Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 May 15.
Published in final edited form as: Curr HIV/AIDS Rep. 2024 Jan 26;21(1):1–10. doi: 10.1007/s11904-024-00688-y

Antiretroviral Therapy in Pregnancy: A 2023 Review of the Literature

Alison N Goulding 1, Kasey Meeks 2, Lena Shay 1, Sarah Casey 3, Colton Applegarth 4, Jennifer McKinney 1
PMCID: PMC11095844  NIHMSID: NIHMS1987412  PMID: 38277098

Abstract

Purpose of Review

Selection of antiretroviral therapy during pregnancy must consider maternal physiology and resulting pharmacokinetic changes in pregnancy, resistance and efficacy profiles, tolerability and frequency of adverse effects, teratogenicity, and maternal, neonatal, and pregnancy outcomes. The objective of this review is to summarize the underlying data that informs the current clinical perinatal guidelines in the USA.

Recent Findings

Data now supports the use of dolutegravir at all stages of pregnancy with no significant increase in neural tube defects. Safety and pharmacokinetic data on newer antiretroviral medications in pregnancy continue to lag behind the general population.

Summary

While there are multiple safety and tolerability concerns with older regimens, there are now multiple options of regimens that are highly efficacious and have good safety data in pregnancy. Most pregnant patients who are virally suppressed on a well-tolerated regimen are able to safely continue those medications during pregnancy.

Keywords: Pregnancy, Antiretroviral therapy, Teratogenicity

Introduction

Recent data demonstrate that women account for 18% and 22% of new and total human immunodeficiency virus (HIV) diagnoses respectively in the USA and 46% of new and 53% of total diagnoses worldwide [1, 2]. Women within reproductive age categories account for the vast majority of new infections [1, 2]. Please note that while gender-inclusive language will be used throughout this paper wherever possible, descriptions of primary data will utilize language from the original publication or data source. About 5000 people with HIV in the USA and 1.2 million worldwide give birth every year in the USA [2, 3]. While efforts to prevent initial HIV infection in persons of reproductive age are important in preventing perinatal transmission, equally important are efforts to ensure that people with HIV receive appropriate treatment. While not all reproductive-age people are interested in trying to conceive, healthcare providers must prioritize discussing reproductive goals with patients and consider these desires in the choice of antiretroviral (ARV) therapy. The Perinatal HIV Clinical Guidelines, written by the US Panel on Treatment of HIV During Pregnancy and Prevention of Perinatal Transmission, assess the available evidence and outline evidence-based recommendations for ARV therapy during the pre-conception, pregnancy, and postpartum periods [4]. Development of guidelines for ARV in pregnancy includes consideration of pharmacokinetics in pregnancy, efficacy, simplicity and tolerability of regimens, drug-drug interactions, teratogenicity, and maternal and neonatal safety data. The purpose of this article is not to summarize existing clinical recommendations but instead to briefly review the primary data that inform clinical guidelines for the use of ARV in pregnancy.

Nucleoside Reverse Transcriptase Inhibitors

Nucleoside reverse transcriptase inhibitors (NRTIs) are the backbone of HIV treatment and prevention, both in pregnancy and in the general population. Zidovudine, a NRTI, was the first medication made available for HIV treatment in 1987 [5]. NRTIs work by inhibiting the reverse transcriptase enzyme, effectively preventing replication of the virus [6, 7]. Despite renal excretion of NRTIs and changes in renal physiology during pregnancy, studies have shown that the pharmacokinetic profiles of NRTIs do not significantly change during pregnancy [8, 9]. There are some data suggesting that genetic differences could affect NRTI levels in pregnancy, but evidence does not currently support the need for dose adjustment [10].

NRTIs are not metabolized through the CYP 450 system; therefore, fewer drug-drug interactions exist compared to non-nucleoside reverse transcriptase inhibitors (NNRTIs) and protease inhibitors (PIs). There are certain tuberculosis medications (including rifampin) and antiepileptic drugs (carbamazepine) however that can affect levels of certain NRTIs, so checking drug-drug interactions prior to coadministration of these drugs is recommended [11].

Some NRTI resistance mutations, with M184V being one of the most common, confer resistance to zidovudine and stavudine, and these mutations can potentially induce cross-resistance with newer NRTIs, such as abacavir and tenofovir [12]. Estimates vary for NRTI resistance in the pregnant and postpartum population, with one study from Cameroon showing NRTI resistance during breastfeeding to be approximately 15% [13]. Resistance, especially to older NRTIs, should be considered when starting ARV medication.

Currently, no significant increase in birth defects has been noted with first-trimester exposure to regimens containing NRTIs by the Antiretroviral Pregnancy Registry. Data are sufficient to rule out a twofold increase in birth defects for didanosine, stavudine, tenofovir alafenamide, and a 1.5-fold increase for abacavir, emtricitabine, lamivudine, tenofovir disoproxil fumarate, and zidovudine [14••].

Historically, an association was noted between NRTI-containing regimens and poor obstetric outcomes [8]; however, current research shows conflicting results with most studies showing no significant association between prenatal exposure to NRTIs and risk of spontaneous abortion, small for gestational age, or prematurity. Some increased risk of low birth weight is seen in patients exposed to NRTI alone, but this regimen is no longer the standard of care [8, 9, 15, 16]. In general, tenofovir disoproxil fumarate/emtricitabine may be associated with larger neonates, fewer neonates that are small for gestational age, and less placental oxidative stress when compared to abacavir/lamivudine or zidovudine/lamivudine, but again, data are mixed [17, 18]. The mechanism of the adverse effects of NRTIs is thought to be through mitochondrial toxicity and oxidative stress, but the exact effect of NRTIs on the long-term health of the fetus and neonate needs more investigation [17, 19].

In terms of potential adverse effects on the pregnant person, tenofovir alafenamide has fewer renal effects than tenofovir disoproxil in the general population, and this trend is also seen in a pregnant cohort with lower creatinine at delivery for patients on tenofovir alafenamide–containing regimens as compared to efavirenz, emtricitabine, and tenofovir disoproxil fumarate containing regimens [8]. Other adverse effects such as myopathy, peripheral neuropathy, and lactic acidosis with hepatic steatosis are more common with use of zidovudine, lamivudine, stavudine, and didanosine, leading to current preference for regimens containing tenofovir/emtricitabine [7].

In summary, NRTIs, especially newer medications, are the backbone of HIV treatment in pregnancy and are generally well tolerated by both mothers and fetuses.

Non-nucleoside Reverse Transcriptase Inhibitors

NNRTIs noncompetitively inhibit reverse transcriptase by altering the binding of the enzyme to DNA [20]. NNRTIs have high oral bioavailability and undergo primarily hepatic metabolism via the cytochrome P450 (CYP 450) system, most commonly by CYP 2C and 3A enzyme families [2124]. Because the enzymatic activity of the CYP 450 system is altered during pregnancy, plasma levels of NNRTIs differ among pregnant and non-pregnant patients. Plasma concentrations of efavirenz and rilpivirine decrease during pregnancy, while concentrations of nevirapine remain unchanged [2527] and concentrations of etravirine increase [28]. There is insufficient data to draw conclusions about the pharmacokinetics of doravirine during pregnancy. Despite the change in plasma levels, dose adjustment is not currently recommended in pregnant patients [21, 24, 25, 29], although more frequent monitoring of HIV viral load may be beneficial for pregnant patients on ARV regimens containing efavirenz, nevirapine, and rilpivirine due to the potential decrease. Data regarding the pharmacokinetics of injectable rilpivirine (in combination with cabotegravir) is based on a few case reports only; preliminary data suggest that plasma and washout concentrations may be similar in pregnant and non-pregnant people [30]. Because of metabolism via the CYP 450 system, there are also drug-drug interactions including with multiple medications that are commonly used in pregnancy: mood medications, including selective serotonin reuptake inhibitors (SSRIs), and certain antimicrobials, including metronidazole and fluconazole [31]. While these medications are not contraindicated, awareness of changing drug levels (both of ARV and the co-administered medication) is important.

NNRTIs have high rates of placental transfer and effectively prevent vertical transmission of HIV [21, 29, 32]. However, they are generally reserved for second- or third-line treatment due to their high susceptibility to drug resistance, as well as decreased efficacy compared to other ARV classes [21, 33]. The prevalence of HIV drug resistance to NNRTIs has been estimated to be as high as 11–15%, occurring more frequently than both NRTIs and protease inhibitors (PIs) [22, 34, 35]. The first-generation NNRTIs, nevirapine and efavirenz, have a very low threshold for resistance, and cross-resistance between these drugs is common. The second-generation NNRTIs, etravirine, rilpivirine, and doravirine have a higher threshold for resistance [22]. Previous studies have shown that several HIV strains resistant to first-generation NNRTIs are still susceptible to a second-generation formulation [21, 23, 24, 36, 37]. In terms of efficacy, regimens containing NNRTIs have been shown to be less effective in suppressing viral load at delivery than dolutegravir, an integrase strand transfer inhibitor (INSTI) [8, 38], although the rates of perinatal HIV transmission do not differ [32].

Historically, there has been concern that NNRTIs may be associated with fetal anomalies. Recent studies have shown no consistent significant association between exposure to nevirapine or rilpivirine and incidence of birth defects, although existing data are inconclusive regarding the effect of efavirenz on neural tube defects specifically or the effects of etravirine or doravirine on any birth defect [24, 29, 39, 40]. Efavirenz has been associated with microcephaly and neurodevelopmental delay in some neonates [39, 4143]. The presence of conflicting results is one reason that efavirenz is classified as an alternative medication for initiation during pregnancy.

There are conflicting data regarding the association between in utero exposure to NNRTIs and adverse fetal and infant outcomes. Some studies have found that low birthweight may be more common among infants exposed to NNRTIs than those exposed to PIs or INSTIs, although the differences were not statistically significant [16, 44]. When compared with dolutegravir-containing regimens, efavirenz- and nevirapine-containing regimens have been shown to have a significantly higher risk of neonatal mortality [8, 39]. Nevirapine-containing regimens in particular have been associated with an increased risk of adverse birth outcomes (stillbirth, preterm delivery, low birth weight), even when compared with ARV regimens containing other NNRTIs [39, 4547]. Data are contradictory when directly comparing NNRTIs to PI-based regimens with some studies showing increased and some studies showing decreased incidence of poor perinatal outcomes [16, 48, 49•]. There are currently insufficient data to establish fetal and infant safety for etravirine, rilpivirine, and doravirine [21, 24, 29].

In terms of obstetric outcomes, NNRTI-based regimens have been associated with an increased risk of gestational diabetes compared to NRTI-based regimens [50]. Nevirapine-based regimens may additionally be associated with an increased risk of hypertensive disorders of pregnancy compared to regimens containing ARV from other classes [37, 39]. Pregnant patients on efavirenz-based regimens have been found to have lower total weight gain than pregnant patients on dolutegravir-based regimens [8, 39]. However, no difference in total weight gain has been found between other regimens containing NNRTIs, INSTIs, or PIs [44].

Pregnancy may increase the risk of adverse events among patients on NNRTI-based ARV regimens. Two of the most common severe adverse effects associated with NNRTIs are hepatic toxicity and skin rash, both of which may occur more frequently in pregnant patients [51]. While hepatotoxicity is theoretically possible with all NNRTIs, it seems to be primarily a concern with first-generation NNRTIs (nevirapine and efavirenz) [52]. Because there is an overlap between the symptoms of pregnancy and early hepatic toxicity, including nausea and vomiting, pregnant patients who continue NNRTI-containing ARV regimens should be monitored closely for hepatotoxicity. Efavirenz has additionally been shown to be associated with an increased risk of neuropsychiatric side effects, including insomnia, depression, and encephalopathy, especially in patients with other psychiatric conditions [39, 53, 54]. In light of these findings, mood should be monitored in pregnant patients on efavirenz-containing regimens, especially in those at increased risk for postpartum depression.

In summary, NNRTIs are not recommended as first-line therapy in pregnant patients due to the high prevalence of drug resistance, possible association with adverse birth outcomes, and insufficient data on fetal and maternal safety.

Protease Inhibitors

Protease inhibitors (PIs) as a class were first introduced in 1995 and have long been used in combination therapies with NRTIs to treat HIV. After their initial introduction, the use of PIs was particularly popular during pregnancy due to concerns that efavirenz, an NNRTI, was linked with an increased risk of neural tube defects, an association that has not been borne out in further studies [39, 55]. However, their use has since fallen out of favor, especially in high-resource settings, due to increasing associations with preterm birth and small-for-gestational-age infants, though they are often still used in low-resource areas [56•].

Pharmacokinetic studies of PIs have shown variable intracellular and plasma drug concentrations in the third trimester of pregnancy relative to the non-pregnant person. These alterations in drug concentration are largely attributable to physiologic and metabolic changes of pregnancy, although some evidence also suggests that genetic alterations affecting CYP 450 3A4 metabolism may be responsible for some reduction in bioavailability [55, 57]. Outside of pregnancy, cobicistat is used to boost the antiretroviral effects of atazanavir and darunavir; however, multiple pharmacokinetic studies showing markedly reduced concentrations of the drug limit its use during pregnancy [5860]. Because of these observed changes in pregnancy, the preferred PIs are administered with ritonavir, another PI, to boost third-trimester bioavailability. Dosing adjustments are recommended for darunavir and lopinavir, while atazanavir requires only once-daily dosing [6163]. Drug interactions are extremely common between PIs and multiple classes of medications, including many that are commonly used in pregnancy such as H2-antagonists, proton-pump inhibitors, macrolide antibiotics, anti-epileptics, anti-depressants, anti-psychotics, and corticosteroids.

Challenges with drug resistance plague PIs as with many other antiretroviral drug classes. A 2022 study of PI resistance found that up to a third of patients failing second-line PI therapy had significant resistance mutations [64]. However, available data looking at the development of resistance in pregnancy suggests no significant increase in the development of PI resistance with short-term use during gestation [64, 65].

Regarding virologic efficacy, studies have demonstrated similar viral suppression and tolerability between PI-based regimens but have suggested decreased viral suppression with lopinavir compared to NNRTIs. Despite differences in viral suppression, rates of perinatal HIV transmission have not been found to differ [66, 67].

Studies of drug concentrations within cord blood have shown low transplacental passage of PIs (likely related to large molecular size and high degree of protein binding), contributing to the low risk of teratogenicity observed with this medication class [39, 55]. Concerning adverse pregnancy outcomes, the most pressing concern is the association between PI use, preterm birth, and small-for-gestational-age infants. Evidence both in vitro and in vivo suggests that PIs broadly affect progesterone, human chorionic gonadotropin, sex-hormone binding globulin, prolactin, and estrogen concentrations in pregnancy, effects which are postulated to contribute mechanistically to increased rates of preterm birth and small for gestational age infants [56•]. Meta-analyses including one published in 2023 have upheld these associations [49•, 68•, 69]. Additionally, evidence from animal models suggests that PI exposure in early pregnancy can also result in dysregulation of angiogenesis within the placenta and alterations in spiral artery remodeling and decidualization, which may contribute to the development of intrauterine growth restriction, preterm birth, or early onset severe preeclampsia [56•]. Many of these studies have been carried out using lopinavir, with less evidence existing for atazanavir or darunavir. When examining other adverse perinatal effects of PIs, a systematic review performed in 2019 found three studies that supported a correlation between PI use and hypertensive diseases of pregnancy [70].

Though generally well tolerated, side effects of PI use (both during and outside of pregnancy) include an increased risk of gastrointestinal upset with diarrhea, nausea, vomiting, and abdominal pain. Clinicians should monitor for such side effects after initiation of PIs although it may be difficult to determine which are side effects of medication versus attributable to pregnancy itself. Atazanavir carries an increased risk of maternal hyperbilirubinemia which could potentially lead to an increased risk of hyperbilirubinemia in the neonate [61].

In summary, due to possible association with adverse pregnancy outcomes (specifically preterm birth and small for gestational age infants), as well as significant drug interactions and the need for alteration in dosing in pregnancy, PIs are less often recommended as first-line therapy in high-resource settings where other regimens are available.

Entry and Attachment Inhibitors

Entry and attachment inhibitors target the HIV-1 entry process. There are several advantages to targeting the entry process: [1] The virus is blocked before integration of the viral genome into the host cell genome; [2] there is no requirement to enter cells, unlike NRTIs, NNRTIs, INSTIs, and PIs; and [3] given that the entry process comprises multiple distinct steps, there are multiple potential targets for entry inhibitors [71]. The main drugs in this class include fostemsavir, maraviroc, and ibalizumab, and they are used to treat patients with multidrug-resistant infections.

Fostemsavir is a prodrug of the active drug temsavir, which is a gp120-directed attachment inhibitor. No pharmacokinetic studies in human pregnancy have been performed on fostemsavir. There are no human data available regarding the placental passage of fostemsavir, though a study in rats demonstrated placental passage of temsavir [72]. Regarding teratogenicity, there is no data available in humans, but no evidence of teratogenicity in rat or rabbit studies at doses comparable to those used in humans [72]. Given limitations in available data, fostemsavir is not a recommended ARV agent in pregnancy, although it can be used in cases of multidrug resistant HIV in which other options are limited.

Maraviroc is a small molecule drug that inhibits the binding of HIV-1 to a co-receptor, cysteine-cysteine chemokine receptor 5 (CCR5), to prevent attachment and entry into the cell [73]. One study measured the pharmacokinetic profiles in the third trimester and postpartum in 18 individuals taking maraviroc. They concluded that standard adult dosing adjusting for concomitant ARV drugs is appropriate in pregnancy. The same study measured median cord blood-to-maternal delivery plasma drug ratios and found moderate placental transfer of maraviroc to the fetus [74]. Data in humans are insufficient to assess for potential teratogenicity of maraviroc. There is no evidence of teratogenicity in studies of rats or rabbits at dosages comparable to those used in humans [75]. Notably, a retrospective study of 857 pregnant people showed increased maternal hepatotoxicity among the 492 individuals starting ARV regimens including maraviroc in pregnancy; the adjusted hazard ratio for hepatotoxicity with maraviroc was 4.19 (95% confidence interval 1.34–13.1, p = 0.01) [76]. In light of limited available data, maraviroc is not recommended for use in pregnancy.

Ibalizumab is a recombinant humanized monoclonal antibody. It has a novel mechanism of action as a cluster of differentiation 4 (CD4)-directed post-attachment inhibitor, and its pharmacokinetic profile allows for dosing every 14 days after the initial loading dose [77]. There are no pharmacokinetic studies on ibalizumab in human pregnancy. No human data are available, but in general, placenta transfer of monoclonal antibodies does occur, so transfer of ibalizumab is possible [78]. Based on data collected in monkeys, there is potential for reversible immunosuppression (specifically CD4 T cell and B cell lymphocytopenia) in infants born to people taking ibalizumab during pregnancy, similar to that seen with use of other monoclonal antibodies during pregnancy. There are no data regarding the teratogenicity of ibalizumab in humans, though a study in monkeys did not report any malformations or premature births [78]. Given insufficient available data in humans, ibalizumab is not a recommended ARV agent in pregnancy.

Overall, for these ARV medications targeting the HIV-1 entry process, there is limited data in pregnancy, and consequently, no members of these newer drug classes are currently recommended in pregnancy.

Integrase Strand Transfer Inhibitors

Integrase strand transfer inhibitors (INSTIs) were first approved for the treatment of HIV in 2007. The class, which includes raltegravir, elvitegravir, dolutegravir, bictegravir, and cabotegravir, has become common in first-line ARV regimens [79]. Notably, the World Health Organization (WHO) has recommended dolutegravir as first-line ARV for all people with HIV, including those who are pregnant or breastfeeding [80]. INSTIs prevent viral integration by blocking the integrase enzyme active site and preventing target cell infection [81]. Pharmacokinetic studies have found that physiologic changes during pregnancy decrease levels of INSTIs during the second and third trimesters [82]. For example, elvitegravir trough levels have been found to be decreased in most pregnant people, which could lead to suboptimal viral suppression, and therefore, it is not recommended for use in pregnancy [83]. Raltegravir is recommended to have twice daily dosing during pregnancy due to decreased plasma levels, while standard dosing of dolutegravir has shown appropriate target levels [8487]. Bictegravir is a newer INSTI and is only available in a fixed-dose formulation; although data are limited, preliminary results suggest adequate levels in pregnancy [88, 89]. Due to the limited data, it is considered an alternative regimen for ARV-naïve patients during pregnancy and should be continued for people already well suppressed prior to pregnancy.

INSTIs have fewer drug-drug interactions than other classes of ARV. However, interactions with several commonly used drugs should be considered before prescribing. These include metformin, rifampin, St. John’s Wort, and various antacids [85, 90]. It is important to counsel regarding the separation of the prenatal vitamin and any antacids containing cations by at least 2 h before or 6 h after taking ARV-containing INSTIs. Lack of separation of prenatal vitamin and ARV is a common reason for viral blips in a patient with otherwise well-controlled HIV and excellent adherence during pregnancy [90].

As with other ARV classes, INSTIs have documented resistance. However, they are known for having a higher resistance barrier as compared to other ARV medications, such as NRTIs and NNRTIs [81]. First-generation drugs, such as raltegravir and elvitegravir, have several known resistance-conferring mutations and demonstrate cross-resistance [86]. Dolutegravir and bictegravir have been shown to have a greater resistance barrier than first-generation options and remain effective against HIV-1 strains that are resistant to raltegravir and elvitegravir [81]. While pharmacokinetic variations and decreased INSTI levels could theoretically lead to increased levels of resistance in pregnant people taking INSTIs, no studies have shown altered resistance profiles among pregnant patients [87].

Regarding the teratogenic effects of INSTIs, the most well-documented concern is that of dolutegravir and neural tube defects. In 2018, an observational study in Botswana resulted in a safety signal being released by WHO after investigators found higher rates of neural tube defects among women taking dolutegravir from conception, as compared to women taking non-dolutegravir ARV regimens [80, 91]. However, analysis of final data from this cohort demonstrated a non-significant increased rate of only 0.2%, after which WHO reversed its statement and recommended dolutegravir as a first-line treatment for all individuals, including pregnant people [80, 92••]. Other INSTIs, such as raltegravir and elvitegravir, have not been found to increase rates of congenital anomalies [93].

Overall, INSTIs are considered safe for use in pregnancy. A few studies have found increased rates of hypertensive disease during pregnancy compared to pregnant people on ARV regimens that did not contain INSTIs, but these findings have not been replicated [94, 95].

While INSTIs are a well-tolerated antiretroviral class, there are some common side effects. All INSTIs can cause mild creatinine elevations, which should be considered when used in patients with chronic kidney disease [96]. Another documented side effect of INSTIs is increased weight gain among non-pregnant people [97]; this trend has also been seen among pregnant women, with one study finding 2 kg of excess weight gain comparing people taking dolutegravir versus efavirenz [98]. Notably, several studies have shown increased rates of neuropsychiatric effects, such as depressive symptoms and anxiety in people taking dolutegravir, so this should be considered for patients with a history of neuropsychiatric illness [99].

In summary, dolutegravir, bictegravir, and raltegravir are the only INSTIs recommended for use in pregnant people, with dolutegravir being preferred in light of its once-daily dosing, higher barrier to resistance, and increased volume and duration of pregnancy safety data.

HIV-1 Capsid Inhibitors

HIV-1 capsid inhibitors are a new class of drugs that interfere with HIV capsid, a protein which acts as a shell to protect HIV’s genetic material (HIV RNA), proteins (nucleocapsid), and enzymes (reverse transcriptase and integrase) required for replication. This class of drugs can interfere with HIV capsid at multiple points in the viral life cycle including nuclear transport, virus assembly and release, and capsid assembly [100]. Currently, lenacapavir [101] is the only HIV capsid inhibitor approved by the US Food and Drug Administration (FDA). It was approved in December 2022 to be used in combination with other ARV drugs for the treatment of multidrug-resistant HIV-1 infection [102]. Currently, there are no pregnancy-specific data available for lenacapavir. The ongoing Women’s HIV Prevention Study is evaluating the use of lenacapavir for pre-exposure prophylaxis in adolescent girls and young women living in sub-Saharan Africa. Participants who become pregnant during the study can remain study participants after a reconsent process. Results from this study are pending [103]. There are currently insufficient human data to recommend or guide the use of lenacapavir in pregnancy.

Conclusion

Selecting ARV regimens in pregnancy is complex and requires consideration of physiologic changes of pregnancy, pharmacokinetic studies, resistance and efficacy profiles, tolerability and frequency of adverse effects, teratogenicity, and maternal, neonatal, and pregnancy outcomes. Based on these factors, medications are classified for use in pregnancy. Preferred and alternative ARV regimens for initiation in ARV-naïve pregnant patients are included in Table 1 as an example. A balance must be attempted between choosing the “safest” regimen (with the acknowledgment that research including pregnant people is limited and often delayed compared to the general population) and understanding the risks of changing regimens during an extremely sensitive and high-risk time for perinatal transmission. Regimens with more data may be selected for patients initiating ARV for the first time, but for the vast majority of pregnant people who are on well-tolerated ARV and have achieved viral suppression, medications should be continued. Data is constantly evolving, and providers who manage ARV in pregnancy should liberally utilize national resources like the national guidelines (https://clinicalinfo.hiv.gov/en/guidelines/perinatal/whats-new) and contact the perinatal HIV hotline (888–448–8765, https://nccc.ucsf.edu/clinician-consultation/perinatal-hiv-aids/) to discuss complicated cases.

Table 1.

Recommended antiretroviral regimens for initiation in antiretroviral-naive pregnant patients

NRTI* NNRTI PI INSTI
ABC/3TC EFV + preferred dual NRTI DRV/r BID + preferred dual NRTI DTG + preferred dual NRTI
TAF/FTC, TAF/3TC, TDF/FTC, TDF/3TC RPV + preferred dual NRTI ATV/r + preferred dual NRTI RAL BID + preferred dual NRTI
ZDV/3TC BID BIC/FTC/TAF

Bold: preferred regimens, italic: alternative regimens.

Adapted from Table 6 What to Start: Initial Antiretroviral Regimens During Pregnancy for People Who Are Antiretroviral-Naïve. https://clinicalinfo.hiv.gov/sites/default/files/guidelines/documents/perinatal-hiv/tables-perinatal.pdf

BID twice daily dosing in pregnancy, ABC abacavir, ATV/r atazanavir/ritonavir, BIC bictegravir, DRV/r darunavir/ritonavir, DTG dolutegravir, EFV efavirenz, FTC emtricitabine, RAL raltegravir, RPV rilpivirine, TAF tenofovir alafenamide, TDF tenofovir disoproxil fumarate, ZDV zidovudine, 3TC lamivudine

*

Dual NRTI combinations must be combined with a third agent from another category

Footnotes

Declarations

Competing Interests The authors declare no competing interests.

Conflict of Interest The authors declare no competing interests.

Human and Animal Rights and Informed Consent This article does not contain any studies with human or animal subjects performed by any of the authors.

Data Availability

There is no primary data for this paper.

References

Papers of particular interest, published recently, have been highlighted as:

• Of importance

•• Of major importance

  • 1.Division of HIV Prevention, Viral Hepatitis, STD, and TB Prevention. HIV and women [Internet]. Atlanta (GA): Centers for Disease Control and Prevention; [reviewed 2023 Feb 16]. Available from: https://www.cdc.gov/hiv/group/gender/women/index.html. [Google Scholar]
  • 2.UNAIDS. Global HIV & AIDS statistics — Fact sheet: UNAIDS; 2024. [updated 2024. https://www.unaids.org/en/resources/fact-sheet.
  • 3.Nesheim SR, FitzHarris LF, Lampe MA, Gray KM. Reconsidering the number of women with HIV infection who give birth annually in the United States. Public Health Rep. 2018;133(6):637–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Panel on treatment of HIV during pregnancy and prevention of perinatal transmission, recommendations for the use of antiretroviral drugs during pregnancy and interventions to reduce perinatal HIV transmission in the United States [Internet]. Department of Health and Human Services. Available at https://clinicalinfo.hiv.gov/en/guidelines/perinatal. Accessed 2023 Oct 15. [Google Scholar]
  • 5.Cihlar T, Ray AS. Nucleoside and nucleotide HIV reverse transcriptase inhibitors: 25 years after zidovudine. Antiviral Res. 2010;85(1):39–58. [DOI] [PubMed] [Google Scholar]
  • 6.Delviks-Frankenberry KA, Nikolenko GN, Boyer PL, Hughes SH, Coffin JM, Jere A, et al. HIV-1 reverse transcriptase connection subdomain mutations reduce template RNA degradation and enhance AZT excision. Proc Natl Acad Sci. 2008;105(31):10943–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Patel PH, Zulfiqar H. Reverse transcriptase inhibitors. [Updated 2023 Jun 23]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; [reviewed 2023 Aug]. Available from https://pubmed.ncbi.nlm.nih.gov/31855348/. [PubMed] [Google Scholar]
  • 8.Lockman S, Brummel SS, Ziemba L, Stranix-Chibanda L, McCarthy K, Coletti A, et al. Efficacy and safety of dolutegravir-and tenofovir alafenamide fumarate-containing HIV antiretroviral treatment regimens started in pregnancy: a randomised controlled trial. Lancet (London, England). 2021;397(10281):1276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Thimm MA, Eke AC. Tenofovir, pregnancy and renal function changes in pregnant women living with HIV. AIDS (London, England). 2021;35(8):1319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Gini J, Olagunju A, Dickinson L, Waitt C, Neary M, Else LJ, et al. Impact of pharmacogenetics and pregnancy on tenofovir and emtricitabine pharmacokinetics. Pharmacogenomics. 2019;20(04):217–23. [DOI] [PubMed] [Google Scholar]
  • 11.US Panel on Treatment of HIV During Pregnancy and Prevention of Perinatal Transmission. Guidelines for the use of antiretroviral agents in adults and adolescents with HIV [Internet] Department of Health and Human Services. Available at https://clinicalinfo.hiv.gov/en/guidelines/perinatal. Accessed 2023 Oct 15. [Google Scholar]
  • 12.Cecchini D, Sfalcin J, Zapiola I, Gómez A, Giuliano SF, Mammana L, et al. Reverse transcriptase and protease inhibitors mutational viral load in HIV infected pregnant women with transmitted drug resistance in Argentina. Rev Esp Quimioter. 2021;34(4):371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Torimiro JN, Nanfack A, Takang W, Keou CK, Joyce AN, Njefi K, et al. Rates of HBV, HCV, HDV and HIV type 1 among pregnant women and HIV type 1 drug resistance-associated mutations in breastfeeding women on antiretroviral therapy. BMC Pregnancy Childbirth. 2018;18(1):1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.••. Antiretroviral Pregnancy Registry Steering Committee. Antiretroviral pregnancy registry interim report for 1 January 1989 through 31 January 2023 [Internet]. Morrisville, NC: Registry Coordinating Center; 2023. Available: www.APRegistry.com. (This interim report summarizes the current state of evidence for major teratogenic effects for pregnancies exposed to antiretroviral medications in the USA.)
  • 15.Group S, Collaboration PHC. Nucleoside reverse transcriptase inhibitor backbones and pregnancy outcomes. AIDS (London, England). 2019;33(2):295–304. [DOI] [PubMed] [Google Scholar]
  • 16.Saint-Lary L, Diallo A, de Monteynard LA, Paul C, Marchand L, Tubiana R, et al. In utero exposure to antiretroviral drugs and pregnancy outcomes: analysis of the French ANRS pharmacovigilance database. Br J Clin Pharmacol. 2022;88(3):942–64. [DOI] [PubMed] [Google Scholar]
  • 17.Balogun K, Serghides L. Comparison of the effects of three dual-nucleos (t) ide reverse transcriptase inhibitor backbones on placenta mitochondria toxicity and oxidative stress using a mouse pregnancy model. Pharmaceutics. 2022;14(5):1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Siemieniuk RA, Foroutan F, Mirza R, Ming JM, Alexander PE, Agarwal A, et al. Antiretroviral therapy for pregnant women living with HIV or hepatitis B: a systematic review and meta-analysis. BMJ Open. 2017;7(9):e019022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Poirier MC, Gibbons AT, Rugeles MT, Andre-Schmutz I, Blanche S. Fetal consequences of maternal antiretroviral nucleoside reverse transcriptase inhibitor (NRTI) use in human and nonhuman primate pregnancy. Curr Opin Pediatr. 2015;27(2):233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bec G, Meyer B, Gerard M-A, Steger J, Fauster K, Wolff P, et al. Thermodynamics of HIV-1 reverse transcriptase in action elucidates the mechanism of action of non-nucleoside inhibitors. J Am Chem Soc. 2013;135(26):9743–52. [DOI] [PubMed] [Google Scholar]
  • 21.Lê MP, Pencolé L, Peytavin G, Bouchet-Crivat F, Mandelbrot L. Placental transfer of doravirine, a recent HIV-1 NNRTI in the ex vivo human cotyledon perfusion model. J Antimicrob Chemother. 2021;76(9):2364–7. [DOI] [PubMed] [Google Scholar]
  • 22.Usach I, Melis V, Peris JE. Non-nucleoside reverse transcriptase inhibitors: a review on pharmacokinetics, pharmacodynamics, safety and tolerability. J Int AIDS Soc. 2013;16(1):18567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Blevins SR, Hester EK, Chastain DB, Cluck DB. Doravirine: a return of the NNRTI class? Ann Pharmacother. 2020;54(1):64–74. [DOI] [PubMed] [Google Scholar]
  • 24.Havens JP, Podany AT, Scarsi KK, Fletcher CV. Clinical pharmacokinetics and pharmacodynamics of etravirine: an updated review. Clin Pharmacokinet. 2020;59(2):137–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lartey M, Kenu E, Lassey A, Ntumy M, Ganu V, Sam M, et al. Pharmacokinetics of efavirenz 600 mg once daily during pregnancy and post partum in Ghanaian women living with HIV. Clin Ther. 2020;42(9):1818–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Schalkwijk S, Colbers A, Konopnicki D, Gingelmaier A, Lambert J, Van Der Ende M, et al. Lowered rilpivirine exposure during the third trimester of pregnancy in human immunodeficiency virus type 1–infected women. Clin Infect Dis. 2017;65(8):1335–41. [DOI] [PubMed] [Google Scholar]
  • 27.Capparelli E, Aweeka F, Hitti J, Stek A, Hu C, Burchett S, et al. Chronic administration of nevirapine during pregnancy: impact of pregnancy on pharmacokinetics. HIV Med. 2008;9(4):214–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ramgopal M, Osiyemi O, Zorrilla C, Crauwels HM, Ryan R, Brown K, et al. Pharmacokinetics of total and unbound etravirine in HIV-1–infected pregnant women. JAIDS J Acquir Immune Defic Syndr. 2016;73(3):268–74. [DOI] [PubMed] [Google Scholar]
  • 29.Osiyemi O, Yasin S, Zorrilla C, Bicer C, Hillewaert V, Brown K, et al. Pharmacokinetics, antiviral activity, and safety of rilpivirine in pregnant women with HIV-1 infection: results of a phase 3b, multicenter, open-label study. Infectious diseases and therapy. 2018;7:147–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Patel P, Ford SL, Baker M, Meyer C, Garside L, D’Amico R, et al. Pregnancy outcomes and pharmacokinetics in pregnant women living with HIV exposed to long-acting cabotegravir and rilpivirine in clinical trials. HIV Med. 2023;24(5):568–79. [DOI] [PubMed] [Google Scholar]
  • 31.Gilani B, Cassagnol M. Biochemistry, Cytochrome P450. [Updated 2023 Apr 24]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; [reviewed 2023 Oct]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK557698/. [PubMed] [Google Scholar]
  • 32.Davey S, Ajibola G, Maswabi K, Sakoi M, Bennett K, Hughes MD, et al. Mother-to-child HIV transmission with in-utero dolutegravir vs. efavirenz in Botswana. J Acquir Immune Defic Syndr. 2020;84(3):235–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Gay CL, Mayo AJ, Mfalila CK, Chu H, Barry AC, Kuruc JD, et al. Efficacy of NNRTI-based antiretroviral therapy initiated during acute HIV infection. AIDS. 2011;25(7):941–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bello G, Kagoli M, Chipeta S, Auld A, Chang JC, DeVos JR, et al. Resistance levels to non-nucleoside reverse transcriptase inhibitors among pregnant women with recent HIV infection in Malawi. Antivir Ther. 2022;27(4):13596535221121224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Guerrero-Beltrán C, Martínez-Sanz J, Álvarez M, Olalla J, García-Álvarez M, Iribarren JA, et al. The algorithm used for the interpretation of doravirine transmitted drug resistance strongly influences clinical practice and guideline recommendations. J Antimicrob Chemother. 2020;75(5):1294–300. [DOI] [PubMed] [Google Scholar]
  • 36.Ayitewala A, Kyeyune F, Ainembabazi P, Nabulime E, Kato CD, Nankya I. Comparison of HIV drug resistance profiles across HIV-1 subtypes A and D for patients receiving a tenofovir-based and zidovudine-based first line regimens in Uganda. AIDS Res Ther. 2020;17:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Anderson MS, Gilmartin J, Cilissen C, De Lepeleire I, Van Bortel L, Dockendorf MF, et al. Safety, tolerability and pharmacokinetics of doravirine, a novel HIV non-nucleoside reverse transcriptase inhibitor, after single and multiple doses in healthy subjects. Antivir Ther. 2015;20(4):397–405. [DOI] [PubMed] [Google Scholar]
  • 38.Kintu K, Malaba TR, Nakibuka J, Papamichael C, Colbers A, Byrne K, et al. Dolutegravir versus efavirenz in women starting HIV therapy in late pregnancy (DolPHIN-2): an open-label, randomised controlled trial. The Lancet HIV. 2020;7(5):e332–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Eke AC, Mirochnick M, Lockman S. Antiretroviral therapy and adverse pregnancy outcomes in people living with HIV. New Engl J Med. 2023;388(4):344–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ford N, Mofenson L, Shubber Z, Calmy A, Andrieux-Meyer I, Vitoria M, et al. Safety of efavirenz in the first trimester of pregnancy: an updated systematic review and meta-analysis. AIDS. 2014;28:S123–31. [DOI] [PubMed] [Google Scholar]
  • 41.Crowell CS, Williams PL, Yildirim C, Van Dyke RB, Smith R, Chadwick EG, et al. Safety of in-utero antiretroviral exposure: neurologic outcomes in children who are HIV-exposed but uninfected. AIDS. 2020;34(9):1377–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Williams PL, Yildirim C, Chadwick EG, Van Dyke RB, Smith R, Correia KF, et al. Association of maternal antiretroviral use with microcephaly in children who are HIV-exposed but uninfected (SMARTT): a prospective cohort study. The Lancet HIV. 2020;7(1):e49–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Cassidy AR, Williams PL, Leidner J, Mayondi G, Ajibola G, Makhema J, et al. In utero efavirenz exposure and neurodevelopmental outcomes in HIV-exposed uninfected children in Botswana. Pediatr Infect Dis J. 2019;38(8):828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Floridia M, Masuelli G, Tassis B, Franceschetti L, Savasi VM, Spinillo A, et al. Weight gain during pregnancy in women with HIV receiving different antiretroviral regimens. Antivir Ther. 2020;25(6):315–25. [DOI] [PubMed] [Google Scholar]
  • 45.Fennell C, Seage GR III, Zash R, Phiri K, Diseko M, Mayondi G, et al. Adverse birth outcomes in Botswana among women with vertically or horizontally acquired human immunodeficiency virus. Journal of the Pediatric Infectious Diseases Society. 2021;10(3):252–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Caniglia EC, Zash R, Jacobson DL, Diseko M, Mayondi G, Lockman S, et al. Emulating a target trial of antiretroviral therapy regimens started before conception and risk of adverse birth outcomes. AIDS. 2018;32(1):113–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zash R, Jacobson DL, Diseko M, Mayondi G, Mmalane M, Essex M, et al. Comparative safety of antiretroviral treatment regimens in pregnancy. JAMA pediatrics. 2017;171(10):e172222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Masaba R, Borkowf CB, Girde S, Zeh C, Ndivo R, Nyang’au I, et al. Adverse fetal and infant outcomes among HIV-infected women who received either nonnucleoside reverse transcriptase inhibitor-based or protease inhibitor-based antiretroviral therapy for prevention of mother-to-child transmission. AIDS. 2018;32(12):1625–32. [DOI] [PubMed] [Google Scholar]
  • 49•. Saint-Lary L, Benevent J, Damase-Michel C, Vayssière C, Leroy V, Sommet A. Adverse perinatal outcomes associated with prenatal exposure to protease-inhibitor-based versus non-nucleoside reverse transcriptase inhibitor-based antiretroviral combinations in pregnant women with HIV infection: a systematic review and meta-analysis. BMC Pregnancy and Childbirth. 2023;23(1):80. 10.1186/s12884-023-05347-5. (This recent systematic review and meta-analysis reviewed existing literature regarding PI-associated adverse pregnancy outcomes and then compared those outcomes to the more commonly used NNRTI regimens to determine which outcomes occur more frequently with PI use.•)
  • 50.Mmasa KN, Powis K, Sun S, Makhema J, Mmalane M, Kgole S, et al. Gestational diabetes in women living with HIV in Botswana: lower rates with dolutegravir-than with efavirenz-based antiretroviral therapy. HIV Med. 2021;22(8):715–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Sibiude J, Warszawski J, Tubiana R, Le Chenadec J, Meier F, Faye A, et al. Liver enzyme elevation in pregnant women receiving antiretroviral therapy in the ANRS-French perinatal cohort. JAIDS J Acquir Immune Defic Syndr. 2019;81(1):83–94. [DOI] [PubMed] [Google Scholar]
  • 52.Benedicto AM, Fuster-Martínez I, Tosca J, Esplugues JV, Blas-García A, Apostolova N. NNRTI and liver damage: evidence of their association and the mechanisms involved. Cells. 2021;10(7):1687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Maughan D, Sonderup M, Gogela N, Locketz M, Wainwright H, Setshedi M, et al. A natural history of efavirenz drug-induced liver injury. S Afr Med J. 2021;111(12):1190–6. [DOI] [PubMed] [Google Scholar]
  • 54.Mollan KR, Smurzynski M, Eron JJ, Daar ES, Campbell TB, Sax PE, et al. Association between efavirenz as initial therapy for HIV-1 infection and increased risk for suicidal ideation or attempted or completed suicide: an analysis of trial data. Ann Intern Med. 2014;161(1):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Andany N, Loutfy MR. HIV protease inhibitors in pregnancy: pharmacology and clinical use. Drugs. 2013;73:229–47. [DOI] [PubMed] [Google Scholar]
  • 56.•. Dunk CE, Serghides L. Protease inhibitor-based antiretroviral therapy in pregnancy: effects on hormones, placenta, and decidua. The Lancet HIV. 2022;9(2):e120–9. 10.1016/S2352-3018(21)00249-6. (This paper provides an updated review of existing evidence supportive of a physiologic explanation for the clinical associations seen between PI use and adverse pregnancy outcomes of preterm birth and small for gestational age infants.)
  • 57.Focà E, Calcagno A, Bonito A, Cusato J, Domenighini E, D’Avolio A, et al. Pharmacokinetic changes during pregnancy according to genetic variants: a prospective study in HIV-infected patients receiving atazanavir-ritonavir. Antimicrobial Agents and Chemotherapy. 2018;62(7): 10.1128/aac.00309-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Momper JD, Wang J, Stek A, Shapiro DE, Scott GB, Paul ME, et al. Pharmacokinetics of darunavir and cobicistat in pregnant and postpartum women with HIV. AIDS. 2021;35(8):1191–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Momper JD, Wang J, Stek A, Shapiro DE, Powis KM, Paul ME, et al. Pharmacokinetics of atazanavir boosted with cobicistat in pregnant and postpartum women with HIV. J Acquir Immune Defic Syndr. 2022;89(3):303–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Crauwels HM, Osiyemi O, Zorrilla C, Bicer C, Brown K. Reduced exposure to darunavir and cobicistat in HIV-1-infected pregnant women receiving a darunavir/cobicistat-based regimen. HIV Med. 2019;20(5):337–43. [DOI] [PubMed] [Google Scholar]
  • 61.Eley T, Bertz R, Hardy H, Burger D. Atazanavir pharmacokinetics, efficacy and safety in pregnancy: a systematic review. Antivir Ther. 2013;18(3):361–75. [DOI] [PubMed] [Google Scholar]
  • 62.Best BM, Stek AM, Mirochnick M, Hu C, Li H, Burchett SK, et al. Lopinavir tablet pharmacokinetics with an increased dose during pregnancy. JAIDS J Acquir Immune Defic Syndr. 2010;54(4):381–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Conradie F, Zorrilla C, Josipovic D, Botes M, Osiyemi O, Vandeloise E, et al. Safety and exposure of once-daily ritonavirboosted atazanavir in HIV-infected pregnant women. HIV Med. 2011;12(9):570–9. [DOI] [PubMed] [Google Scholar]
  • 64.Chimukangara B, Lessells RJ, Sartorius B, Gounder L, Manyana S, Pillay M, et al. HIV-1 drug resistance in adults and adolescents on protease inhibitor-based antiretroviral therapy in KwaZulu-Natal Province, South Africa. J Global Antimicrob Resist. 2022;29:468–75. [DOI] [PubMed] [Google Scholar]
  • 65.Gingelmaier A, Eberle J, Kost BP, Bogner JR, Hofmann J, Weissenbacher T, et al. Protease inhibitor–based antiretroviral prophylaxis during pregnancy and the development of drug resistance. Clin Infect Dis. 2010;50(6):890–4. [DOI] [PubMed] [Google Scholar]
  • 66.Perry M, Taylor G, Sabin C, Conway K, Flanagan S, Dwyer E, et al. Lopinavir and atazanavir in pregnancy: comparable infant outcomes, virological efficacies and preterm delivery rates. HIV Med. 2016;17(1):28–35. [DOI] [PubMed] [Google Scholar]
  • 67.Cohan D, Natureeba P, Koss CA, Plenty A, Luwedde F, Mwesigwa J, et al. Efficacy and safety of lopinavir/ritonavir versus efavirenz-based antiretroviral therapy in HIV-infected pregnant Ugandan women. AIDS. 2015;29(2):183–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.•. Cowdell I, Beck K, Portwood C, Sexton H, Kumarendran M, Brandon Z, et al. Adverse perinatal outcomes associated with protease inhibitor-based antiretroviral therapy in pregnant women living with HIV: a systematic review and meta-analysis. EClinicalMedicine. 2022: 101368. 10.1016/j.eclinm.2022.101368. (This recent meta-analysis provides more robust evidence regarding the association of PI use and small for gestational age infants by analyzing 34 cohort studies with the inclusion of newer studies and a wider variety of PI-containing regimens compared to past meta-analyses.)
  • 69.Mesfin YM, Kibret KT, Taye A. Is protease inhibitors based antiretroviral therapy during pregnancy associated with an increased risk of preterm birth? Systematic review and a meta-analysis. Reprod Health. 2016;13(1):1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Premkumar A, Dude AM, Haddad LB, Yee LM. Combined antiretroviral therapy for HIV and the risk of hypertensive disorders of pregnancy: a systematic review. Pregnancy hypertension. 2019;17:178–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Eggink D, Bontjer I, de Taeye SW, Langedijk JP, Berkhout B, Sanders RW. HIV-1 anchor inhibitors and membrane fusion inhibitors target distinct but overlapping steps in virus entry. J Biol Chem. 2019;294(15):5736–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Fostemsavir (Rukobia) [package insert]. Food and Drug Administration. 2020. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/212950s000lbl.pdf
  • 73.Louten JV. Antivirals, and the beneficial uses of viruses. Essential human virology: Academic Press; 2016. p. 49. [Google Scholar]
  • 74.Colbers A, Best B, Schalkwijk S, Wang J, Stek A, Hidalgo Tenorio C, et al. Maraviroc pharmacokinetics in HIV-1–infected pregnant women. Clin Infect Dis. 2015;61(10):1582–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Maraviroc (Selzentry) [package insert]. U.S. Food and Drug Administration. 2020. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/022128Orig1s019 Osl.
  • 76.Huntington S, Thorne C, Anderson J, Newell M-L, Taylor G, Pillay D, et al. Does pregnancy increase the risk of ART-induced hepatotoxicity among HIV-positive women. J Int AIDS Soc. 2014;17(4 Suppl 3):19486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Beccari MV, Mogle BT, Sidman EF, Mastro KA, Asiago-Reddy E, Kufel WD. Ibalizumab, a novel monoclonal antibody for the management of multidrug-resistant HIV-1 infection. Antimicrobial agents and chemotherapy. 2019;63(6): 10.1128/aac.00110-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Trogarzo (ibalizumab-uiyk) [package insert]. Food and Drug Administration. 2020 Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/761065s008lbl.pdf.
  • 79.Latham AH, Nissim OA, Spitznagel MC, Kirk SE, Tarleton JL, Lazenby GB. Impact of integrase strand transfer inhibitor use during pregnancy on viral suppression at delivery and infant outcomes: a statewide retrospective cohort study. JAIDS J Acquir Immune Defic Syndr. 2022;89(4):448–53. [DOI] [PubMed] [Google Scholar]
  • 80.World Health Organization. Policy brief: update of recommendations on first-and second-line antiretroviral regimens. [Updated 2019 Jul 17]. Geneva: World Health Organization; [reviewed 2023 Aug]. Available at: https://iris.who.int/bitstream/handle/10665/325892/WHO-CDS-HIV-19.15-eng.pdf?sequence=1. [Google Scholar]
  • 81.Mbhele N, Chimukangara B, Gordon M. HIV-1 integrase strand transfer inhibitors: a review of current drugs, recent advances and drug resistance. Int J Antimicrob Agents. 2021;57(5):106343. [DOI] [PubMed] [Google Scholar]
  • 82.van der Galiën R, Ter Heine R, Greupink R, Schalkwijk SJ, van Herwaarden AE, Colbers A, et al. Pharmacokinetics of HIV-integrase inhibitors during pregnancy: mechanisms, clinical implications and knowledge gaps. Clin Pharmacokinet. 2019;58:309–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Momper JD, Best BM, Wang J, Capparelli EV, Stek A, Barr E, et al. Elvitegravir/cobicistat pharmacokinetics in pregnant and postpartum women with HIV. AIDS. 2018;32(17):2507–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Mulligan N, Best BM, Wang J, Capparelli EV, Stek A, Barr E, et al. Dolutegravir pharmacokinetics in pregnant and postpartum women living with HIV. AIDS. 2018;32(6):729–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Lu C-H, Bednarczyk EM, Catanzaro LM, Shon A, Xu J-C, Ma Q. Pharmacokinetic drug interactions of integrase strand transfer inhibitors. Current Res Pharmacol Drug Discov. 2021;2:100044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Garrido C, Villacian J, Zahonero N, Pattery T, Garcia F, Gutierrez F, et al. Broad phenotypic cross-resistance to elvitegravir in HIV-infected patients failing on raltegravir-containing regimens. Antimicrob Agents Chemother. 2012;56(6):2873–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Buckoreelall K, Cressey TR, King JR. Pharmacokinetic optimization of antiretroviral therapy in pregnancy. Clin Pharmacokinet. 2012;51:639–59. [DOI] [PubMed] [Google Scholar]
  • 88.Zhang H, Hindman JT, Lin L, Davis M, Shang J, Xiao D, Avihingsanon A, et al. A study of the pharmacokinetics, safety, and efficacy of bictegravir/emtricitabine/tenofovir alafenamide in virologically suppressed pregnant women with HIV. AIDS. 2024;38(1):F1–F9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Bukkems VE, Hidalgo-Tenorio C, Garcia C, Van Hulzen AG, Richel O, Burger DM, et al. First pharmacokinetic data of bictegravir in pregnant women living with HIV. AIDS. 2021;35(14):2405–6. [DOI] [PubMed] [Google Scholar]
  • 90.Federspiel J, Bukhari MJ, Hamill MM. Interactions between highly active antiretroviral therapy and over-the-counter agents: a cautionary note. BMJ Case Rep. 2021;14(1):e236655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Zash R, Makhema J, Shapiro RL. Neural-tube defects with dolutegravir treatment from the time of conception. N Engl J Med. 2018;379(10):979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.••. Zash R, Holmes L, Diseko M, Jacobson DL, Brummel S, Mayondi G, et al. Neural-tube defects and antiretroviral treatment regimens in Botswana. New Engl J Med. 2019;381(9):827–40. 10.1056/NEJMoa1905230. (Updated data from Botswana on the association between dolutegravir and neural tube defects. This update found a non-significant, minimal relationship, and supported the inclusion of dolutegravir as the preferred regimen for patients who are trying to conceive or pregnant.)
  • 93.Rasi V, Cortina-Borja M, Peters H, Sconza R, Thorne C. Brief report: surveillance of congenital anomalies after exposure to raltegravir or elvitegravir during pregnancy in the United Kingdom and Ireland, 2008–2018. JAIDS J Acquir Immune Defic Syndr. 2019;80(3):264–8. [DOI] [PubMed] [Google Scholar]
  • 94.Smith C, Fought AJ, Sung JF, McKinney JR, Metz TD, Fetters KB, et al. Congenital malformations and preeclampsia associated with integrase inhibitor use in pregnancy: a singlecenter analysis. PLoS ONE. 2023;18(6):e0276473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Zash R, Caniglia E, Mayondi G, Diseko M, Mabuka J, Jacobson D, et al. The risk of gestational hypertension with use of dolutegravir at conception. Abstract presented at: Conference on Retroviruses and Opportunistic Infections, March 6, 2021; Virtual. Session P2: Abstract number 576. [Google Scholar]
  • 96.Raffi F, Rachlis A, Stellbrink H-J, Hardy WD, Torti C, Orkin C, et al. Once-daily dolutegravir versus raltegravir in antiretroviral-naive adults with HIV-1 infection: 48 week results from the randomised, double-blind, non-inferiority SPRING-2 study. The Lancet. 2013;381(9868):735–43. [DOI] [PubMed] [Google Scholar]
  • 97.Asundi A, Olson A, Jiang W, Varshney SP, White LF, Sagar M, et al. Integrase inhibitor use associated with weight gain in women and incident diabetes mellitus. AIDS Res Hum Retroviruses. 2022;38(3):208–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Group NAS. Dolutegravir-based or low-dose efavirenz–based regimen for the treatment of HIV-1. New Engl J Med. 2019;381(9):816–26. [DOI] [PubMed] [Google Scholar]
  • 99.Senneker T, Tseng A. An update on neuropsychiatric adverse effects with second-generation integrase inhibitors and nonnucleoside reverse transcriptase inhibitors. Curr Opin HIV AIDS. 2021;16(6):309–20. [DOI] [PubMed] [Google Scholar]
  • 100.Thenin-Houssier S, Valente TS. HIV-1 capsid inhibitors as antiretroviral agents. Current HIV research. 2016;14(3):270–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Link JO, Rhee MS, Tse WC, Zheng J, Somoza JR, Rowe W, et al. Clinical targeting of HIV capsid protein with a long-acting small molecule. Nature. 2020;584(7822):614–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.FDA approves new HIV drug for adults with limited treatment options [Internet]. 2022. December 22. Available: https://www.fda.gov/news-events/press-announcements/fda-approves-new-hiv-drug-adults-limited-treatment-options.
  • 103.Gilead announces new arm of HIV women’s prevention study to evaluate the investigational long-acting HIV-1 capsid inhibitor lenacapavir in addition to Descovy for PrEP [Internet]. 2020. December 21. Available: https://www.gilead.com/news-and-press/company-statements/gilead-announces-new-arm-of-hiv-womens-prevention-study.

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

There is no primary data for this paper.

RESOURCES