Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jul 1.
Published in final edited form as: Obstet Gynecol Surv. 2023 Jul;78(7):411–428. doi: 10.1097/OGX.0000000000001159

Cannabis and Pregnancy: A review

Sarena Hayer 1, Ava D Mendelbaum 2, Lester Watch 3, Kimberly S Ryan 4, Madeline A Hedges 5, Jennifer A Manuzak 6, Charles A Easley IV 7, Danny Schust 8,**, Jamie Lo 9,**
PMCID: PMC10372687  NIHMSID: NIHMS1892281  PMID: 37480292

Abstract

Importance:

Prenatal cannabis use is rising and is a major public health issue. Cannabis use in pregnancy and during lactation has been associated with increased maternal and offspring morbidity and mortality.

Objective:

This review aims to summarize the existing literature and current recommendations for cannabis use during pregnancy or lactation.

Evidence Acquisition:

A PubMed, Cochrane Library, and Google Scholar literature search using the following terms was performed to gather relevant data: “cannabis,” “cannabinoid,” “delta-9-tetrahydrocannabinol,” “THC,” “cannabidiol,” “fetal outcomes,” “perinatal outcomes,” “pregnancy,” and “lactation.”

Results:

Available studies on cannabis use in pregnancy and during lactation were reviewed and support an association with increased risk of preterm birth, neonatal intensive care unit admission, low birth weight, and small for gestational age infants.

Conclusion and Relevance:

There is a critical need for research on the effects of cannabis use in pregnancy and during lactation. This is a necessary first step prior to furthering patient education, developing interventions, and targeting antenatal surveillance to ameliorate the adverse impacts on maternal and fetal health.

Target Audience:

Obstetricians and gynecologists, family physicians, pediatricians

Learning Objectives:

After participating in this activity, the learner will be better able to describe options for treatment of cannabis use disorder, explain common forms of cannabinoids available for use, and discuss screening options for cannabis use in pregnancy.

Keywords: cannabis, fetal, cannabinoid, neonatal, lactation, THC, cannabis use disorder, delta-9-tetrahydrocannabinol, pregnancy

MeSH Headings: Cannabinoids, fetal, neonatal, lactation, Cannabis use disorder, pregnancy

INTRODUCTION

Cannabis use in the United States is rising steeply, especially amongst adults of reproductive age. In the last decade, past-year prevalence of cannabis use amongst reproductive-age adults increased from 10.4% to 18.0% and daily or almost daily use (e.g., 300 or more days per year) rose from 1.3% to 3.9%1. Similarly, cannabis use in pregnancy has also increased from 3.4% in 2002 to 7.0% in 20172, and by 25% during the COVID-19 pandemic3. Prevalence of use is highest amongst pregnant individuals between 18 and 25 years of age4 and in the first trimester when the fetus is most sensitive to adversity2.

These trends are in contrast to patterns of alcohol and tobacco use in pregnancy, which have decreased over time5. This difference is partly a result of changing cannabis legislation, which has increased the availability of cannabis and decreased perceived risks with use. Studies have shown that legalization of cannabis in states such as Colorado and California have been associated with increased use in pregnancy6. Prior reports have also highlighted that the true prevalence of cannabis use in pregnancy is likely underreported in survey data7.

The American College of Obstetricians and Gynecologists has advised individuals to abstain from cannabis when pregnant and breastfeeding, but these individuals often continue to use partly because current safety data is limited8. There is concern for adverse pregnancy outcomes given that delta-9-tetrahydrocannabinol (THC, the main psychoactive component of cannabis) can readily cross the placenta8. The limited available evidence suggests that prenatal cannabis exposure is associated with stillbirth, fetal growth restriction, impaired fetal neurodevelopment, preterm delivery, increased neonatal intensive care unit admissions, and small for gestational age infants912.

However, there is still a paucity of knowledge related to the safety of using cannabis in pregnancy because the majority of published studies are limited by a retrospective or observational study design, rely primarily on patient self-report, are confounded by polysubstance use, have a small sample size, lack biological drug testing, have inconsistent dose information, and mostly focus on the effects of smoking cannabis rather than other contemporary forms of cannabis use13. In addition, it is difficult to compare the different modes of delivery and concentration of cannabis consumption. Unlike alcohol where a shot is equivalent to a beer and a glass of wine, smoking cannabis versus vaping, dabbing, or consuming edibles achieve different THC concentrations and there is a wide range in onset and duration of effect. This is also complicated by issues with poor labeling accuracy of available cannabis products14, 15.

The purpose of this review is to provide an updated overview regarding the association between prenatal cannabis exposure, and pregnancy and offspring outcomes.

HEALTH POLICY

Currently, in the United States, 37 states and Washington DC have legalized medical cannabis use and of those, 21 states have legalized non-medical cannabis use. Despite these recent moves at the state-level towards legalization, in the last two decades there has also been a trend towards more punitive policies on cannabis use in pregnancy that mandate health professional reporting and child welfare notification16. These measures stand in contrast to the legalization of cannabinoids for recreational and medical use at the state level and the implied safety with legislative acceptance of cannabis use17. In response, some states have implemented supportive policies aimed at reducing barriers to treatment16.

PROFESSIONAL GUIDELINES

American College of Obstetricians and Gynecologists

The American College of Obstetricians and Gynecologists (ACOG) recommends discontinuing cannabis use in individuals who are contemplating pregnancy or are currently pregnant18. ACOG guidelines recommend that pregnant individuals should be asked about cannabis use and counseled on the potential risks of continued use in pregnancy. These recommendations include encouraging pregnant individuals who use cannabis for medicinal purposes to consider alternative therapies with more sufficient pregnancy-specific safety data. ACOG also discourages cannabis use when breastfeeding given the absence of sufficient data to evaluate the effects of cannabis use on infants during lactation.

American Academy of Pediatrics

The American Academy of Pediatrics (AAP) recommends against cannabis use during pregnancy. In addition, the AAP recommends that pregnant individuals should be informed about the lack of definitive safety information and counseled on the potential adverse effects of cannabis use on fetal and child development19. The AAP also discourages use of any cannabis during breastfeeding due to insufficient evidence regarding the potential risks associated with exposure during lactation. Clinicians are encouraged to include cannabis use when discussing the use of other substances, such as tobacco and alcohol, during pregnancy and breastfeeding.

Centers for Disease Control and Prevention

The Centers for Disease Control and Prevention (CDC) advise pregnant individuals to refrain from cannabis use in pregnancy, citing potential birth complications and adverse impacts on fetal development20. Overall, the CDC recognizes the need for more research on the potential long-term consequences of cannabis use in pregnancy.

Academy of Breastfeeding Medicine

The Academy of Breastfeeding Medicine (ABM) protocol highlights the need for research on the long-term effects of cannabis use during breastfeeding because the available information remains limited21. The ABM advises healthcare providers to counsel lactating individuals that use cannabis to avoid or reduce use while breastfeeding21.

PERCEPTIONS OF CANNABIS USE

Amongst reproductive-aged women in the United States, the perception that cannabis has no associated risks has increased three-fold from 2005 to 201522. An analysis of the National Survey of Drug Use and Health from 2007 to 2012 demonstrated that 70% of pregnant and non-pregnant females believe there is a slight or no risk of harm from using cannabis one to two times per week23. Cannabis use prior to pregnancy has been associated with increased acceptability of use during pregnancy22, 24, and cannabis legalization has also influenced these attitudes and beliefs. In a study surveying 306 pregnant individuals, 10% reported that they would use cannabis more if it were legalized24. In states where cannabis has been legalized, women were more likely to use cannabis in the preconception, prenatal, and postpartum periods25.

Currently, information regarding the risks of cannabis use in pregnancy are obtained from different sources that may not consistently reflect evidence-based recommendations. A study conducting a series of 26 semi-structured interviews with pregnant participants found that commonly cited sources of information included the internet or anecdotal advice, with few reporting receiving useful information from a healthcare provider22, 26. Another study examining recommendations given by randomly selected Colorado cannabis dispensaries when approached by a pregnant mystery caller reported that 69% of dispensaries recommended the treatment of morning sickness with cannabis, one third stated that cannabis use is safe in pregnancy, and that these recommendations were based largely on personal opinion27.

Prior research has demonstrated that healthcare providers are not always consistently counseling patients, nor do they feel comfortable with counseling on the risks associated with cannabis use in pregnancy28. Several studies have assessed the counseling given by healthcare providers regarding prenatal cannabis use. A prior study sent a questionnaire to healthcare providers in France and demonstrated that 68.1% did not feel well informed about the risks of cannabis use in pregnancy29. In addition, another study using semi-structured interviews with 51 providers in the United States found that most were not familiar with the risks of cannabis use in pregnancy and that counseling practices focused more on legal risks than safety of use30, 31. This study also recorded the initial prenatal visits with these providers and in almost half of the visits, healthcare providers did not respond to disclosures of cannabis use or counsel patients30, 31. Crowley et al., conducted a survey-based assessment of postpartum mothers and found that most were aware that cannabis use while breastfeeding may be harmful to their newborn, but very few received counseling about the risks of use with lactation32.

FACTORS ASSOCIATED WITH CANNABIS USE IN PREGNANCY

Cannabis use in pregnancy is more prevalent in individuals who are younger, and with lower levels of education and household income33, 34. In pregnant individuals who co-use other substances such as tobacco, alcohol, and illicit drugs, these disparities are more pronounced, indicating that socioeconomic factors are associated with these higher-risk behaviors35. For example, individuals involved in the United States criminal justice system who use cannabis in pregnancy are also more likely to co-use cocaine and opioids36. In addition, there is a high incidence of cannabis use among pregnant and postpartum individuals with HIV. Yee et al., found that 11.7% of pregnant and 23.7% of postpartum individuals with HIV used cannabis in 2019 compared to 7.1% and 10.2% respectively in 200837.

In addition to socioeconomic factors, mental health also plays a role in patterns of use in pregnancy. Individuals with depression are more than three times more likely to use cannabis during pregnancy38. Prenatal cannabis use has also been associated with anxiety and trauma diagnoses39.

CANNABIS USE DISORDER

According to the Diagnostic and Statistical Manual of Mental Disorders, 5th edition (DSM-5), cannabis use disorder is defined as a pattern of continued use despite clinically significant impairment or distress40. The prevalence of cannabis use disorder among adults in the United States who have past-year cannabis use ranges from 9% to 31% depending on the different survey method used41. The National Surveys on Drug Use and Health collected data from 2007–2012 and reported that the prevalence of cannabis use disorder was 18.1% amongst past-year cannabis users who were pregnant23. A recent study by Wicken et al., surveying postpartum individuals who tested positive for cannabis use by urine toxicology at time of admission for delivery noted that 26.1% of survey respondents met criteria for cannabis use disorder42.

There are also notable gender-based differences associated with cannabis use disorder. While treatment-seeking rates are low for both men and women, women with cannabis use disorder are more likely to have concurrent mood and anxiety disorders43. Specific to pregnancy, the effects of prenatal cannabis use disorder are relatively understudied, but studies have shown that cannabis use disorder is associated with a greater risk of major adverse neonatal and infant outcomes44, 45. A retrospective cohort study of 4.83 million individuals who delivered a singleton birth from 2001 to 2012 reported that prenatal cannabis use disorder was associated with higher odds of small for gestational age, preterm birth, low birth weight, and death within one year of birth45.

COMMON FORMS OF CANNABIS USE IN PREGNANCY

Inhaled marijuana

This is the most common form46, 47 of cannabis use and includes smoking, vaporizing and dabbing (Figure 1). Smoking and vaporizing are the quickest method for THC to enter systemically, and provides a rapid onset (1–3 minutes) and short duration (1–3 hours) of symptoms48, 49. Dabbing refers to the inhalation of extremely concentrated THC made from hash oil that is vaporized48, 50.

Figure 1.

Figure 1.

Different formulations of cannabis

Oral or gastrointestinal absorption of marijuana

This is the second most popular mode of cannabis use amongst pregnant individuals and those of reproductive age46. Oral absorption includes drops, tinctures, sprays, lollipops, or breath strips with symptoms occurring after approximately 20 minutes that can last 1 to 3 hours48, 50 (Figure 1). Gastrointestinal absorption includes edibles, candies, drinks, snacks and capsules with an onset of action ranging from 30 to 90 minutes after consumption and persisting for 6 to 8 hours48 (Figure 1). As a result of the delayed onset of symptoms, these methods have a higher likelihood of overconsumption48, 50.

Skin absorption of marijuana

There are two types of skin-applied formulations of cannabis, they include topicals and transdermals (Figure 1). Topicals are absorbed through the epidermis or hair follicles and bind to cannabinoid receptors to provide localized effects. These include balms, salves, lotions, liniments, and bath soaks. Their onset of action is approximately 15 to 30 minutes after application with effects lasting for 3 to 6 hours48, 50. Topicals will not result in a positive urine drug screen because they do not enter the bloodstream. Transdermals are absorbed into the bloodstream with a similar onset of action of 15 to 30 minutes after applying and can persist for 6 to 8 hours49. These include gels and patches that are time-released48, 50.

Suppositories

Suppositories can be inserted rectally or vaginally and are especially suitable for individuals who cannot or do not want to ingest or inhale cannabis (Figure 1). Vaginal suppositories have an onset time of approximately 20 minutes and last for 3 to 6 hours48. Rectal suppositories are less psychoactive because they are not as readily absorbed into the bloodstream48, 50.

OTHER CANNABINOIDS USED IN PREGNANCY

Cannabidiol (CBD)

Cannabidiol, or CBD, is a component of the Cannabis sativa plant that does not have psychotropic activity51. Its mainstream use has increased in the United States due to its therapeutic potential through anxiolytic, anti-inflammatory, and antiemetic effects52. Data on the effects of cannabidiol use in pregnancy however, are extremely limited despite increasing use within this population for the treatment of symptoms such as nausea and chronic pain53. In an in vitro study examining the effects of cannabidiol on trophoblast cell and placental function, cannabidiol was found to interfere with trophoblast turnover and placental remodeling51. A prior study in mice noted that the transfer of cannabidiol from maternal blood to fetal blood was rapid, with accumulations of compound found in the fetal brain, liver, and gastrointestinal tract, as well as within amniotic fluid54.

Delta-8-tetrahydrocannabinol and delta-10- tetrahydrocannabinol

There is increasing interest in less commonly known cannabinoid formulations. In addition to delta-9-THC, there are other analogues of THC, including delta-8-THC and delta-10-THC, that are molecularly different due to the location of the double-bonded carbon chain (Figure 2). Although lawmakers have specified that hemp is only federally legal if it contains no more than 0.3% delta-9-THC, delta-8-THC and delta-10-THC are less regulated and can be synthesized using CBD from legal hemp. Currently, delta-8-THC is commonly labeled as a hemp product and can be purchased online or in stores, but has not been approved by the FDA.

Figure 2.

Figure 2.

Popular types of cannabinoid formulations available.

Delta-8-THC has psychoactive and intoxicating effects, similar to delta-9-THC, but studies have shown that delta-8-THC differs mechanistically in how it binds to the cannabinoid receptors in the body55 which may lead to differing effects55. Currently, delta-8-THC is used for its potential relaxing, euphoric, antiemetic, anxiolytic, appetite stimulating, and analgesic effects56. There are reportedly fewer adverse effects associated with use of delta-8-THC versus delta-9-THC, but studies have shown that individuals can still experience difficulty concentrating, issues with short-term memory, and an altered sense of time with use of delta-8-THC56. Limited research has been conducted on delta-10-THC, but some studies have indicated that it may have less potent psychoactive effects than delta-9-THC or delta-8-THC. Similar to delta-8-THC, delta-10-THC is not regulated or approved for use by the FDA.

Synthetic Cannabinoids

Different than delta-8-, delta-9- and delta-10-THC, synthetic cannabinoids (e.g., K2, spice, AK-47, Mr. Happy, Scooby Snax, Kush, or Kronic) are human-made mind-altering chemicals that are either in a liquid form that can be vaporized and inhaled in e-cigarettes, or are sprayed on a dried plant material that can be smoked. Synthetic cannabinoids were first synthesized in the 1970s, and initially were structurally similar to THC. Since then, a wide variety of compounds with structures differing from THC have been produced that also readily interact with cannabinoid receptors.

Despite their differences from THC, they are referred to as “cannabinoids” because they are similar to chemicals found in the cannabis plant. Synthetic cannabinoids are often marketed as “safe, legal alternatives” to cannabis, but can have more intense mind-altering effects that are unpredictable, more dangerous, and potentially life-threatening. In mice, synthetic cannabinoids are also reported to have adverse reproductive and pregnancy consequences, including an impact on preimplantation embryonal growth, and placental transport and development57. Of note, these synthetic cannabinoid compounds were not originally synthesized for intended human consumption, have not undergone necessary safety testing, and little is known about the scope of their effects in humans.

CANNABIS AND THE ENDOCANNABINOID SYSTEM (ECS)

The endocannabinoid system regulates aspects of the human nervous and immune systems via five different cannabinoid receptors, the two best-studied cannabinoid receptors are CB1 and CB258, 59. CB1 and CB2 receptors are present throughout the body, but are primarily found in central nervous system cells and immune cells, respectively. Endocannabinoids, or endogenous cannabinoids, and other factors are involved in complex human processes, such as thermoregulation, sensory and time perception, sleep, pain, pleasure, memory, appetite and more60. Through activation and inactivation of these receptors, cannabinoid use results in the classic symptoms of polyphagia, dry eyes and neuronal depression or activation depending on the strain, potency and route used61. Furthermore, the use of cannabis and exogenous cannabinoid formulations can cause disruption of the complex processes regulated by the endocannabinoid system. This is concerning because THC can bind to CB1 and CB2 receptors in the placenta, as well as cross the placenta and bind to these receptors in cells and tissues of the developing fetus.

Understanding how cannabinoids affect the endocannabinoid system is important as more people continue to use cannabis. There is recent evidence showing that the effects of cannabinoids, THC and CBD may not be as straightforward as previously understood. Studies have noted that CBD has a low affinity for the known CB1/CB2 receptors and THC can behave as an agonist or antagonist depending on the type of receptor and its location49. This highlights the complexity and heterogeneity of the endocannabinoid system and the potential impact of exogenous cannabinoid use.

PHARMACOKINETICS IN PREGNANCY

Existing human studies on the pharmacokinetics of cannabis have largely focused on non-pregnant adults and consumption of cannabis products through smoking6264. Prior human and animal studies have demonstrated that THC crosses the placenta resulting in fetal blood concentrations similar to maternal concentrations in some species6568. Older studies, using rodent and rabbit models, have reported on the pharmacokinetics and placental transfer of THC, but at a lower THC dose than that commonly experienced today, as most of this prior research was performed in the 1960s-1980s64, 6976. In the rat, fetal blood concentrations are one-tenth of maternal concentrations after maternal ingestion of THC11,37 and one-third of maternal concentrations following maternal inhalation or intravenous use of THC67, 68.

As the potency of cannabis has increased significantly the last few decades, there is limited data regarding the quantity of THC transferred in relation to the concentration of THC in contemporary cannabis products, frequency of use, and maternal blood concentrations. The route of use (e.g., ingestion, inhalation) can affect the amount of THC absorbed. Oral cannabis use is associated with THC absorption greater than 90%77, but due to significant first pass hepatic metabolism, bioavailability is less than 20%78. In contrast, smoking cannabis avoids first pass hepatic metabolism, but there is loss of THC in sidestream smoke, the cigarette butt, and from pyrolysis, resulting in low THC absorption and variability in bioavailability from 2 to 56%79, 80. Studies have also suggested variability in placental transport and physiology of THC81. Prior dizygotic twin studies, where both fetuses theoretically are exposed to similar maternal drug levels, have demonstrated large disparities in cannabinoid concentrations of hair and meconium samples between twins, including undetectable levels in one twin while positive in the other82.

CANNABIS USE IN PREGNANCY

Common Uses of Cannabis in Pregnancy

Despite the growing evidence demonstrating adverse outcomes associated with cannabis in pregnancy, many pregnant individuals continue to use. Thus, understanding the reasons why pregnant individuals use cannabis is an opportunity to provide additional support and targeted therapies. The most common reasons for cannabis use during pregnancy include treatment of hyperemesis gravidarum, anxiety, insomnia, nausea, and chronic pain83, 84. Importantly, many of these reasons have alternative treatments that are better studied in pregnancy. A prior study surveying pregnant and lactating individuals using cannabis identified three categories of reasons for use during pregnancy and lactation84. These include: 1) sensation-seeking for fun and enjoyment, 2) symptom management of chronic conditions and conditions related to pregnancy, and 3) coping with the unpleasant, but nonpathologized, experiences of life. Prior to pregnancy, individuals endorsed reasons for cannabis use in these three categories with similar proportions84. However, during pregnancy, the reason for use was largely for symptom management; during lactation, reasons for use resembled those expressed preconception84.

Screening for Cannabis Use in Pregnancy

ACOG recommends universal screening for substance use in pregnancy through validated questionnaires18. A variety of drug use screening tools exist and have been validated in pregnancy85, including the Screening, Brief Intervention, and Referral to Treatment approach (SBIRT), which involves assessing the severity of substance use, risk stratification, with subsequent counseling, intervention, or referral to specialty care86.

Self-reported cannabis use can underestimate the prevalence of use in pregnancy83, 87, 88. In a prior study, when compared to screening through urine drug testing, only 36% of patients who tested positive for cannabis disclosed current use to their healthcare provider89. Given the likelihood of underreporting, one study aimed to develop a more sensitive tool for identifying women using cannabis in pregnancy. In this study, 400 participants who had recently given birth were surveyed on direct assessment of drug use (directly asking about recent drug use), semi-indirect assessment (asking only about use prior to pregnancy), or indirect assessment (no mention of drug use); survey answers were then compared with urine and hair sample testing90. The study found that the indirect screening tool predicted toxicology results more accurately than direct questioning, although the clinical utility of indirect screening has not been elucidated, especially in the setting of false positive results85.

Despite underreporting through screening, ACOG does not recommend routine urine drug screening18. Biological sampling should only be obtained when indicated for clinical care and after obtaining maternal consent, especially when considering mandatory reporting91. In addition, patients are often selected for biological sampling in a biased fashion. In a retrospective cohort study of all deliveries at a tertiary care center from 2015 to 2019, after adjusting for commonly used diagnoses that prompted screening, non-Hispanic Black patients were significantly more likely to have urine drug screening performed (OR 2.0, 95% CI 1.6–2.4) despite being as likely to have a negative screen result as White patients92.

When biologic sampling is indicated, it may be performed through testing of maternal urine, serum, saliva, or hair (Figure 3). The most common method is urine toxicology, which identifies use in the preceding 3–5 days through detection of the most stable cannabis metabolite, 11-nor-delta-9-tetrahydrocannabinol-9-carboxylic acid (THC-COOH)93. Urine toxicology cannot capture use outside of an assay’s detection window93. Thus, a single urine toxicology result likely underestimates prevalence and may be biased towards individuals with heavier or longer-term use, as an urinalysis in this population is more likely to be positive87. If screening immunoassay tests are positive, a confirmatory test using gas chromatography-mass spectrometry analysis is recommended due to the high false-positive rate of screening point-of-care tests91.

Figure 3.

Figure 3.

Different methods for maternal cannabis drug testing

Infant testing involves sampling of umbilical cord homogenate, meconium, serum, or urine. Umbilical cord homogenate testing for THC-COOH can detect use from the third trimester onwards83, 94 (Figure 4). Immunoassay screening of fetal meconium extracts measures exposure from the end of the second trimester through the third trimester, but may be difficult to collect due to early passage in utero or requiring several days after birth for complete passage95 (Figure 4).

Figure 4.

Figure 4.

Postnatal methods for detecting prenatal cannabis exposure.

Treatment

Although admissions for cannabis use in pregnancy have risen from 1992 to 201296, there remains no current evidence-based interventions for cannabis use in pregnancy. There are limited existing studies examining the feasibility of interventions. One prior study randomized 45 pregnant individuals to electronic screening and brief intervention, tailored text messaging, or both; this study found that both intervention strategies had strong feasibility and modest to high acceptability90. Another study performed a randomized controlled trial of 50 pregnant individuals and examined the acceptability and preliminary efficacy of a computer-based, single-session, brief motivational intervention followed by a booster session in the reduction of both substance use and sexually transmitted infection risk97. This study also found high acceptability and significant reduction in cannabis or alcohol use in the intervention group.

Despite an increased prevalence of cannabis use disorder, treatment of cannabis use disorder in pregnancy has not been well studied. In the general adult and adolescent population, psychotherapeutic interventions remain first-line for treatment of cannabis use disorder as no pharmacological agent has been found to be clinically effective98. The evidence most consistently supports a combined approach of cognitive-behavioral therapy, motivational enhancement therapy, and potentially abstinence-based incentives98. When referring to specialty care in pregnancy, it is important to note that pregnant individuals using substances face particular barriers to accessing care. These challenges include difficulty finding treatment, a paucity of suitable treatment options, and skipping treatment due to a fear of being detected by health or criminal justice authorities99.

Treatment of cannabis use in pregnancy can also include addressing withdrawal symptoms. The Diagnostic and Statistical Manual of Mental Disorders, 5th edition (DSM-5), defines cannabis withdrawal syndrome as having at least three of the following symptoms occurring within seven days of reduced use: 1) irritability, anger, or aggression; 2) nervousness or anxiety; 3) sleep disturbance; 4) appetite or weight disturbance; 5) restlessness; 6) depressed mood; and 7) somatic symptoms (e.g., nausea, vomiting, headaches)40. In a recent meta-analysis, the prevalence of cannabis withdrawal syndrome amongst regular cannabis users was reported to be 47%100. Additionally, concurrent tobacco or other substance use and daily cannabis use were associated with a higher incidence of cannabis withdrawal syndrome100. Given the high prevalence of withdrawal symptoms in regular cannabis users, treatment of pregnant individuals should incorporate consideration for supportive therapies to manage withdrawal symptoms.

Pharmacologic Treatment for Withdrawal

In regular and heavy cannabis users, symptoms of withdrawal can occur after abrupt cessation or reduction in the quantity of cannabis used. Most commonly people identify symptoms of anxiety, irritability, anger or aggression, disturbed sleep, depressed mood and loss of appetite101. Symptom onset occurred at 24–48 hours after cessation and peak symptoms at 2 to 6 days101. Some studies have explored pharmacologic intervention for cannabis withdrawal, but most are limited by small sample size. Within the studies that used a standardized scoring system, for withdrawal, such as the 16-item marijuana withdrawal check list or the 19-item cannabis withdrawal scale, six reported greater improvement of symptoms in individuals administered a cannabinoid medication versus placebo. These medications included dronabinol102, quetiapine103, nabiximols104, gabapentin105, and oral THC106. Pharmacologic intervention can be tailored to individual withdrawal symptoms.

Intrapartum Analgesic Considerations

Cannabis use in pregnancy should not affect clinical decision-making regarding timing or mode of delivery. With regards to anesthesia however, cannabis use may enhance central nervous system depression if combined with other sedative hypnotic drugs107. Significant preoperative cannabis use should prompt consideration of additional postoperative nausea and vomiting prophylaxis, possible greater depth of anesthesia during induction and maintenance of anesthesia, and increased postoperative analgesia requirements108.

MATERNAL OUTCOMES FROM PRENATAL CANNABIS USE

The data on prenatal cannabis use on maternal health are limited, but overall does not consistently support any pregnancy-related adverse maternal outcomes1012, 109. Several prior studies have examined the association between cannabis use in pregnancy and hypertensive disorders with conflicting results. Some studies have reported no association110112, while others, including a retrospective cohort study of singleton deliveries in California from 2011 to 2017, noted that cannabis-related diagnoses were associated with an increased odds of hypertensive disorders (OR 1.2, 95% CI 1.2–1.3)113. Similarly, another retrospective cohort study also found increased odds of gestational hypertension (aOR 1.19, 95% CI 1.06–1.34), preeclampsia (aOR 1.16, 95% CI 1.0–1.28), and severe maternal morbidity (aOR 1.22, 95% CI 1.02–1.47) with maternal cannabis use114. Co-use of cannabis with cigarette smoking in pregnancy has also been linked to increased odds of preeclampsia (aOR 2.5, 95% CI 1.4–5.0) and maternal asthma (aOR 2.4, 95% CI 1.0–5.9)112.

Data on associations of cannabis use with gestational diabetes are similarly mixed109. With regards to other maternal outcomes, a previous retrospective cohort study of births in the United States from 1999 to 2013 found increased odds of prolonged hospital stay (OR 1.17, CI 1.11–1.23)115 and a former meta-analysis of 24 studies determined higher odds of anemia with cannabis use during pregnancy (pooled OR 1.36, 95% CI 1.10–1.69)9.

PLACENTAL OUTCOMES FROM CANNABIS EXPOSURE IN PREGNANCY

Components of the endocannabinoid system have been documented in the placenta starting at mid-gestation and play a role in placenta development, including placentation and trophoblast differentiation, and placental function116. At term, CB1 receptor expression has also been demonstrated in the amniotic epithelium, reticular and decidual cells117. Prior human studies have reported that THC can affect amnion development by inhibiting migration of the epithelial layer of placental amnion tissue, which may increase the risk for preterm labor and other adverse pregnancy outcomes118. Other human studies have also reported THC-induced altered placental steroidogenesis and estrogen signaling119, diminished placental blood flow120, 121, and histologic studies demonstrating increased syncytiotrophoblast knotting and fibrin deposition in the placental villi of cannabis users122.

Non-human primate studies have similarly reported that chronic prenatal THC exposure adversely impacts placental development and function123. Roberts et al., showed that THC-exposed pregnancies had significantly decreased placental perfusion and fetal oxygen availability suggestive of placental insufficiency. In addition, placental histological analyses demonstrated evidence of ischemic injury with microinfarctions in THC-exposed placentas only. Placental bulk RNA-sequencing was also notable for THC-induced alterations in the placental transcriptome and pathway analysis was suggestive of dysregulated vascular development and angiogenesis. Prior rodent studies have also reported that prenatal THC use is associated with an increased fetal/placental weight ratio due to observed fetal growth restriction and increased placental weight122, 124, 125. Lastly, in vitro studies have demonstrated that THC can negatively impact placental development by impairing cytotrophoblast fusion and biochemical differentiation, and inhibiting trophoblast cell turnover126.

FETAL OUTCOMES FROM MATERNAL CANNABIS USE

Prenatal Cannabis Exposure and Neurodevelopment

There is growing evidence of THC-associated adverse effects for fetal and neonatal developmental outcomes.127 In utero exposure to THC has been found to impact fetal brain development, including interruption of normal brain maturation, that may increase the risk for later in life neurocognitive and neuropsychiatric disorders128130. Maternal cannabis use has also been associated with “withdrawal”-like syndrome in newborns131 and increased aggressive behavior and attention deficits in offspring as early as 18 months of age131, 132.

Currently, the underlying mechanisms for these observations are not fully understood. The neurotransmitter serotonin (5-hydroxytryptamine, 5-HT) is present early in the developing fetal brain in humans and works as hormone/growth and differentiation factor133. Serotonin plays a central role in brain development, mood regulation, stress reactivity and risk of neuropsychiatric disorders, such as schizophrenia, affective disorders and anxiety134. Therefore, alterations in serotonin signaling during intrauterine life has critical implications for behavior and mental health later in life.

Expression of cannabinoid receptors in the human placenta has been associated with the regulation of the serotonin transporter activity. Prenatal cannabis may affect the placental clearance of serotonin through the serotonin transporter leading to hyperserotoninemia135. Due to the immaturity of the blood brain barrier in the fetus, serotonin can reach the developing brain. It has been shown that increased levels of serotonin during fetal development results in a dysfunction of the hypothalamic-pituitary axis that affects the amygdala as well as the regulation of oxytocin, a prosocial hormone, leading to neuropsychiatric disorders. To this point, high levels of serotonin are seen in the blood of over 30% of children with autism spectrum disorder, highlighting the role of serotonin in neuropsychiatric disorders136, 137.

Offspring Epigenetic Regulation

A growing body of literature has demonstrated that environmental stimuli, including maternal substance use, can alter epigenetic regulation and impact gene transcription138. Epigenetic mechanisms are increasingly recognized as a key link between early life experiences and risk of longer-term psychopathology.139 Cannabis exposure preconception or during pregnancy has been shown to alter epigenetic processes (e.g., DNA methylation and histone modifications) with functional consequences for gene expression including genes involved in the development of autism spectrum disorder, attention deficit hyperactivity disorder, schizophrenia, addiction, and other psychiatric diseases140.

Congenital anomalies

At this time, the literature does not consistently support an association of maternal cannabis use with an increased risk of fetal anomalies. A prior study found that cannabis use was associated with higher rates of birth defects involving the cardiac, genitourinary, gastrointestinal, and cerebral nervous system in addition to cleft lip and/or palate and Trisomy 21141. Other studies also support an increased risk of fetal cardiac defects, specifically ventricular septal defects142, neural tube defect143, minor physical anomalies144, gastroschisis145, diaphragmatic hernias145, and esophageal atresia145. However, most studies have not found a positive association between periconceptional cannabis use and birth defects146149, including fetal gastroschisis150, 151 and Ebstein anomaly152. A future review on this topic might consider a meta-analysis of these outcomes.

Fetal growth

Currently, the evidence suggests that prenatal cannabis exposure is associated with small for gestational age, defined as a birth weight less than the 10th percentile912. Although some studies did not adjust for confounders such as prenatal nicotine exposure, the most recently published systematic review observed that prenatal cannabis use significantly increased the likelihood of small for gestational age10. Lo et al., reported in unadjusted (18 studies; N=1,774,485) and adjusted (6 studies; N=686,336) meta-analyses that prenatal cannabis use significantly increased the odds of small for gestational age (unadjusted OR, 2.06; 95% CI, 1.76 to 2.41; aOR, 1.76; 95% CI, 1.52 to 2.05; I2, 86%)10. There is less consistent literature supporting the association between maternal cannabis use and fetal growth restriction, defined as an overall estimated fetal weight or an abdominal circumference measurement of less than the 10th percentile.

Preterm birth

Many studies have consistently reported that prenatal cannabis use is associated with an increased risk of preterm birth, defined as delivery prior to 37 weeks gestation, even after adjusting for confounders such as tobacco use912, 153. In general, most studies reported the results of any preterm birth, but only a few stratified results by spontaneous versus indicated preterm births.

Apgar score

The Apgar (Appearance, Pulse, Grimace, Activity, and Respiration) score is a quick assessment of newborn health at 1 and 5 minutes after birth to determine the need for resuscitation. While some studies reported a decreased Apgar scores at 1 minute154, overall, the majority of studies comparing Apgar scores between pregnancies with and without prenatal cannabis exposure found no significant differences between groups10, 12.

Neonatal Intensive Care Unit Admission (NICU)

The literature consistently supports an association between prenatal cannabis exposure and a significantly higher likelihood of neonatal intensive care unit (NICU) admissions1012, 110, 111, 154, 155. Overall, results were homogenous with little variability in findings between studies.

Perinatal Death

There is no consistent association amongst existing studies between prenatal cannabis use and perinatal death9, 10, 12, 156. However, the definition of perinatal mortality varied across studies from stillbirth to the inclusion of miscarriage, spontaneous abortions, or death before hospital discharge. In addition, some studies did not adjust for confounders such as tobacco use. Varner et al., reported the findings of the Stillbirth Collaborative Research Network, a case-control study from 2006 to 2008, and noted that cannabis use was associated with an increased risk of stillbirth (OR 2.34, 95% CI 1.13–4.81), but the effect was partially confounded by smoking157.

CHILDHOOD OUTCOMES

Cognitive Development and Behavior

The extant literature consists of three large, longitudinal prospective studies examining the effects of prenatal cannabis exposure on offspring neurocognitive development; all controlling for several different covariates. The oldest is the Ottawa Prenatal Prospective Study (OPPS), a landmark study initiated in 1978 that examined the effects of cannabis use in pregnancy amongst white, predominantly low-risk middle class families (n=583)158. Offspring were followed annually until at least 6 years of age and less frequently through 14 years of age. In 1982, the Maternal Health Practices and Child Development (MHPCD) study (n=763) was conducted to examine the effects of prenatal alcohol and cannabis exposure on offspring up to 16 years of age. Participants of this study were primarily lower income, and half of the cohort identified as African American159. The most recent is the Generation R study, a population-based birth cohort study that followed 7,452 participants in the Netherlands launched in 2002 with follow up of offspring that is still ongoing160.

Overall, these three studies demonstrated an association between prenatal cannabis exposure and adverse offspring neurodevelopmental and behavioral outcomes, despite a relatively small sample size of exposed offspring and limited data on the amount and potency of cannabis exposure in each study. There were some inconsistent findings across all three studies that may be partly attributed to maternal characteristics that differ amongst the studies. The MHPCD study reported that prenatal cannabis use was associated with decreased infant development at 9 and 19 months of age161. In contrast, a more recent observational cohort study of 3,435 individuals that used cannabis in pregnancy found no association with abnormal development screens in exposed infants at 3 and 9 months of age, but was associated with an increased risk for abnormal developmental screens at 12 months of age (aRR 1.90, 95% CI 0.92–3.91)162. At 36 months of age, the MHPCD study found that offspring exposure to cannabis in the first and second trimesters of pregnancy had significantly lower childhood performance on the Stanford-Binet Intelligence Scale163. By 6 years old, children exposed to heavy cannabis use in utero, defined as consumption of one or more cigarettes per day, had significant deficits in short-term memory and lower verbal reasoning scores164. These findings persisted in prenatally exposed children through 10 and 14 years of age and included deficits in reading and spelling scores165, 166, as well as lower ratings on teacher’s evaluations, negative effects on memory, and increased impulsivity, hyperactivity, inattention, and delinquency165, 167, 168.

Some of these findings of the MHPCD study were also noted in the earlier OPPS, which found that at 4 years of age and older, children exposed to cannabis in utero were found to have increased behavioral problems, as well as decreased language comprehension, decreased sustained attention and memory, and lower performance on visual perceptual tasks169. At 9 to 12 years of age, prenatal cannabis exposure was noted to be negatively associated with executive function tasks requiring impulse control, but not with global intelligence scores170. Similar findings were reported in a more recent secondary analysis of two multi-center randomized-controlled trials, which found that prenatal cannabis exposure was not associated with a difference in offspring intelligence quotient (IQ) at 60 months of age, but was associated with worse attention scores91.

In the Generation R study, female offspring at 18 months of age with prenatal cannabis exposure were found to have increased aggression and attention problems87. At 7 to 10 years of age, offspring exposed to cannabis in utero were noted to have behavioral problems, but not emotional problems, suggesting that the association was due to residual confounding171. Both the Generation R study as well as the OPPS specifically examined brain morphology in children with prior prenatal cannabis exposure. 54 children from the Generational R study, ages 6 to 8 years old, underwent brain magnetic resonance imaging and were found to have no change in global brain volumes172. However, when compared to unexposed children, offspring with in utero cannabis exposure had thicker frontal cortices172. This suggests that prenatal cannabis exposure may be associated with changes in brain morphology. Of note however, most of the children in the cannabis-exposed study group also had prenatal tobacco exposure. 16 participants from OPPS were also examined at 18–22 years of age and compared to 15 non-exposed adolescents; exposed individuals were found to have variations in neural activity while performing visuospatial tasks173.

Other studies have also noted associations between prenatal cannabis exposure and autism spectrum disorder and sleep disturbance. A retrospective cohort study of all births in Ontario, Canada from 2007 to 2012 found an association between cannabis use in pregnancy and autism spectrum disorder in offspring (aHR 1.51, 95% CI 1.17–1.96)174. Another prospective study found that prenatal cannabis exposure was associated with disturbed nocturnal sleep in exposed children at 3 years of age175. However, this study was limited by a small cohort size of 18 exposed children and 20 controls.

Mental Health

There have been several studies examining the association between prenatal cannabis exposure and the subsequent development of psychiatric disorders or symptoms in exposed children. One of these studies, the Adolescent Brain and Cognitive Development Study (ABCD Study), was a cross-sectional study of 11,489 children starting at ages 9 to 11 to track their biological and behavioral development through adolescence into young adulthood. Drawing data from the ABCD Study, a prior cohort study included 4,361 children that completed a questionnaire designed to assess psychotic-like experiences to understand the association between prenatal cannabis exposure and psychosis-proneness in offspring176. This cohort study showed that maternal cannabis use after knowledge of pregnancy was associated with a small increase in psychosis proneness (p=0.004)176.

The ABCD Study also determined that cannabis exposure after maternal knowledge of pregnancy was associated with a small increased risk of psychopathology in children at middle age (e.g., 9 to 11 years old), in addition to attention and social problems177. More recent published findings from the ABCD Study by Baranger, et al., similarly noted that prenatal cannabis exposure was associated with persistent vulnerability to psychopathology in offspring throughout early adolescence that was not ameliorated by age178. Increased psychopathology may result in greater risk for later-in-life psychiatric disorders and problematic substance use during peak periods of vulnerability during late adolescence. In contrast, a prior longitudinal cohort study of 6,356 children who completed a semi-structured interview for psychotic symptoms at 12 years of age reported that maternal cannabis use in pregnancy was not shown to have an association with psychotic symptoms in offspring at age 12179.

In addition to examining the associations between prenatal cannabis exposure and psychotic symptoms, a systematic review of studies on maternal use of cannabis in pregnancy and subsequent development of psychiatric disorders in offspring found that prenatal exposure may be associated with attention-deficit hyperactivity disorder and affective symptoms180. These findings are consistent with the above-mentioned longitudinal studies that demonstrated associations with decreased attention an increased impulsivity and hyperactivity, as well as an increase in offspring depressive symptoms at 10 years of age181. A prior meta-analysis also found that maternal cannabis use was associated with increased odds of offspring conduct disorder problems (OR 1.29, 95% CI 0.93–1.81)182.

Increased Risk of Substance Use and Addiction Vulnerability

There is evidence that prenatal cannabis exposure may increase the risk of offspring cannabis and tobacco use later in life, and addiction vulnerability. A prior human study examined the neurobiology underlying the risk of addiction vulnerability in mothers using cannabis that underwent elective abortions between 18 and 22 weeks of gestation183. This study reported reduced dopamine receptor D2 mRNA expression in the nucleus accumbens of fetal brains exposed to cannabis prenatally compared to controls The nucleus accumbens has is an important component of motor and reward circuits184, and disruptions in the dopamine signaling pathways can increase the risk of adverse psychiatric outcomes.185

Drawing on data from the OPPS, 152 offspring, ages 16 to 21 years old, completed a Drug History Questionnaire (DHQ) to determine if maternal cannabis use in pregnancy was associated with offspring cannabis and cigarette smoking186. This study found that maternal cannabis use was associated with both offspring cannabis use (OR=2.76) and daily tobacco cigarette smoking (OR=2.36) and that this use increased in a dose-responsive pattern186. A secondary analysis from the MHPCD study examined 590 offspring at 22 years old who completed a structured diagnostic interview to assess for an association between prenatal cannabis exposure and the development of cannabis use disorder in offspring187. Although there was no association between cannabis exposure and the development of cannabis use disorder, offspring who were prenatally exposed had increased risk of initiating cannabis use prior to 16 years of age (RR=1.16)187. The association between prenatal cannabis exposure and early onset of cannabis use in offspring was also demonstrated by another study that utilized MHPCD data188.

CANNABIS USE DURING LACTATION

Incidence of Use

Postnatal infant exposure to cannabis through breastfeeding remains poorly understood and may confer further risk to the infant. This topic is of particular importance as cannabis is the most commonly used recreational drug among lactating individuals83, 189, 190. Data from a recent cross-sectional study in Colorado estimated the prevalence of cannabis use in the breastfeeding postpartum period to be 5%191. Another study surveying lactation professionals attending the 2014 Vermont Lactation Consultant Association conference estimated that 15% of their breastfeeding clients used cannabis192.

Concentration of THC in Breast Milk

Individuals who use cannabis during breastfeeding risk infant exposure to the drug and its metabolites in breast milk. Studies have demonstrated that THC is secreted into breast milk and absorbed and metabolized by the breastfed infant50, 193. A prior study found that THC concentration are eight times higher in maternal milk relative to plasma, and infants are exposed to 0.8% of the mother’s consumed dose in a single feeding193. Baker et al., similarly studied the transfer of THC into the breast milk of eight individuals after smoking cannabis and detected a mean of 2.5% of the maternal dose within four hours after use50. While further research is needed to minimize confounders and with larger sample sizes, these studies provide preliminary evidence of the transfer and accumulation of THC in breast milk.

The concentration of THC in breast milk is partly influenced by maternal dose and the frequency of use. A study of 50 breastfeeding individuals who reported cannabis use while lactating found the greatest concentration of THC was one hour after consumption and was highest in mothers with daily use190. This study also demonstrated that THC was measurable in breast milk up to six days after maternal use.

Offspring Outcomes from Cannabis Use While Breastfeeding

There remains a paucity of clinical evidence regarding the developmental impact of exposure to cannabis through breastfeeding. However, as THC is highly lipophilic, it can be inferred that THC may be stored in infant brain tissue, which may have long-term effects on neurodevelopment. Astley and Little examined the impact of cannabis use on infant psychomotor development at one year of age194. Using a matched cohort study of 68 infant-mother pairs, the study found that offspring cannabis exposure in the first month postpartum was associated with decreased motor development at 1 year of age. However, this study was limited by the inability to control for the effects of prenatal THC exposure. In contrast, a study of 27 breastfed infants exposed to cannabis postnatally demonstrated no differences in motor and mental development relative to 35 unexposed infants195.

In the absence of definitive data on infant safety, professional guidelines encourage reducing or eliminating cannabis use while breastfeeding. The ABM suggest that breastfeeding mothers be counseled to reduce or eliminate use to avoid possible neurobehavioral effects on the infant21. The ACOG and AAP recommend refraining from cannabis use during lactation18, 19. Despite these recommendations, cannabis use remains prevalent and is often perceived as safe among postpartum populations189.

CONCLUSION

In this review, we have summarized the available data on prenatal cannabis exposure. Cannabis use in pregnancy is rising in part due to increased availability and greater societal acceptance. Of concern is the increasing potency of cannabis products and the growing body of evidence suggesting an increased risk of adverse prenatal and offspring outcomes associated with cannabis use in pregnancy1012, 83. In addition, most healthcare providers lack evidence-based knowledge regarding the potential effects of cannabis use and are unable to advise or counsel patients adequately. With many cannabis-based products now on the market, this is an area of significant public health relevance where additional research is needed to delineate the effects of different formulations (e.g., CBD, synthetic cannabinoids, delta-8-, delta-9-, and delta-10-THC) and methods of use (e.g., inhalation versus ingestion). An improved understanding of the issues surrounding maternal cannabis use is necessary to better educate and counsel individuals regarding the safety of use preconception, in pregnancy, and during breastfeeding, and to develop targeted antenatal surveillance strategies to mitigate maternal and offspring morbidity and mortality.

Funding:

NIH R01 OD028223-01, NIH/NIDA DP1 DA056493-01

Footnotes

The authors report no conflicts of interest

BIBLIOGRAPHY

  • 1.Center for Behavioral Health Statistics and Quality. 2019 National Survey on Drug Use and Health: Detailed Tables. Substance Abuse and Mental Health Services Administration. September 11, 2020. Accessed February 19, 2023. https://www.samhsa.gov/data/report/2019-nsduh-detailed-tables. [Google Scholar]
  • 2.Volkow ND, Han B, Compton WM, McCance-Katz EF. Self-reported Medical and Nonmedical Cannabis Use Among Pregnant Women in the United States. Jama. 2019;322(2):167–9. doi: 10.1001/jama.2019.7982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Young-Wolff KC, Ray GT, Alexeeff SE, Adams SR, Does MB, Ansley D, Avalos LA. Rates of Prenatal Cannabis Use Among Pregnant Women Before and During the COVID-19 Pandemic. Jama. 2021;326(17):1745–7. doi: 10.1001/jama.2021.16328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Brown QL, Sarvet AL, Shmulewitz D, Martins SS, Wall MM, Hasin DS. Trends in Marijuana Use Among Pregnant and Nonpregnant Reproductive-Aged Women, 2002–2014. Jama. 2017;317(2):207–9. doi: 10.1001/jama.2016.17383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Agrawal A, Rogers CE, Lessov-Schlaggar CN, Carter EB, Lenze SN, Grucza RA. Alcohol, Cigarette, and Cannabis Use Between 2002 and 2016 in Pregnant Women From a Nationally Representative Sample. JAMA Pediatr. 2019;173(1):95–6. doi: 10.1001/jamapediatrics.2018.3096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Gnofam M, Allshouse AA, Stickrath EH, Metz TD. Impact of Marijuana Legalization on Prevalence of Maternal Marijuana Use and Perinatal Outcomes. Am J Perinatol. 2020;37(1):59–65. Epub 20190906. doi: 10.1055/s-0039-1696719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Beatty JR, Svikis DS, Ondersma SJ. Prevalence and Perceived Financial Costs of Marijuana versus Tobacco use among Urban Low-Income Pregnant Women. J Addict Res Ther. 2012;3(4). doi: 10.4172/2155-6105.1000135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Committee Opinion No. 637: Marijuana Use During Pregnancy and Lactation. Obstet Gynecol. 2015;126(1):234–8. Epub 2015/08/05. doi: 10.1097/01.AOG.0000467192.89321.a6. [DOI] [PubMed] [Google Scholar]
  • 9.Gunn JK, Rosales CB, Center KE, Nuñez A, Gibson SJ, Christ C, Ehiri JE. Prenatal exposure to cannabis and maternal and child health outcomes: a systematic review and meta-analysis. BMJ Open. 2016;6(4):e009986. Epub 20160405. doi: 10.1136/bmjopen-2015-009986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lo JO, Shaw B, Robalino S, Ayers CK, Durbin S, Rushkin MC, Olyaei A, Kansagara D, Harrod CS. Cannabis Use in Pregnancy and Neonatal Outcomes: A Systematic Review and Meta-Analysis. Cannabis Cannabinoid Res. 2023. Epub 20230201. doi: 10.1089/can.2022.0262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Conner SN, Bedell V, Lipsey K, Macones GA, Cahill AG, Tuuli MG. Maternal Marijuana Use and Adverse Neonatal Outcomes: A Systematic Review and Meta-analysis. Obstet Gynecol. 2016;128(4):713–23. doi: 10.1097/aog.0000000000001649. [DOI] [PubMed] [Google Scholar]
  • 12.Marchand G, Masoud AT, Govindan M, Ware K, King A, Ruther S, Brazil G, Ulibarri H, Parise J, Arroyo A, Coriell C, Goetz S, Karrys A, Sainz K. Birth Outcomes of Neonates Exposed to Marijuana in Utero: A Systematic Review and Meta-analysis. JAMA Netw Open. 2022;5(1):e2145653. Epub 20220104. doi: 10.1001/jamanetworkopen.2021.45653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lo JO, Hedges JC, Girardi G. Impact of cannabinoids on pregnancy, reproductive health, and offspring outcomes. Am J Obstet Gynecol. 2022;227(4):571–81. Epub 20220531. doi: 10.1016/j.ajog.2022.05.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Bonn-Miller MO, Loflin MJE, Thomas BF, Marcu JP, Hyke T, Vandrey R. Labeling Accuracy of Cannabidiol Extracts Sold Online. JAMA. 2017;318(17):1708–9. doi: 10.1001/jama.2017.11909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Vandrey R, Raber JC, Raber ME, Douglass B, Miller C, Bonn-Miller MO. Cannabinoid Dose and Label Accuracy in Edible Medical Cannabis Products. JAMA. 2015;313(24):2491–3. doi: 10.1001/jama.2015.6613. [DOI] [PubMed] [Google Scholar]
  • 16.Faherty LJ, Stein BD, Terplan M. Consensus Guidelines and State Policies: The Gap Between Principle and Practice at the Intersection of Substance Use and Pregnancy. Am J Obstet Gynecol MFM. 2020;2(3):100137. Epub 20200517. doi: 10.1016/j.ajogmf.2020.100137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Mark K, Terplan M. Cannabis and pregnancy: Maternal child health implications during a period of drug policy liberalization. Prev Med. 2017;104:46–9. Epub 20170518. doi: 10.1016/j.ypmed.2017.05.012. [DOI] [PubMed] [Google Scholar]
  • 18.Braillon A, Bewley S. Committee Opinion No. 722: Marijuana Use During Pregnancy and Lactation. Obstet Gynecol. 2018;131(1):164. doi: 10.1097/aog.0000000000002429. [DOI] [PubMed] [Google Scholar]
  • 19.Ryan SA, Ammerman SD, O’Connor ME. Marijuana Use During Pregnancy and Breastfeeding: Implications for Neonatal and Childhood Outcomes. Pediatrics. 2018;142(3). Epub 20180827. doi: 10.1542/peds.2018-1889. [DOI] [PubMed] [Google Scholar]
  • 20.Ko JY, Coy KC, Haight SC, Haegerich TM, Williams L, Cox S, Njai R, Grant AM. Characteristics of Marijuana Use During Pregnancy - Eight States, Pregnancy Risk Assessment Monitoring System, 2017. MMWR Morb Mortal Wkly Rep. 2020;69(32):1058–63. Epub 20200814. doi: 10.15585/mmwr.mm6932a2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Reece-Stremtan S, Marinelli KA. ABM clinical protocol #21: guidelines for breastfeeding and substance use or substance use disorder, revised 2015. Breastfeed Med. 2015;10(3):135–41. doi: 10.1089/bfm.2015.9992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Jarlenski M, Koma JW, Zank J, Bodnar LM, Bogen DL, Chang JC. Trends in perception of risk of regular marijuana use among US pregnant and nonpregnant reproductive-aged women. Am J Obstet Gynecol. 2017;217(6):705–7. Epub 20170824. doi: 10.1016/j.ajog.2017.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ko JY, Farr SL, Tong VT, Creanga AA, Callaghan WM. Prevalence and patterns of marijuana use among pregnant and nonpregnant women of reproductive age. Am J Obstet Gynecol. 2015;213(2):201.e1–.e10. Epub 20150312. doi: 10.1016/j.ajog.2015.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Mark K, Gryczynski J, Axenfeld E, Schwartz RP, Terplan M. Pregnant Women’s Current and Intended Cannabis Use in Relation to Their Views Toward Legalization and Knowledge of Potential Harm. J Addict Med. 2017;11(3):211–6. doi: 10.1097/adm.0000000000000299. [DOI] [PubMed] [Google Scholar]
  • 25.Skelton KR, Hecht AA, Benjamin-Neelon SE. Recreational Cannabis Legalization in the US and Maternal Use during the Preconception, Prenatal, and Postpartum Periods. Int J Environ Res Public Health. 2020;17(3). Epub 20200201. doi: 10.3390/ijerph17030909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lebron CN, Morales V, Saenz S, Vidot DC. “Ganja Mamas”: Online discussions about cannabis use in pregnancy. Drug Alcohol Depend. 2022;241:109689. Epub 20221111. doi: 10.1016/j.drugalcdep.2022.109689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Dickson B, Mansfield C, Guiahi M, Allshouse AA, Borgelt LM, Sheeder J, Silver RM, Metz TD. Recommendations From Cannabis Dispensaries About First-Trimester Cannabis Use. Obstet Gynecol. 2018;131(6):1031–8. doi: 10.1097/aog.0000000000002619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bayrampour H, Zahradnik M, Lisonkova S, Janssen P. Women’s perspectives about cannabis use during pregnancy and the postpartum period: An integrative review. Prev Med. 2019;119:17–23. Epub 20181212. doi: 10.1016/j.ypmed.2018.12.002. [DOI] [PubMed] [Google Scholar]
  • 29.Gérardin M, Victorri-Vigneau C, Louvigné C, Rivoal M, Jolliet P. Management of cannabis use during pregnancy: an assessment of healthcare professionals’ practices. Pharmacoepidemiol Drug Saf. 2011;20(5):464–73. Epub 20110110. doi: 10.1002/pds.2095. [DOI] [PubMed] [Google Scholar]
  • 30.Holland CL, Nkumsah MA, Morrison P, Tarr JA, Rubio D, Rodriguez KL, Kraemer KL, Day N, Arnold RM, Chang JC. “Anything above marijuana takes priority”: Obstetric providers’ attitudes and counseling strategies regarding perinatal marijuana use. Patient Educ Couns. 2016;99(9):1446–51. Epub 20160604. doi: 10.1016/j.pec.2016.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Holland CL, Rubio D, Rodriguez KL, Kraemer KL, Day N, Arnold RM, Tarr JA, Chang JC. Obstetric Health Care Providers’ Counseling Responses to Pregnant Patient Disclosures of Marijuana Use. Obstet Gynecol. 2016;127(4):681–7. doi: 10.1097/aog.0000000000001343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Crowley HR, Goyal NK, Chung EK. Marijuana and Breastfeeding: A Pilot Survey of Mothers. Hosp Pediatr. 2022;12(7):e255–e60. doi: 10.1542/hpeds.2021-006420. [DOI] [PubMed] [Google Scholar]
  • 33.van Gelder MM, Reefhuis J, Caton AR, Werler MM, Druschel CM, Roeleveld N. Characteristics of pregnant illicit drug users and associations between cannabis use and perinatal outcome in a population-based study. Drug Alcohol Depend. 2010;109(1–3):243–7. Epub 20100218. doi: 10.1016/j.drugalcdep.2010.01.007. [DOI] [PubMed] [Google Scholar]
  • 34.Mark K, Desai A, Terplan M. Marijuana use and pregnancy: prevalence, associated characteristics, and birth outcomes. Arch Womens Ment Health. 2016;19(1):105–11. Epub 20150419. doi: 10.1007/s00737-015-0529-9. [DOI] [PubMed] [Google Scholar]
  • 35.Coleman-Cowger VH, Schauer GL, Peters EN. Marijuana and tobacco co-use among a nationally representative sample of US pregnant and non-pregnant women: 2005–2014 National Survey on Drug Use and Health findings. Drug Alcohol Depend. 2017;177:130–5. Epub 20170524. doi: 10.1016/j.drugalcdep.2017.03.025. [DOI] [PubMed] [Google Scholar]
  • 36.Washio Y, Mark K, Terplan M. Characteristics of Pregnant Women Reporting Cannabis Use Disorder at Substance Use Treatment Entry. J Addict Med. 2018;12(5):395–400. doi: 10.1097/adm.0000000000000424. [DOI] [PubMed] [Google Scholar]
  • 37.Yee LM, Kacanek D, Brightwell C, Haddad LB, Jao J, Powis KM, Yao T-J, Barr E, Broadwell C, Siminski S, Seage GR III, Chadwick EG, Study PHAC. Marijuana, Opioid, and Alcohol Use Among Pregnant and Postpartum Individuals Living With HIV in the US. JAMA Network Open. 2021;4(12):e2137162–e. doi: 10.1001/jamanetworkopen.2021.37162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Goodwin RD, Zhu J, Heisler Z, Metz TD, Wyka K, Wu M, Das Eiden R. Cannabis use during pregnancy in the United States: The role of depression. Drug Alcohol Depend. 2020;210:107881. Epub 20200303. doi: 10.1016/j.drugalcdep.2020.107881. [DOI] [PubMed] [Google Scholar]
  • 39.Young-Wolff KC, Sarovar V, Tucker LY, Goler NC, Alexeeff SE, Ridout KK, Avalos LA. Association of Depression, Anxiety, and Trauma With Cannabis Use During Pregnancy. JAMA Netw Open. 2020;3(2):e1921333. Epub 20200205. doi: 10.1001/jamanetworkopen.2019.21333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Sarmiento C, Lau C. Diagnostic and statistical manual of mental disorders: DSM-5. The Wiley Encyclopedia of Personality and Individual Differences: Personality Processes and Individual Differences. 2020:125–9. [Google Scholar]
  • 41.Hasin DS, Saha TD, Kerridge BT, Goldstein RB, Chou SP, Zhang H, Jung J, Pickering RP, Ruan WJ, Smith SM, Huang B, Grant BF. Prevalence of Marijuana Use Disorders in the United States Between 2001–2002 and 2012–2013. JAMA Psychiatry. 2015;72(12):1235–42. doi: 10.1001/jamapsychiatry.2015.1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wicken C, Walia A, Solhjoo S, Mark K. Prevalence of cannabis use disorder among pregnant people who test positive for cannabis at time of delivery. AJOG Glob Rep. 2022;2(4):100095. Epub 20220901. doi: 10.1016/j.xagr.2022.100095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Khan SS, Secades-Villa R, Okuda M, Wang S, Pérez-Fuentes G, Kerridge BT, Blanco C. Gender differences in cannabis use disorders: results from the National Epidemiologic Survey of Alcohol and Related Conditions. Drug Alcohol Depend. 2013;130(1–3):101–8. Epub 20121122. doi: 10.1016/j.drugalcdep.2012.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bandoli G, Delker E, Schumacher BT, Baer RJ, Kelly AE, Chambers CD. Prenatal cannabis use disorder and infant hospitalization and death in the first year of life. Drug Alcohol Depend. 2023;242:109728. Epub 20221209. doi: 10.1016/j.drugalcdep.2022.109728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Shi Y, Zhu B, Liang D. The associations between prenatal cannabis use disorder and neonatal outcomes. Addiction. 2021;116(11):3069–79. Epub 20210422. doi: 10.1111/add.15467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Young-Wolff KC, Adams SR, Brown QL, Weisner C, Ansley D, Goler N, Skelton KR, Satre DD, Foti TR, Conway A. Modes of cannabis administration in the year prior to conception among patients in Northern California. Addict Behav Rep. 2022;15:100416. Epub 20220222. doi: 10.1016/j.abrep.2022.100416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Young-Wolff KC, Adams SR, Wi S, Weisner C, Conway A. Routes of cannabis administration among females in the year before and during pregnancy: Results from a pilot project. Addict Behav. 2020;100:106125. Epub 20190909. doi: 10.1016/j.addbeh.2019.106125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Cannabis Clinicians Colorado. Medical marijuana new patient success guide. Available from: http://coscc.org/wp-content/uploads/2016/07/CCC-General-CannabisAsMedicine-2017DRAFT.pdf Last accessed February 25, 2023.
  • 49.Thompson R, DeJong K, Lo J. Marijuana Use in Pregnancy: A Review. Obstet Gynecol Surv. 2019;74(7):415–28. doi: 10.1097/ogx.0000000000000685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Baker T, Datta P, Rewers-Felkins K, Thompson H, Kallem RR, Hale TW. Transfer of Inhaled Cannabis Into Human Breast Milk. Obstet Gynecol. 2018;131(5):783–8. doi: 10.1097/aog.0000000000002575. [DOI] [PubMed] [Google Scholar]
  • 51.Alves P, Amaral C, Teixeira N, Correia-da-Silva G. Cannabidiol disrupts apoptosis, autophagy and invasion processes of placental trophoblasts. Arch Toxicol. 2021;95(10):3393–406. Epub 20210724. doi: 10.1007/s00204-021-03122-z. [DOI] [PubMed] [Google Scholar]
  • 52.Iffland K, Grotenhermen F. An Update on Safety and Side Effects of Cannabidiol: A Review of Clinical Data and Relevant Animal Studies. Cannabis Cannabinoid Res. 2017;2(1):139–54. Epub 20170601. doi: 10.1089/can.2016.0034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sarrafpour S, Urits I, Powell J, Nguyen D, Callan J, Orhurhu V, Simopoulos T, Viswanath O, Kaye AD, Kaye RJ, Cornett EM, Yazdi C. Considerations and Implications of Cannabidiol Use During Pregnancy. Curr Pain Headache Rep. 2020;24(7):38. Epub 20200610. doi: 10.1007/s11916-020-00872-w. [DOI] [PubMed] [Google Scholar]
  • 54.Ochiai W, Kitaoka S, Kawamura T, Hatogai J, Harada S, Iizuka M, Ariumi M, Takano S, Nagai T, Sasatsu M, Sugiyama K. Maternal and Fetal Pharmacokinetic Analysis of Cannabidiol during Pregnancy in Mice. Drug Metab Dispos. 2021;49(4):337–43. Epub 20210202. doi: 10.1124/dmd.120.000270. [DOI] [PubMed] [Google Scholar]
  • 55.Tagen M, Klumpers LE. Review of delta-8-tetrahydrocannabinol (Δ(8) -THC): Comparative pharmacology with Δ(9) -THC. Br J Pharmacol. 2022;179(15):3915–33. Epub 20220601. doi: 10.1111/bph.15865. [DOI] [PubMed] [Google Scholar]
  • 56.Kruger JS, Kruger DJ. Delta-8-THC: Delta-9-THC’s nicer younger sibling? J Cannabis Res. 2022;4(1):4. Epub 20220104. doi: 10.1186/s42238-021-00115-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Sun X, Dey SK. Synthetic cannabinoids and potential reproductive consequences. Life Sci. 2014;97(1):72–7. Epub 20130701. doi: 10.1016/j.lfs.2013.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Jaques SC, Kingsbury A, Henshcke P, Chomchai C, Clews S, Falconer J, Abdel-Latif ME, Feller JM, Oei JL. Cannabis, the pregnant woman and her child: weeding out the myths. J Perinatol. 2014;34(6):417–24. Epub 20140123. doi: 10.1038/jp.2013.180. [DOI] [PubMed] [Google Scholar]
  • 59.Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 1990;346(6284):561–4. doi: 10.1038/346561a0. [DOI] [PubMed] [Google Scholar]
  • 60.National Institute on Drug Abuse; National Institutes of Health: U.S. Department of Health and Human Services. The science of drug abuse and addiction. Marijuana 2017:1–55. [Google Scholar]
  • 61.Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. Br J Pharmacol. 2008;153(2):199–215. Epub 20070910. doi: 10.1038/sj.bjp.0707442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Joya X, Pujadas M, Falcón M, Civit E, Garcia-Algar O, Vall O, Pichini S, Luna A, de la Torre R. Gas chromatography-mass spectrometry assay for the simultaneous quantification of drugs of abuse in human placenta at 12th week of gestation. Forensic Sci Int. 2010;196(1–3):38–42. Epub 20100106. doi: 10.1016/j.forsciint.2009.12.044. [DOI] [PubMed] [Google Scholar]
  • 63.Blackard C, Tennes K. Human placental transfer of cannabinoids. N Engl J Med. 1984;311(12):797. doi: 10.1056/nejm198409203111213. [DOI] [PubMed] [Google Scholar]
  • 64.Marchetti D, Di Masi G, Cittadini F, La Monaca G, De Giovanni N. Placenta as alternative specimen to detect in utero cannabis exposure: A systematic review of the literature. Reprod Toxicol. 2017;73:250–8. Epub 20170620. doi: 10.1016/j.reprotox.2017.06.049. [DOI] [PubMed] [Google Scholar]
  • 65.Hutchings DE, Martin BR, Gamagaris Z, Miller N, Fico T. Plasma concentrations of delta-9-tetrahydrocannabinol in dams and fetuses following acute or multiple prenatal dosing in rats. Life Sci. 1989;44(11):697–701. doi: 10.1016/0024-3205(89)90380-9. [DOI] [PubMed] [Google Scholar]
  • 66.Bailey JR, Cunny HC, Paule MG, Slikker W Jr. Fetal disposition of delta 9-tetrahydrocannabinol (THC) during late pregnancy in the rhesus monkey. Toxicol Appl Pharmacol. 1987;90(2):315–21. doi: 10.1016/0041-008x(87)90338-3. [DOI] [PubMed] [Google Scholar]
  • 67.Abrams RM, Cook CE, Davis KH, Niederreither K, Jaeger MJ, Szeto HH. Plasma delta-9-tetrahydrocannabinol in pregnant sheep and fetus after inhalation of smoke from a marijuana cigarette. Alcohol Drug Res. 1985;6(5):361–9. [PubMed] [Google Scholar]
  • 68.Martin BR, Dewey WL, Harris LS, Beckner JS. 3H-delta9-tetrahydrocannabinol distribution in pregnant dogs and their fetuses. Res Commun Chem Pathol Pharmacol. 1977;17(3):457–70. [PubMed] [Google Scholar]
  • 69.Agurell S, Nilsson IM, Ohlsson A, Sandberg F. Elimination of tritium-labelled cannabinols in the rat with special reference to the development of tests for the identification of cannabis users. Biochem Pharmacol. 1969;18(5):1195–201. doi: 10.1016/0006-2952(69)90122-1. [DOI] [PubMed] [Google Scholar]
  • 70.Agurell S, Nilsson IM, Ohlsson A, Sandberg F. On the metabolism of tritium-labelled 1-tetrahydrocannabinol in the rabbit. Biochem Pharmacol. 1970;19(4):1333–9. doi: 10.1016/0006-2952(70)90048-1. [DOI] [PubMed] [Google Scholar]
  • 71.Christensen HD, Freudenthal RI, Gidley JT, Rosenfeld R, Boegli G, Testino L, Brine DR, Pitt CG, Wall ME. Activity of delta8- and delta9-tetrahydrocannabinol and related compounds in the mouse. Science. 1971;172(3979):165–7. doi: 10.1126/science.172.3979.165. [DOI] [PubMed] [Google Scholar]
  • 72.Freudenthal RI, Martin J, Wall ME. Distribution of delta 9-tetrahydrocannabinol in the mouse. Br J Pharmacol. 1972;44(2):244–9. doi: 10.1111/j.1476-5381.1972.tb07260.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Harbison RD, Mantilla-Plata B. Prenatal toxicity, maternal distribution and placental transfer of tetrahydrocannabinol. J Pharmacol Exp Ther. 1972;180(2):446–53. [PubMed] [Google Scholar]
  • 74.Klausner HA, Dingell JV. The metabolism and excretion of delta 9-tetrahydrocannabinol in the rat. Life Sci I. 1971;10(1):49–59. doi: 10.1016/0024-3205(71)90245-1. [DOI] [PubMed] [Google Scholar]
  • 75.Layman JM, Milton AS. Distribution of tritium labelled delta 1-tetrahydrocannabinol in the rat brain following intraperitoneal administration. Br J Pharmacol. 1971;42(2):308–10. doi: 10.1111/j.1476-5381.1971.tb07114.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Vardaris RM, Weisz DJ, Fazel A, Rawitch AB. Chronic administration of delta-9-tetrahydrocannabinol to pregnant rats: studies of pup behavior and placental transfer. Pharmacol Biochem Behav. 1976;4(3):249–54. doi: 10.1016/0091-3057(76)90236-7. [DOI] [PubMed] [Google Scholar]
  • 77.Parikh N, Kramer WG, Khurana V, Cognata Smith C, Vetticaden S. Bioavailability study of dronabinol oral solution versus dronabinol capsules in healthy volunteers. Clin Pharmacol. 2016;8:155–62. Epub 20161012. doi: 10.2147/cpaa.S115679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Wall ME, Sadler BM, Brine D, Taylor H, Perez-Reyes M. Metabolism, disposition, and kinetics of delta-9-tetrahydrocannabinol in men and women. Clin Pharmacol Ther. 1983;34(3):352–63. doi: 10.1038/clpt.1983.179. [DOI] [PubMed] [Google Scholar]
  • 79.Ohlsson A, Lindgren JE, Wahlen A, Agurell S, Hollister LE, Gillespie HK. Plasma delta-9 tetrahydrocannabinol concentrations and clinical effects after oral and intravenous administration and smoking. Clin Pharmacol Ther. 1980;28(3):409–16. doi: 10.1038/clpt.1980.181. [DOI] [PubMed] [Google Scholar]
  • 80.Huestis MA. Human cannabinoid pharmacokinetics. Chem Biodivers. 2007;4(8):1770–804. doi: 10.1002/cbdv.200790152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Tortoriello G, Morris CV, Alpar A, Fuzik J, Shirran SL, Calvigioni D, Keimpema E, Botting CH, Reinecke K, Herdegen T, Courtney M, Hurd YL, Harkany T. Miswiring the brain: Δ9-tetrahydrocannabinol disrupts cortical development by inducing an SCG10/stathmin-2 degradation pathway. Embo j. 2014;33(7):668–85. Epub 20140127. doi: 10.1002/embj.201386035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Boskovic R, Klein J, Woodland C, Karaskov T, Koren G. The role of the placenta in variability of fetal exposure to cocaine and cannabinoids: a twin study. Can J Physiol Pharmacol. 2001;79(11):942–5. [PubMed] [Google Scholar]
  • 83.Metz TD, Borgelt LM. Marijuana Use in Pregnancy and While Breastfeeding. Obstet Gynecol. 2018;132(5):1198–210. doi: 10.1097/aog.0000000000002878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Vanstone M, Taneja S, Popoola A, Panday J, Greyson D, Lennox R, McDonald SD. Reasons for cannabis use during pregnancy and lactation: a qualitative study. Cmaj. 2021;193(50):E1906–e14. doi: 10.1503/cmaj.211236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Terplan M Commentary on Ondersma et al. (2012): beyond the quest for the perfect test-drug use screening in pregnancy. Addiction. 2012;107(12):2107–8. doi: 10.1111/j.1360-0443.2012.04094.x. [DOI] [PubMed] [Google Scholar]
  • 86.Wright TE, Terplan M, Ondersma SJ, Boyce C, Yonkers K, Chang G, Creanga AA. The role of screening, brief intervention, and referral to treatment in the perinatal period. Am J Obstet Gynecol. 2016;215(5):539–47. Epub 20160701. doi: 10.1016/j.ajog.2016.06.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.El Marroun H, Tiemeier H, Jaddoe VW, Hofman A, Verhulst FC, van den Brink W, Huizink AC. Agreement between maternal cannabis use during pregnancy according to self-report and urinalysis in a population-based cohort: the Generation R Study. Eur Addict Res. 2011;17(1):37–43. Epub 20101026. doi: 10.1159/000320550. [DOI] [PubMed] [Google Scholar]
  • 88.Lozano J, García-Algar O, Marchei E, Vall O, Monleon T, Giovannandrea RD, Pichini S. Prevalence of gestational exposure to cannabis in a Mediterranean city by meconium analysis. Acta Paediatr. 2007;96(12):1734–7. Epub 20071022. doi: 10.1111/j.1651-2227.2007.00535.x. [DOI] [PubMed] [Google Scholar]
  • 89.Chang JC, Holland CL, Tarr JA, Rubio D, Rodriguez KL, Kraemer KL, Day N, Arnold RM. Perinatal Illicit Drug and Marijuana Use. Am J Health Promot. 2017;31(1):35–42. Epub 20161117. doi: 10.4278/ajhp.141215-QUAL-625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ondersma SJ, Svikis DS, LeBreton JM, Streiner DL, Grekin ER, Lam PK, Connors-Burge V. Development and preliminary validation of an indirect screener for drug use in the perinatal period. Addiction. 2012;107(12):2099–106. Epub 20120810. doi: 10.1111/j.1360-0443.2012.03982.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Smid MC, Metz TD, McMillin GA, Mele L, Casey BM, Reddy UM, Wapner RJ, Thorp JM, Saade GR, Tita ATN, Miller ES, Rouse DJ, Sibai B, Costantine MM, Mercer BM, Caritis SN. Prenatal Nicotine or Cannabis Exposure and Offspring Neurobehavioral Outcomes. Obstet Gynecol. 2022;139(1):21–30. doi: 10.1097/aog.0000000000004632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Winchester ML, Shahiri P, Boevers-Solverson E, Hartmann A, Ross M, Fitzgerald S, Parrish M. Racial and Ethnic Differences in Urine Drug Screening on Labor and Delivery. Matern Child Health J. 2022;26(1):124–30. Epub 20220106. doi: 10.1007/s10995-021-03258-5. [DOI] [PubMed] [Google Scholar]
  • 93.Yonkers KA, Howell HB, Gotman N, Rounsaville BJ. Self-report of illicit substance use versus urine toxicology results from at-risk pregnant women. J Subst Use. 2011;16(5):372–89. doi: 10.3109/14659891003721133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Metz TD, Silver RM, McMillin GA, Allshouse AA, Jensen TL, Mansfield C, Heard K, Kinney GL, Wymore E, Binswanger IA. Prenatal Marijuana Use by Self-Report and Umbilical Cord Sampling in a State With Marijuana Legalization. Obstet Gynecol. 2019;133(1):98–104. doi: 10.1097/aog.0000000000003028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Colby JM. Comparison of umbilical cord tissue and meconium for the confirmation of in utero drug exposure. Clin Biochem. 2017;50(13–14):784–90. Epub 20170311. doi: 10.1016/j.clinbiochem.2017.03.006. [DOI] [PubMed] [Google Scholar]
  • 96.Martin CE, Longinaker N, Mark K, Chisolm MS, Terplan M. Recent trends in treatment admissions for marijuana use during pregnancy. J Addict Med. 2015;9(2):99–104. doi: 10.1097/adm.0000000000000095. [DOI] [PubMed] [Google Scholar]
  • 97.Tzilos Wernette G, Plegue M, Kahler CW, Sen A, Zlotnick C. A Pilot Randomized Controlled Trial of a Computer-Delivered Brief Intervention for Substance Use and Risky Sex During Pregnancy. J Womens Health (Larchmt). 2018;27(1):83–92. Epub 20171005. doi: 10.1089/jwh.2017.6408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Sherman BJ, McRae-Clark AL. Treatment of Cannabis Use Disorder: Current Science and Future Outlook. Pharmacotherapy. 2016;36(5):511–35. doi: 10.1002/phar.1747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Stone R Pregnant women and substance use: fear, stigma, and barriers to care. Health & Justice. 2015;3(1):2. doi: 10.1186/s40352-015-0015-5. [DOI] [Google Scholar]
  • 100.Bahji A, Stephenson C, Tyo R, Hawken ER, Seitz DP. Prevalence of Cannabis Withdrawal Symptoms Among People With Regular or Dependent Use of Cannabinoids: A Systematic Review and Meta-analysis. JAMA Netw Open. 2020;3(4):e202370. Epub 20200401. doi: 10.1001/jamanetworkopen.2020.2370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Connor JP, Stjepanović D, Budney AJ, Le Foll B, Hall WD. Clinical management of cannabis withdrawal. Addiction. 2022;117(7):2075–95. Epub 20220110. doi: 10.1111/add.15743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Levin FR, Mariani JJ, Brooks DJ, Pavlicova M, Cheng W, Nunes EV. Dronabinol for the treatment of cannabis dependence: a randomized, double-blind, placebo-controlled trial. Drug Alcohol Depend. 2011;116(1–3):142–50. Epub 20110218. doi: 10.1016/j.drugalcdep.2010.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Mariani JJ, Pavlicova M, Jean Choi C, Basaraba C, Carpenter KM, Mahony AL, Brooks DJ, Bisaga A, Naqvi N, Nunes EV, Levin FR. Quetiapine treatment for cannabis use disorder. Drug Alcohol Depend. 2021;218:108366. Epub 20201020. doi: 10.1016/j.drugalcdep.2020.108366. [DOI] [PubMed] [Google Scholar]
  • 104.Trigo JM, Lagzdins D, Rehm J, Selby P, Gamaleddin I, Fischer B, Barnes AJ, Huestis MA, Le Foll B. Effects of fixed or self-titrated dosages of Sativex on cannabis withdrawal and cravings. Drug Alcohol Depend. 2016;161:298–306. Epub 20160223. doi: 10.1016/j.drugalcdep.2016.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Mason BJ, Crean R, Goodell V, Light JM, Quello S, Shadan F, Buffkins K, Kyle M, Adusumalli M, Begovic A, Rao S. A proof-of-concept randomized controlled study of gabapentin: effects on cannabis use, withdrawal and executive function deficits in cannabis-dependent adults. Neuropsychopharmacology. 2012;37(7):1689–98. Epub 20120229. doi: 10.1038/npp.2012.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Budney AJ, Vandrey RG, Hughes JR, Moore BA, Bahrenburg B. Oral delta-9-tetrahydrocannabinol suppresses cannabis withdrawal symptoms. Drug Alcohol Depend. 2007;86(1):22–9. Epub 20060612. doi: 10.1016/j.drugalcdep.2006.04.014. [DOI] [PubMed] [Google Scholar]
  • 107.Hernandez M, Birnbach DJ, Van Zundert AA. Anesthetic management of the illicit-substance-using patient. Curr Opin Anaesthesiol. 2005;18(3):315–24. doi: 10.1097/01.aco.0000169241.21680.0b. [DOI] [PubMed] [Google Scholar]
  • 108.Ladha KS, McLaren-Blades A, Goel A, Buys MJ, Farquhar-Smith P, Haroutounian S, Kotteeswaran Y, Kwofie K, Le Foll B, Lightfoot NJ, Loiselle J, Mace H, Nicholls J, Regev A, Rosseland LA, Shanthanna H, Sinha A, Sutherland A, Tanguay R, Yafai S, Glenny M, Choi P, Ladak SSJ, Leroux TS, Kawpeng I, Samman B, Singh R, Clarke H. Perioperative Pain and Addiction Interdisciplinary Network (PAIN): consensus recommendations for perioperative management of cannabis and cannabinoid-based medicine users by a modified Delphi process. Br J Anaesth. 2021;126(1):304–18. Epub 20201029. doi: 10.1016/j.bja.2020.09.026. [DOI] [PubMed] [Google Scholar]
  • 109.Singh S, Filion KB, Abenhaim HA, Eisenberg MJ. Prevalence and outcomes of prenatal recreational cannabis use in high-income countries: a scoping review. Bjog. 2020;127(1):8–16. Epub 20191024. doi: 10.1111/1471-0528.15946. [DOI] [PubMed] [Google Scholar]
  • 110.Warshak CR, Regan J, Moore B, Magner K, Kritzer S, Van Hook J. Association between marijuana use and adverse obstetrical and neonatal outcomes. J Perinatol. 2015;35(12):991–5. Epub 20150924. doi: 10.1038/jp.2015.120. [DOI] [PubMed] [Google Scholar]
  • 111.Metz TD, Allshouse AA, Hogue CJ, Goldenberg RL, Dudley DJ, Varner MW, Conway DL, Saade GR, Silver RM. Maternal marijuana use, adverse pregnancy outcomes, and neonatal morbidity. Am J Obstet Gynecol. 2017;217(4):478.e1–.e8. Epub 20170531. doi: 10.1016/j.ajog.2017.05.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Chabarria KC, Racusin DA, Antony KM, Kahr M, Suter MA, Mastrobattista JM, Aagaard KM. Marijuana use and its effects in pregnancy. Am J Obstet Gynecol. 2016;215(4):506.e1–7. Epub 20160602. doi: 10.1016/j.ajog.2016.05.044. [DOI] [PubMed] [Google Scholar]
  • 113.Bandoli G, Jelliffe-Pawlowski L, Schumacher B, Baer RJ, Felder JN, Fuchs JD, Oltman SP, Steurer MA, Marienfeld C. Cannabis-related diagnosis in pregnancy and adverse maternal and infant outcomes. Drug Alcohol Depend. 2021;225:108757. Epub 20210521. doi: 10.1016/j.drugalcdep.2021.108757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Prewitt KC, Hayer S, Garg B, Benson AE, Hedges MA, Caughey AB, Lo JO. Impact of Prenatal Cannabis Use Disorder on Perinatal Outcomes. J Addict Med. 2022. Epub 20221214. doi: 10.1097/adm.0000000000001123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Petrangelo A, Czuzoj-Shulman N, Balayla J, Abenhaim HA. Cannabis Abuse or Dependence During Pregnancy: A Population-Based Cohort Study on 12 Million Births. J Obstet Gynaecol Can. 2019;41(5):623–30. Epub 20181115. doi: 10.1016/j.jogc.2018.09.009. [DOI] [PubMed] [Google Scholar]
  • 116.Sun X, Dey SK. Endocannabinoid signaling in female reproduction. ACS Chemical Neuroscience. 2012;3(5):349–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Park B, Gibbons H, Mitchell M, Glassa M. Identification of the CB1 cannabinoid receptor and fatty acid amide hydrolase (FAAH) in the human placenta. Placenta. 2003;24(5):473–8. [DOI] [PubMed] [Google Scholar]
  • 118.Yao J, He Q, Liu M, Chang X, Wu J, Duan T, Wang K. Effects of Δ (9)-tetrahydrocannabinol (THC) on human amniotic epithelial cell proliferation and migration. Toxicology. 2018;394:19–26. [DOI] [PubMed] [Google Scholar]
  • 119.Maia J, Almada M, Midão L, Fonseca BM, Braga J, Gonçalves D, Teixeira N, Correia-da-Silva G. The Cannabinoid Delta-9-tetrahydrocannabinol Disrupts Estrogen Signaling in Human Placenta. Toxicological Sciences. 2020;177(2):420–30. [DOI] [PubMed] [Google Scholar]
  • 120.Carter RC, Wainwright H, Molteno CD, Georgieff MK, Dodge NC, Warton F, Meintjes EM, Jacobson JL, Jacobson SW. Alcohol, Methamphetamine, and Marijuana Exposure Have Distinct Effects on the Human Placenta. Alcohol Clin Exp Res. 2016;40(4):753–64. doi: 10.1111/acer.13022. [DOI] [PubMed] [Google Scholar]
  • 121.El Marroun H, Tiemeier H, Steegers EA, Roos-Hesselink JW, Jaddoe VW, Hofman A, Verhulst FC, van den Brink W, Huizink AC. A prospective study on intrauterine cannabis exposure and fetal blood flow. Early Hum Dev. 2010;86(4):231–6. Epub 20100506. doi: 10.1016/j.earlhumdev.2010.03.006. [DOI] [PubMed] [Google Scholar]
  • 122.Chang X, Bian Y, He Q, Yao J, Zhu J, Wu J, Wang K, Duan T. Suppression of STAT3 signaling by Δ9-tetrahydrocannabinol (THC) induces trophoblast dysfunction. Cellular Physiology and Biochemistry. 2017;42(2):537–50. [DOI] [PubMed] [Google Scholar]
  • 123.Roberts VHJ, Schabel MC, Boniface ER, D’Mello RJ, Morgan TK, Terrobias JJD, Graham JA, Borgelt LM, Grant KA, Sullivan EL, Lo JO. Chronic prenatal delta-9-tetrahydrocannabinol exposure adversely impacts placental function and development in a rhesus macaque model. Sci Rep. 2022;12(1):20260. Epub 20221124. doi: 10.1038/s41598-022-24401-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Benevenuto SG, Domenico MD, Martins MAG, Costa NS, de Souza ARL, Costa JL, Tavares MF, Dolhnikoff M, Veras MM. Recreational use of marijuana during pregnancy and negative gestational and fetal outcomes: an experimental study in mice. Toxicology. 2017;376:94–101. [DOI] [PubMed] [Google Scholar]
  • 125.Natale BV, Gustin KN, Lee K, Holloway AC, Laviolette SR, Natale DR, Hardy DB. Δ9-tetrahydrocannabinol exposure during rat pregnancy leads to symmetrical fetal growth restriction and labyrinth-specific vascular defects in the placenta. Scientific reports. 2020;10(1):1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Costa M, Fonseca B, Marques F, Teixeira N, Correia-da-Silva G. The psychoactive compound of Cannabis sativa, Δ9-tetrahydrocannabinol (THC) inhibits the human trophoblast cell turnover. Toxicology. 2015;334:94–103. [DOI] [PubMed] [Google Scholar]
  • 127.Grzeskowiak LE, Grieger JA, Andraweera P, Knight EJ, Leemaqz S, Poston L, McCowan L, Kenny L, Myers J, Walker JJ. The deleterious effects of cannabis during pregnancy on neonatal outcomes. Medical Journal of Australia. 2020;212(11):519–24. [DOI] [PubMed] [Google Scholar]
  • 128.Nashed MG, Hardy DB, Laviolette SR. Prenatal Cannabinoid Exposure: Emerging Evidence of Physiological and Neuropsychiatric Abnormalities. Frontiers in psychiatry. 2021;11:1577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Brummelte S, Mc Glanaghy E, Bonnin A, Oberlander T. Developmental changes in serotonin signaling: Implications for early brain function, behavior and adaptation. Neuroscience. 2017;342:212–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Wu C-S, Jew CP, Lu H-C. Lasting impacts of prenatal cannabis exposure and the role of endogenous cannabinoids in the developing brain. Future neurology. 2011;6(4):459–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Huizink A. Prenatal cannabis exposure and infant outcomes: overview of studies. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2014;52:45–52. [DOI] [PubMed] [Google Scholar]
  • 132.El Marroun H, Hudziak JJ, Tiemeier H, Creemers H, Steegers EA, Jaddoe VW, Hofman A, Verhulst FC, van den Brink W, Huizink AC. Intrauterine cannabis exposure leads to more aggressive behavior and attention problems in 18-month-old girls. Drug and alcohol dependence. 2011;118(2–3):470–4. [DOI] [PubMed] [Google Scholar]
  • 133.Sundström E, Kölare S, Souverbie F, Samuelsson EB, Pschera H, Lunell NO, Seiger A. Neurochemical differentiation of human bulbospinal monoaminergic neurons during the first trimester. Brain Res Dev Brain Res. 1993;75(1):1–12. doi: 10.1016/0165-3806(93)90059-j. [DOI] [PubMed] [Google Scholar]
  • 134.Whitaker-Azmitia PM, Druse M, Walker P, Lauder JM. Serotonin as a developmental signal. Behav Brain Res. 1996;73(1–2):19–29. doi: 10.1016/0166-4328(96)00071-x. [DOI] [PubMed] [Google Scholar]
  • 135.Kenney SP, Kekuda R, Prasad PD, Leibach FH, Devoe LD, Ganapathy V. Cannabinoid receptors and their role in the regulation of the serotonin transporter in human placenta. Am J Obstet Gynecol. 1999;181(2):491–7. doi: 10.1016/s0002-9378(99)70583-1. [DOI] [PubMed] [Google Scholar]
  • 136.Hadjikhani N. Serotonin, pregnancy and increased autism prevalence: is there a link? Med Hypotheses. 2010;74(5):880–3. Epub 20091216. doi: 10.1016/j.mehy.2009.11.015. [DOI] [PubMed] [Google Scholar]
  • 137.Bonnin A, Levitt P. Placental source for 5-HT that tunes fetal brain development. Neuropsychopharmacology. 2012;37(1):299–300. doi: 10.1038/npp.2011.194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Lester BM, Conradt E, Marsit CJ. Are epigenetic changes in the intrauterine environment related to newborn neurobehavior? Epigenomics. 2014;6(2):175–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Roth TL, David Sweatt J. Annual Research Review: Epigenetic mechanisms and environmental shaping of the brain during sensitive periods of development. Journal of Child Psychology and Psychiatry. 2011;52(4):398–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Smith A, Kaufman F, Sandy MS, Cardenas A. Cannabis exposure during critical windows of development: Epigenetic and molecular pathways implicated in neuropsychiatric disease. Current environmental health reports. 2020;7:325–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Forrester MB, Merz RD. Risk of selected birth defects with prenatal illicit drug use, Hawaii, 1986–2002. J Toxicol Environ Health A. 2007;70(1):7–18. [DOI] [PubMed] [Google Scholar]
  • 142.Williams LJ, Correa A, Rasmussen S. Maternal lifestyle factors and risk for ventricular septal defects. Birth Defects Res Part A Clin Mol Teratol. 2004;70(2):59–64. [DOI] [PubMed] [Google Scholar]
  • 143.Shaw GM, Velie EM, Morland KB. Parental recreational drug use and risk for neural tube defects. Am J Epidemiol. 1996;144(12):1155–60. [DOI] [PubMed] [Google Scholar]
  • 144.Cornelius MD, Taylor PM, Geva D, Day NL. Prenatal tobacco and marijuana use among adolescents: effects on offspring gestational age, growth, and morphology. Pediatrics. 1995;95(5):738–43. [PubMed] [Google Scholar]
  • 145.van Gelder MMHJ, Donders ART, Devine O, Roeleveld N, Reefhuis J, National Birth Defects Prevention S. Using bayesian models to assess the effects of under-reporting of cannabis use on the association with birth defects, national birth defects prevention study, 1997–2005. Paediatr Perinat Epidemiol. 2014;28(5):424–33. Epub 2014/08/26. doi: 10.1111/ppe.12140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.van Gelder MM, Reefhuis J, Caton AR, Werler MM, Druschel CM, Roeleveld N, National Birth Defects Prevention S. Maternal periconceptional illicit drug use and the risk of congenital malformations. Epidemiology. 2009;20(1):60–6. doi: 10.1097/EDE.0b013e31818e5930. [DOI] [PubMed] [Google Scholar]
  • 147.Gibson GT, Baghurst PA, Colley DP. Maternal alcohol, tobacco and cannabis consumption and the outcome of pregnancy. Aust N Z J Obstet Gynaecol. 1983;23(1):15–9. [DOI] [PubMed] [Google Scholar]
  • 148.Kharbanda EO, Vazquez-Benitez G, Kunin-Batson A, Nordin JD, Olsen A, Romitti PA. Birth and early developmental screening outcomes associated with cannabis exposure during pregnancy. J Perinatol. 2020;40(3):473–80. doi: 10.1038/s41372-019-0576-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Petrangelo A, Czuzoj-Shulman N, Balayla J, Abenhaim HA. Cannabis Abuse or Dependence During Pregnancy: A Population-Based Cohort Study on 12 Million Births. J Obstet Gynaecol Can. 2019;41(5):623–30. doi: 10.1016/j.jogc.2018.09.009. [DOI] [PubMed] [Google Scholar]
  • 150.Forrester MB, Merz RD. Comparison of trends in gastroschisis and prenatal illicit drug use rates. J Toxicol Environ Health A. 2006;69(13):1253–9. [DOI] [PubMed] [Google Scholar]
  • 151.Lam PK, Torfs CP. Interaction between maternal smoking and malnutrition in infant risk of gastroschisis. Birth Defects Res Part A Clin Mol Teratol. 2006;76(3):182–6. [DOI] [PubMed] [Google Scholar]
  • 152.Downing KF, Riehle-Colarusso T, Gilboa SM, Lin AE, Oster ME, Tinker SC, Farr SL, National Birth Defects Prevention S. Potential risk factors for Ebstein anomaly, National Birth Defects Prevention Study, 1997–2011. Cardiol Young. 2019;29(6):819–27. doi: 10.1017/S1047951119000970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Duko B, Dachew BA, Pereira G, Alati R. The effect of prenatal cannabis exposure on offspring preterm birth: a cumulative meta-analysis. Addiction. 2023;118(4):607–19. Epub 20221115. doi: 10.1111/add.16072. [DOI] [PubMed] [Google Scholar]
  • 154.Bailey BA, Wood DL, Shah D. Impact of pregnancy marijuana use on birth outcomes: results from two matched population-based cohorts. J Perinatol. 2020;40(10):1477–82. Epub 20200305. doi: 10.1038/s41372-020-0643-z. [DOI] [PubMed] [Google Scholar]
  • 155.Rodriguez CE, Sheeder J, Allshouse AA, Scott S, Wymore E, Hopfer C, Hermesch A, Metz TD. Marijuana use in young mothers and adverse pregnancy outcomes: a retrospective cohort study. Bjog. 2019;126(12):1491–7. Epub 20190825. doi: 10.1111/1471-0528.15885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Mumford SL, Flannagan KS, Radoc JG, Sjaarda LA, Zolton JR, Metz TD, Plowden TC, Perkins NJ, DeVilbiss EA, Andriessen VC, A CP, Kim K, Yisahak SF, Freeman JR, Alkhalaf Z, Silver RM, Schisterman EF. Cannabis use while trying to conceive: a prospective cohort study evaluating associations with fecundability, live birth and pregnancy loss. Hum Reprod. 2021;36(5):1405–15. doi: 10.1093/humrep/deaa355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Varner MW, Silver RM, Rowland Hogue CJ, Willinger M, Parker CB, Thorsten VR, Goldenberg RL, Saade GR, Dudley DJ, Coustan D, Stoll B, Bukowski R, Koch MA, Conway D, Pinar H, Reddy UM. Association between stillbirth and illicit drug use and smoking during pregnancy. Obstet Gynecol. 2014;123(1):113–25. doi: 10.1097/aog.0000000000000052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Fried PA, Watkinson B, Willan A. Marijuana use during pregnancy and decreased length of gestation. Am J Obstet Gynecol. 1984;150(1):23–7. doi: 10.1016/s0002-9378(84)80103-9. [DOI] [PubMed] [Google Scholar]
  • 159.Day N, Cornelius M, Goldschmidt L, Richardson G, Robles N, Taylor P. The effects of prenatal tobacco and marijuana use on offspring growth from birth through 3 years of age. Neurotoxicol Teratol. 1992;14(6):407–14. doi: 10.1016/0892-0362(92)90051-b. [DOI] [PubMed] [Google Scholar]
  • 160.El Marroun H, Tiemeier H, Steegers EA, Jaddoe VW, Hofman A, Verhulst FC, van den Brink W, Huizink AC. Intrauterine cannabis exposure affects fetal growth trajectories: the Generation R Study. J Am Acad Child Adolesc Psychiatry. 2009;48(12):1173–81. doi: 10.1097/CHI.0b013e3181bfa8ee. [DOI] [PubMed] [Google Scholar]
  • 161.Richardson GA, Day NL, Goldschmidt L. Prenatal alcohol, marijuana, and tobacco use: infant mental and motor development. Neurotoxicol Teratol. 1995;17(4):479–87. doi: 10.1016/0892-0362(95)00006-d. [DOI] [PubMed] [Google Scholar]
  • 162.Kharbanda EO, Vazquez-Benitez G, Kunin-Batson A, Nordin JD, Olsen A, Romitti PA. Birth and early developmental screening outcomes associated with cannabis exposure during pregnancy. J Perinatol. 2020;40(3):473–80. Epub 20200107. doi: 10.1038/s41372-019-0576-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Day NL, Richardson GA, Goldschmidt L, Robles N, Taylor PM, Stoffer DS, Cornelius MD, Geva D. Effect of prenatal marijuana exposure on the cognitive development of offspring at age three. Neurotoxicol Teratol. 1994;16(2):169–75. doi: 10.1016/0892-0362(94)90114-7. [DOI] [PubMed] [Google Scholar]
  • 164.Goldschmidt L, Richardson GA, Willford J, Day NL. Prenatal marijuana exposure and intelligence test performance at age 6. J Am Acad Child Adolesc Psychiatry. 2008;47(3):254–63. doi: 10.1097/CHI.0b013e318160b3f0. [DOI] [PubMed] [Google Scholar]
  • 165.Goldschmidt L, Richardson GA, Cornelius MD, Day NL. Prenatal marijuana and alcohol exposure and academic achievement at age 10. Neurotoxicol Teratol. 2004;26(4):521–32. doi: 10.1016/j.ntt.2004.04.003. [DOI] [PubMed] [Google Scholar]
  • 166.Goldschmidt L, Richardson GA, Willford JA, Severtson SG, Day NL. School achievement in 14-year-old youths prenatally exposed to marijuana. Neurotoxicol Teratol. 2012;34(1):161–7. Epub 20110822. doi: 10.1016/j.ntt.2011.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Goldschmidt L, Day NL, Richardson GA. Effects of prenatal marijuana exposure on child behavior problems at age 10. Neurotoxicol Teratol. 2000;22(3):325–36. doi: 10.1016/s0892-0362(00)00066-0. [DOI] [PubMed] [Google Scholar]
  • 168.Richardson GA, Ryan C, Willford J, Day NL, Goldschmidt L. Prenatal alcohol and marijuana exposure: effects on neuropsychological outcomes at 10 years. Neurotoxicol Teratol. 2002;24(3):309–20. doi: 10.1016/s0892-0362(02)00193-9. [DOI] [PubMed] [Google Scholar]
  • 169.Fried PA. The Ottawa Prenatal Prospective Study (OPPS): methodological issues and findings--it’s easy to throw the baby out with the bath water. Life Sci. 1995;56(23–24):2159–68. doi: 10.1016/0024-3205(95)00203-i. [DOI] [PubMed] [Google Scholar]
  • 170.Fried PA, Watkinson B, Gray R. Differential effects on cognitive functioning in 9- to 12-year olds prenatally exposed to cigarettes and marihuana. Neurotoxicol Teratol. 1998;20(3):293–306. doi: 10.1016/s0892-0362(97)00091-3. [DOI] [PubMed] [Google Scholar]
  • 171.El Marroun H, Bolhuis K, Franken IHA, Jaddoe VWV, Hillegers MH, Lahey BB, Tiemeier H. Preconception and prenatal cannabis use and the risk of behavioural and emotional problems in the offspring; a multi-informant prospective longitudinal study. Int J Epidemiol. 2019;48(1):287–96. doi: 10.1093/ije/dyy186. [DOI] [PubMed] [Google Scholar]
  • 172.El Marroun H, Tiemeier H, Franken IH, Jaddoe VW, van der Lugt A, Verhulst FC, Lahey BB, White T. Prenatal Cannabis and Tobacco Exposure in Relation to Brain Morphology: A Prospective Neuroimaging Study in Young Children. Biol Psychiatry. 2016;79(12):971–9. Epub 20150901. doi: 10.1016/j.biopsych.2015.08.024. [DOI] [PubMed] [Google Scholar]
  • 173.Smith AM, Fried PA, Hogan MJ, Cameron I. Effects of prenatal marijuana on visuospatial working memory: an fMRI study in young adults. Neurotoxicol Teratol. 2006;28(2):286–95. Epub 20060213. doi: 10.1016/j.ntt.2005.12.008. [DOI] [PubMed] [Google Scholar]
  • 174.Corsi DJ, Donelle J, Sucha E, Hawken S, Hsu H, El-Chaâr D, Bisnaire L, Fell D, Wen SW, Walker M. Maternal cannabis use in pregnancy and child neurodevelopmental outcomes. Nat Med. 2020;26(10):1536–40. Epub 20200810. doi: 10.1038/s41591-020-1002-5. [DOI] [PubMed] [Google Scholar]
  • 175.Dahl RE, Scher MS, Williamson DE, Robles N, Day N. A longitudinal study of prenatal marijuana use. Effects on sleep and arousal at age 3 years. Arch Pediatr Adolesc Med. 1995;149(2):145–50. doi: 10.1001/archpedi.1995.02170140027004. [DOI] [PubMed] [Google Scholar]
  • 176.Fine JD, Moreau AL, Karcher NR, Agrawal A, Rogers CE, Barch DM, Bogdan R. Association of Prenatal Cannabis Exposure With Psychosis Proneness Among Children in the Adolescent Brain Cognitive Development (ABCD) Study. JAMA Psychiatry. 2019;76(7):762–4. doi: 10.1001/jamapsychiatry.2019.0076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Paul SE, Hatoum AS, Fine JD, Johnson EC, Hansen I, Karcher NR, Moreau AL, Bondy E, Qu Y, Carter EB, Rogers CE, Agrawal A, Barch DM, Bogdan R. Associations Between Prenatal Cannabis Exposure and Childhood Outcomes: Results From the ABCD Study. JAMA Psychiatry. 2021;78(1):64–76. doi: 10.1001/jamapsychiatry.2020.2902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Baranger DAA, Paul SE, Colbert SMC, Karcher NR, Johnson EC, Hatoum AS, Bogdan R. Association of Mental Health Burden With Prenatal Cannabis Exposure From Childhood to Early Adolescence: Longitudinal Findings From the Adolescent Brain Cognitive Development (ABCD) Study. JAMA Pediatr. 2022;176(12):1261–5. doi: 10.1001/jamapediatrics.2022.3191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Zammit S, Thomas K, Thompson A, Horwood J, Menezes P, Gunnell D, Hollis C, Wolke D, Lewis G, Harrison G. Maternal tobacco, cannabis and alcohol use during pregnancy and risk of adolescent psychotic symptoms in offspring. Br J Psychiatry. 2009;195(4):294–300. doi: 10.1192/bjp.bp.108.062471. [DOI] [PubMed] [Google Scholar]
  • 180.Roncero C, Valriberas-Herrero I, Mezzatesta-Gava M, Villegas JL, Aguilar L, Grau-López L. Cannabis use during pregnancy and its relationship with fetal developmental outcomes and psychiatric disorders. A systematic review. Reprod Health. 2020;17(1):25. Epub 20200217. doi: 10.1186/s12978-020-0880-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Gray KA, Day NL, Leech S, Richardson GA. Prenatal marijuana exposure: effect on child depressive symptoms at ten years of age. Neurotoxicol Teratol. 2005;27(3):439–48. doi: 10.1016/j.ntt.2005.03.010. [DOI] [PubMed] [Google Scholar]
  • 182.Ruisch IH, Dietrich A, Glennon JC, Buitelaar JK, Hoekstra PJ. Maternal substance use during pregnancy and offspring conduct problems: A meta-analysis. Neurosci Biobehav Rev. 2018;84:325–36. Epub 20170825. doi: 10.1016/j.neubiorev.2017.08.014. [DOI] [PubMed] [Google Scholar]
  • 183.DiNieri JA, Wang X, Szutorisz H, Spano SM, Kaur J, Casaccia P, Dow-Edwards D, Hurd YL. Maternal cannabis use alters ventral striatal dopamine D2 gene regulation in the offspring. Biological psychiatry. 2011;70(8):763–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Zahm DS. An integrative neuroanatomical perspective on some subcortical substrates of adaptive responding with emphasis on the nucleus accumbens. Neuroscience & Biobehavioral Reviews. 2000;24(1):85–105. [DOI] [PubMed] [Google Scholar]
  • 185.Zhu Z, Wang G, Ma K, Cui S, Wang J-H. GABAergic neurons in nucleus accumbens are correlated to resilience and vulnerability to chronic stress for major depression. Oncotarget. 2017;8(22):35933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Porath AJ, Fried PA. Effects of prenatal cigarette and marijuana exposure on drug use among offspring. Neurotoxicol Teratol. 2005;27(2):267–77. Epub 20050108. doi: 10.1016/j.ntt.2004.12.003. [DOI] [PubMed] [Google Scholar]
  • 187.Sonon K, Richardson GA, Cornelius J, Kim KH, Day NL. Developmental pathways from prenatal marijuana exposure to Cannabis Use Disorder in young adulthood. Neurotoxicol Teratol. 2016;58:46–52. Epub 20160518. doi: 10.1016/j.ntt.2016.05.004. [DOI] [PubMed] [Google Scholar]
  • 188.Goldschmidt L, Richardson GA, Larkby C, Day NL. Early marijuana initiation: The link between prenatal marijuana exposure, early childhood behavior, and negative adult roles. Neurotoxicol Teratol. 2016;58:40–5. Epub 20160601. doi: 10.1016/j.ntt.2016.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Coy KC, Haight SC, Anstey E, Grant AM, Ruffo N, Ko JY. Postpartum Marijuana Use, Perceptions of Safety, and Breastfeeding Initiation and Duration: An Analysis of PRAMS Data From Seven States, 2017. J Hum Lact. 2021;37(4):803–12. Epub 20210213. doi: 10.1177/0890334421993466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Bertrand KA, Hanan NJ, Honerkamp-Smith G, Best BM, Chambers CD. Marijuana Use by Breastfeeding Mothers and Cannabinoid Concentrations in Breast Milk. Pediatrics. 2018;142(3). doi: 10.1542/peds.2018-1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Crume TL, Juhl AL, Brooks-Russell A, Hall KE, Wymore E, Borgelt LM. Cannabis Use During the Perinatal Period in a State With Legalized Recreational and Medical Marijuana: The Association Between Maternal Characteristics, Breastfeeding Patterns, and Neonatal Outcomes. J Pediatr. 2018;197:90–6. Epub 20180328. doi: 10.1016/j.jpeds.2018.02.005. [DOI] [PubMed] [Google Scholar]
  • 192.Bergeria CL, Heil SH. Surveying Lactation Professionals Regarding Marijuana Use and Breastfeeding. Breastfeed Med. 2015;10(7):377–80. Epub 20150807. doi: 10.1089/bfm.2015.0051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Perez-Reyes M, Wall ME. Presence of delta9-tetrahydrocannabinol in human milk. N Engl J Med. 1982;307(13):819–20. doi: 10.1056/nejm198209233071311. [DOI] [PubMed] [Google Scholar]
  • 194.Astley SJ, Little RE. Maternal marijuana use during lactation and infant development at one year. Neurotoxicol Teratol. 1990;12(2):161–8. doi: 10.1016/0892-0362(90)90129-z. [DOI] [PubMed] [Google Scholar]
  • 195.Tennes K, Avitable N, Blackard C, Boyles C, Hassoun B, Holmes L, Kreye M. Marijuana: prenatal and postnatal exposure in the human. NIDA Res Monogr. 1985;59:48–60. [PubMed] [Google Scholar]

RESOURCES