U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

National Academies of Sciences, Engineering, and Medicine; Health and Medicine Division; Board on Health Sciences Policy; Committee on Developing a Framework to Address Legal, Ethical, Regulatory, and Policy Issues for Research Specific to Pregnant and Lactating Persons; March A, Helman A, Riley MF, editors. Advancing Clinical Research with Pregnant and Lactating Populations: Overcoming Real and Perceived Liability Risks. Washington (DC): National Academies Press (US); 2024 May 24.

Cover of Advancing Clinical Research with Pregnant and Lactating Populations

Advancing Clinical Research with Pregnant and Lactating Populations: Overcoming Real and Perceived Liability Risks.

Show details

3Strategies to Reduce Harm Through Clinical Research

The most straightforward way to mitigate liability is to prevent harm in the first place. While it is impossible to eliminate any possibility of injury during clinical research, and therefore impossible to completely avoid any liability risk, it is possible to limit the likelihood of injury. The regulatory system and required ethics oversight, review, and approval of medical products are designed to protect the rights and welfare of human participants in clinical research and ensure that medical products are generally safe and effective before they go onto the market for general use. A key component of that review and evaluation is ensuring that risk of harm—which encompasses the probability and magnitude of harm—to research participants is minimized to the extent possible. Not only does the regulatory system and ethics oversight aim to reduce the risk of harm to research participants, but it also ensures that the risk of harm is reasonable in relation to the anticipated benefits of participating in clinical research.

In clinical research involving pregnant and lactating women, reducing harm to the fetus and child is of paramount concern to research participants, sponsors, investigators, and institutional review boards (IRBs) (Wada et al., 2018; Wang et al., 2017; Zhao et al., 2018). However, it should be noted that the risk of harm in lactation studies is different than in pregnancy studies. Risk of harm during lactation is much lower to the offspring, as the majority of medications on the market (about 90 percent) are considered safe for breastfeeding because the drug concentration in the milk is low enough to be considered safe for breastfeeding children (Newton and Hale, 2015). As discussed in Chapter 2, liability for pregnant women and lactating women often is conflated. This applies to risk of harm as well, resulting in a dearth of information on dosing, safety, and efficacy of medical products in lactating women.

As summarized in Chapter 1, the desire to avoid harm to pregnant and lactating research participants, as well as their fetuses and children, is a central factor that has resulted in decisions to exclude them from participating in clinical research entirely. However, their exclusion from clinical research can result in harm in the clinical practice setting from inadequate or inappropriate treatment in the absence of evidence from clinical studies. There are two types of potential harm that require mitigation strategies: harm from exclusion from research—which can manifest in multiple ways including lack of treatment options, lack of evidence to make informed decisions, and outdated treatment regimens—and harm from participation in research, which can involve harm to research participants and their fetus or child and harm to the general populations when access to medical products are delayed. This chapter seeks to reduce both types of harms through the conduct of research involving pregnant and lactating women in a way that reduces harm to the participant and their fetus or child.

This chapter is divided into four sections. An introduction to the medical product development pathway is followed by discussions of strategies to reduce harm through the application of Food and Drug Administration (FDA) guidance and regulations, the application of U.S. Department of Health and Human Services (HHS) and FDA protections for human subjects, and through research design. The first section, a review of the medical product development pathway, is meant to provide a necessary and important background for the current system of development, including the studies required to move along this pathway. The second section, reducing harm through FDA guidance and regulations, provides an overview of the current rules and guidance documents applicable to conducting research with pregnant and lactating women and offers suggestions for potential improvements to current FDA guidance and regulations. The third section, reducing harm through HHS and FDA protections for human subjects, describes federal regulations that guide the ethical conduct of human subjects research to protect research participants from potential harms. And lastly, the fourth section examines reducing harm through research design and how different study designs and methodologies could be employed to improve data collection and reduce harm to research participants. Importantly, this chapter provides evidence that although the current drug development pathway and regulations go a long way towards reducing harm, there are opportunities to make them better for pregnant and lactating women.

INTRODUCTION TO THE MEDICAL PRODUCT DEVELOPMENT PATHWAY

As discussed in Chapter 2, thalidomide has shaped the United States’ approach to inclusion of pregnant women in clinical research in meaningful ways. In 1962, in response to the effects of thalidomide, Congress passed the Kefauver-Harris Drug Efficacy Amendments to the Food Drug and Cosmetic (FD&C) Act, which strengthened the licensure system for new drugs, giving FDA authority to refuse approval of any new drug application that did not meet safety, effectiveness, and labeling requirements.

The current medical product development pathway is designed to ensure that risk of harm is minimized for the individuals who participate in clinical research and for the individuals who may use the product once it is on the market (Berlin et al., 2008). Rigorous preclinical and then clinical studies must be conducted prior to product approval to demonstrate that the product has a favorable benefit–risk balance for use in its intended population. Despite these requirements, pregnant and lactating women often must use FDA-approved products on-label without accompanying safety, efficacy, and dosage data tailored to the pregnant and lactating population (Byrne et al., 2020). This section provides a brief overview of the current U.S. medical development pathway, which is a critical precursor to understanding how to improve current systems to safely include pregnant and lactating women in critical research studies.

Preclinical Development Studies

The goal of preclinical development studies is to serve as a bridge between initial laboratory findings that hold promise for a therapeutic target and use of the experimental product in a clinical setting. Preclinical studies include the development of animal models that are predictive of the pharmaceutical agent’s activity, toxicokinetic and nonclinical pharmacokinetic studies, identification of biomarkers that quantify the activity of interest and potential safety parameters of the therapy, establishment of a dose–response relationship for the product, construction of an initial dosing schedule for human pharmacokinetics and pharmacodynamics (PK/PD) studies, and optimization of the dosing regimen, including route of administration. This section explores three broad areas of preclinical studies. This section applies to most medical products in development, although FDA does have product-specific guidance for vaccines for infectious diseases and for oncology products.1

Pharmacokinetic and Pharmacodynamic Studies

Preclinical PK/PD studies anticipate the kinetics (how a drug moves throughout the body) and dynamics (biochemical, physiologic, and molecular effects of the product on the body) to be expected when studies are subsequently conducted in humans. These are single-dose and multidose escalation studies that integrate activities collectively known as ADME into the process:

1.

Absorption of the experimental product following different routes of administration,

2.

Distribution of the experimental product to organ systems,

3.

Metabolic pathways of the experimental agent, and

4.

Excretion mechanisms of the experimental product through organ systems.

Genetic Toxicity and Carcinogenicity Studies

Genetic toxicity studies involve examining the potential for gene mutation in bacteria and assessing the potential for chromosomal damage in mammalian cells or in an vitro assay (FDA, 2006). These results inform the determination of whether the product development process may proceed to human studies.

Lifetime carcinogenicity studies in rodents are intended for experimental agents that are expected to be administered to patients on regular schedules for substantial parts of their lives (FDA, 1996). These studies are often conducted in conjunction with genotoxicity studies, toxicokinetic studies, and mechanistic studies to form a more detailed picture of carcinogenic potential. Results of these studies help to contextualize the eventual formation of the benefit–risk picture. Because low percentages of new molecular entities at this stage of development eventually reach the stage of submission of a New Drug Application (NDA), carcinogenicity studies are generally not conducted until much later in the development cycle.

Developmental and Reproductive Toxicology (DART) Studies

DART studies identify the experimental product’s adverse effects seen in animal species that may portend the types of toxicities that could occur in humans, including evaluation for teratogenicity. The results of these preclinical studies aid in selecting an initial starting dose and a potential dose titration schedule, and the results aid in estimating the probable highest safe dose for human clinical trials, while also initially characterizing potential adverse effects that might occur in humans (ICH, n.d.). DART studies are categorized into three segments according to the stage of development during which the experimental product is administered (Table 3-1). A fourth category (Juvenile) provides data on the experimental product’s potential effect in the pediatric population.

Table Icon

TABLE 3-1

Developmental and Reproductive Toxicology (DART) Studies.

Clinical Development Studies

Prior to the initiation of a clinical study of a new investigational product in humans, the sponsor must submit an Investigational New Drug Application (IND) to FDA (FDA, 2018e). In the application, the clinical investigator includes PK/PD and toxicology data from the preclinical animal studies, manufacturing information, clinical protocols for intended studies to be conducted in humans, data from any prior human research, and information about the investigator. This section provides a description of the phases of drug development and in following sections specifies the FDA guidance and special considerations for research with pregnant and lactating populations.

Phase I Studies

Once the sponsor completes preclinical studies that demonstrate the product is anticipated to be generally safe when used in humans, development can proceed to Phase I studies in human volunteers. The standard approach to Phase I studies begins with a single-dose escalation study (Figure 3-1). Usually, these studies begin by dosing three to five volunteers, at a dose anticipated to have no observable effect, determined during preclinical studies. The amount of the single dose is gradually increased over days or weeks, noting gradual changes in symptoms, physical exams, and laboratory values. This dose escalation schedule continues until a critical value of tolerance is reached, identified as the dose-limiting toxicity. Thereafter, the next lower dose level is identified as the maximum tolerated dose.

FIGURE 3-1. Phase 1: Dose escalation.

FIGURE 3-1

Phase 1: Dose escalation. SOURCE: Adapted from Beninger, 2024.

The range of the dose amounts between those associated with the first onset of observable effects and the maximum tolerated dose is known as the therapeutic window, which serves as the range of doses to be evaluated during Phase II studies. Analysis of the PK/PD assists in narrowing the dose range of interest, setting the dosing interval, helping to estimate the duration of dosing for later studies when appropriate, and proposing the features of the safety profile. Multidose studies generally follow with a separate cohort of volunteers, based on the now available human PK parameters.

Phase II and Phase III Studies

Phase II studies generally begin in volunteers either with, or at risk for, the disease of interest. The goal is dose selection, the process is dose finding, and the numbers generally range from 100 to 300 volunteers (Table 3-2). Depending on whether treatment is anticipated to be time limited (e.g., for treatments in oncology and infectious diseases) or indefinite (e.g., for cardiovascular diseases and diabetes mellitus), the dosing schedule of amount and duration considers the effects on the markers of benefit and the accompanying safety profile. The Phase II studies may include two or more dose amounts and are often conducted with control arms for placebos or active comparators.2

Table Icon

TABLE 3-2

Common Parameters of Studies Conducted DuringClinical Development.

Phase III studies are conducted in thousands of patients with the doses (amount and schedule) expected to be marketed, guided by the results of the Phase II studies. These studies are conducted to collect the quantitative statistical data that will support the commercial sponsor’s New Drug Application (NDA). Phase III is also commonly a time to conduct the increasingly important pharmacogenomic studies (FDA, 2021), as well as expected drug–drug interaction studies.

Product Review and Approval

Once the phased studies are complete, the commercial sponsor then submits an NDA or a Biologics License Application (BLA) to FDA, the approval of which is required before marketing the product is permitted. FDA is the regulatory agency with the statutory authority and the responsibility for determining whether an NDA or BLA provides substantial evidence to support the safety and effectiveness of the therapeutic product under consideration. FDA approval means that the data on the use of the therapeutic product is determined to provide benefits that outweigh its known and potential risks for the intended population (FDA, 2018b, 2022).

Prescription labeling represents what FDA determines to be the relevant experimental evidence that supports the safety and effectiveness of the use of the agent in patients with diseases, conditions, or circumstances indicated for prophylactic use, organized and presented in a standardized format. Labeling for prescription medicines is required for all FDA-approved prescription drugs and biological products and contains a summary of the essential scientific information needed for the safe and effective use of the medicine.3

When a prescription product is approved for use in adults, the product is also approved for use in pregnant and lactating women by default unless there is a clear contraindication or warnings against the product’s use during pregnancy or lactation, which must then be included on the product label. This is because pregnant and lactating women are considered a subpopulation of the adult population and, therefore, absent a contraindication or warnings against the product’s use during pregnancy or lactation, they are not excluded from the approved population when a drug or biological product is approved for use in adults.

REDUCING HARM THROUGH FDA REGULATIONS AND GUIDANCE

FDA has developed several guidance documents relevant to clinical research that includes pregnant or lactating women. “Pregnant Women: Scientific and Ethical Considerations for Inclusion in Clinical Trials” provides considerations for when to include pregnant women in clinical research (FDA, 2018d). “Pharmacokinetics in Pregnancy—Study Design, Data Analysis, and Impact on Dosing and Labeling” details the agency’s recommendations for designing and conducting PK studies in pregnant women (FDA, 2004). “Postapproval Pregnancy Safety Studies” provides guidance on conducting observational studies in pregnant women after a product has received approval from FDA (FDA, 2019b). And “Clinical Lactation Studies: Considerations for Study Design,” offers guidance on conducting studies that evaluate the safety and efficacy of drugs in lactating women (FDA, 2019a). As mentioned in Chapter 2, rules and regulations are legally enforceable, whereas guidance documents are not. However, there is generally little practical difference in how industry sponsors adhere to the two types of regulatory information (Seiguer and Smith, 2005). This section discusses different types of FDA regulatory information relevant to pregnant, potentially pregnant, and lactating women and offers suggestions for how this guidance might be improved to reduce harm for pregnant and lactating women, and their fetuses and children.

FDA Guidance on Preclinical Studies for Pregnant Women and Potentially Pregnant Women

The International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use (ICH) strives to have better regulatory harmonization worldwide to ensure safe and effective development of medications. FDA is a founding member of ICH and plays a major role in the development of ICH guidelines, which FDA then adopts and issues as guidance to industry.

As mentioned above, genetic toxicity studies are critical for examining the potential for a product to cause gene changes in mammalian cells. According to ICH guidelines, all preclinical female reproduction toxicity studies and standard genotoxicity tests should be completed before the inclusion of potentially pregnant women not using highly effective birth control in any clinical trial (ICH, n.d.).

Highly relevant to the participation of women who are pregnant and potentially pregnant is the completion of DART studies. ICH recommends conducting DART studies to characterize the risk of the experimental product and take appropriate precautions during exposure of potentially pregnant women in clinical trials, or to limit the risk by taking precautions to prevent pregnancy during clinical trials (ICH, n.d.). In the United States, embryo-fetal development studies (Segment II) for most products can be deferred until the initiation of Phase III trials, the final phase of clinical research prior to submitting marketing applications if precautions are taken to prevent pregnancy in potentially pregnant women (exceptions exist for oncology products to treat advanced cancer). FDA does not require that pre- and postnatal development studies (Segment III) be submitted until the sponsor seeks approval of the product.

If DART studies were conducted earlier in the product development pathway, it could provide an opportunity to begin clinical phases of research with pregnant women sooner (Sewell et al., 2022). Earlier DART studies could shorten the time that pregnant patients and their health care providers must wait for high-quality evidence generated in later stages of clinical research. Further, earlier completion of DART studies would allow for earlier detection of potential harmful pharmaceutical and biopharmaceutical products for pregnant and lactating women and their offspring. Timely conduct of DART studies could also enable women who become pregnant over the course of a clinical trial to remain in the trial once pregnant, which would provide critical information about the use of the product in early pregnancy.

A critique of conducting DART studies earlier in the clinical development process is that doing so before having defined the dose in the general human population could increase the cost of research and delay timelines for approval of the product (Sewell et al., 2022). Although this is a valid concern, waiting to conduct relevant DART studies can delay later stages of research by requiring sponsors to update protocols according to DART study results. Additionally, DART studies can feasibly be conducted without delaying product approval by beginning to conduct DART studies as soon as the dose to be used in Phase III studies for the general population is determined.

Preclinical studies, in general, may not be highly predictive of human responses. For a study of 108 oncology drugs, animal toxicity did not show a strong predictive correlation with human toxicity; the median positive predictive value was 0.65 and the negative predictive value was 0.50 (Atkins et al., 2020). It is not known how well DART studies predict potential outcomes in humans, nor are DART studies routinely designed to capture specific outcomes that may be relevant to later studies in humans. For example, in utero fetal exposure to products may affect immune system development, higher-order learning and memory, and endocrine functioning, but fetal exposure concentrations of drugs are not typically assessed in DART studies (Sewell et al., 2022); yet, such data are critically important to further demonstrate the safety of a product in pregnant women and can only be captured in specially designed studies. Thus, investigators must be attentive to, and discerning with, the results from DART studies. One potential way to exercise greater care in the conduct of these studies is with the selective use of data safety monitoring boards (DSMBs) during the conduct of Phase I studies (see Box 3-1). While few adverse events may be detected during Phase I studies owing to the small number of enrolled participants, use of a DSMB could rapidly evaluate adverse events that do arise during this early stage of clinical development and halt exposure of the product to additional participants if necessary.

Box Icon

BOX 3-1

Data Safety Monitoring Boards.

FDA Guidance on the Inclusion of Pregnant and Potentially Pregnant Women in Clinical Studies

For sponsors who plan to include pregnant women in clinical trials of their investigational drug, biological product, or medical device, FDA recommends that sponsors be prepared to discuss such plans with the appropriate FDA review division early in the development phase, and such discussions should involve FDA experts in bioethics and maternal health (FDA, 2018c). However, these discussions are sponsor initiated and not required when pregnant women are not included in the clinical trial.

At the time of IND application submission for products to treat conditions that are not specific to pregnancy, there is rarely any information or consideration of the dosing, efficacy, or safety of the investigational therapeutic agent in pregnant women at this stage. However, according to FDA’s draft guidance for drug developers, “Formal Meetings Between the FDA and Sponsors or Applicants of PDUFA Products,” pre-IND and later clinical-stage meetings between FDA and sponsors may include discussion of trial populations as well as design plans (FDA, 2023d). Therefore, these meetings could provide opportunities for sponsors to discuss plans for involving pregnant and lactating women in their clinical development programs.

FDA’s draft guidance, “Pregnant Women: Scientific and Ethical Considerations for Inclusion in Clinical Trials,” published in 2018, provides the most expansive current guidance to industry on how and when to include pregnant women in clinical trials for drugs and biological products. This guidance discusses both the scientific and ethical issues that sponsors should address when considering the inclusion of pregnant women in clinical trials.

This 2018 draft guidance states that pregnant women may be enrolled in clinical trials that involve greater than minimal risk to the fetuses. When a trial offers the potential for direct clinical benefit to the enrolled pregnant women and/or their fetuses, it can be acceptable to expose a fetus to greater than minimal risk. FDA provides examples of when such exposure would be acceptable, which include when a trial offers a needed but otherwise unavailable therapy or when a drug or biological product being studied reduces the risk of acquiring a serious health condition (FDA, 2018c). However, the guidance does not define “greater than minimal risk” clearly or provide many examples, which can cause confusion in interpreting the guidance for sponsors and IRBs.

The 2018 draft guidance also includes information on the timing of enrollment for involving pregnant women in clinical trials. According to the draft guidance, Phase I and Phase II clinical trials in nonpregnant women, including potentially pregnant women, should be completed before enrolling pregnant women into later phases. The guidance also lists situations that might affect the decision of when to enroll pregnant women in a trial, including if there are other approved treatments available, if there are limited therapeutic options, or if safety data are available for a drug that has been studied for other indications or for use in other populations (FDA, 2018c).

However, current guidance does not set clear expectations for when pregnant women should ideally be included in clinical studies, other than what studies must be completed in nonpregnant women first. The lack of clear expectations results in most sponsors never conducting studies with pregnant women. Further, FDA does not provide guidance on conducting clinical studies for conditions specific to pregnancy, particularly when the study is for a new product without human safety data. These studies pose a different set of considerations from products to treat general conditions and warrant a specific discussion in guidance documents, especially for study design.

FDA Guidance on Conducting Clinical Studies with Pregnant Women

PK/PD Studies

FDA’s 2004 final guidance, “Pharmacokinetics in Pregnancy, Study Design, Data Analysis, and Impact on Dosing and Labeling,” provides specific recommendations for designing and conducting PK/PD studies in pregnant women and lays out a framework to stimulate further study and research to assist in rational therapeutics for pregnant patients. FDA acknowledges that (1) pregnant women are “actively excluded” from clinical trials, (2) data in product labels regarding PK and dose adjustments during pregnancy rarely provide information for appropriate prescribing in pregnancy, and (3) there has been a significant amount of pharmacological research conducted to improve the quality and quantity of data available for other altered physiologic states (e.g., patients with renal and hepatic disease) and subpopulations (e.g., pediatric patients). Because of that, FDA has stated that “The need for PK/PD studies in pregnancy is no less than for these populations, nor is the need for the development of therapeutic treatments for pregnant women” (FDA, 2004).

This guidance specifies that pregnant women may be involved in PK studies if the following conditions are met:4

  • Preclinical studies, including studies on pregnant animals, and clinical studies, including studies on nonpregnant women, have been conducted and provide data for assessing potential risk to pregnant women and fetuses (FDA, 2004).
  • The risk to the fetus is not greater than minimal, and the purpose of the research is the development of important biomedical knowledge, which cannot be obtained by any other means (FDA, 2004).

The guidance also recommends that PK studies be conducted if the following occurs:

1.

the drug is known to be prescribed in or used by pregnant women, especially in the second and third trimesters,

2.

for a new drug or indication if there is anticipated or actual use of the drug in pregnancy,

3.

use is expected to be rare but the consequences of the uninformed dosages are great (e.g., for drugs with narrow therapeutic ranges and for cancer chemotherapeutic agents), and

4.

if pregnancy is likely to alter significantly the PK of a drug (e.g., for drugs that are renally excreted) (FDA, 2004).

FDA further states in this guidance:

Although PK studies in pregnancy can be considered in Phase III development programs depending on anticipated use in pregnancy and the results of reproductive toxicity studies, FDA anticipates that most PK studies in pregnant women will occur in the postmarketing period and will be conducted using pregnant women who have already been prescribed the drug as therapy by their own physician. (FDA, 2004)

Given the known physiological changes that occur during pregnancy, it is critical that clinical studies investigate PK/PD during all phases of pregnancy. Despite this, PK/PD studies are often not available for common medications used during pregnancy and rarely are PK/PD studies conducted in all phases of pregnancy (Coppola et al., 2022). It is critical that more PK/PD studies are conducted throughout pregnancy and the postpartum period to better understand appropriate dosing. Therefore, although current FDA guidance recommends PK studies be conducted if the product is going to be used by pregnant women, more concrete time lines for conducting PK/PD studies and additional guidelines on completing studies throughout stages of pregnancy may encourage sponsors to complete more of these studies.

Data Collection

When pregnant women are enrolled in a clinical trial, FDA’s draft guidance, “Pregnant Women: Scientific and Ethical Considerations for Inclusion in Clinical Trials,” provides that data collection elements should include (at a minimum): (1) gestational age at enrollment; (2) gestational timing and duration of drug exposure; and (3) pregnancy outcomes including adverse maternal, fetal, and neonatal events. Further, the draft guidance states while all clinical trials require monitoring, clinical trials that involve pregnant women should include a data monitoring plan that includes members with relevant specialty and perinatal expertise to permit ongoing recognition and evaluation of safety concerns that arise during the course of the trial (FDA, 2018c). However, the guidance provides few details on the content or format of such a plan, including few details on the duration of infant follow-up or infant outcomes or acceptable research designs to collect such information. More detailed guidance on the proper monitoring and follow-up that industry could conduct for research involving pregnant women would help minimize harm for these populations.

Finally, while PK studies can provide information on appropriate dosing and basic safety, they are not capable of determining the efficacy of a medical product or describing the safety of long-term exposure that might be expected from routine clinical use. While it is understandable that FDA would not want to be overly prescriptive in how to evaluate the safety and efficacy of a product in pregnant women given the potential diversity of products and therapeutic targets, additional information that is currently not discussed in FDA guidance to establish a product’s safety profile and efficacy in pregnant women could be helpful for sponsors. For example, information on what data FDA considers sufficient evidence to describe safety in pregnant participants would facilitate this research and could help sponsors develop labeling that is more useful for pregnant patients and their providers.

FDA Guidance Addressing Lactating Women in Clinical Research

Drug exposure and risks during the lactation period are generally lower than pregnancy and depend on whether the child is breastfeeding; the transfer of the drug into human milk; and the absorption, metabolism, and elimination of the drug by the breastfed child (Newton and Hale, 2015). In 2019, FDA updated its draft guidance, “Clinical Lactation Studies: Considerations for Study Design,” which provides recommendations for sponsors conducting pre- or postmarketing clinical lactation studies. The guidance clarifies that although FDA has required lactation studies under Section 505(o)(3) of the FD&C Act under certain circumstances to inform breastfeeding with drug use recommendations included in the “Lactation” subsection of labeling, the guidance states that FDA “is considering additional circumstances in which lactation studies may be required” (FDA, 2019a).

According to FDA guidance, lactating women may participate in clinical lactation studies if they are prescribed a medically necessary drug in the postapproval setting as part of standard clinical care and are allowed to continue breastfeeding while taking the drug. For new investigational drugs, lactating women who are administered an investigational drug for a disorder or condition must discontinue breastfeeding because of the potential or unknown risk to the infant and milk must be discarded for a duration depending on the half-life of the medication. For healthy volunteers who participate in a trial of an investigational drug for research, breastfeeding must be discontinued for the duration of the study.

There are three main types of study designs for clinical lactation studies:

1.

Lactating women (milk only) study: Human milk is collected, and drug concentrations determined. These may be used to estimate drug transfer to the milk to determine whether there are clinically relevant concentrations in human milk, and to evaluate effects of the drug on milk production. Most published studies are in this category.

2.

Lactating women (milk and plasma) study: Milk and plasma are collected from lactating women, and pharmacokinetic data is gathered as well as data regarding any effects on milk production. These are conducted when there may be concern for drug accumulation (i.e., long half-life).

3.

Mother–infant pair: Mother and infant provide blood to determine drug concentrations and pharmacokinetics in the lactating woman and infant as well as drug excretion in human milk. These studies can also include an assessment of the drug effects in the infants. These studies are typically done if there is evidence that there is substantial drug transfer into human milk, placing the infant at risk.

FDA’s clinical lactation studies guidance encourages sponsors to consider conducting clinical lactation studies even when not required, such as

1.

when a drug under review for approval is expected to be used by women of reproductive age,

2.

use of a drug in lactating women becomes evident after approval,

3.

the sponsor is seeking a new indication for an approved drug and provides evidence of use or anticipated use of the drug by lactating women, and

4.

when marketed medications are commonly used by women of reproductive age (FDA, 2019a).

Although the lactation guidance is relatively thorough in outlining research considerations for types of lactation studies and ethical considerations for the participation of lactating women in clinical studies, the guidance falls short in many of the same ways the pregnancy guidance does. First, although lactation studies are encouraged, FDA guidance does not set clear expectations or timelines for when these studies should be completed. Further, as with the guidance on pregnancy, FDA guidance on lactation recommends follow-up examination and testing of the breastfed infant in clinical studies of approved medications postmarket, but it does not provide details on expectations for the duration of follow-up and what types of outcomes should be measured. Nor does it provide guidance on the continuum of care from pregnancy to lactation and consideration for breastfeeding if enrolled in a clinical trial while pregnant.

For studies of new investigational products, FDA guidance on lactation dissuades lactating women from continuing to breastfeed, which could have detrimental health effects on the children receiving human milk, could affect their milk supply, maternal–infant bonding, and could be considered a harm in and of itself. While the mother may express milk to maintain her milk supply, particularly for a short-term study, the discontinuation of breastfeeding presents a major barrier for studies of medications to treat conditions not specific to breastfeeding as well as breastfeeding conditions, such as mastitis, low milk supply, or breastfeeding-associated pain. Discontinuation of breastfeeding and the resultant increased risk of infectious disease and other morbidities may outweigh the potential (and likely low) risk of the investigational product. Human milk transfers beneficial bacteria, immune cells, and nutrients to the nursing child that improve the immunological health of the child (Camacho-Morales et al., 2021).

Patients seeking treatment for breastfeeding conditions typically want to continue breastfeeding. In most cases, requirements to discontinue breastfeeding are not scientifically justified. One common method for estimating the risk of drug exposure to the child is the estimate of the relative infant dose (RID), which standardizes the exposure by weight. A RID of less than 10 percent is generally considered safe for use in lactation and safe for breastfeeding the healthy child (Newton and Hale, 2015). Currently, approximately 90 percent of marketed drugs have a RID in the “acceptable” range considered safe for breastfeeding (less than 10 percent). Therefore, these restrictions present sometimes overly burdensome barriers for participants in these studies and create challenges with recruitment for these studies, as suspending breastfeeding to participate in a study may dissuade many potential research participants from enrolling.

Further, when the study protocol calls for the cessation of breastfeeding, there is no requirement to provide participants with any alternatives or remuneration (i.e., pumping supplies, free formula for supplementation) to offset the monetary and potential emotional costs of discontinuation requirements. This may place an undue burden and greater barriers to entry for low-income populations. Lastly, FDA guidance on lactation suggests sponsors consider conducting an assessment on the effect of the product on milk production, which may include both volume and composition. However, the guidance provides few details on how this assessment might be conducted or how this assessment may take into account the different stages of breastfeeding.

FDA Rule on Labeling for Pregnancy and Lactation

In 2014, FDA amended its regulations through the finalization of its Pregnancy and Lactation Labeling Rule (the PLLR) (initially proposed in 2008), which created a consistent format for providing information about the risks and benefits of prescription drug and biological product use during pregnancy and lactation and by females and males of reproductive potential. For human prescription drug and biological products approved on or after June 30, 2001, the PLLR required that the labeling be revised to include

1.

a summary of the risks of using a drug during pregnancy (Section 8.1 of the labeling) or lactation (Section 8.2 of the labeling), and for females and males of reproductive potential (Section 8.3 of the labeling);

2.

a discussion of the data supporting that summary; and

3.

relevant information to provide health care providers and patients with the best available evidence to make informed decisions regarding the use of medications during pregnancy and lactation.

Under the PLLR, both the pregnancy and lactation sections of a drug or biological product’s labeling must include summaries of the pertinent available evidence providing information about the safety and use of the drug in pregnancy and lactation. Information on pregnancy exposure registries, if available, including how to enroll or to obtain more information, must also be included. A risk summary is also required that provides, as a narrative summary, a statement of background risk if there are data demonstrating that the product is systemically absorbed or if there are data on the product’s presence in human milk. This includes a separate summary based on human data, animal data, and pharmacologic data that describes the risk of adverse developmental outcomes, if such data are available (FDA, 2018b).

Additionally, the PLLR requires statements acknowledging when data on any of the labeling requirements are not available or do not establish the presence or absence of drug- or vaccine-associated risk.5 Lastly, the PLLR requires the label to be updated to include clinically relevant information as it becomes available to prevent the label from becoming “inaccurate, false, or misleading.”6

Although the PLLR was designed to provide more relevant summary information for health care providers and patients, there still is not much information in product labels about their use during pregnancy or while lactating. A cross-sectional labeling analysis of 290 newly FDA-approved medications from January 2010 to December 2019 indicated that

All products submitted after June 20, 2015, were in compliance with the Pregnancy and Lactation Labeling Rule (PLLR); however, of those submitted between 2010 and 2015, 32.6 percent were not in PLLR format by the designated date of June 30, 2019. Human data on pregnancy and lactation were available in less than 20 percent of new product labeling. (Byrne et al., 2020)

Postmarketing Commitments and Requirements

Phase IV postmarketing studies are commonly required by FDA (2016). These are clinical studies, epidemiologic studies, and registries that focus on specific questions of safety and/or effectiveness for various related conditions of the disease of interest for related demographic populations, such as pediatrics (FDA, 2023c), related diseases, special populations, safety issues, and long-term use. These studies are known as postmarketing commitments or postmarketing requirements. Postmarketing commitments involve preclinical studies or clinical trials that a sponsor agrees to conduct postapproval but are not legally required to be performed (FDA, 2016). Postmarketing requirements, however, are preclinical studies or clinical trials that a sponsor is required to conduct in order to comply with certain laws and/or regulations, or to assess a known serious risk related to the use of the drug, assess signals of serious risk related to the use of a drug, or identify an unexpected serious risk when available data indicate the potential for a serious risk (FDA, 2016). FDA may also impose postmarketing requirements on manufacturers of certain Class II or Class III medical devices that are approved by FDA.

Data for these types of studies can come from institutional electronic medical records, health insurance claim data, and registries. These observational studies may aid in (1) assessing the relative risk of a serious adverse event occurring with the use of a particular drug or biologic; (2) identifying certain risk factors that make the occurrence of a serious adverse event among a particular patient population more likely; and (3) obtaining data over a significant period of time, which may help identify rare serious adverse events, among others (FDA, 2011). Observational studies of pregnancy may aid in informing pregnancy or child outcomes following drug exposure, in comparison to a group that has not been exposed to the drug product (FDA, 2011).

FDA has the authority to require postmarketing studies and clinical trials to (1) “assess a known serious risk related to the use of the drug,” (2) “assess signals of serious risk related to the use of the drug,” and (3) “identify an unexpected serious risk when available data indicate the potential for a serious risk.”7 FDA also has the authority to require postapproval studies or trials if it becomes aware of new safety information (FDA, 2011). However, if human pregnancy or lactation data has not been collected at the time of FDA approval, FDA may not have the information required to determine whether there are potential risks to pregnant and lactating women to evaluate through postmarketing studies. Expanding FDA’s authority to request postmarketing studies could help fill important clinical knowledge gaps on the safety and efficacy of products in pregnant and lactating women, particularly to better identify long-term effects of a product.

Section 505(o)(3)(E)(ii) of the FD&C Act requires a sponsor to “periodically report,” and in any event at least annually, on the status of preclinical studies or clinical trials, regardless of whether or not the sponsor was required to conduct a clinical trial or study as part of a postmarketing requirement or voluntarily chose to do so. A sponsor must report on the preclinical study or clinical trial’s status to comply with this section.8 The status report must include a timetable for the completion of specific target goals, along with a status update of the study or trial (FDA, 2011). Unfortunately, the annual reports do not provide details on the ongoing status of the trial such as enrollment updates, any adverse events reported to FDA, or any information that might assist health care providers make prescribing decisions, although any adverse events that inform the labelling should be reflected in the label under FDA requirements.

In 2019, FDA issued its draft guidance, “Postapproval Pregnancy Safety Studies.” This guidance describes three postapproval approaches to assessing drug safety in pregnant women who have been exposed to a drug or biological product: (1) pharmacovigilance, (2) pregnancy registries, and (3) complementary data sources (FDA, 2019b).9 Based on an approach’s relative strengths and limitations and its potential application to a particular drug or biological product, FDA may recommend or require a particular approach or combination of approaches to be used by a sponsor for a particular drug or biologic product (FDA, 2019b).

Pharmacovigilance

The goal of pharmacovigilance is “to protect patients from unnecessary harm by identifying previously unrecognized drug hazards, elucidating predisposing factors, refuting false safety signals, and quantifying risk in relation to benefit” (Talbot and Nilsson, 1998). Pharmacovigilance takes place throughout the life cycle of the pharmaceutical product, including the entire drug development pathway and postmarketing surveillance.

Pharmacovigilance has three core functions: case management, signal management, and risk–benefit management (Beninger, 2020).

1.

Case management is concerned primarily with the input of adverse event information. Such information is collected throughout all stages of a product’s life cycle, including all phases of product development, and filed into relevant safety databases in a standardized way and reported to pertinent regulatory authorities in a timely way to meet compliance requirements.

2.

Signal management is concerned with querying the safety database to answer internal sponsor questions and external regulatory agency issues in light of newly available safety information. Although signal management can be done prior to product approval, the importance of signal management increases significantly postapproval, with the growth of the patient population exposed to the product.

3.

Benefit–risk management is concerned with maintaining a favorable benefit–risk balance across the range of patient populations and labeled uses through the appropriate use of labeling categories and other related regulatory and administrative mechanisms (Beninger, 2020; FDA, 2023a).

As pointed out in the FDA guidance, individual case safety reports are the most common source of reports of adverse pregnancy outcomes, but they can be challenging to interpret owing to incomplete information or additional risk factors for the adverse event, which might not be addressed in the case report (FDA, 2019b). Further, FDA guidance notes there are limitations to spontaneous marketing reports, including under-reporting, lack of a denominator, and incompleteness of reported information. Therefore, FDA guidance recommends using additional sources to evaluate product safety, such as observational studies.

Pregnancy Registries

Pregnancy registries are a common study design that may be used to collect safety data in the postapproval setting and can help inform decision making among health care providers and their patients (FDA, 2023e). Pregnancy registries involve the prospective enrollment of women who have been exposed to a drug or biologic product and are usually followed through delivery and postpartum to evaluate the effects of exposure on the newborn (FDA, 2019b). Such registries may be led by sponsors, government, or institutions; they can be product specific or cover multiple products; they can also involve multiple institutions and other collaborative stakeholders and include more than one country. Registries are an important and potentially powerful safety tool because of their ability to prospectively capture detailed patient data over a long period of time (FDA, 2019b). Because of difficulties in enrollment and retention, however, pregnancy registry data often may not provide sufficient statistical power to assess the safety of drug and biological products during pregnancy (FDA, 2019b).

A large portion of the draft guidance “Postapproval Pregnancy Safety Studies” discusses recommendations for the design and implementation of pregnancy registries. Pregnant women who have been exposed to a drug or biological product may volunteer to participate in a registry during their pregnancy and be followed through delivery (FDA, 2019b). Because a pregnancy registry follows a pregnant woman over the course of their pregnancy and following the birth of their newborn, it may allow assessment of “maternal, obstetrical, fetal, and infant outcomes, including pregnancies that do not result in a live birth” (FDA, 2019b). Although the guidance points to a number of strengths in using pregnancy registries, it highlights some limitations for such registries: analyses may result in insufficient statistical power in detecting associations for rare pregnancy outcomes, registries may not address more specific or rare congenital malformations or congenital anomalies, there may be significant challenges to recruitment and retention, and the data from a registry alone may not be able to adequately assess the safety of a drug or biological product taken during pregnancy (FDA, 2019b).

FDA also provides guidance on the potential duration of a pregnancy registry. It recommends that pregnancy registries collect data until there is sufficient information gathered to meet the registry’s scientific objectives; conversely, if the registry is not able to collect sufficient information to meet its objectives, consideration should be given to discontinuing the registry (FDA, 2019b).

FDA may also require that a lactation study be added to a pregnancy registry to capture potential drug exposure data during breastfeeding (FDA, 2019b). Such lactation data are gathered to assess the safety of drugs and biological products that women may take while breastfeeding, which may or may not have been taken while pregnant (FDA, 2019b). While there is a human milk research biorepository that evaluates the transfer and effects of medication in human milk (Mommy’s Milk, 2024), there appear to be no existing lactation-specific registries.

Complementary Database Studies

FDA guidance on postmarketing studies also discusses complementary studies that may be conducted alongside pregnancy registries to address the “specific effects” of a drug or biological product during pregnancy (FDA, 2019b). These studies may be retrospective in their design and use secondary data sources, such as electronic health records, population-based surveillance, and national registries or registers.

For studies that use electronic health care data, FDA provides recommendations for identifying pregnancies in health care records and acknowledges the challenges of identifying pregnancies that do not result in a live birth. The ability to link the records of the pregnant woman to the offspring is critical to evaluating fetal outcomes related to in utero exposure (Johnson et al., 2013). These linkages can be developed through a number of records, including birth certificates, health record identifiers, and congenital malformation surveillance registries. The guidance also describes methods for estimating gestational age to understand the window during which a fetus would have been exposed to a medical product used by a pregnant woman.

Another form of complementary studies is the case-control study, which can be useful for obtaining additional information or long-term follow-up once a safety signal has been identified. While the guidance notes that these studies can be affected by recall bias from self-reported outcomes, case-control studies have the benefit of being able to capture detailed exposure and outcome assessments through follow-up interviews with the pregnant individual and to collect biospecimens. FDA guidance outlines the necessary considerations and the existing challenges for selecting and validating cases and matched controls to be included in the analysis. FDA goes on to note that it is important for cases and controls to be from the same disease population whenever possible to facilitate comparisons.

In addition, FDA has made a number of commitments in the Prescription Drug User Fee Act (PDUFA) VII Commitment Letter that focuses on pregnancy postmarketing requirements. As described in Box 3-2, these initiatives will result in updated guidance for sponsors conducting postmarketing studies with pregnant women.

Box Icon

BOX 3-2

FDA’s PDUFA VII Commitments.

REDUCING HARM THROUGH FEDERAL PROTECTIONS FOR HUMAN SUBJECTS

The Federal Policy for the Protection of Human Subjects outlines basic provisions for the oversight, ethics review, and approval of research with human participants. In 1991, it was revised and codified by 15 federal departments and agencies and became known as the “Common Rule.” Research funded or conducted by HHS is subject to additional regulatory protections, including provisions specific to the conduct of research involving pregnant women. HHS regulations for the protection of the rights and welfare of human participants in research are codified in title 45 of the Code of Federal Regulations, part 46, including Subparts A through E. Subpart A is the codification of the Common Rule. Subparts B, C, and D provide rules for specific subpopulations in research funded or conducted by HHS. Subpart B provides additional protections for pregnant women, human fetuses, and neonates; Subpart C provides additional protections for incarcerated populations; Subpart D provides additional protections for children; and Subpart E covers registration of IRBs with HHS (HHS, 2022a).

Under the requirements of Subpart B, pregnant women or fetuses may be involved in research if all of the following conditions are met:

  • Preclinical studies, including studies on pregnant animals, and clinical studies, including studies on nonpregnant women, have been conducted and provide data for assessing potential risks to pregnant women and fetuses.
  • The risk to the fetus is caused solely by interventions or procedures that hold out the prospect of direct benefit for the woman or the fetus; or, if there is no such prospect of benefit, the risk to the fetus is not greater than minimal and the purpose of the research is the development of important biomedical knowledge that cannot be obtained by any other means.
  • Any risk is the least possible for achieving the objectives of the research.
  • If the research holds out the prospect of direct benefit to the pregnant woman, the prospect of a direct benefit both to the pregnant woman and the fetus, or no prospect of benefit for the woman nor the fetus when risk to the fetus is not greater than minimal and the purpose of the research is the development of important biomedical knowledge that cannot be obtained by any other means, then informed consent of the mother is required.
  • If the research holds the prospect of direct benefit solely to the fetus, then the consent of the pregnant woman and the father is required. The father’s consent need not be obtained if he is unable to consent because of unavailability, incompetence, temporary incapacity, or the pregnancy resulted from rape or incest.
  • Each consenting individual is fully informed regarding the reasonably foreseeable effect of the research on the fetus or neonate.
  • For children who are pregnant, assent and permission are obtained in accord with the provisions of Subpart D.
  • No inducements, monetary or otherwise, will be offered to terminate a pregnancy.
  • Individuals engaged in the research will have no part in any decisions as to the timing, method, or procedures used to terminate a pregnancy.
  • Individuals engaged in the research will have no part in determining the viability of a neonate.

Subpart B is an important factor for the conduct of research involving pregnant women and is discussed in greater detail in Chapter 4. HHS regulations for human subject protections do not specifically address considerations for lactating women, nor do they clarify whether Subpart D, additional protections for children, apply when a child is exposed to the milk of a lactating woman participating in clinical research.

Despite this regulatory ambiguity, the Secretary’s Advisory Committee on Human Research Protections (SACHRP) released recommendations in 2022 that address the protection of nonsubjects from research-related harms (HHS, 2022c). SACHRP acknowledges that IRBs vary in whether breastfeeding children of lactating research participants are themselves considered research participants—a determination that would require the application of Subpart D, which pertains to research with children. Even in circumstances when the IRB does not consider breastfeeding children of lactating research participants to themselves be research participants, SACHRP recommends that IRBs consider the risks to such children as nonsubjects who may be at risk because of their direct physical contact with the research participant.

FDA has not adopted the HHS regulations but has its own regulations for the protection of human subjects and institutional review boards.10,11 While FDA’s human subject regulations are not identical to those from HHS, they are similar (FDA, 2018a). Additionally, FDA released a proposed rule in 2022 that would harmonize certain sections of FDA’s regulations on the protection of human subjects and IRBs with HHS regulations.12 FDA’s regulations for human subject protections and IRBs do not have specific considerations for pregnancy, other than to note that IRBs are to implement additional safeguards for clinical studies that include pregnant women, and that IRBs might consider including among its membership individuals with relevant expertise if they regularly review protocols that include pregnant women.

FDA regulations for the protection of human subjects do include considerations specific for children (Subpart D), which FDA notes do apply when an infant is exposed to the milk of an individual enrolled in a clinical lactation study (FDA, n.d.a). However, in some contexts, IRBs may determine that collection of outcome data for the breastfeeding children of a lactating research participant does not constitute research, and that under either HHS or FDA definitions,13,14 FDA and HHS regulations specific for children therefore do not apply (HHS, 2022c). In such cases, SACHRP also recommends IRBs consider the risks to breastfeeding children as nonsubjects. For postmarket lactation studies where breastfeeding is permitted, it is important that children are monitored for adverse events and that these data are collected and reported.

Office for Human Research Protections (OHRP)

The HHS Office for Human Research Protections (OHRP) was established in 2000 to oversee HHS efforts to protect human research participants in clinical research and to provide leadership for all federal agencies that support human subjects research under the Common Rule (HHS, 2020). OHRP’s core functions include providing regulatory guidance and clarity, developing educational materials, administering IRB registration programs, and maintaining regulatory oversight of IRBs. OHRP’s guidance documents are not legally binding but help to inform and guide IRBs. OHRP has several guidance documents addressing populations mentioned in HHS regulations for the protection of the rights and welfare of human participants in research, including children and prisoners, but it has not issued any guidance on pregnant and lactating women as research subjects.

OHRP could develop guidance focused on pregnant and lactating women, including guidance to IRBs on the interpretation of Subpart B for clinical research including pregnant women and the applicability of Subpart D for clinical research including lactating women in which the child is exposed to human milk. Various research institutions have developed guidance and standard operating procedures specific to clinical research including pregnant women (CU Denver, 2022; MCW, 2023; Purdue, 2019; University of Utah, n.d.; UW, 2021, 2023). A study at the University of Washington found that such materials facilitated research that includes pregnant women at the institution (Mastroianni et al., 2020). OHRP guidance applicable to the inclusion of pregnant and lactating women in clinical research could help IRBs provide feedback to protocols and ultimately approve more protocols for human subject research involving pregnant and lactating women.

SACHRP was created in 2003 to “provide expert advice and recommendations to the Secretary [of Health and Human Services], through the Assistant Secretary for Health, on issues and topics pertaining to or associated with the protection of human research subjects” (HHS, 2022b). SACHRP is composed of appointed experts and heads of various HHS agencies. As a committee, it provides advice on improving protections for research participants. SACHRP occasionally establishes subcommittees that are formed of experts on special topics of interest to the committee (HHS, 2016). For example, in the early 2000s, SACHRP created a subcommittee to provide advice on pediatric research “to help ensure that children who participate in research are neither underprotected nor overprotected” (HHS, 2016). To date, there is no record that SACHRP has provided advice on the inclusion of pregnant and lactating women in clinical trials. It does have the authority to establish a subcommittee focused on research with pregnant and lactating women and to request recommendations for HHS to ensure that pregnant and lactating women included in research are similarly neither underprotected nor overprotected.

Institutional Review Boards

Federal regulations require that institutions engaged in clinical research involving human participants must use an IRB (HHS, 2018). IRBs are tasked with determining whether the research protocols before them are ethically justifiable and with ensuring that the researchers involved are not bound to certain interests that might pose a conflict with the ethical conduct of the research (Grady, 2015). Importantly, IRBs also serve a critical role in interpreting federal and state laws and regulations relevant to the protection of human subjects from exploitation and undue risk of harm. As a result, IRB members must be familiar with the Common Rule, FDA, HHS, and other agency and department regulations on human subject protections, as well as OHRP guidance. Given that IRBs are responsible for promoting a favorable balance of risks and benefits to research participants and that there is regulatory ambiguity for the protection of pregnant and lactating participants (van der Zande et al., 2017), clearer guidance from OHRP could help IRBs uphold their duties to minimize harm to pregnant and lactating participants.

For an institution to receive federal support for research involving human subjects, it must register its IRB with OHRP and renew its registration every 3 years to ensure compliance with federal regulations. IRBs that review HHS-supported research must apply the Common Rule and other HHS regulations for the protection of human subjects to that research, including those addressing the inclusion of pregnant research participants. In addition, IRBs that review FDA-regulated research must apply FDA regulations for the protection of human subjects to that research, which are similar but not identical to the Common Rule.

In ensuring compliance with federal regulations, IRBs aim to ensure that the research is conducted ethically, which includes minimizing harm to research participants and balancing risk with commensurate benefit to the research participants or the broader population through the creation of generalizable knowledge. It also ensures that informed consent is adequate and that there is equitable selection of research participants.

REDUCING HARM THROUGH RESEARCH DESIGN

Postmarket observational studies are informative, but they delay the generation of safety data for pregnant and lactating women until the product is already being broadly used by the public, thus amplifying the potential for harm. To move beyond a reliance on such data, consideration of how to reduce harm to research participants through research design is important (Huybrechts et al., 2019). Such research can be done safely and ethically, but as noted earlier in the chapter, FDA draft guidance relevant to clinical research in pregnant and lactating women provides few details on the appropriate design of these studies. The design and methods of a study are a crucial element of reducing harm to research participants (IOM, 2003). Research on human immunodeficiency virus and acquired immunodeficiency syndrome (HIV/AIDS) is one area that has had success in conducting clinical research with pregnant and lactating women, as described in Box 3-3. Although there are still improvements to be made to HIV/AIDS research involving pregnant and lactating women, the lessons learned from HIV/AIDS can inform broader research involving pregnant and lactating women.

Box Icon

BOX 3-3

Successes of HIV Research in Pregnant and Lactating Women.

Staging of Clinical Research

An important consideration for designing clinical studies that include pregnant and lactating women is the timing of enrollment of these populations. Regulators and sponsors must strike a balance between including pregnant and lactating women in a sufficiently timely manner as to provide quality evidence on the use of the product in these populations, without rushing their inclusion in the absence of adequate safety information or planning. Moreover, a potential form of harm resulting from the inclusion of pregnant and lactating women in clinical research is that FDA approval of a medical product could be delayed, slowing the general population’s access to the product. Delays in regulatory approval could result from more complex data analyses and identification of false-positive safety signals, which are discussed in more detail in Chapter 5. Considering the most appropriate stages of clinical development to include pregnant women and lactating women (likely different for each population, given the differences in risk) could mitigate the risk of this form of harm. Importantly, the committee emphasizes that it is possible to include pregnant and lactating women in clinical research without expanding the risk of harm from delayed product approval, but careful consideration for which staging approach fits the research needs of the project being conducted is important. Nothing in current FDA regulations or the committee’s recommendations would require clinical studies in pregnant and lactating women to be complete before product approval for the general adult population.

A staggered approach to the enrollment of pregnant and lactating women entails conducting PK/PD clinical studies specific to the population of interest concurrently with the conduct of Phase III trials in the general adult population (Eke et al., 2019). Staggered enrollment would allow for the development of study protocols specific to pregnancy and promote the involvement of researchers with expertise in conducting research in these populations (Baylis and Halperin, 2012). Enrollment of pregnant participants could also be tailored to begin recruitment with individuals in their third trimester of pregnancy, when fetal organogenesis is complete, gradually enrolling participants in earlier stages of pregnancy. Lactating women could be included earlier than pregnant women for investigational products, especially since FDA guidance suggests that women cease breastfeeding for investigational products. Therefore, there is no risk to the baby and lactating women could be enrolled earlier in the trial. However, to avoid breastfeeding cessation, single-dose PK studies, where human milk is collected at time intervals until the drug has been excreted from the body, could be done early to determine potential for infant exposure. Once these single-dose PK studies are completed, if the medication is deemed safe for breastfeeding, as is the case for 90 percent of on-market medications, then breastfeeding could continue.

In embedded enrollment, the strategy would involve enhanced safety and pharmacokinetic evaluations in pregnant and lactating women who enroll in Phase III or late Phase II studies for the general adult population (Eke et al., 2019). According to this approach, pregnant and lactating women would be evaluated as subpopulations of the broader adult population enrolled in the clinical trials. Because pregnant and lactating women would be enrolled in Phase II or Phase III trials, researchers would be able to collect data relevant to these phases of research simultaneously with the basic safety data being collected (Baylis and Halperin, 2012). An embedded approach to enrollment would also facilitate comparisons between pregnant or lactating participants and nonpregnant or nonlactating participants enrolled in the same trial.

A potential concern with this approach is that embedding pregnant and lactating women in the same trials as the general adult population could delay interpretation of overall trial results when there are challenges with the recruitment of pregnant and lactating participants, which could delay drug approval. It may also present risks to pregnant populations, given that there may not be sufficient safety and efficacy data before enrolling them in the trial. Therefore, this approach may be best for lifesaving medications for conditions with no available alternative therapies, but not for non-life-threatening acute and chronic conditions for which alternate therapies are available.

The opportunistic approach to enrollment is a useful way of collecting data on pregnant and lactating women who become pregnant while already enrolled in a trial (Eke et al., 2019). Once the participant is unblinded and reconsented, pharmacokinetic and safety data can be collected for these individuals before therapy is discontinued and potentially throughout all trimesters of pregnancy and postpartum. However, an opportunistic enrollment approach is likely to be slow and may not accomplish recruitment goals while clinical trials are actively being conducted. Such an approach also raises issues of whether the investigators involved in these trials will have sufficient expertise in pregnancy and lactation. Regardless, an opportunistic approach to enrollment can be an effective strategy for collecting data on the use of the product in early stages of pregnancy.

Pragmatic Study Designs

A pragmatic study design, specifically when focusing on pregnant and lactating women as the study population, is pivotal in advancing the understanding of medical products within real-world clinical settings (Eke et al., 2019). At their core, pragmatic study designs aim to evaluate the real-world effect of interventions or strategies that are already part of clinical practice (Patsopoulos, 2011; CU Denver, 2019). Unlike traditional clinical trials with tightly controlled settings, these studies strive to replicate the conditions encountered in routine patient care. This fundamental difference is crucial for gauging how interventions perform when applied to diverse groups of pregnant and lactating women during their specific health care journeys, which can be particularly useful in reducing harm in real-world use of the product (Eke et al., 2019).

One of the defining features of pragmatic trials in this context is the inclusion of a diverse and representative range of participants (Eke et al., 2019). By encompassing such diversity, pragmatic studies can provide valuable insights into how interventions function across different subgroups, considering such factors as age, ethnicity, socioeconomic status, and underlying health conditions, which often influence health care outcomes.

Pragmatic study designs in pregnant and lactating women would be bolstered by encompassing longitudinal data collection. Recognizing that pregnancy and lactation are dynamic processes with evolving needs and experiences, these trials could span extended periods to capture a comprehensive view of the effects of the intervention over time. This longitudinal approach enables researchers to assess not only short-term outcomes but also the sustainability and long-term effect of the intervention. Observing the long-term effects of medical product exposure can facilitate the identification of latent adverse effects. In addition, pragmatic study designs are attuned to minimizing disruptions to routine clinical care (CU, Denver, 2019). This consideration is essential to ensure that both pregnant and lactating patients and health care providers can participate in the study without an undue burden or disruption to their usual health care activities. By integrating seamlessly into clinical practice, pragmatic trials can gather data in a nonintrusive manner that respects the demands of health care delivery.

Opportunistic Studies

Nonrandomized opportunistic studies, a subset of observational research, provide a unique way to study pregnant and lactating women already using specific medical products or interventions (Sheffield et al., 2014). Opportunistic studies thus reduce harm to research participants by only studying the product in individuals who would already be using the product. However, conducting such studies requires careful consideration to ensure ethical and appropriate practices, with informed consent and IRB oversight being ethical cornerstones. Participants must fully understand the research objectives, potential risks, and benefits. An opportunistic study could be a useful study design when it would otherwise be ethically or logistically challenging to conduct an interventional study, such as chemotherapy treatment during pregnancy.

Innovative Methodologies

In striving toward increasing research in pregnant and lactating women earlier in the drug development process, several innovative approaches can be undertaken to conduct and analyze such clinical research safely while lessening the risk of harm to the pregnant woman and developing fetus. Although many of these methods are still evolving, they present novel ways to evaluate the safety and efficacy of medical products before they are used in humans. Further exploration of these methodologies could align with FDA’s Broad Agency Announcement for regulatory science innovation, advancing the ability of FDA regulators to assess clinical studies that use methods to predict exposure to the medical product under evaluation (FDA, 2024a). In fact, FDA recently funded a project studying the use of physiologically based pharmacokinetic models (FDA, n.d.b).

Fetal–Placental Transport

Innovative approaches that facilitate the understanding of the fetal–placental interface—a barrier that limits drug delivery to the developing fetus—are important to predict fetal exposure to medical products in utero. Innovative techniques such as in vitro, ex vivo human cotyledon perfusion, placental drug transport-on-a-chip, and in silico models are increasingly being used to evaluate maternal–fetal medication transfer across the fetal–placental interface prior to human dosing during pregnancy to predict a drug’s safety. These approaches represent promising methods for generating necessary data (Eke et al., 2020). Microengineered models of the human placenta (placental drug transport-on-a-chip models) are currently being used to simulate and explore drug transfer between the maternal–fetal circulation, with the goal of reducing the risk of fetal harm while conducting research safely (Eke et al., 2020). Further exploration of these technologies can enhance their predictive capabilities and have the potential to advance their readiness for use in regulatory decision making.

In Vivo Exposure Assessment Methods

The use of methods that minimize exposure to the medical product being studied, specifically microdosing and short-course (targeted) PK study approaches that have been increasingly employed, could generate early data for pregnant and lactating women (van Nuland et al., 2019). Microdosing involves administering a dose that stimulates a cellular response, but it is a small fraction of the dose that is anticipated to produce any therapeutic effect. Given the success of microdosing strategies in reducing drug development times for pediatric patients, this strategy may also be successfully applied to pregnant individuals (Burt et al., 2016).

Population Pharmacokinetic Modeling

A population PK analysis estimates standard values for PK parameters in a specified population (Avram, 2020). Population PK models are simultaneously capable of explaining interindividual variability, intraindividual variability, and variability attributable to demographic or clinical characteristics. A benefit of a population PK approach is that it involves collecting fewer samples from a larger study population, and it can incorporate data from various sources, even if the data are incomplete (Sheffield et al., 2014). Population PK modeling can be particularly useful for determining dosing in pregnancy after an initial PK study in a small group of research participants has indicated that pregnancy alters the PK of the medical product (Coppola, 2022). However, since it requires a larger number of subjects to complete the study, it is only realistic for common conditions with a large population taking the same medication during pregnancy.

Physiologically Based Pharmacokinetic Modeling

Physiologically based PK (PBPK) models integrate preclinical and clinical data that have been collected to predict drug concentrations in multiple tissues following the administration of a medical product (Avram, 2020; Eke et al., 2021). These mathematical models are capable of producing reliable predictions of drug exposure in pregnant women and can account for the stages of pregnancy and drug–drug interactions, as well as predict any adverse effects of the drug (Eke et al., 2020). PBPK models also hold promise for predicting infant exposure to a drug through human milk. In addition, modeling can help predict the safety of exposing a child to the drug through human milk. However, PBPK models have so far been unable to reliably predict fetal PK parameters for in utero exposure (Eke and Gebreyohannes, 2020).

Human Milk Transport Modeling

Predicting drug exposure in human milk using in vitro approaches could help inform in silico models that simulate newborn exposure to a drug through human milk. Milk-to-plasma ratio of drugs—an important parameter in predicting breastfed drug exposure to a child through human milk—through passive diffusion and the directionality of drug transport have been studied using an in vitro mouse mammary epithelial cell culture model that mimics the secretory and tight-junction properties of human mammary epithelium (Eke et al., 2020).

CONCLUSIONS

Conclusion 3-1: The U.S. drug development regulatory process is designed to minimize harm for research participants and for those who use approved medical products. Pregnant and lactating women, as well as their fetuses and children, are not able to benefit from the harm minimization strategies that are incorporated into medical product development, review, and approval processes. This is because pregnant and lactating women are often excluded from clinical research, which leaves them and their health care providers with insufficient safety and efficacy data to make informed decisions about using medical products.

Conclusion 3-2: Current FDA guidance on clinical studies with pregnant and lactating women describes limited aspects of study design, research time lines, safeguards, and product-specific monitoring. Greater clarity and specificity of regulatory guidance for conducting research with pregnant and lactating women outside of postmarketing commitments would help further reduce and prevent harm for these populations.

Conclusion 3-3: Guidance from the Office for Human Research Protections (OHRP) on involving pregnant women, lactating women, and breastfeeding infants and children in clinical research could help inform institutional review boards on how to safely oversee this research, including how to interpret Subparts B and D of 45 CFR 46 and how to properly assess and reduce risk in clinical research that involves pregnant and lactating women and their offspring.

REFERENCES

Footnotes

1

Appendix C provides a full overview of these product-specific guidances and is available at https://nap​.nationalacademies​.org/catalog/27595.

2

Phases of an Investigation, 21 CFR 312.21.

3

New Drugs, 21 USC § 355, (Jan. 7, 2011).

4

Research Involving Pregnant Women or Fetuses, 45 CFR Subpart B, § 46.204 (Nov. 3, 2001).

5

Specific requirements on content and format of labeling for human prescription drug and biological products described in § 201.56(b), 21 CFR § 201.57(c)(9).

6

Specific requirements on content and format of labeling for human prescription drug and biological products described in § 201.56(b), 21 CFR § 201.57(c)(9).

7

New Drugs, 21 USC § 355(o)(3)(B).

8

New Drugs, 21 USC § 355(o)(3)(E)(ii).

9

The three postapproval approaches can be used alone or in combination with each other.

10

Protection of Human Subjects, 21 CFR 50.

11

Institutional Review Boards, 21 CFR 56.

12

Protection of Human Subjects and Institutional Review Boards, 87 Federal Register 58733, (Sep. 28, 2022).

13

Definitions, 21 CFR 50.3.

14

Definitions, 45 CFR 164.501.

Copyright 2024 by the National Academy of Sciences. All rights reserved.
Bookshelf ID: NBK605205

Views

  • PubReader
  • Print View
  • Cite this Page
  • PDF version of this title (2.0M)

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...