Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2020 Aug 1;35(8):1781-1796.
doi: 10.1093/humrep/deaa151.

Periconceptional exposure to lopinavir, but not darunavir, impairs decidualization: a potential mechanism leading to poor birth outcomes in HIV-positive pregnancies

Affiliations

Periconceptional exposure to lopinavir, but not darunavir, impairs decidualization: a potential mechanism leading to poor birth outcomes in HIV-positive pregnancies

Smriti Kala et al. Hum Reprod. .

Abstract

Study question: Does HIV protease inhibitor (PI)-based combination antiretroviral therapy (cART) initiated at periconception affect key events in early pregnancy, i.e. decidualization and spiral artery remodeling?

Summary answer: Two PIs, lopinavir and darunavir, currently offered as cART options in HIV-positive pregnancies were evaluated, and we found that lopinavir-based cART, but not darunavir-based cART, impaired uterine decidualization and spiral artery remodeling in both human ex vivo and mouse in vivo experimental models.

What is known already: Early initiation of cART is recommended for pregnant women living with HIV. However, poor birth outcomes are frequently observed in HIV-positive pregnancies exposed to PI-based cART, especially when it is initiated prior to conception. The correlation between early initiation of PI-cART and adverse birth outcomes is poorly understood, due to lack of data on the specific effects of PI-cART on the early stages of pregnancy involving uterine decidualization and spiral artery remodeling.

Study design, size, duration: Lopinavir and darunavir were evaluated in clinically relevant combinations using an ex vivo human first-trimester placenta-decidua explant model, an in vitro human primary decidual cell culture system, and an in vivo mouse pregnancy model. The first-trimester (gestational age, 6-8 weeks) human placenta-decidua tissue was obtained from 11 to 15 healthy women undergoing elective termination of pregnancy. C57Bl/6 female mice (four/treatment group) were administered either lopinavir-cART, darunavir-cART or water by oral gavage once daily starting on the day of plug detection until sacrifice.

Participants/materials, setting, methods: Human: Spiral artery remodeling was assessed by immunohistochemical analysis of first-trimester placenta-decidua explant co-culture system. Trophoblast migration was measured using a placental explant culture. A primary decidual cell culture was used to evaluate the viability of immune cell populations by flow cytometry. Soluble factors, including biomarkers of decidualization and angiogenesis, were quantified by ELISA and Luminex assay using decidua-conditioned media. Mouse: In the mouse pregnancy model, gestational day 6.5 or 9.5 implantation sites were used to assess decidualization, spiral artery remodeling and uterine natural killer (uNK) cell numbers by immunohistochemistry. Transcription factor STAT3 was assayed by immunohistochemistry in both human decidua and mouse implantation sites.

Main results and the role of chance: Lopinavir-cART, but not darunavir-cART, impaired uterine decidualization and spiral artery remodeling in both experimental models. Lopinavir-cART treatment was also associated with selective depletion of uNK cells, reduced trophoblast migration and defective placentation. The lopinavir-associated decidualization defects were attributed to a decrease in expression of transcription factor STAT3, known to regulate decidualization. Our results suggest that periconceptional initiation of lopinavir-cART, but not darunavir-cART, causes defective maturation of the uterine endometrium, leading to impairments in spiral artery remodeling and placentation, thus contributing to the poor birth outcomes.

Large scale data: N/A.

Limitations, reasons for caution: The human first-trimester placenta/decidua samples could only be obtained from healthy females undergoing elective termination of pregnancy. As biopsy is the only way to obtain first-trimester decidua from pregnant women living with HIV on PI-cART, ethics approval and participant consent are difficult to obtain. Furthermore, our animal model is limited to the study of cART and does not include HIV. HIV infection is also associated with immune dysregulation, inflammation, alterations in angiogenic factors and complement activation, all of which could influence decidual and placental vascular remodeling and modify any cART effects.

Wider implications of the findings: Our findings provide mechanistic insight with direct clinical implications, rationalizing why the highest adverse birth outcomes are reported in HIV-positive pregnancies exposed to lopinavir-cART from conception. We demonstrate that dysregulation of decidualization is the mechanism through which lopinavir-cART, but not darunavir-cART, use in early pregnancy leads to poor birth outcomes. Although lopinavir is no longer a first-line regimen in pregnancy, it remains an alternate regimen and is often the only PI available in low resource settings. Our results highlight the need for reconsidering current guidelines recommending lopinavir use in pregnancy and indicate that lopinavir should be avoided especially in the first trimester, whereas darunavir is safe to use and should be the preferred PI in pregnancy.Further, in current times of the COVID-19 pandemic, lopinavir is among the top drug candidates which are being repurposed for inclusion in clinical trials world-over, to assess their therapeutic potential against the dangerous respiratory disease. Current trials are also testing the efficacy of lopinavir given prophylactically to protect health care workers and people with potential exposures. Given the current extraordinary numbers, these might include women with early pregnancies, who may or may not be cognizant of their gestational status. This is a matter of concern as it could mean that women with early pregnancies might be exposed to this drug, which can cause decidualization defects. Our findings provide evidence of safety concerns surrounding lopinavir use in pregnancy, that women of reproductive age considering participation in such trials should be made aware of, so they can make a fully informed decision.

Study funding/competing interest(s): This work was supported by funding from the Canadian Institutes of Health Research (CIHR) (PJT-148684 and MOP-130398 to L.S.). C.D. received support from CIHR Foundation (FDN143262 to Stephen Lye). S.K. received a TGHRI postdoctoral fellowship. The authors declare that there are no conflicts of interest. L.S. reports personal fees from ViiV Healthcare for participation in a Women and Transgender Think Tank.

Keywords: COVID-19; HIV protease inhibitors; birth outcomes; darunavir; decidualization; drug safety; lopinavir; spiral artery remodeling; trophoblasts; uterine natural killer cells.

PubMed Disclaimer

Figures

Figure 1.
Figure 1.
Lopinavir treatment impairs spiral artery remodeling in a human placenta-decidua explant co-culture system. Immunohistochemical assessment of remodeling vessels in serial cross-sections of placental-decidual co-cultures, treated with protease inhibitors (PIs) or DMSO control as indicated. Representative images of immunostaining with cytokeratin-7 (CK-7; marker for trophoblasts), CD31 and smooth muscle actin (SMA) (markers for spiral arteries), and CD45 (marker for leukocytes) are shown. (A–D) Complete vascular transformation in co-cultures treated with DMSO (control) and (I–L) ritonavir-boosted darunavir (DRV/r) as indicated. (A, I) Placenta tissue (#) is in close contact with the decidua, and CK-7 positive extravillous trophoblasts (EVTs; *) were released from the placenta. Endovascular EVTs (EnEVTs) were present within the lumen of spiral arteries (arrows). (B, J) The invaded arteries displayed loss of CD31 positive endothelial cells and (C, K) complete removal of SMA positive cells lining the vessel walls. (D, L) CD45 staining showed leukocyte enrichment at the site of vascular remodeling. (E–H) Impairment of vascular remodeling in co-culture treated with ritonavir-boosted lopinavir (LPV/r). (E) The placenta (#) is in close contact with the decidua and CK-7 positive EVTs (*) were released, but there was a complete lack of EnEVT invasion of spiral arteries (arrows). (F) Non-invaded arteries retained CD31 positive endothelium and (G) intact rings of SMA. (H) CD45 positive leukocytes were evenly distributed, but sparsely surrounded the vessels. (M–P) Decidua parietalis cultured in the absence of placenta. (M) The tissue was negative for CK-7. (N) Non-invaded arteries stained robustly for CD31 and (O) SMA. (P) CD45 positive leukocytes were spread evenly throughout the tissue. Inset is negative control: non-immune mouse IgG. Scale bars, 100 μm. (Q) Quantification of the depth of remodeling in co-cultures. Results are the mean ± SD. *** indicates P < 0.0001, calculated using ANOVA with Tukey’s post hoc analysis. Each data point represents the average of two to three measurements per experiment; n = 5 independent experiments.
Figure 2.
Figure 2.
Lopinavir treatment inhibits trophoblast migration in a human placental explant culture. (A) Explants from first-trimester placentas treated with DMSO (control) or protease inhibitors (PIs) as indicated, and cultured in the presence of decidua-conditioned medium (DCM) obtained from primary decidual cell culture also treated with the same drugs (or DMSO). Representative images are shown. Scale bars, 1 cm. (B) Extravillous trophoblast (EVT) outgrowth was measured at the termination of the experiment (66 h). Results are the mean ± SD. *** indicates P < 0.0001, calculated using ANOVA with Tukey’s post hoc analysis. Each data point represents the average of three replicates per experiment; n = 7 independent experiments. cART, combination antiretroviral therapy; DRV/r, ritonavir-boosted darunavir; LPV/r, ritonavir-boosted lopinavir.
Figure 3.
Figure 3.
Lopinavir treatment leads to depletion of uterine NK cells in a primary culture of human decidual cells. (A) Quantification of flow cytometry data showing total live cells and leukocyte subsets from the primary human decidual cell cultures treated with DMSO (control) or protease inhibitors (PIs) as indicated. Results are the mean ± SD. *** indicates P-value < 0.0001, calculated using ANOVA with Tukey’s post hoc analysis. For T cells, macrophages and stroma cells, P-value was not significant between the treatment groups. Each data point represents the average of two replicates per experiment; n = 11 independent experiments. (B) Representative images of CD56 immunostaining of uterine NK (uNK) cells in first-trimester decidua treated with DMSO (control) or PIs as indicated. In the ritonavir-boosted lopinavir (LPV/r) treatment group, uNK cells display a condensed appearance (arrows), characteristic of apoptotic cells. Scale bar, 20 µm. (C) Stereological quantification of CD56 positive uNK cells. Results are the mean ± SD. *** indicates P < 0.0001, calculated using ANOVA with Tukey’s post hoc analysis. Each data point represents the average of two to three replicates per experiment; n = 6 independent experiments. cART, combination antiretroviral therapy; DRV/r, ritonavir-boosted darunavir.
Figure 4.
Figure 4.
Lopinavir treatment causes decidualization defects and impairs spiral artery remodeling in a mouse pregnancy model. (A) Representative images of Ki67 immunostaining of proliferative cells in midsaggital sections from GD 6.5 implantation sites of control, ritonavir-boosted lopinavir (LPV/r)-combination antiretroviral therapy (cART) and ritonavir-boosted darunavir (DRV/r)-cART-treated mice. E, embryo; PDZ, primary decidual zone; SDZ, secondary decidual zone; M, mesometrium; AM, anti-mesometrium. * indicates uterine lumen. Scale bar, 600 µm. (B) Quantification of staining intensity (pixels) of Ki67 positive cells. Results are the mean ± SD. *** indicates P < 0.0001, calculated using ANOVA with Tukey’s post hoc analysis. Each data point represents an implantation site, three implantation sites were analyzed per mouse (n = 4 mice/treatment group). (C) Representative images of H&E-stained midsaggital sections from GD9.5 implantation sites of control, LPV/r-cART and DRV/r-cART-treated mice. Dec, decidua; Emb, embryo; Pla, placenta. Lower panels show a magnified view of the decidua basalis to highlight the spiral arteries (indicated by arrows). Scale bar, 1 mm (upper panels), 200 µm (lower panels). (D) Quantification of lumen areas of spiral arteries. Results are the mean ± SD. *** indicates P < 0.0001, calculated using ANOVA with Tukey’s post hoc analysis. Each data point represents an implantation site, and is the average of 15 measurements (5 measurements per section, 3 sections per implantation site); 2–3 implantation sites were analyzed per mouse (n = 4 mice/treatment group). (E) Representative images of smooth muscle actin (SMA) immunostaining of the spiral arteries (indicated by arrows) in decidua basalis region of GD9.5 implantation sites of control, LPV/r-cART and DRV/r-cART-treated mice. Scale bar, 200 µm. (F) Quantification of staining intensity (pixels) of SMA positive cells. Results are the mean ± SD. *** indicates P < 0.0001, calculated using ANOVA with Tukey’s post hoc analysis. Each data point represents an implantation site, three implantation sites were analyzed per mouse (n = 4 mice/treatment group).
Figure 5.
Figure 5.
Lopinavir treatment leads to depletion of uterine NK cells in a mouse pregnancy model. (A) Representative images of DBA-lectin immunostaining of the uterine NK (uNK) cells in decidua basalis region of GD9.5 implantation sites of control, ritonavir-boosted lopinavir (LPV/r)-combination antiretroviral therapy (cART) and ritonavir-boosted darunavir (DRV/r)-cART-treated mice. Lower panels show a magnified view of the decidua basalis to highlight the uNK cell infiltration of the spiral arteries (indicated by arrows). The condensed apoptotic appearance of uNK cells in the LPV/r-cART treatment is indicated by *. Scale bar, 1 mm (upper panels), 200 µm (lower panels). (B) Quantification of staining intensity (pixels) of DBA-lectin positive uNK cells and (C) uNK cell counts for each treatment group. Results are the mean ± SD. *** indicates P < 0.0001, calculated using ANOVA with Tukey’s post hoc analysis. Each data point represents an implantation site, three implantation sites were analyzed per mouse (n = 4 mice/treatment group).
Figure 6.
Figure 6.
Lopinavir treatment causes defective placentation in a mouse pregnancy model. Representative images of pan-cytokeratin immunostaining (green) of trophoblasts in the placentas of GD 9.5 implantation sites of control, ritonavir-boosted lopinavir (LPV/r)-combination antiretroviral therapy (cART) and ritonavir-boosted darunavir (DRV/r)-cART-treated mice. DAPI, used for nuclear staining is blue. Parietal trophoblast giant cells (P-TGCs) with large, polyploid nuclei are indicated. Scale bar, 200 µm.
Figure 7.
Figure 7.
Lopinavir treatment inhibits the transcription factor STAT3. (A) Representative images of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) immunostaining of stroma cells in first-trimester human decidua treated with DMSO (control) or protease inhibitor (PI)-combination antiretroviral therapy (cART) as indicated. Inset is negative control. Scale bar, 50 µm. (B) Quantification of staining intensity (pixels) of p-STAT3 positive stroma cells. Results are the mean ± SD. *** indicates P < 0.0001, calculated using ANOVA with Tukey’s post hoc analysis. Each data point represents the average of two to three replicates per experiment; n = 10 independent experiments. (C) Representative images of p-STAT3 immunostaining of GD 6.5 implantation sites of control, ritonavir-boosted lopinavir (LPV/r)-cART and ritonavir-boosted darunavir (DRV/r)-cART treated mice. E, embryo; PDZ, primary decidual zone; SDZ, secondary decidual zone; M, mesometrium; AM, anti-mesometrium. * indicates uterine lumen. Scale bar, 600 µm. (D) Quantification of staining intensity (pixels) of p-STAT3 positive cells. Results are the mean ± SD. *** indicates P < 0.0001, calculated using ANOVA with Tukey’s post hoc analysis. Each data point represents an implantation site, three implantation sites were analyzed per mouse (n = 4 mice/treatment group).

Similar articles

Cited by

References

    1. Balogun KA, Guzman Lenis MS, Papp E, Loutfy M, Yudin MH, MacGillivray J, Walmsley SL, Silverman M,, Serghides L.. Elevated levels of estradiol in human immunodeficiency virus-infected pregnant women on protease inhibitor-based regimens. Clin Infect Dis 2018;66:420–427. - PMC - PubMed
    1. Bhurke AS, Bagchi IC, Bagchi MK.. Progesterone-regulated endometrial factors controlling implantation. Am J Reprod Immunol 2016;75:237–2. - PMC - PubMed
    1. Burton GJ, Woods AW, Jauniaux E, Kingdom JC.. Rheological and physiological consequences of conversion of the maternal spiral arteries for uteroplacental blood flow during human pregnancy. Placenta 2009;30:473–4. - PMC - PubMed
    1. Cahill LS, Zhou YQ, Hoggarth J, Yu LX, Rahman A, Stortz G, Whitehead CL, Baschat A, Kingdom JC, Macgowan CK. et al. Placental vascular abnormalities in the mouse alter umbilical artery wave reflections. Am J Physiol Heart Circ Physiol 2019;316:H664–H672. - PMC - PubMed
    1. Carlino C, Stabile H, Morrone S, Bulla R, Soriani A, Agostinis C, Bossi F, Mocci C, Sarazani F, Tedesco F. et al. Recruitment of circulating NK cells through decidual tissues: a possible mechanism controlling NK cell accumulation in the uterus during early pregnancy. Blood 2008;111:3108–31. - PubMed

Publication types

MeSH terms