Skip to main content

Can complement fix placental malaria?

The Original Article was published on 24 August 2021

The problem of placental malaria

Placental malaria (PM) is a deadly public health problem caused by the human parasite Plasmodium falciparum, and this scourge will get worse as existing control measures lose potency. Our understanding of PM pathogenesis suggests a vaccine is feasible, but first-generation candidates yielded only modest variant-specific activity in early trials. In this issue of BMC Medicine, Opi and colleagues provide evidence for a heretofore unrecognized mechanism of protective immunity, whereby the neutralizing activity of antibody against PM parasites is enhanced by fixing the complement component C1q [1].

Recent estimates [2] hold that up to 50,000 maternal deaths and 200,000 stillbirths result from PM in Africa annually, with additional mortality during infancy related to preterm and low birthweight deliveries. Despite public health advances including chemoprevention using intermittent sulfadoxine-pyrimethamine (SP) treatments, as well as insecticide-treated bednets, the terrible toll on mothers and their offspring continues. Deaths will increase as parasite resistance to SP intensifies and spreads.

The underlying cause of PM is well-established: P. falciparum-infected erythrocytes (IEs) adhere to receptors, mainly chondroitin sulfate A (CSA), and sequester in intervillous spaces (maternal blood) of the placenta, eliciting an inflammatory response associated with poor maternal and fetal outcomes. The parasite ligand for CSA, VAR2CSA, is a member of the P. falciparum erythrocyte membrane protein 1 (PfEMP1) family of variant surface antigens. Its large size and extensive sequence variation make VAR2CSA a challenging target for vaccine developers, and insights into protective antibodies and epitopes are needed to focus immunogen design.

Nature suggests the solution to the PM problem. Over successive pregnancies, pregnant women in areas of endemicity develop resistance and simultaneously acquire antibodies against placental IEs, including antibody that blocks IE binding to CSA [3] or mediates IE opsonization/phagocytosis [4]. These functional antibodies are believed to target VAR2CSA, and antibodies to VAR2CSA increase over successive pregnancies, but the association of VAR2CSA antibodies to improved clinical outcomes has been inconsistent [5].

A new role for complement in serum anti-adhesion activity

In their study, Opi and colleagues examine additional antibody mechanisms that might contribute to PM immunity. The immunoglobulins IgG1 and IgG3 dominate the naturally acquired response to placental IEs and to VAR2CSA, and both subclasses fix complement, which prompted the authors to examine antibody for complement-dependent functional activities [1]. Using samples from a longitudinal cohort of pregnant women in Papua New Guinea, the authors first demonstrated that serum antibodies fix complement on the IE surface of CSA-binding parasites. Levels of antibody-mediated complement fixation increased with gravidity and also predicted lower risk of PM at delivery among the subset of women who were infected at enrollment. This seroepidemiology suggests complement fixation might play a role in protective immunity.

The team examined possible mechanisms by which complement might neutralize parasites. They found that serum antibodies that bound recombinant VAR2CSA domains can fix the complement components C1q and C3. Using the CSA-binding laboratory isolate CS2 that maintains VAR2CSA on its IE surface, however, the group observed no evidence that complement fixation led to the formation of membrane attack complex or to inhibition of growth in vitro. These findings echo earlier studies with IEs from non-pregnant individuals, where the absence of complement activity was attributed to complement regulatory protein on the red cell surface [6, 7]. Instead, and surprisingly, fixation of C1q enhanced serum anti-adhesion activity against CSA-binding IEs.

Levels of complement on IE were reduced after CS2 was engineered to block PfEMP-1 surface expression, further implicating VAR2CSA as a target. Taken together, the data support the idea that VAR2CSA antibodies fix complement which enhances their anti-adhesion activity against CSA-binding IE and thereby might prevent placental sequestration. Of note, modest levels of complement were still detected on IEs without surface PfEMP1, and therefore, other surface antigens may also be targets of complement-fixing antibody.

The contribution of complement to protective immunity

In contrast to these new findings, a recent study of Ghanaian gravidae [8] observed complement fixation by serum antibody on recombinant VAR2CSA but not on the IE surface of CSA-binding IT4/FCR3 parasites (that are isogenic to the CS2 parasite studied here). Opi and colleagues confirmed C3 fixation on the IE surface of an additional maternal isolate from Papua New Guinea and also showed that serum antibody bound to recombinant VAR2CSA fragments from different alleles fixed C1q or C3. Nevertheless, studies are warranted in different geographical regions using diverse parasites to understand how host or parasite heterogeneity impacts complement-dependent functional activity, and different alleles of full-length recombinant VAR2CSA ectodomain [9] should be assessed as reagents to measure complement-fixing antibody.

Future studies will also benefit by enrolling women earlier in gestation, as Opi et al. suggest, since C1q-binding serum antibody did not predict protection in women without infection at enrollment, a majority of the study population. Capturing the many parasitemia events that appear early in second trimester might strengthen the associations beyond those seen in this study. Finally, larger studies or meta-analyses will be needed to discriminate the independent contributions made by anti-adhesion antibodies, complement-dependent anti-adhesion activity, opsonizing antibodies, and other immune measures, to protection from PM.

Conclusions

Opi and colleagues describe a role for complement to enhance protective anti-adhesion antibody activity during PM. Thus, VAR2CSA vaccine activity may similarly be enhanced by inducing antibodies that fix complement. The human response to the VAR2CSA vaccine PRIMVAC was dominated by IgG1 and IgG3, but nevertheless, serum anti-adhesion activity measured in vitro was modest against different parasite variants [10]. Given the findings of Opi et al., antisera from past and future trials should henceforth be examined with careful attention to complement in the assays. Vaccine design and formulation should also consider the potential contribution of complement-fixing antibodies to protective efficacy, alongside the contributions of opsonizing/phagocytosing and anti-adhesion antibodies. Ultimately, vaccine trials might provide the clearest evidence for the individual and combined benefits of these different immune mechanisms for preventing PM.

Availability of data and materials

Not applicable

References

  1. Opi H, Boyle M, McLean A, Reiling L, Chan J, Stanisic D, et al. Reduced risk of placental parasitaemia associated with complement fixation on Plasmodium falciparum by antibodies among pregnant women. BMC Med. 2021; Article in press.

  2. McGready R, Nosten F, Barnes KI, Mokuolu O, White NJ. Why is WHO failing women with falciparum malaria in the first trimester of pregnancy? Lancet. 2020;395(10226):779. https://0-doi-org.brum.beds.ac.uk/10.1016/S0140-6736(20)30161-6.

    Article  PubMed  Google Scholar 

  3. Fried M, Nosten F, Brockman A, Brabin BJ, Duffy PE. Maternal antibodies block malaria. Nature. 1998;395(6705):851–2. https://0-doi-org.brum.beds.ac.uk/10.1038/27570.

    Article  CAS  PubMed  Google Scholar 

  4. Keen J, Serghides L, Ayi K, Patel SN, Ayisi J, van Eijk A, et al. HIV impairs opsonic phagocytic clearance of pregnancy-associated malaria parasites. PLoS Med. 2007;4(5):e181. https://0-doi-org.brum.beds.ac.uk/10.1371/journal.pmed.0040181.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Cutts JC, Agius PA, Zaw L, Powell R, Moore K, Draper B, et al. Pregnancy-specific malarial immunity and risk of malaria in pregnancy and adverse birth outcomes: a systematic review. BMC Med. 2020;18(1):14. https://0-doi-org.brum.beds.ac.uk/10.1186/s12916-019-1467-6.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Stanley HA, Mayes JT, Cooper NR, Reese RT. Complement activation by the surface of Plasmodium falciparum infected erythrocytes. Mol Immunol. 1984;21(2):145–50. https://0-doi-org.brum.beds.ac.uk/10.1016/0161-5890(84)90129-9.

    Article  CAS  PubMed  Google Scholar 

  7. Wiesner J, Jomaa H, Wilhelm M, Tony HP, Kremsner PG, Horrocks P, et al. Host cell factor CD59 restricts complement lysis of Plasmodium falciparum-infected erythrocytes. Eur J Immunol. 1997;27(10):2708–13. https://0-doi-org.brum.beds.ac.uk/10.1002/eji.1830271034.

    Article  CAS  PubMed  Google Scholar 

  8. Larsen MD, Quintana MDP, Ditlev SB, Bayarri-Olmos R, Ofori MF, Hviid L, et al. Evasion of classical complement pathway activation on Plasmodium falciparum-infected erythrocytes opsonized by PfEMP1-specific IgG. Front Immunol. 2018;9:3088.

    Article  CAS  Google Scholar 

  9. Renn JP, Doritchamou JYA, Duffy PE. Expression of large full-length PfEMP1 proteins in HEK293 cells. Methods Mol Biol. 2021; Article in press.

  10. Sirima SB, Richert L, Chene A, Konate AT, Campion C, Dechavanne S, et al. PRIMVAC vaccine adjuvanted with Alhydrogel or GLA-SE to prevent placental malaria: a first-in-human, randomised, double-blind, placebo-controlled study. Lancet Infect Dis. 2020;20(5):585–97. https://0-doi-org.brum.beds.ac.uk/10.1016/S1473-3099(19)30739-X.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank J. Patrick Gorres for editing the manuscript.

Funding

Authors JYAD and PED are supported by the Intramural Research Program of the National Institute of Allergy and Infectious Diseases, National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

All authors read and approved the final manuscript

Corresponding author

Correspondence to Patrick E. Duffy.

Ethics declarations

Ethics approval and consent to participate

Not applicable

Competing interests

The authors declare no conflicts of interest are associated with this manuscript.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Doritchamou, J.Y.A., Duffy, P.E. Can complement fix placental malaria?. BMC Med 19, 231 (2021). https://0-doi-org.brum.beds.ac.uk/10.1186/s12916-021-02083-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12916-021-02083-5

Keywords