sigma-Aldrich) for cells expanded with PHA. Spots were counted using an automated AID ELISPOT reader system (AutoImmun Diagnostika GmbH). Results are presented as net spots per million PBMCs calculated according to the following formula: (mean number of spots/million PBMCs with antigen) – (mean number of spots/million PBMCs with culture medium).

Cytokine Flow Cytometry

Based on the number of PBMCs available, 9 samples were also analyzed by flow cytometry. After expansion, 0.2–0.4 × 106 cells were transferred to a 96-well round-bottom plate in 40 µL culture medium without fetal calf serum, in the presence of the corresponding peptide pool or SEB or culture medium alone. Following 1 hour incubation at 37°C in a humidified 5% CO2, 140 µL of culture medium supplemented with 10% heat-inactivated fetal calf serum containing brefeldin A (Sigma-Aldrich) at a final concentration of 10 µg/mL was added. After overnight incubation, cells were washed in phosphate-buffered saline (PBS) ethylenediaminetetraacetic acid 2mM and incubated with the Live/Dead Fixable Far Red dead cell stain kit (Thermo Fisher Scientific) for 30 minutes at 4°C. Cells were then washed with PBS, fixed, and permeabilized with BD Cytofix/Cytoperm (BD Biosciences) according to the manufacturer’s instructions for intracellular staining with the following monoclonal antibodies: CD3 PC5, CD4 ECD, CD8 PC7, IFN-γ FITC (all from Beckman Coulter). Finally, cells were resuspended in 1% paraformaldehyde and analyzed with a Navios flow cytometer (Beckman Coulter).

Statistical Analysis

Categorical variables are described as count and percentage. Fisher exact test was used to compare the difference in frequencies. Quantitative variables are described as median and range; comparisons between primary and remote infections or between HCMV transmitting and nontransmitting women were made with Mann-Whitney U test. Receiver operating characteristic (ROC) analysis was performed to establish the best cutoff to identify transmitting and nontransmitting women. (Prism 6 software, GraphPad Software). Furthermore, in addition to cultured ELISPOT responses, different parameters were considered in relation to the transmission of HCMV to the fetus (DNAemia, AI, IgG directed to gB, gH/gL/gO, and the pentameric complex gH/gL/pUL128L and antibodies neutralizing the infection of epithelial cells ARPE-19 and HELF). Parameters showing different levels in transmitting and nontransmitting women (with a P value <.10) were subsequently analyzed in a multivariate logistic regression model; P < .05 was considered statistically significant. All tests were 2-sided. The data analysis was performed with the Stata statistical package (release 14.0, 2015, StataCorp LLC) and NCSS (release 2006, NCSS LLC).

RESULTS

Human Cytomegalovirus-Specific T-Cell Response in Immunocompetent Pregnant Women Measured by Cultured Enzyme-Linked Immunospot

Memory T cells with proliferative capacity directed against pp65, IE-1, and IE-2 were measured in 44 pregnant women in the second month after onset of primary HCMV infection and in 8 pregnant women with remote infection (Figure 1). The median number of net spots/million PBMCs in response to pp65, IE-1, and IE-2 was significantly lower in primary than remote infection. In addition, a significantly higher response to pp65 than to IE-1 (P < .05) and IE-2 (P = .01) was observed in primary infections. No significant difference was observed between primary and remote infections in response to SEB. Flow cytometry analysis showed that, among 9 women with primary infection examined, 6 (67%) had a detectable response against at least 1 of the 3 proteins. Most IFN-γ producing antigen-specific T cells were CD4+ (Figure 2).

Figure 1.

T-cell response to staphylococcal enterotoxin B (SEB) and human cytomegalovirus proteins pp65, IE-1, and IE-2, detected by cultured enzyme-linked immunospot assay, in pregnant women with primary or remote infection. Results are expressed as net spots/million peripheral blood mononuclear cells (PBMCs); P values, determined by Mann-Whitney U test, are reported; Abbreviation: ns, not significant (P > .05).

Figure 2.

Flow cytometry detection of interferon gamma (IFN-γ) produced by memory T cells with proliferative capacity in the presence of culture medium alone or after stimulation with pp65, IE-1, and IE-2. Percentages of IFN-γ–producing CD4+ and CD8+ T cells examined in a representative pregnant woman with primary human cytomegalovirus infection are reported.

Human Cytomegalovirus-Specific Memory T-Cell Response and Virus Transmission to the Fetus

Memory T cells with proliferative capacity specific for pp65, IE-1, and IE-2 were compared between 15 women who transmitted and 29 women who did not transmit the infection to the fetus (Figure 3). The memory T-cell response specific for pp65 was significantly lower (P < .01) in transmitting (median, 13 [range, 0–1150] spots/million PBMCs) than in nontransmitting mothers (median, 388 [range, 0–8867] spots/million PBMCs). No differences were observed in the response to IE-1 or IE-2, between transmitting and nontransmitting women (both groups showed low number of net spots/million), as well as for the response to SEB.

Figure 3.

T-cell response to staphylococcal enterotoxin B (SEB) and human cytomegalovirus proteins pp65, IE-1, and IE-2, detected by cultured enzyme-linked immunospot assay in 29 nontransmitting and 15 transmitting women; P values, determined by Mann-Whitney U test, are reported; Abbreviations: ns, not significant (P > .05); PBMC, peripheral blood mononuclear cell.

Receiver Operating Characteristic Analysis and Risk of Human Cytomegalovirus Transmission to the Fetus

ROC analysis showed that a pp65 specific T-cell response of 20 net spots/million PBMCs (Figure 4) was the best cutoff to discriminate between transmitting and nontransmitting women (area under the curve = 0.74, sensitivity 53%, specificity 86%, positive predictive value = 67%, negative predictive value = 78%). Among women with a response ≤20 spots/million, 4 of 12 (33.3%) did not transmit the infection to the fetus, whereas 25 of 32 (78.1%) women with a response >20 spots/million PBMCs did not transmit the infection (P = .01; Figure 5). On the basis of this cutoff, an odds ratio of 7.14 (95% confidence interval, 1.65–30.89) for nontransmission of the virus to the fetus was associated with a pp65-specific T-cell response >20 spots/million PBMCs.

Figure 4.

Receiver operating characteristic curve for the establishment of the best cutoff to discriminate transmitting and nontransmitting women. Black dots represent values of cultured enzyme-linked immunospot assay corresponding to different sensitivity and specificity. White dot represents the cutoff of 20 spots/million peripheral blood mononuclear cells. Abbreviation: AUC, area under the curve.

Figure 5.

Cultured enzyme-linked immunospot assay response and number of nontransmitting (white histogram) or transmitting (black histogram) women divided in 2 groups: >20 or ≤20 spots/million peripheral blood mononuclear cells (PBMCs); P value, determined by Fisher exact test, is reported.

Immunological and Virological Parameters Associated With Human Cytomegalovirus Transmission to the Fetus

In addition to the T-cell response, we analyzed the possible association of antibody response with virus transmission to the fetus. The kinetics of IgG, IgM, and IgG AI to total HCMV antigen and IgG directed to envelope glycoprotein complexes (gB, gH/gL/gO, gH/gL/pUL128L) and the neutralizing antibody response vs HCMV infection of ARPE-19 and HELF during primary infection was investigated elsewhere [24]. In the 44 pregnant women analyzed in the present study, none of these parameters was found to be significantly different between transmitting and nontransmitting women (data not shown), with the exception of AI—that is, nontransmitting women showed higher levels of the IgG AI to whole virus antigen (P = .06; Figure 6A). Then, we examined DNAemia levels in transmitting and nontransmitting women, and we found that DNAemia levels were significantly lower in nontransmitting women (P = .04; Figure 6B).

Figure 6.

Immunoglobulin G avidity index (A) and DNAemia (B) in 29 nontransmitting and 15 transmitting women. P values, determined by Mann-Whitney U test, are reported.

The 3 parameters detected as significantly different between transmitting and nontransmitting mothers were analyzed in a multivariate logistic regression model, and high cultured ELISPOT response to pp65, high AI, and low DNAemia were independently associated with lack of HCMV transmission (Table 2). However, cultured ELISPOT to pp65 showed the highest association.

Table 2.

Multivariate Logistic Regression Analysis

ParametersP ValueOdds Ratio (95% CI)
pp65 cultured ELISPOT (>20 spots/million PBMCs)<.0133.15 (2.96–371.96)
Avidity index (for each 10% increase).031.93 (1.07–3.47)
DNAemia (log10 copies/mL).010.19 (.05–.70)
ParametersP ValueOdds Ratio (95% CI)
pp65 cultured ELISPOT (>20 spots/million PBMCs)<.0133.15 (2.96–371.96)
Avidity index (for each 10% increase).031.93 (1.07–3.47)
DNAemia (log10 copies/mL).010.19 (.05–.70)

Abbreviations: CI, confidence interval; ELISPOT, enzyme-linked immunospot assay; PBMC, peripheral blood mononuclear cell.

Table 2.

Multivariate Logistic Regression Analysis

ParametersP ValueOdds Ratio (95% CI)
pp65 cultured ELISPOT (>20 spots/million PBMCs)<.0133.15 (2.96–371.96)
Avidity index (for each 10% increase).031.93 (1.07–3.47)
DNAemia (log10 copies/mL).010.19 (.05–.70)
ParametersP ValueOdds Ratio (95% CI)
pp65 cultured ELISPOT (>20 spots/million PBMCs)<.0133.15 (2.96–371.96)
Avidity index (for each 10% increase).031.93 (1.07–3.47)
DNAemia (log10 copies/mL).010.19 (.05–.70)

Abbreviations: CI, confidence interval; ELISPOT, enzyme-linked immunospot assay; PBMC, peripheral blood mononuclear cell.

DISCUSSION

In this study, memory HCMV-specific T cells with proliferative capacity were investigated in pregnant women experiencing primary HCMV infection. We used a cultured ELISPOT and 15-mer peptide pools that allow the detection of CD4+ and CD8+ T cells, regardless of human leukocyte antigen [27, 28]. At 1–2 months after infection onset, specific T cells detected by the cultured ELISPOT (mostly CD4+ T cells) were mainly directed to the HCMV pp65. A higher cultured ELISPOT response, associated with a higher AI and low DNAemia, was associated with a lower risk of transmission to the fetus.

While the standard ELISPOT allows detection of effector or effector-memory T cells, the cultured ELISPOT (through antigen-specific expansion of T cells for 12 days) detects memory T cells with proliferative capacity [17, 29]; these cells display characteristics of central memory T cells [18]. Pregnant women with remote infection exhibited memory T cells with proliferative capacity specific for pp65, IE-1, and IE-2. Pregnant women examined in the second month after onset of primary HCMV infection had a significantly lower response to the 3 proteins examined with respect to remote infections, and the pp65-specific T-cell response was significantly greater with respect to IE-1 and IE-2. This may be due to the fact that the IE-specific T cells are mainly CD8+, and this subset is poorly expandable in vitro during primary infection [7]. On the contrary, CD4+ T cells are the major responders in the cultured ELISPOT during primary infection and recognize pp65 better than IE antigens. This response is different from that observed after T-cell stimulation for a few hours. With the latter assay, effector-memory CD4+ and CD8+ T cells specific for pp65 and IE-1 are already detectable within a few weeks after infection onset [30]. While effector T cells, which promptly limit the infection, have a lower proliferative potential in vitro (thus, are not detected by the cultured ELISPOT), memory T cells with proliferative capacity appear much later.

By taking advantage of the cultured ELISPOT assay, we observed a significantly higher proliferative T-cell response (to pp65 but not to IE-1 or IE-2) in nontransmitting women. Conversely, a recent study adopting the standard ELISPOT reported apparently discordant results, as higher T-cell response, most likely CD8+, was associated with HCMV transmission to the fetus [15]. However, as mentioned above, the standard ELISPOT detects effector T cells (regardless of their proliferative capacity). In addition, in the aforementioned study, primary and remote infections were grouped together. When considering only primary infections, we found no significant difference for CD4+ and CD8+ IE-1– or pp65-specific effector T-cell responses (detected by flow cytometry) between transmitting and nontransmitting women [31]. Instead, the more rapid development of a particular effector memory T-cell subset, reexpressing CD45RA (TEMRA), was associated with the control of infection [8, 9, 32]. HCMV-specific T-cell responses in women transmitting the infection was also examined by Quantiferon [14]. In this case, the amount of IFN-γ released by HCMV-specific CD8+ T cells (normalized on the IFN-γ response to PHA) was lower in nontransmitting women. As it was observed that transmitting women had a delayed clearance of HCMV DNA in blood [33], it is possible that a strong, specific T-cell response is induced by higher amounts of antigen. In these previous studies, only the “effector-like” response (IFN-γ production) of HCMV-specific T cells was determined, regardless of their “memory-like” response (ie, proliferation). In the present study, we investigated a particular subset of T cells that are important for the establishment of long-term memory. Thus, it seems that higher levels of the effector T-cell response and lower levels of T cells with proliferative capacity are associated with transmission to the fetus.

During primary infection, HCMV-specific T cells have a decreased ability to proliferate and secrete interleukin 2 [6, 7, 34]. Most important, and in agreement with the present results, a delayed HCMV-specific CD4+ lymphoproliferative response was observed in women transmitting the infection to the fetus [6, 7]. In the present study, we observed that memory T cells with proliferative capacity detected by cultured ELISPOT within 2 months after onset of infection were mainly CD4+. The crucial role of maternal CD4+ T cells in preventing CMV transmission to the fetus is supported by a study of Bialas and colleagues [35], in which the depletion of CD4+ T cells in the rhesus macaque model was associated with a reduced virus-specific CD8+ T-cell response, delayed production of neutralizing antibodies, and higher plasma and amniotic fluid viral load. Additionally, in transplant recipients who do not spontaneously control HCMV infection, significantly lower levels of CD4+ T cells were observed [12].

In addition to the high proliferative T-cell response, we observed that high IgG AI was associated with a lower risk of transmission to the fetus. The appearance of early antibody with high avidity was previously correlated with a lower risk of HCMV transmission to the fetus [36]. On the contrary, neither neutralization titer nor IgG to glycoprotein complexes was found to be associated with transmission in the second month after infection, although in a previous study we found higher titers of IgG to gH/gL/gO and gH/gL/pUL128L in nontransmitting mothers at an earlier time-point [5]. Finally, as already reported, a lower DNAemia was detected in nontransmitting mothers [5, 33]. Multivariate logistic regression analysis showed that the 3 parameters (high proliferative T-cell response, high AI, and low DNAemia) were all independently associated with a lower risk of congenital infection, although the T-cell response showed the highest association.

Our observation that both T-cell and antibody response correlate with the outcome of maternal primary infection may suggest that an ideal candidate vaccine should be able to elicit both humoral and T-cell response against HCMV. Accordingly, in a recent study in the guinea pig model, the combined vaccination with gB and pp65 was able to induce both humoral and cellular response and conferred better protection against maternal viremia than monovalent vaccines [37].

Limitations of our study are the small number of patients examined and the adoption of nonstandardized techniques. In particular, it should be verified also whether the cultured ELISPOT assay could be exported outside the research setting. However, results of the logistic regression analysis indicate that the combination of different parameters (T-cell response, serology, and DNAemia) could provide useful information for the assessment of risk of HCMV congenital infection. In conclusion, confirmation of results reported in a larger population should be achievable in the next future.

Notes

Acknowledgments. We thank Daniela Sartori for editing the manuscript; Laurene Kelly for revision of the English; and Daniele Lilleri for helpful discussion and revision of the manuscript.

Financial support. This work was supported by the Ministero della Salute, Ricerca Corrente (grant number 80207) and Fondazione Cariplo (grant number 2012-0626).

Potential conflicts of interest. All authors: No reported conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

References

1.

Kenneson
A
,
Cannon
MJ
.
Review and meta-analysis of the epidemiology of congenital cytomegalovirus (CMV) infection
.
Rev Med Virol
2007
;
17
:
253
76
.

2.

Dollard
SC
,
Grosse
SD
,
Ross
DS
.
New estimates of the prevalence of neurological and sensory sequelae and mortality associated with congenital cytomegalovirus infection
.
Rev Med Virol
2007
;
17
:
355
63
.

3.

Britt
W
.
Controversies in the natural history of congenital human cytomegalovirus infection: the paradox of infection and disease in offspring of women with immunity prior to pregnancy
.
Med Microbiol Immunol
2015
;
204
:
263
71
.

4.

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
.

5.

Lilleri
D
,
Kabanova
A
,
Revello
MG
et al. 
Fetal human cytomegalovirus transmission correlates with delayed maternal antibodies to gH/gL/pUL128-130-131 complex during primary infection
.
PLoS One
2013
;
8
:
e59863
.

6.

Revello
MG
,
Lilleri
D
,
Zavattoni
M
et al. 
Lymphoproliferative response in primary human cytomegalovirus (HCMV) infection is delayed in HCMV transmitter mothers
.
J Infect Dis
2006
;
193
:
269
76
.

7.

Lilleri
D
,
Fornara
C
,
Furione
M
,
Zavattoni
M
,
Revello
MG
,
Gerna
G
.
Development of human cytomegalovirus-specific T cell immunity during primary infection of pregnant women and its correlation with virus transmission to the fetus
.
J Infect Dis
2007
;
195
:
1062
70
.

8.

Fornara
C
,
Lilleri
D
,
Revello
MG
et al. 
Kinetics of effector functions and phenotype of virus-specific and γδ T lymphocytes in primary human cytomegalovirus infection during pregnancy
.
J Clin Immunol
2011
;
31
:
1054
64
.

9.

Fornara
C
,
Furione
M
,
Arossa
A
,
Gerna
G
,
Lilleri
D
.
Comparative magnitude and kinetics of human cytomegalovirus-specific CD4⁺ and CD8⁺ T-cell responses in pregnant women with primary versus remote infection and in transmitting versus non-transmitting mothers: its utility for dating primary infection in pregnancy
.
J Med Virol
2016
;
88
:
1238
46
.

10.

Meyers
JD
,
Flournoy
N
,
Thomas
ED
.
Cytomegalovirus infection and specific cell-mediated immunity after marrow transplant
.
J Infect Dis
1980
;
142
:
816
24
.

11.

Gamadia
LE
,
Remmerswaal
EB
,
Weel
JF
,
Bemelman
F
,
van Lier
RA
,
Ten Berge
IJ
.
Primary immune responses to human CMV: a critical role for IFN-gamma-producing CD4+ T cells in protection against CMV disease
.
Blood
2003
;
101
:
2686
92
.

12.

Gabanti
E
,
Bruno
F
,
Lilleri
D
et al. 
Human cytomegalovirus (HCMV)-specific CD4+ and CD8+ T cells are both required for prevention of HCMV disease in seropositive solid-organ transplant recipients
.
PLoS One
2014
;
9
:
e106044
.

13.

Gabanti
E
,
Lilleri
D
,
Ripamonti
F
et al. 
Reconstitution of human cytomegalovirus-specific CD4+ T cells is critical for control of virus reactivation in hematopoietic stem cell transplant recipients but does not prevent organ infection
.
Biol Blood Marrow Transplant
2015
;
21
:
2192
202
.

14.

Eldar-Yedidia
Y
,
Bar-Meir
M
,
Hillel
M
et al. 
Low interferon relative-response to cytomegalovirus is associated with low likelihood of intrauterine transmission of the virus
.
PLoS One
2016
;
11
:
e0147883
.

15.

Saldan
A
,
Forner
G
,
Mengoli
C
,
Gussetti
N
,
Palù
G
,
Abate
D
.
Strong cell-mediated immune response to human cytomegalovirus is associated with increased risk of fetal infection in primarily infected pregnant women
.
Clin Infect Dis
2015
;
61
:
1228
34
.

16.

Sallusto
F
,
Lenig
D
,
Förster
R
,
Lipp
M
,
Lanzavecchia
A
.
Two subsets of memory T lymphocytes with distinct homing potentials and effector functions
.
Nature
1999
;
401
:
708
12
.

17.

Calarota
SA
,
Chiesa
A
,
Zelini
P
,
Comolli
G
,
Minoli
L
,
Baldanti
F
.
Detection of Epstein-Barr virus-specific memory CD4+ T cells using a peptide-based cultured enzyme-linked immunospot assay
.
Immunology
2013
;
139
:
533
44
.

18.

Todryk
SM
,
Pathan
AA
,
Keating
S
et al. 
The relationship between human effector and memory T cells measured by ex vivo and cultured ELISPOT following recent and distal priming
.
Immunology
2009
;
128
:
83
91
.

19.

Revello
MG
,
Percivalle
E
,
Zannino
M
,
Rossi
V
,
Gerna
G
.
Development and evaluation of a capture ELISA for IgM antibody to the human cytomegalovirus major DNA binding protein
.
J Virol Methods
1991
;
35
:
315
29
.

20.

Revello
MG
,
Genini
E
,
Gorini
G
,
Klersy
C
,
Piralla
A
,
Gerna
G
.
Comparative evaluation of eight commercial human cytomegalovirus IgG avidity assays
.
J Clin Virol
2010
;
48
:
255
9
.

21.

Furione
M
,
Rognoni
V
,
Cabano
E
,
Baldanti
F
.
Kinetics of human cytomegalovirus (HCMV) DNAemia in transplanted patients expressed in international units as determined with the Abbott RealTime CMV assay and an in-house assay
.
J Clin Virol
2012
;
55
:
317
22
.

22.

Revello
MG
,
Furione
M
,
Rognoni
V
,
Arossa
A
,
Gerna
G
.
Cytomegalovirus DNAemia in pregnant women
.
J Clin Virol
2014
;
61
:
590
2
.

23.

Revello
MG
,
Zavattoni
M
,
Furione
M
,
Lilleri
D
,
Gorini
G
,
Gerna
G
.
Diagnosis and outcome of preconceptional and periconceptional primary human cytomegalovirus infections
.
J Infect Dis
2002
;
186
:
553
7
.

24.

Lilleri
D
,
Gerna
G
,
Furione
M
,
Zavattoni
M
,
Spinillo
A
.
Neutralizing and ELISA IgG antibodies to human cytomegalovirus glycoprotein complexes may help date the onset of primary infection in pregnancy
.
J Clin Virol
2016
;
81
:
16
24
.

25.

Gerna
G
,
Sarasini
A
,
Patrone
M
et al. 
Human cytomegalovirus serum neutralizing antibodies block virus infection of endothelial/epithelial cells, but not fibroblasts, early during primary infection
.
J Gen Virol
2008
;
89
:
853
65
.

26.

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
.

27.

Kiecker
F
,
Streitz
M
,
Ay
B
et al. 
Analysis of antigen-specific T-cell responses with synthetic peptides—what kind of peptide for which purpose?
Hum Immunol
2004
;
65
:
523
36
.

28.

Zandvliet
ML
,
van Liempt
E
,
Jedema
I
et al. 
Co-ordinated isolation of CD8(+) and CD4(+) T cells recognizing a broad repertoire of cytomegalovirus pp65 and IE1 epitopes for highly specific adoptive immunotherapy
.
Cytotherapy
2010
;
12
:
933
44
.

29.

Calarota
SA
,
Foli
A
,
Maserati
R
et al. 
HIV-1-specific T cell precursors with high proliferative capacity correlate with low viremia and high CD4 counts in untreated individuals
.
J Immunol
2008
;
180
:
5907
15
.

30.

Khan
N
,
Best
D
,
Bruton
R
,
Nayak
L
,
Rickinson
AB
,
Moss
PA
.
T cell recognition patterns of immunodominant cytomegalovirus antigens in primary and persistent infection
.
J Immunol
2007
;
178
:
4455
65
.

31.

Lilleri
D
,
Zelini
P
,
Fornara
C
,
Comolli
G
,
Revello
MG
,
Gerna
G
.
Human cytomegalovirus-specific CD4+ and CD8+ T cell responses in primary infection of the immunocompetent and the immunocompromised host
.
Clin Immunol
2009
;
131
:
395
403
.

32.

Lilleri
D
,
Fornara
C
,
Revello
MG
,
Gerna
G
.
Human cytomegalovirus-specific memory CD8+ and CD4+ T cell differentiation after primary infection
.
J Infect Dis
2008
;
198
:
536
43
.

33.

Zavattoni
M
,
Furione
M
,
Lanzarini
P
et al. 
Monitoring of human cytomegalovirus DNAemia during primary infection in transmitter and non-transmitter mothers
.
J Clin Virol
2016
;
82
:
89
93
.

34.

Antoine
P
,
Olislagers
V
,
Huygens
A
et al. 
Functional exhaustion of CD4+ T lymphocytes during primary cytomegalovirus infection
.
J Immunol
2012
;
189
:
2665
72
.

35.

Bialas
KM
,
Tanaka
T
,
Tran
D
et al. 
Maternal CD4+ T cells protect against severe congenital cytomegalovirus disease in a novel nonhuman primate model of placental cytomegalovirus transmission
.
Proc Natl Acad Sci U S A
2015
;
112
:
13645
50
.

36.

Furione
M
,
Rognoni
V
,
Sarasini
A
et al. 
Slow increase in IgG avidity correlates with prevention of human cytomegalovirus transmission to the fetus
.
J Med Virol
2013
;
85
:
1960
7
.

37.

Schleiss
MR
,
Berka
U
,
Watson
E
et al. 
Additive protection against congenital cytomegalovirus conferred by combined glycoprotein B/pp65 vaccination using a lymphocytic choriomeningitis virus vector
.
Clin Vaccine Immunol
2017
;
24
:
e00300
16
.

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.