Browsing by Author "Permar, Sallie R"
- Results Per Page
- Sort Options
Item Open Access Association of HIV-1 Envelope-Specific Breast Milk IgA Responses with Reduced Risk of Postnatal Mother-to-Child Transmission of HIV-1.(J Virol, 2015-10) Pollara, Justin; McGuire, Erin; Fouda, Genevieve G; Rountree, Wes; Eudailey, Josh; Overman, R Glenn; Seaton, Kelly E; Deal, Aaron; Edwards, R Whitney; Tegha, Gerald; Kamwendo, Deborah; Kumwenda, Jacob; Nelson, Julie AE; Liao, Hua-Xin; Brinkley, Christie; Denny, Thomas N; Ochsenbauer, Christina; Ellington, Sascha; King, Caroline C; Jamieson, Denise J; van der Horst, Charles; Kourtis, Athena P; Tomaras, Georgia D; Ferrari, Guido; Permar, Sallie RUNLABELLED: Infants born to HIV-1-infected mothers in resource-limited areas where replacement feeding is unsafe and impractical are repeatedly exposed to HIV-1 throughout breastfeeding. Despite this, the majority of infants do not contract HIV-1 postnatally, even in the absence of maternal antiretroviral therapy. This suggests that immune factors in breast milk of HIV-1-infected mothers help to limit vertical transmission. We compared the HIV-1 envelope-specific breast milk and plasma antibody responses of clade C HIV-1-infected postnatally transmitting and nontransmitting mothers in the control arm of the Malawi-based Breastfeeding Antiretrovirals and Nutrition Study using multivariable logistic regression modeling. We found no association between milk or plasma neutralization activity, antibody-dependent cell-mediated cytotoxicity, or HIV-1 envelope-specific IgG responses and postnatal transmission risk. While the envelope-specific breast milk and plasma IgA responses also did not reach significance in predicting postnatal transmission risk in the primary model after correction for multiple comparisons, subsequent exploratory analysis using two distinct assay methodologies demonstrated that the magnitudes of breast milk total and secretory IgA responses against a consensus HIV-1 envelope gp140 (B.con env03) were associated with reduced postnatal transmission risk. These results suggest a protective role for mucosal HIV-1 envelope-specific IgA responses in the context of postnatal virus transmission. This finding supports further investigations into the mechanisms by which mucosal IgA reduces risk of HIV-1 transmission via breast milk and into immune interventions aimed at enhancing this response. IMPORTANCE: Infants born to HIV-1-infected mothers are repeatedly exposed to the virus in breast milk. Remarkably, the transmission rate is low, suggesting that immune factors in the breast milk of HIV-1-infected mothers help to limit transmission. We compared the antibody responses in plasma and breast milk of HIV-1-transmitting and -nontransmitting mothers to identify responses that correlated with reduced risk of postnatal HIV-1 transmission. We found that neither plasma nor breast milk IgG antibody responses were associated with risk of HIV-1 transmission. In contrast, the magnitudes of the breast milk IgA and secretory IgA responses against HIV-1 envelope proteins were associated with reduced risk of postnatal HIV-1 transmission. The results of this study support further investigations of the mechanisms by which mucosal IgA may reduce the risk of HIV-1 transmission via breastfeeding and the development of strategies to enhance milk envelope-specific IgA responses to reduce mother-to-child HIV transmission and promote an HIV-free generation.Item Open Access Combined HIV-1 Envelope Systemic and Mucosal Immunization of Lactating Rhesus Monkeys Induces a Robust Immunoglobulin A Isotype B Cell Response in Breast Milk.(Journal of virology, 2016-05) Nelson, Cody S; Pollara, Justin; Kunz, Erika L; Jeffries, Thomas L; Duffy, Ryan; Beck, Charles; Stamper, Lisa; Wang, Minyue; Shen, Xiaoying; Pickup, David J; Staats, Herman F; Hudgens, Michael G; Kepler, Thomas B; Montefiori, David C; Moody, M Anthony; Tomaras, Georgia D; Liao, Hua-Xin; Haynes, Barton F; Ferrari, Guido; Fouda, Genevieve GA; Permar, Sallie RUnlabelled
Maternal vaccination to induce anti-HIV immune factors in breast milk is a potential intervention to prevent postnatal HIV-1 mother-to-child transmission (MTCT). We previously demonstrated that immunization of lactating rhesus monkeys with a modified vaccinia Ankara (MVA) prime/intramuscular (i.m.) protein boost regimen induced functional IgG responses in milk, while MVA prime/intranasal (i.n.) boost induced robust milk Env-specific IgA responses. Yet, recent studies have suggested that prevention of postnatal MTCT may require both Env-specific IgA and functional IgG responses in milk. Thus, to investigate whether both responses could be elicited by a combined systemic/mucosal immunization strategy, animals previously immunized with the MVA prime/i.n. boost regimen received an i.n./i.m. combined C.1086 gp120 boost. Remarkably, high-magnitude Env-specific IgA responses were observed in milk, surpassing those in plasma. Furthermore, 29% of vaccine-elicited Env-specific B cells isolated from breast milk were IgA isotype, in stark contrast to the overwhelming predominance of IgG isotype Env-specific B cells in breast milk of chronically HIV-infected women. A clonal relationship was identified between Env-specific blood and breast milk B cells, suggesting trafficking of that cell population between the two compartments. Furthermore, IgA and IgG monoclonal antibodies isolated from Env-specific breast milk B cells demonstrated diverse Env epitope specificities and multiple effector functions, including tier 1 neutralization, antibody-dependent cellular cytotoxicity (ADCC), infected cell binding, and inhibition of viral attachment to epithelial cells. Thus, maternal i.n./i.m. combined immunization is a novel strategy to enhance protective Env-specific IgA in milk, which is subsequently transferred to the infant via breastfeeding.Importance
Efforts to increase the availability of antiretroviral therapy to pregnant and breastfeeding women in resource-limited areas have proven remarkably successful at reducing HIV vertical transmission rates. However, more than 200,000 children are infected annually due to failures in therapy implementation, monitoring, and adherence, nearly half by postnatal HIV exposure via maternal breast milk. Intriguingly, in the absence of antiretroviral therapy, only 10% of breastfed infants born to HIV-infected mothers acquire the virus, suggesting the existence of naturally protective immune factors in milk. Enhancement of these protective immune factors through maternal vaccination will be a critical strategy to reduce the global pediatric AIDS epidemic. We have previously demonstrated that a high magnitude of HIV Env-specific IgA in milk correlates with reduced risk of infant HIV acquisition. In this study, we describe a novel HIV vaccine regimen that induces potent IgA responses in milk and therefore could potentially protect against breast milk HIV MTCT.Item Open Access Congenital human cytomegalovirus infection is associated with decreased transplacental IgG transfer efficiency due to maternal hypergammaglobulinemia.(Clinical infectious diseases : an official publication of the Infectious Diseases Society of America, 2021-07-14) Semmes, Eleanor C; Li, Shuk Hang; Hurst, Jillian H; Yang, Zidanyue; Niedzwiecki, Donna; Fouda, Genevieve G; Kurtzberg, Joanne; Walsh, Kyle M; Permar, Sallie RBackground
Placentally-transferred maternal IgG protects against pathogens in early life, yet vertically-transmitted infections can interfere with transplacental IgG transfer. Although human cytomegalovirus (HCMV) is the most common placentally-transmitted viral infection worldwide, the impact of congenital HCMV (cCMV) infection on transplacental IgG transfer has been underexplored.Methods
We evaluated total and antigen-specific maternal and cord blood IgG levels and transplacental IgG transfer efficiency in a U.S-based cohort of 93 mother-infant pairs including 27 cCMV-infected and 66 cCMV-uninfected pairs, of which 29 infants were born to HCMV-seropositive non-transmitting mothers and 37 to HCMV-seronegative mothers. Controls were matched on sex, race/ethnicity, maternal age, and delivery year.Results
Transplacental IgG transfer efficiency was decreased by 23% (95% CI 10-36%, p=0.0079) in cCMV-infected pairs and 75% of this effect (95% CI 28-174%, p=0.0085) was mediated by elevated maternal IgG levels (i.e., hypergammaglobulinemia) in HCMV-transmitting women. Despite reduced transfer efficiency, IgG levels were similar in cord blood from infants with and without cCMV infection.Conclusions
Our results indicate that cCMV infection moderately reduces transplacental IgG transfer efficiency due to maternal hypergammaglobulinemia; however, infants with and without cCMV infection had similar antigen-specific IgG levels, suggesting comparable protection from maternal IgG acquired via transplacental transfer.Item Open Access Conjugation of HIV-1 envelope to hepatitis B surface antigen alters vaccine responses in rhesus macaques.(NPJ vaccines, 2023-11) Nettere, Danielle; Unnithan, Shakthi; Rodgers, Nicole; Nohara, Junsuke; Cray, Paul; Berry, Madison; Jones, Caroline; Armand, Lawrence; Li, Shuk Hang; Berendam, Stella J; Fouda, Genevieve G; Cain, Derek W; Spence, Taylor N; Granek, Joshua A; Davenport, Clemontina A; Edwards, Robert J; Wiehe, Kevin; Van Rompay, Koen KA; Moody, M Anthony; Permar, Sallie R; Pollara, JustinAn effective HIV-1 vaccine remains a critical unmet need for ending the AIDS epidemic. Vaccine trials conducted to date have suggested the need to increase the durability and functionality of vaccine-elicited antibodies to improve efficacy. We hypothesized that a conjugate vaccine based on the learned response to immunization with hepatitis B virus could be utilized to expand T cell help and improve antibody production against HIV-1. To test this, we developed an innovative conjugate vaccine regimen that used a modified vaccinia virus Ankara (MVA) co-expressing HIV-1 envelope (Env) and the hepatitis B virus surface antigen (HBsAg) as a prime, followed by two Env-HBsAg conjugate protein boosts. We compared the immunogenicity of this conjugate regimen to matched HIV-1 Env-only vaccines in two groups of 5 juvenile rhesus macaques previously immunized with hepatitis B vaccines in infancy. We found expansion of both HIV-1 and HBsAg-specific circulating T follicular helper cells and elevated serum levels of CXCL13, a marker for germinal center activity, after boosting with HBsAg-Env conjugate antigens in comparison to Env alone. The conjugate vaccine elicited higher levels of antibodies binding to select HIV Env antigens, but we did not observe significant improvement in antibody functionality, durability, maturation, or B cell clonal expansion. These data suggests that conjugate vaccination can engage both HIV-1 Env and HBsAg specific T cell help and modify antibody responses at early time points, but more research is needed to understand how to leverage this strategy to improve the durability and efficacy of next-generation HIV vaccines.Item Open Access Cultivating Research Skills During Clinical Training to Promote Pediatric-Scientist Development.(Pediatrics, 2019-08) Hurst, Jillian H; Barrett, Katherine J; Kelly, Matthew S; Staples, Betty B; McGann, Kathleen A; Cunningham, Coleen K; Reed, Ann M; Gbadegesin, Rasheed A; Permar, Sallie RPhysician-scientists represent a critical component of the biomedical and health research workforce. However, the proportion of physicians who spend a significant amount of effort on scientific research has declined over the past 40 years. This trend has been particularly noticeable in pediatrics despite recent scientific work revealing that early life influences, exposures, and health status play a significant role in lifelong health and disease. To address this problem, the Duke University Department of Pediatrics developed the Duke Pediatric Research Scholars Program for Physician-Scientist Development (DPRS). The DPRS is focused on research training during pediatric residency and fellowship. We aim to provide sufficient research exposure and support to help scholars develop a research niche and scholarly products as well as identify the career pathways that will enable them to achieve their research goals. Herein, we describe the DPRS's organizational structure, core components, recruitment strategies, and initial results, and we discuss implementation challenges and solutions. Additionally, we detail the program's integration with the department's residency and fellowship training programs (with particular reference to the challenges of integrating research into small- to medium-sized residency programs) and describe the development and integration of related initiatives across Duke University School of Medicine. The program served as the basis for 2 successful National Institutes of Health Stimulating Access to Research in Residency (R38) applications, and we hope it will serve as a model to integrate formalized research training for residents and fellows who wish to pursue research careers in academic medicine.Item Open Access Defining the Role of Antibodies in Protection Against Cytomegalovirus Acquisition and Congenital Disease for Rational Vaccine Design(2018) Nelson, Cody ShawHuman cytomegalovirus (HCMV) is the most common cause of congenital infection worldwide, impacting 1 in 150 live-born infants. Children afflicted by congenital HCMV frequently suffer from lifelong, debilitating neurologic sequelae including microcephaly, sensorineural hearing loss, and cognitive impairment. Natural maternal immunity to HCMV reduces the frequency of congenital infection, but does not prevent the disease altogether. Thus, a vaccine to reduce the incidence and severity of infant infection is a public health priority. Employing a nonhuman primate model of congenital CMV transmission as well as clinical samples from a partially-efficacious HCMV vaccine trial, we sought to examine both the attributes of anti-HCMV immune responses that provide protective immunity as well as the impact of vaccine-elicited immunity on the in vivo HCMV viral population.
First, we used a nonhuman primate model of congenital CMV infection to investigate the ability of preexisting antibodies to protect against placental CMV transmission in the setting of primary maternal infection and subsequent viremia, which is required for placental virus exposure. Pregnant, CD4+ T cell-depleted, rhesus CMV (RhCMV)-seronegative rhesus monkeys were treated with either standardly-produced hyperimmune globulin (HIG) from RhCMV-seropositive macaques or dose-optimized, potently RhCMV-neutralizing HIG prior to intravenous challenge with an RhCMV swarm. HIG passive infusion provided complete protection against fetal loss in both groups. The dose-optimized, RhCMV-neutralizing HIG additionally inhibited placental transmission of RhCMV and reduced viral replication and diversity. Our findings suggest that the presence of durable and potently-neutralizing antibodies at the time of primary infection can prevent transmission of systemically-replicating maternal RhCMV to the developing fetus.
Next, we assessed the properties of antibody responses elicited by glycoprotein B (gB) + MF59 adjuvant subunit vaccination – the most successful HCMV vaccine tested clinicaly to-date, which demonstrated approximately 50% efficacy in preventing HCMV acquisition in multiple phase 2 trials. Plasma from 33 gB/MF59 vaccinees at peak immunogenicity was tested for gB epitope specificity as well as neutralizing and non-neutralizing anti-HCMV effector functions, and compared to an HCMV-seropositive cohort. gB/MF59 vaccination elicited IgG responses with gB-binding magnitude and avidity comparable to natural infection. Additionally, IgG subclass distribution was similar with predominant IgG1 and IgG3 responses induced by gB vaccination and HCMV infection. However, vaccine-elicited antibodies exhibited limited neutralization of the autologous virus, negligible neutralization of multiple heterologous strains, and limited binding responses against gB structural motifs targeted by neutralizing antibodies including AD-1, AD-2, and Domain I. Interestingly, vaccinees had high-magnitude IgG responses against AD-3 linear epitopes, demonstrating immunodominance against this non-neutralizing, cytosolic region. Finally, vaccine-elicited IgG robustly bound trimeric, membrane-associated gB on the surface of transfected or HCMV-infected cells and mediated virion phagocytosis, raising the possibility that non-neutralizing antibody effector functions contributed to the partial protection against HCMV acquisition observed in gB/MF59 vaccinees.
Lastly, we evaluated the impact of gB/MF59-elicited immune responses on the population of viruses acquired by trial participants. In this analysis, we employed quantitative PCR as well as two distinct next-generation sequencing strategies (short amplicon and whole gene) to interrogate genetic differences between the HCMV populations infecting gB/MF59 vaccinees and placebo recipients. For the majority of subject-specific viral populations analyzed, we identified 1 or 2 dominant viral variants, as well as a large number of minor variants present at very low frequency. This finding suggests that the intrahost viral population constitutes a heterogeneous swarm of genetically-distinct virus quasi-species. Additionally, we identified several distinctions between the viral populations of acutely-infected vaccinees and placebo recipients. First, there was reduced magnitude viral shedding in the saliva of gB vaccinees compared to placebo. Furthermore, we noted evidence of genetic compartmentalization at the gB locus in 3 of 4 vaccinees, though only in 1 of 7 placebo recipients. Finally, we observed an enrichment of gB1 genotype HCMV variants among placebo recipients compared to vaccinees, and hypothesize that the gB1 genotype vaccine immunogen might have elicited genotype-specific protection that accounts for the efficacy observed in clinical trial.
Thus, we have made several observations that will inform rational design of the next generation of HCMV vaccines. First, our data suggests that preexisting antibodies can protect against congenital CMV transmission in a rhesus monkey model, and thus that antibodies could be a primary target of vaccines to eliminate congenital HCMV infection. Secondly, our analysis of antibody responses elicited by gB/MF59 vaccination indicates that non-neutralizing antibody functions contributed to the observed 50% vaccine protection and therefore should be a consideration in future vaccine design. Finally, our examination of viral populations in gB/MF59 vaccinees indicates that gB-elicited antibodies had a measurable impact on viral intrahost population dynamics and that gB immunogen strain-specific responses may have defined vaccine protection, suggesting that immunogen strain breadth may be an important factor to consider for future vaccine design.
Item Open Access Envelope-specific IgG Responses in HIV-infected Women(2018) Martinez, David RafaelA better understanding of 1) maternal HIV Envelope (Env)-specific IgG responses that are partially protective against vertical HIV transmission, and 2) factors that mediate the transplacental transfer of maternal protective IgG is needed to improve infant health in early life, in which maternal passively-acquired IgG mediates protection against neonatal infections. To understand maternal factors and IgG characteristics that mediate transplacental IgG transfer, we examined transplacental transfer efficiency determinants of maternal HIV and standard vaccine-antigen-specific IgG in a population of HIV-infected women, which have disrupted transplacental IgG transfer. Our findings suggest that maternal health factors and maternal IgG characteristics, such as binding to placentally expressed Fc receptors, IgG subclass frequency, and Fc region glycan profiles all mediate transplacental IgG transfer efficiency. We also identified maternal linear variable loop 3 (V3)-specific IgG binding and neutralizing responses targeting the C terminal region as partially protective against vertical transmission of HIV. These novel findings provide a roadmap of maternal factors and IgG characteristics as targets that can be harnessed to improve the transplacental IgG transfer of routinely-administered maternal vaccines and benchmarks for assessing maternal HIV vaccines that target the V3 loop.
Item Unknown Evaluating Humoral Immune Responses Against HLA and Cytomegalovirus in Human Lung Transplantation(2024) Harnois, MelissaIn the past 30 years, the global prevalence of chronic respiratory diseases has increased by 40%. Importantly, lung transplantation remains the only definitive treatment for patients with advanced lung disease. Despite the rapidly growing need for lung transplantation, lung transplant recipients have by far the worst survival rates among all solid organ transplant types, with a current median survival of only 6 years post transplantation. Chronic lung allograft dysfunction (CLAD) is the leading cause of limited long-term survival of lung transplant recipients, affecting approximately 50% of patients within 5 years post-transplantation. Previous studies have identified humoral alloimmune responses against mismatched donor HLA and cytomegalovirus (CMV) infection as primary independent risk factors for chronic deterioration of the transplanted allograft and the fatal diagnosis of CLAD. This dissertation is comprised of work that aims to address critical gaps in knowledge in the fields of transplant immunology and infectious disease. The first component focuses on CMV-specific humoral immunity in naturally infected healthy blood donors (Chapter 2) and in CMV seropositive lung transplant recipients (Chapter 3). The second area of study focuses on humoral alloimmune responses and current methods used to evaluate and characterize donor HLA-specific alloantibodies (Chapter 4), as well as factors contributing to HLA-DQ immunogenicity (Chapter 5). These projects utilized clinical data and longitudinally collected human plasma samples from the completed multicenter Clinical Trials in Organ Transplantation (CTOT)-20 and CTOT-22 consortium studies. The findings from Chapter 2 contribute to our knowledge of naturally acquired immunity against CMV, which informs strategies for antibody-based therapeutics to help treat CMV infection as well as CMV vaccine development. The results from Chapter 3 indicate that CMV-specific antibodies have the potential to be used as novel biomarkers for viremia risk, possibly in combination with other known risk factors for viremia including CMV-specific T cell responses. These tools may help inform antiviral treatment duration in clinical practice. Chapter 4 of this dissertation represents the first in-depth technical comparison across traditional and modified single antigen bead Luminex assays using IgG enriched sera for donor-HLA specific post-transplant monitoring. These findings may be useful for clinical workflows to help improve efficiency and cost by stratifying patient samples that may benefit from additional testing in modified assays. Finally, Chapter 5 of this dissertation examines the specificity and timing of donor HLA-specific antibodies (DSA), as well as other risk factors for DSA development. In this project, we detail novel strategies for calculating HLA-DQ mismatch by accounting for trans-encoded DQ heterodimer formation. The results from this study also reveal elevated risk for developing DSA among patients with high-risk epitope mismatches and suggest that different organs may require different HLA mismatch calculation strategies. Overall, the results from these projects advance our understanding of the human immune response to alloantigens and cytomegalovirus in lung transplantation and push the field forward to help improve patient outcomes.
Item Embargo Fc-mediated Immunity in Congenital Human Cytomegalovirus Infection(2023) Semmes, Eleanor CaldwellHuman cytomegalovirus (HCMV) is a ubiquitous, host-restricted β-herpesvirus that has co-evolved with humans over millennia. In healthy individuals, HCMV infection is usually mild or potentially beneficial, as seropositivity can boost host heterologous immunity. Conversely, infection in immunocompromised individuals or in utero can cause severe disease. Congenital HCMV infection (cCMV), acquired via transplacental transmission from mother to fetus, is the most common vertically transmitted infection worldwide and is associated with stillbirth, neurodevelopmental impairment, hearing loss, and childhood leukemia. There are no vaccines or therapeutics to prevent cCMV and our knowledge of the impacts of infection on neonatal immunity remain limited. In my dissertation work, we aimed to discover protective immune responses against cCMV infection and to define how cCMV modulates neonatal immunity. To investigate these aims, we established a case-control cohort of mother-infant dyads with and without cCMV infection using the Carolinas Cord Blood Bank. We leveraged paired maternal and cord blood samples to define 1) how HCMV modifies placental IgG transfer, 2) protective antibody responses against cCMV transmission, and 3) the impact of cCMV on neonatal cellular immunity. First, we found that placental IgG transfer efficiency was reduced in cCMV infection because HCMV transmitting mothers had high rates of hypergammaglobulinemia, leading to saturation of the neonatal Fc receptor (FcRn) and decreased IgG transfer ratios. However, neonates with and without cCMV infection had similar levels of antigen-specific IgG, suggesting equivalent protection conferred by FcRn-mediated IgG transfer. When comparing HCMV transmitting to HCMV seropositive non-transmitting dyads, we observed that placental transfer of high avidity HCMV-specific IgG was reduced in cCMV infection but that cCMV-infected infants had high levels of neutralizing, HCMV-specific IgG. Next, we found that antibody-dependent cellular phagocytosis (ADCP), a Fc-mediated antibody effector function, was associated with protection against cCMV transmission. We determined that HCMV-specific IgG activation of FcγRI and FcγRIIA, which mediate ADCP and other innate immune cell effector functions by binding to the Fc region of IgG, was enhanced in mothers who did not transmit HCMV in utero. Finally, we compared cord blood immunophenotypes in infants with and without cCMV infection. We found that activated CD8+ and CD4+ T cells were increased and that regulatory T cells were reduced in cCMV infection. Moreover, we discovered that a unique population of “NKT-like” CD8+ T cells expressing NK cell markers such as FcγRIIIA/CD16 and NKG2A/C was significantly expanded in cord blood from cCMV-infected neonates. Cumulatively, our data highlights the important role of Fc-mediated immunity in cCMV infection and reveals that HCMV is a powerful immunomodulator in utero, which can guide future vaccination strategies against HCMV and other infectious diseases.
Item Open Access Geographic and Racial Disparities in Infant Hearing Loss.(Otolaryngology--head and neck surgery : official journal of American Academy of Otolaryngology-Head and Neck Surgery, 2018-10-09) Lantos, Paul M; Maradiaga-Panayotti, Gabriela; Barber, Xavier; Raynor, Eileen; Tucci, Debara; Hoffman, Kate; Permar, Sallie R; Jackson, Pearce; Hughes, Brenna L; Kind, Amy; Swamy, Geeta KObjective Approximately 1 to 2 of every 1000 American newborns has hearing loss identified by newborn screening. This study was designed to determine if infant hearing loss is more common in socioeconomically disadvantaged communities. Study Design In this retrospective study, we analyzed electronic medical record data using geostatistical models. Setting Infants were residents of Durham County, North Carolina, born in 2 hospitals of the Duke University Health System. This county includes the city of Durham and surrounding suburban and rural communities. Subjects and Methods Subjects were hearing-screened newborns, born between 2005 and 2016, whose residential address was in Durham County, North Carolina. This was a retrospective study using medical record data. We used Bayesian regression models with smoothing of coordinate date to identify both spatial and nonspatial predictors of infant hearing loss. Results We identified 19,348 infants from Durham County, of whom 675 had failed initial hearing screening and 191 had hearing loss confirmed on follow-up. Hearing loss was significantly associated with minority race (odds ratio [OR], 2.45; 95% confidence interval, 1.97-3.06), as well as lower gestational age and maternal sexually transmitted infections. We identified significant geographic heterogeneity, with a higher probability of hearing loss in poorer urban neighborhoods (local OR range, 0.59-1.39). Neighborhood disadvantage was a significant predictor of hearing loss, as was high local seroprevalence of cytomegalovirus (CMV) among pregnant women. Conclusions Urban, low-income neighborhoods have a high prevalence of infant hearing loss compared with more affluent surrounding communities, particularly among minorities. This distribution may be attributable to congenital CMV infection.Item Open Access Innovations in MD-only physician-scientist training: experiences from the Burroughs Wellcome Fund physician-scientist institutional award initiative.(The Journal of clinical investigation, 2021-05) McElvaine, Allison T; Hawkins-Salsbury, Jacqueline A; Arora, Vineet M; Gladwin, Mark T; Goldenring, James R; Huston, David P; Krakow, Deborah; Rhee, Kyu; Solway, Julian; Steinman, Richard A; Towler, Dwight A; Utz, Paul J; Yokoyama, Wayne M; Simpson, Rolly L; Muglia, Louis J; Permar, Sallie R; Gbadegesin, Rasheed AItem Open Access Isolation of HIV-1-neutralizing mucosal monoclonal antibodies from human colostrum.(PLoS One, 2012) Friedman, James; Alam, S Munir; Shen, Xiaoying; Xia, Shi-Mao; Stewart, Shelley; Anasti, Kara; Pollara, Justin; Fouda, Genevieve G; Yang, Guang; Kelsoe, Garnett; Ferrari, Guido; Tomaras, Georgia D; Haynes, Barton F; Liao, Hua-Xin; Moody, M Anthony; Permar, Sallie RBACKGROUND: Generation of potent anti-HIV antibody responses in mucosal compartments is a potential requirement of a transmission-blocking HIV vaccine. HIV-specific, functional antibody responses are present in breast milk, and these mucosal antibody responses may play a role in protection of the majority of HIV-exposed, breastfeeding infants. Therefore, characterization of HIV-specific antibodies produced by B cells in milk could guide the development of vaccines that elicit protective mucosal antibody responses. METHODS: We isolated B cells from colostrum of an HIV-infected lactating woman with a detectable neutralization response in milk and recombinantly produced and characterized the resulting HIV-1 Envelope (Env)-specific monoclonal antibodies (mAbs). RESULTS: The identified HIV-1 Env-specific colostrum mAbs, CH07 and CH08, represent two of the first mucosally-derived anti-HIV antibodies yet to be reported. Colostrum mAb CH07 is a highly-autoreactive, weakly-neutralizing gp140-specific mAb that binds to linear epitopes in the gp120 C5 region and gp41 fusion domain. In contrast, colostrum mAb CH08 is a nonpolyreactive CD4-inducible (CD4i) gp120-specific mAb with moderate breadth of neutralization. CONCLUSIONS: These novel HIV-neutralizing mAbs isolated from a mucosal compartment provide insight into the ability of mucosal B cell populations to produce functional anti-HIV antibodies that may contribute to protection against virus acquisition at mucosal surfaces.Item Open Access Lack of B cell dysfunction is associated with functional, gp120-dominant antibody responses in breast milk of simian immunodeficiency virus-infected African green monkeys.(J Virol, 2013-10) Amos, Joshua D; Wilks, Andrew B; Fouda, Genevieve G; Smith, Shannon D; Colvin, Lisa; Mahlokozera, Tatenda; Ho, Carrie; Beck, Krista; Overman, R Glenn; DeMarco, C Todd; Hodge, Terry L; LaBranche, Celia C; Montefiori, David C; Denny, Thomas N; Liao, Hua-Xin; Tomaras, Georgia D; Moody, M Anthony; Permar, Sallie RThe design of an effective vaccine to reduce the incidence of mother-to-child transmission (MTCT) of human immunodeficiency virus (HIV) via breastfeeding will require identification of protective immune responses that block postnatal virus acquisition. Natural hosts of simian immunodeficiency virus (SIV) sustain nonpathogenic infection and rarely transmit the virus to their infants despite high milk virus RNA loads. This is in contrast to HIV-infected women and SIV-infected rhesus macaques (RhMs), nonnatural hosts which exhibit higher rates of postnatal virus transmission. In this study, we compared the systemic and mucosal B cell responses of lactating, SIV-infected African green monkeys (AGMs), a natural host species, to that of SIV-infected RhMs and HIV-infected women. AGMs did not demonstrate hypergammaglobulinemia or accumulate circulating memory B cells during chronic SIV infection. Moreover, the milk of SIV-infected AGMs contained higher proportions of naive B cells than RhMs. Interestingly, AGMs exhibited robust milk and plasma Env binding antibody responses that were one to two logs higher than those in RhMs and humans and demonstrated autologous neutralizing responses in milk at 1 year postinfection. Furthermore, the plasma and milk Env gp120-binding antibody responses were equivalent to or predominant over Env gp140-binding antibody responses in AGMs, in contrast to that in RhMs and humans. The strong gp120-specific, functional antibody responses in the milk of SIV-infected AGMs may contribute to the rarity of postnatal transmission observed in natural SIV hosts.Item Open Access Maternal B Cell and Antibody Responses to Zika Virus for Design of Immune Interventions in Pregnancy(2021) Singh, TulikaZika virus (ZIKV) re-emerged in the 2015-2016 epidemic in the Americas, when it was recognized that this mosquito transmitted virus can also be transmitted congenitally. One in 10 infants born to ZIKV-infected pregnancies presented with congenital defects including microcephaly, brain and ocular damage, neurodevelopmental delays, and mobility defects. In the past outbreak 11,000 children in Brazil were born with microcephaly and even more have lifelong disability. To prevent congenital transmission, a vaccine that is effective in pregnancy is urgently needed. Developing such an intervention requires an understanding of the targeted immune responses that mediates protection in pregnancy. Therefore, we investigated maternal B cell and antibody immunity to ZIKV.
First, we characterized ZIKV immunity in mothers with natural ZIKV infection and found that all mounted high ZIKV neutralizing antibody titers within 10 days of symptoms and maintained this throughout gestation. We then evaluated transplacental transfer of IgG and found that maternal ZIKV infection did not impair transfer of vaccine-elicited and flavivirus-neutralizing IgG, indicating that maternal immunization is a viable option to transfer immune protection to the newborn. However, a potential risk of transferred IgG is antibody-dependent enhancement of a heterologous flavivirus in early life. Consequently, we next tested the role of vertically transferred cross-reactive IgG in infant cord blood and found that transferred IgG may mediate in vitro flavivirus enhancement in absence of high ZIKV-neutralizing titers.
Next, we evaluated the role of IgM antibodies in the maternal immune response to ZIKV, as IgM do not cross the placenta during gestation and were found for an unusually long time in many cohorts. We found that plasma IgM contributes to early ZIKV neutralization across several ZIKV-infected pregnant women. We then isolated a potently ZIKV-neutralizing IgM monoclonal antibody, DH1017.IgM, which demonstrates 39-fold increased neutralization activity than a recombinant IgG with the same antigen binding sites, suggesting that the multivalency of the IgM may have a role in function. Potency of DH1017.IgM increased in a dose-dependent manner with complement. Structural studies of the epitope revealed a novel E dimer epitope on Domain II. This epitope can be bound by DH1017.Fab at multiple angles and is computationally predicted to have the capacity to be bound by all five IgM monomers simultaneously, defining a novel route of ZIKV neutralization. Importantly, the DH017.IgM protects mice from a lethal challenge of ZIKV and reduced in vitro enhancement otherwise observed with DH1017.IgG, suggesting that DH1017.IgM may be a suitable candidate for prophylactic intervention in pregnancy.
Altogether, we have identified key aspects of maternal immunity that will inform the development of ZIKV vaccines. First, we show that pregnant women can mount a robust B cell response against ZIKV, that is durable throughout pregnancy, and that protective levels of vaccine-elicited IgG can be transferred transplacentally to infants, suggesting that maternal immunization is a viable strategy to prevent infections in pregnancy and protect the newborn. However, with the transfer of flavivirus cross-reactive IgG, there is a need to monitor infants for enhanced flavivirus disease upon maternal immunization. Subsequently, we found that IgM antibodies in plasma contribute to ZIKV neutralization and identified a novel route for virus neutralization by DH1017.IgM that can be leveraged in immunogen design. Thus, vaccine design should assess if inclusion of multimeric immunogens that mimic the conformational surface of the virion supports development of IgM-mediated immunity.
Item Open Access Maternal Fc-mediated non-neutralizing antibody responses correlate with protection against congenital human cytomegalovirus infection.(The Journal of clinical investigation, 2022-08) Semmes, Eleanor C; Miller, Itzayana G; Wimberly, Courtney E; Phan, Caroline T; Jenks, Jennifer A; Harnois, Melissa J; Berendam, Stella J; Webster, Helen; Hurst, Jillian H; Kurtzberg, Joanne; Fouda, Genevieve G; Walsh, Kyle M; Permar, Sallie RHuman cytomegalovirus (HCMV) is the most common congenital infection and a leading cause of stillbirth, neurodevelopmental impairment, and pediatric hearing loss worldwide. Development of a maternal vaccine or therapeutic to prevent congenital HCMV has been hindered by limited knowledge of the immune responses that protect against HCMV transmission in utero. To identify protective antibody responses, we measured HCMV-specific IgG binding and antiviral functions in paired maternal and cord blood sera from HCMV-seropositive transmitting (n = 41) and non-transmitting (n = 40) mother-infant dyads identified via a large, US-based, public cord blood bank. We found that high-avidity IgG binding to HCMV and antibody-dependent cellular phagocytosis (ADCP) were associated with reduced risk of congenital HCMV infection. We also determined that HCMV-specific IgG activation of FcγRI and FcγRII was enhanced in non-transmitting dyads and that increased ADCP responses were mediated through both FcγRI and FcγRIIA expressed on human monocytes. These findings suggest that engagement of FcγRI/FcγRIIA and Fc effector functions including ADCP may protect against congenital HCMV infection. Taken together, these data can guide future prospective studies on immune correlates against congenital HCMV transmission and inform HCMV vaccine and immunotherapeutic development.Item Open Access Maternal HIV-1 Env Vaccination for Systemic and Breast Milk Immunity To Prevent Oral SHIV Acquisition in Infant Macaques.(mSphere, 2018-01) Eudailey, Joshua A; Dennis, Maria L; Parker, Morgan E; Phillips, Bonnie L; Huffman, Tori N; Bay, Camden P; Hudgens, Michael G; Wiseman, Roger W; Pollara, Justin J; Fouda, Genevieve G; Ferrari, Guido; Pickup, David J; Kozlowski, Pamela A; Van Rompay, Koen KA; De Paris, Kristina; Permar, Sallie RMother-to-child transmission (MTCT) of human immunodeficiency virus type 1 (HIV-1) contributes to an estimated 150,000 new infections annually. Maternal vaccination has proven safe and effective at mitigating the impact of other neonatal pathogens and is one avenue toward generating the potentially protective immune responses necessary to inhibit HIV-1 infection of infants through breastfeeding. In the present study, we tested the efficacy of a maternal vaccine regimen consisting of a modified vaccinia virus Ankara (MVA) 1086.C gp120 prime-combined intramuscular-intranasal gp120 boost administered during pregnancy and postpartum to confer passive protection on infant rhesus macaques against weekly oral exposure to subtype C simian-human immunodeficiency virus 1157ipd3N4 (SHIV1157ipd3N4) starting 6 weeks after birth. Despite eliciting a robust systemic envelope (Env)-specific IgG response, as well as durable milk IgA responses, the maternal vaccine did not have a discernible impact on infant oral SHIV acquisition. This study revealed considerable variation in vaccine-elicited IgG placental transfer and a swift decline of both Env-specific antibodies (Abs) and functional Ab responses in the infants prior to the first challenge, illustrating the importance of pregnancy immunization timing to elicit optimal systemic Ab levels at birth. Interestingly, the strongest correlation to the number of challenges required to infect the infants was the percentage of activated CD4+ T cells in the infant peripheral blood at the time of the first challenge. These findings suggest that, in addition to maternal immunization, interventions that limit the activation of target cells that contribute to susceptibility to oral HIV-1 acquisition independently of vaccination may be required to reduce infant HIV-1 acquisition via breastfeeding. IMPORTANCE Without novel strategies to prevent mother-to-child HIV-1 transmission, more than 5% of HIV-1-exposed infants will continue to acquire HIV-1, most through breastfeeding. This study of rhesus macaque dam-and-infant pairs is the first preclinical study to investigate the protective role of transplacentally transferred HIV-1 vaccine-elicited antibodies and HIV-1 vaccine-elicited breast milk antibody responses in infant oral virus acquisition. It revealed highly variable placental transfer of potentially protective antibodies and emphasized the importance of pregnancy immunization timing to reach peak antibody levels prior to delivery. While there was no discernible impact of maternal immunization on late infant oral virus acquisition, we observed a strong correlation between the percentage of activated CD4+ T cells in infant peripheral blood and a reduced number of challenges to infection. This finding highlights an important consideration for future studies evaluating alternative strategies to further reduce the vertical HIV-1 transmission risk.Item Open Access Maternal HIV-1 envelope-specific antibody responses and reduced risk of perinatal transmission.(J Clin Invest, 2015-07-01) Permar, Sallie R; Fong, Youyi; Vandergrift, Nathan; Fouda, Genevieve G; Gilbert, Peter; Parks, Robert; Jaeger, Frederick H; Pollara, Justin; Martelli, Amanda; Liebl, Brooke E; Lloyd, Krissey; Yates, Nicole L; Overman, R Glenn; Shen, Xiaoying; Whitaker, Kaylan; Chen, Haiyan; Pritchett, Jamie; Solomon, Erika; Friberg, Emma; Marshall, Dawn J; Whitesides, John F; Gurley, Thaddeus C; Von Holle, Tarra; Martinez, David R; Cai, Fangping; Kumar, Amit; Xia, Shi-Mao; Lu, Xiaozhi; Louzao, Raul; Wilkes, Samantha; Datta, Saheli; Sarzotti-Kelsoe, Marcella; Liao, Hua-Xin; Ferrari, Guido; Alam, S Munir; Montefiori, David C; Denny, Thomas N; Moody, M Anthony; Tomaras, Georgia D; Gao, Feng; Haynes, Barton FDespite the wide availability of antiretroviral drugs, more than 250,000 infants are vertically infected with HIV-1 annually, emphasizing the need for additional interventions to eliminate pediatric HIV-1 infections. Here, we aimed to define humoral immune correlates of risk of mother-to-child transmission (MTCT) of HIV-1, including responses associated with protection in the RV144 vaccine trial. Eighty-three untreated, HIV-1-transmitting mothers and 165 propensity score-matched nontransmitting mothers were selected from the Women and Infants Transmission Study (WITS) of US nonbreastfeeding, HIV-1-infected mothers. In a multivariable logistic regression model, the magnitude of the maternal IgG responses specific for the third variable loop (V3) of the HIV-1 envelope was predictive of a reduced risk of MTCT. Neutralizing Ab responses against easy-to-neutralize (tier 1) HIV-1 strains also predicted a reduced risk of peripartum transmission in secondary analyses. Moreover, recombinant maternal V3-specific IgG mAbs mediated neutralization of autologous HIV-1 isolates. Thus, common V3-specific Ab responses in maternal plasma predicted a reduced risk of MTCT and mediated autologous virus neutralization, suggesting that boosting these maternal Ab responses may further reduce HIV-1 MTCT.Item Open Access Neighborhood Disadvantage is Associated with High Cytomegalovirus Seroprevalence in Pregnancy.(J Racial Ethn Health Disparities, 2017-08-24) Lantos, Paul M; Hoffman, Kate; Permar, Sallie R; Jackson, Pearce; Hughes, Brenna L; Kind, Amy; Swamy, GeetaBACKGROUND: Cytomegalovirus (CMV) is the most common infectious cause of fetal malformations and childhood hearing loss. CMV is more common among socially disadvantaged groups, and geographically clusters in poor communities. The Area Deprivation Index (ADI) is a neighborhood-level index derived from census data that reflects material disadvantage. METHODS: We performed a geospatial analysis to determine if ADI predicts the local odds of CMV seropositivity. We analyzed a dataset of 3527 women who had been tested for CMV antibodies during pregnancy. We used generalized additive models to analyze the spatial distribution of CMV seropositivity. Adjusted models included individual-level age and race and neighborhood-level ADI. RESULTS: Our dataset included 1955 CMV seropositive women, 1549 who were seronegative, and 23 with recent CMV infection based on low avidity CMV antibodies. High ADI percentiles, representing greater neighborhood poverty, were significantly associated with the nonwhite race (48 vs. 22, p < 0.001) and CMV seropositivity (39 vs. 28, p < 0.001). Our unadjusted spatial models identified clustering of high CMV odds in poor, urban neighborhoods and clustering of low CMV odds in more affluent suburbs (local odds ratio 0.41 to 1.90). Adjustment for both individual race and neighborhood ADI largely eliminated this spatial variability. ADI remained a significant predictor of local CMV seroprevalence even after adjusting for individual race. CONCLUSIONS: Neighborhood-level poverty as measured by the ADI is a race-independent predictor of local CMV seroprevalence among pregnant women.Item Open Access Neonatal Rhesus Macaques Have Distinct Immune Cell Transcriptional Profiles following HIV Envelope Immunization.(Cell reports, 2020-02) Han, Qifeng; Bradley, Todd; Williams, Wilton B; Cain, Derek W; Montefiori, David C; Saunders, Kevin O; Parks, Robert J; Edwards, Regina W; Ferrari, Guido; Mueller, Olaf; Shen, Xiaoying; Wiehe, Kevin J; Reed, Steven; Fox, Christopher B; Rountree, Wes; Vandergrift, Nathan A; Wang, Yunfei; Sutherland, Laura L; Santra, Sampa; Moody, M Anthony; Permar, Sallie R; Tomaras, Georgia D; Lewis, Mark G; Van Rompay, Koen KA; Haynes, Barton FHIV-1-infected infants develop broadly neutralizing antibodies (bnAbs) more rapidly than adults, suggesting differences in the neonatal versus adult responses to the HIV-1 envelope (Env). Here, trimeric forms of HIV-1 Env immunogens elicit increased gp120- and gp41-specific antibodies more rapidly in neonatal macaques than adult macaques. Transcriptome analyses of neonatal versus adult immune cells after Env vaccination reveal that neonatal macaques have higher levels of the apoptosis regulator BCL2 in T cells and lower levels of the immunosuppressive interleukin-10 (IL-10) receptor alpha (IL10RA) mRNA transcripts in T cells, B cells, natural killer (NK) cells, and monocytes. In addition, immunized neonatal macaques exhibit increased frequencies of activated blood T follicular helper-like (Tfh) cells compared to adults. Thus, neonatal macaques have transcriptome signatures of decreased immunosuppression and apoptosis compared with adult macaques, providing an immune landscape conducive to early-life immunization prior to sexual debut.Item Open Access Postnatal cytomegalovirus exposure in infants of antiretroviral-treated and untreated HIV-infected mothers.(Infect Dis Obstet Gynecol, 2014) Meyer, Sarah A; Westreich, Daniel J; Patel, Emily; Ehlinger, Elizabeth P; Kalilani, Linda; Lovingood, Rachel V; Denny, Thomas N; Swamy, Geeta K; Permar, Sallie RHIV-1 and CMV are important pathogens transmitted via breastfeeding. Furthermore, perinatal CMV transmission may impact growth and disease progression in HIV-exposed infants. Although maternal antiretroviral therapy reduces milk HIV-1 RNA load and postnatal transmission, its impact on milk CMV load is unclear. We examined the relationship between milk CMV and HIV-1 load (4-6 weeks postpartum) and the impact of antiretroviral treatment in 69 HIV-infected, lactating Malawian women and assessed the relationship between milk CMV load and postnatal growth in HIV-exposed, breastfed infants through six months of age. Despite an association between milk HIV-1 RNA and CMV DNA load (0.39 log(10) rise CMV load per log(10) rise HIV-1 RNA load, 95% CI 0.13-0.66), milk CMV load was similar in antiretroviral-treated and untreated women. Higher milk CMV load was associated with lower length-for-age (-0.53, 95% CI: -0.96, -0.10) and weight-for-age (-0.40, 95% CI: -0.67, -0.13) Z-score at six months in exposed, uninfected infants. As the impact of maternal antiretroviral therapy on the magnitude of postnatal CMV exposure may be limited, our findings of an inverse relationship between infant growth and milk CMV load highlight the importance of defining the role of perinatal CMV exposure on growth faltering of HIV-exposed infants.