Resisting and tolerating P. falciparum in pregnancy under different malaria transmission intensities

Nicaise Tuikue Ndam, Emmanuel Mbuba, Raquel González, Pau Cisteró, Simon Kariuki, Esperança Sevene, María Rupérez, Ana Maria Fonseca, Anifa Vala, Sonia Maculuve, Alfons Jiménez, Llorenç Quintó, Peter Ouma, Michael Ramharter, John J Aponte, Arsenio Nhacolo, Achille Massougbodji, Valerie Briand, Peter G Kremsner, Ghyslain Mombo-Ngoma, Meghna Desai, Eusebio Macete, Michel Cot, Clara Menéndez, Alfredo Mayor, Nicaise Tuikue Ndam, Emmanuel Mbuba, Raquel González, Pau Cisteró, Simon Kariuki, Esperança Sevene, María Rupérez, Ana Maria Fonseca, Anifa Vala, Sonia Maculuve, Alfons Jiménez, Llorenç Quintó, Peter Ouma, Michael Ramharter, John J Aponte, Arsenio Nhacolo, Achille Massougbodji, Valerie Briand, Peter G Kremsner, Ghyslain Mombo-Ngoma, Meghna Desai, Eusebio Macete, Michel Cot, Clara Menéndez, Alfredo Mayor

Abstract

Background: Resistance and tolerance to Plasmodium falciparum can determine the progression of malaria disease. However, quantitative evidence of tolerance is still limited. We investigated variations in the adverse impact of P. falciparum infections among African pregnant women under different intensities of malaria transmission.

Methods: P. falciparum at delivery was assessed by microscopy, quantitative PCR (qPCR) and placental histology in 946 HIV-uninfected and 768 HIV-infected pregnant women from Benin, Gabon, Kenya and Mozambique. Resistance was defined by the proportion of submicroscopic infections and the levels of anti-parasite antibodies quantified by Luminex, and tolerance by the relationship of pregnancy outcomes with parasite densities at delivery.

Results: P. falciparum prevalence by qPCR in peripheral and/or placental blood of HIV-uninfected Mozambican, Gabonese and Beninese women at delivery was 6% (21/340), 11% (28/257) and 41% (143/349), respectively. The proportion of peripheral submicroscopic infections was higher in Benin (83%) than in Mozambique (60%) and Gabon (55%; P = 0.033). Past or chronic placental P. falciparum infection was associated with an increased risk of preterm birth in Mozambican newborns (OR = 7.05, 95% CI 1.79 to 27.82). Microscopic infections were associated with reductions in haemoglobin levels at delivery among Mozambican women (-1.17 g/dL, 95% CI -2.09 to -0.24) as well as with larger drops in haemoglobin levels from recruitment to delivery in Mozambican (-1.66 g/dL, 95% CI -2.68 to -0.64) and Gabonese (-0.91 g/dL, 95% CI -1.79 to -0.02) women. Doubling qPCR-peripheral parasite densities in Mozambican women were associated with decreases in haemoglobin levels at delivery (-0.16 g/dL, 95% CI -0.29 to -0.02) and increases in the drop of haemoglobin levels (-0.29 g/dL, 95% CI -0.44 to -0.14). Beninese women had higher anti-parasite IgGs than Mozambican women (P < 0.001). No difference was found in the proportion of submicroscopic infections nor in the adverse impact of P. falciparum infections in HIV-infected women from Kenya (P. falciparum prevalence by qPCR: 9%, 32/351) and Mozambique (4%, 15/417).

Conclusions: The lowest levels of resistance and tolerance in pregnant women from areas of low malaria transmission were accompanied by the largest adverse impact of P. falciparum infections. Exposure-dependent mechanisms developed by pregnant women to resist the infection and minimise pathology can reduce malaria-related adverse outcomes. Distinguishing both types of defences is important to understand how reductions in transmission can affect malaria disease.

Trial registration: ClinicalTrials.gov NCT00811421 . Registered 18 December 2008.

Keywords: Immunity; Malaria; Pregnancy; Resistance; Tolerance.

Figures

Fig. 1
Fig. 1
Prevalence of P. falciparum maternal infection according to study area. Prevalence of P. falciparum at delivery in peripheral and/or placental blood as detected by qPCR (a) and microscopy (b). P value according to the multivariate analysis adjusted for type of IPTp drug, season, age, gravidity, gestational age, anaemia, literacy, RPR result and MUAC at recruitment, as well as CD4 + T cell count in the case of HIV-infected women. T bars represent standard errors. B Benin, G Gabon, M Mozambique, K Kenya
Fig. 2
Fig. 2
Impact of microscopic malaria infections at delivery on pregnancy outcomes. Maternal microscopic infections were considered present if P. falciparum parasites were observed in peripheral blood at delivery and/or in the placenta either by microscopy or histology. The dot and T bar represents the mean difference and 95% confidence interval in haemoglobin levels at delivery (a, b), the difference of haemoglobin levels from recruitment to delivery (c, d), or birth weight (e, f) between malaria infected and uninfected women in the multivariate regression analysis adjusted for type of IPTp drug, season, age, gravidity, gestational age, anaemia, literacy, RPR result and MUAC at recruitment, plus CD4 + T cell count at recruitment in the case of HIV-infected women (b, d, f). Modification of the associations by study area (B Benin, G Gabon, K Kenya, M Mozambique) was determined through the inclusion of an interaction term in the regression models, and combination of the coefficients plus the interaction and the standard error was estimated by the delta method
Fig. 3
Fig. 3
Proportion of P. falciparum submicroscopic infections, PfHRP2 concentrations and antibody responses against P. falciparum antigens according to study area. Proportion of infections that are submicroscopic in peripheral (a) and placental (b) blood. P value according to the multivariate analysis adjusted for type of IPTp drug, season, age, gravidity, gestational age, anaemia, literacy, RPR result and MUAC at recruitment, as well as CD4 + T cell count in the case of HIV-infected women. T bars represent standard errors. B Benin, G Gabon, M Mozambique, K Kenya. c PfHRP2 concentrations, indicative of overall parasite biomass, among women with a peripheral qPCR-detected infection in Mozambique (M; n = 13) and Benin (B; n = 42). d Levels of IgG antibodies against MSP1 and DBL3X in Beninese (n = 170) and Mozambican (n = 170) women. e Levels of IgG antibodies against MSP1 and DBL3X by parity (primigravidae (PG) versus multigravidae (MG)), with the proportional increase (Δ) of the antibody levels plus 95% confidence interval in multigravidae versus primigravidae women. P values obtained from regression analysis adjusted for type of IPTp drug, season, gravidity, gestational age, anaemia, literacy, RPR result and MUAC at recruitment, as well as CD4 + T cell count in the case of HIV-infected women. MFI mean fluorescence intensity
Fig. 4
Fig. 4
Relationship of qPCR-detected parasite densities in peripheral blood with pregnancy outcomes. The dot and T bar represent the effect and 95% confidence interval on haemoglobin levels at delivery (a, b), the difference of haemoglobin levels from recruitment to delivery (c, d), or birth weight (e, f) due to a two-fold increase in the parasitemia levels in its natural scale. Modification of the associations by study area (B Benin, G Gabon, K Kenya, M Mozambique) was determined through the inclusion of an interaction term in the regression models, and combination of the coefficients plus the interaction and the standard error was estimated by the delta method. Analyses were adjusted for type of IPTp drug, season, age, gravidity, gestational age, anaemia, literacy, RPR result and MUAC at recruitment, as well as CD4 + T cell count in the case of HIV-infected women (b, d, f)

References

    1. Mayor A, Bardaji A, Macete E, Nhampossa T, Fonseca AM, Gonzalez R, Maculuve S, Cistero P, Ruperez M, Campo J, et al. Changing trends in P. falciparum burden, immunity, and disease in pregnancy. N Engl J Med. 2015;373(17):1607–17. doi: 10.1056/NEJMoa1406459.
    1. Nosten F, Rogerson SJ, Beeson JG, McGready R, Mutabingwa TK, Brabin B. Malaria in pregnancy and the endemicity spectrum: what can we learn? Trends Parasitol. 2004;20(9):425–32. doi: 10.1016/j.pt.2004.06.007.
    1. Ataide R, Mayor A, Rogerson SJ. Malaria, primigravidae, and antibodies: knowledge gained and future perspectives. Trends Parasitol. 2014;30(2):85–94. doi: 10.1016/j.pt.2013.12.007.
    1. Mockenhaupt FP, Rong B, Till H, Eggelte TA, Beck S, Gyasi-Sarpong C, Thompson WN, Bienzle U. Submicroscopic Plasmodium falciparum infections in pregnancy in Ghana. Trop Med Int Health. 2000;5(3):167–73. doi: 10.1046/j.1365-3156.2000.00532.x.
    1. Walker-Abbey A, Djokam RR, Eno A, Leke RF, Titanji VP, Fogako J, Sama G, Thuita LH, Beardslee E, Snounou G, et al. Malaria in pregnant Cameroonian women: the effect of age and gravidity on submicroscopic and mixed-species infections and multiple parasite genotypes. Am J Trop Med Hyg. 2005;72(3):229–35.
    1. Cottrell G, Moussiliou A, Luty AJ, Cot M, Fievet N, Massougbodji A, Deloron P, Tuikue NN. Submicroscopic Plasmodium falciparum infections are associated with maternal anemia, premature births and low birthweight. Clin Infect Dis. 2015;60(10):1481–8. doi: 10.1093/cid/civ122.
    1. Medzhitov R, Schneider DS, Soares MP. Disease tolerance as a defense strategy. Science. 2012;335(6071):936–41. doi: 10.1126/science.1214935.
    1. Raberg L, Sim D, Read AF. Disentangling genetic variation for resistance and tolerance to infectious diseases in animals. Science. 2007;318(5851):812–4. doi: 10.1126/science.1148526.
    1. Zumla A, Rao M, Wallis RS, Kaufmann SH, Rustomjee R, Mwaba P, Vilaplana C, Yeboah-Manu D, Chakaya J, Ippolito G, et al. Host-directed therapies for infectious diseases: current status, recent progress, and future prospects. Lancet Infect Dis. 2016;16(4):e47–63. doi: 10.1016/S1473-3099(16)00078-5.
    1. Raberg L. How to live with the enemy: understanding tolerance to parasites. PLoS Biol. 2014;12(11) doi: 10.1371/journal.pbio.1001989.
    1. Galatas B, Bassat Q, Mayor A. Malaria parasites in the asymptomatic: looking for the hay in the haystack. Trends Parasitol. 2016;32(4):296–308. doi: 10.1016/j.pt.2015.11.015.
    1. Collins WE, Jeffery GM. A retrospective examination of sporozoite- and trophozoite-induced infections with Plasmodium falciparum: development of parasitologic and clinical immunity during primary infection. Am J Trop Med Hyg. 1999;61(1 Suppl):4–19. doi: 10.4269/tropmed.1999.61-04.
    1. Gupta S, Snow RW, Donnelly CA, Marsh K, Newbold C. Immunity to non-cerebral severe malaria is acquired after one or two infections. Nat Med. 1999;5(3):340–3. doi: 10.1038/6560.
    1. Rogier C, Commenges D, Trape JF. Evidence for an age-dependent pyrogenic threshold of Plasmodium falciparum parasitemia in highly endemic populations. Am J Trop Med Hyg. 1996;54(6):613–9. doi: 10.4269/ajtmh.1996.54.613.
    1. Smith T, Killeen G, Lengeler C, Tanner M. Relationships between the outcome of Plasmodium falciparum infection and the intensity of transmission in Africa. Am J Trop Med Hyg. 2004;71(2 Suppl):80–6.
    1. Petersen E, Hogh B, Marbiah NT, David K, Hanson AP. Development of immunity against Plasmodium falciparum malaria: clinical and parasitologic immunity cannot be separated. J Infect Dis. 1991;164(5):949–53. doi: 10.1093/infdis/164.5.949.
    1. Bardaji A, Sigauque B, Romagosa C, Bruni L, Sanz S, Mabunda S, Mandomando I, Aponte J, Sevene E, Alonso PL, et al. Clinical malaria in African pregnant women. Malar J. 2008;7(1):27. doi: 10.1186/1475-2875-7-27.
    1. O’Neil-Dunne I, Achur RN, Agbor-Enoh ST, Valiyaveettil M, Naik RS, Ockenhouse CF, Zhou A, Megnekou R, Leke R, Taylor DW, et al. Gravidity-dependent production of antibodies that inhibit binding of Plasmodium falciparum-infected erythrocytes to placental chondroitin sulfate proteoglycan during pregnancy. Infect Immun. 2001;69(12):7487–92. doi: 10.1128/IAI.69.12.7487-7492.2001.
    1. Gonzalez R, Mombo-Ngoma G, Ouedraogo S, Kakolwa MA, Abdulla S, Accrombessi M, Aponte JJ, Akerey-Diop D, Basra A, Briand V, et al. Intermittent preventive treatment of malaria in pregnancy with mefloquine in HIV-negative women: a multicentre randomized controlled trial. PLoS Med. 2014;11(9) doi: 10.1371/journal.pmed.1001733.
    1. Gonzalez R, Desai M, Macete E, Ouma P, Kakolwa MA, Abdulla S, Aponte JJ, Bulo H, Kabanywanyi AM, Katana A, et al. Intermittent preventive treatment of malaria in pregnancy with mefloquine in HIV-infected women receiving cotrimoxazole prophylaxis: a multicenter randomized placebo-controlled trial. PLoS Med. 2014;11(9) doi: 10.1371/journal.pmed.1001735.
    1. Ballard JL, Khoury JC, Wedig K, Wang L, Eilers-Walsman BL, Lipp R. New Ballard Score, expanded to include extremely premature infants. J Pediatr. 1991;119(3):417–23. doi: 10.1016/S0022-3476(05)82056-6.
    1. Greenwood AM, Armstrong JR, Byass P, Snow RW, Greenwood BM. Malaria chemoprophylaxis, birth weight and child survival. Trans R Soc Trop Med Hyg. 1992;86(5):483–5. doi: 10.1016/0035-9203(92)90078-Q.
    1. Ordi J, Ismail MR, Ventura PJ, Kahigwa E, Hirt R, Cardesa A, Alonso PL, Menendez C. Massive chronic intervillositis of the placenta associated with malaria infection. Am J Surg Pathol. 1998;22(8):1006–11. doi: 10.1097/00000478-199808000-00011.
    1. Joanny F, Lohr SJ, Engleitner T, Lell B, Mordmuller B. Limit of blank and limit of detection of Plasmodium falciparum thick blood smear microscopy in a routine setting in Central Africa. Malar J. 2014;13:234. doi: 10.1186/1475-2875-13-234.
    1. Taylor SM, Mayor A, Mombo-Ngoma G, Kenguele HM, Ouedraogo S, Ndam NT, Mkali H, Mwangoka G, Valecha N, Singh JP, et al. A quality control program within a clinical trial Consortium for PCR protocols to detect Plasmodium species. J Clin Microbiol. 2014;52(6):2144–9. doi: 10.1128/JCM.00565-14.
    1. Dondorp AM, Desakorn V, Pongtavornpinyo W, Sahassananda D, Silamut K, Chotivanich K, Newton PN, Pitisuttithum P, Smithyman AM, White NJ, et al. Estimation of the total parasite biomass in acute falciparum malaria from plasma PfHRP2. PLoS Med. 2005;2(8) doi: 10.1371/journal.pmed.0020204.
    1. Mayor A, Moro L, Aguilar R, Bardaji A, Cistero P, Serra-Casas E, Sigauque B, Alonso PL, Ordi J, Menendez C. How hidden can malaria be in pregnant women? Diagnosis by microscopy, placental histology, polymerase chain reaction and detection of histidine-rich protein 2 in plasma. Clin Infect Dis. 2012;54(11):1561–8. doi: 10.1093/cid/cis236.
    1. Mayor A, Serra-Casas E, Bardaji A, Sanz S, Puyol L, Cistero P, Sigauque B, Mandomando I, Aponte JJ, Alonso PL, et al. Sub-microscopic infections and long-term recrudescence of Plasmodium falciparum in Mozambican pregnant women. Malar J. 2009;8:9. doi: 10.1186/1475-2875-8-9.
    1. Sinton JA. Immunity or tolerance in malarial infections: (section of comparative medicine) Proc R Soc Med. 1938;31(11):1298–302.
    1. Okell LC, Ghani AC, Lyons E, Drakeley CJ. Submicroscopic infection in Plasmodium falciparum-endemic populations: a systematic review and meta-analysis. J Infect Dis. 2009;200(10):1509–17. doi: 10.1086/644781.
    1. Chico RM, Cano J, Ariti C, Collier TJ, Chandramohan D, Roper C, Greenwood B. Influence of malaria transmission intensity and the 581G mutation on the efficacy of intermittent preventive treatment in pregnancy: systematic review and meta-analysis. Trop Med Int Health. 2015;20(12):1621–33. doi: 10.1111/tmi.12595.
    1. Chico RM, Chaponda EB, Ariti C, Chandramohan D. Sulfadoxine-pyrimethamine exhibits dose-response protection against adverse birth outcomes related to malaria and sexually transmitted and reproductive tract infections. Clin Infect Dis. 2017;64(8):1043–51. doi: 10.1093/cid/cix026.
    1. Desai M, Gutman J, Taylor SM, Wiegand RE, Khairallah C, Kayentao K, Ouma P, Coulibaly SO, Kalilani L, Mace KE, et al. Impact of sulfadoxine-pyrimethamine resistance on effectiveness of intermittent preventive therapy for malaria in pregnancy at clearing infections and preventing low birth weight. Clin Infect Dis. 2016;62(3):323–33. doi: 10.1093/cid/civ881.
    1. Naidoo I, Roper C. Drug resistance maps to guide intermittent preventive treatment of malaria in African infants. Parasitology. 2011;138(12):1469–79. doi: 10.1017/S0031182011000746.
    1. Menendez C, Serra-Casas E, Scahill MD, Sanz S, Nhabomba A, Bardaji A, Sigauque B, Cistero P, Mandomando I, Dobano C, et al. HIV and placental infection modulate the appearance of drug-resistant Plasmodium falciparum in pregnant women who receive intermittent preventive treatment. Clin Infect Dis. 2011;52(1):41–8. doi: 10.1093/cid/ciq049.
    1. Mombo-Ngoma G, Oyakhirome S, Ord R, Gabor JJ, Greutelaers KC, Profanter K, Greutelaers B, Kurth F, Lell B, Kun JF, et al. High prevalence of dhfr triple mutant and correlation with high rates of sulphadoxine-pyrimethamine treatment failures in vivo in Gabonese children. Malar J. 2011;10:123. doi: 10.1186/1475-2875-10-123.
    1. Ogouyemi-Hounto A, Ndam NT, Fadegnon G, Azagnandji C, Bello M, Moussiliou A, Chippaux JP, Kinde Gazard D, Massougbodji A. Low prevalence of the molecular markers of Plasmodium falciparum resistance to chloroquine and sulphadoxine/pyrimethamine in asymptomatic children in Northern Benin. Malar J. 2013;12:413. doi: 10.1186/1475-2875-12-413.
    1. Artavanis-Tsakonas K, Tongren JE, Riley EM. The war between the malaria parasite and the immune system: immunity, immunoregulation and immunopathology. Clin Exp Immunol. 2003;133(2):145–52. doi: 10.1046/j.1365-2249.2003.02174.x.
    1. Portugal S, Moebius J, Skinner J, Doumbo S, Doumtabe D, Kone Y, Dia S, Kanakabandi K, Sturdevant DE, Virtaneva K, et al. Exposure-dependent control of malaria-induced inflammation in children. PLoS Pathog. 2014;10(4) doi: 10.1371/journal.ppat.1004079.
    1. Jagannathan P, Eccles-James I, Bowen K, Nankya F, Auma A, Wamala S, Ebusu C, Muhindo MK, Arinaitwe E, Briggs J, et al. IFNgamma/IL-10 co-producing cells dominate the CD4 response to malaria in highly exposed children. PLoS Pathog. 2014;10(1) doi: 10.1371/journal.ppat.1003864.
    1. Tran TM, Jones MB, Ongoiba A, Bijker EM, Schats R, Venepally P, Skinner J, Doumbo S, Quinten E, Visser LG, et al. Transcriptomic evidence for modulation of host inflammatory responses during febrile Plasmodium falciparum malaria. Sci Rep. 2016;6:31291. doi: 10.1038/srep31291.
    1. Cottrell G, Mary JY, Barro D, Cot M. The importance of the period of malarial infection during pregnancy on birth weight in tropical Africa. Am J Trop Med Hyg. 2007;76(5):849–54.
    1. Huynh BT, Fievet N, Gbaguidi G, Dechavanne S, Borgella S, Guezo-Mevo B, Massougbodji A, Ndam NT, Deloron P, Cot M. Influence of the timing of malaria infection during pregnancy on birth weight and on maternal anemia in Benin. Am J Trop Med Hyg. 2011;85(2):214–20. doi: 10.4269/ajtmh.2011.11-0103.
    1. Valea I, Tinto H, Drabo MK, Huybregts L, Sorgho H, Ouedraogo JB, Guiguemde RT, van Geertruyden JP, Kolsteren P, D’Alessandro U, et al. An analysis of timing and frequency of malaria infection during pregnancy in relation to the risk of low birth weight, anaemia and perinatal mortality in Burkina Faso. Malar J. 2012;11:71. doi: 10.1186/1475-2875-11-71.
    1. Gonzalez R, Ataide R, Naniche D, Menendez C, Mayor A. HIV and malaria interactions: where do we stand? Expert Rev Anti Infect Ther. 2012;10(2):153–65. doi: 10.1586/eri.11.167.
    1. Brabin BJ, Romagosa C, Abdelgalil S, Menendez C, Verhoeff FH, McGready R, Fletcher KA, Owens S, D’Alessandro U, Nosten F, et al. The sick placenta-the role of malaria. Placenta. 2004;25(5):359–78. doi: 10.1016/j.placenta.2003.10.019.
    1. Liu M, Hassana S, Stiles JK. Heme-mediated apoptosis and fusion damage in BeWo trophoblast cells. Sci Rep. 2016;6:36193. doi: 10.1038/srep36193.
    1. Higgins SJ, Purcell LA, Silver KL, Tran V, Crowley V, Hawkes M, Conroy AL, Opoka RO, Hay JG, Quaggin SE, et al. Dysregulation of angiopoietin-1 plays a mechanistic role in the pathogenesis of cerebral malaria. Sci Transl Med. 2016;8(358):358ra128. doi: 10.1126/scitranslmed.aaf6812.
    1. Higgins SJ, Kain KC, Liles WC. Immunopathogenesis of falciparum malaria: implications for adjunctive therapy in the management of severe and cerebral malaria. Expert Rev Anti Infect Ther. 2011;9(9):803–19. doi: 10.1586/eri.11.96.

Source: PubMed

3
Iratkozz fel