Abstract
Fecal microbiota transplantation (FMT) has rapidly grown in notoriety and popularity worldwide as a treatment for both recurrent and refractory C. difficile infection (CDI), as well as for a myriad of other indications, with varying levels of evidence to justify its use. At present, FMT use in the U.S. has not received marketing approval from the U.S. Food and Drug Administration (FDA), but is permitted under “enforcement discretion” for CDI not responding to standard therapy. Meanwhile, the rising interest in the gut microbiome throughout mainstream media has paved the way for “do-it-yourself” (DIY) adaptations of the procedure. This access and unregulated use, often outside any clinical supervision, has quickly outpaced the medical community’s research and regulatory efforts. While some studies have been able to demonstrate the success of FMT in treating conditions other than CDI—studies on ulcerative colitis have been particularly promising—little is still known about the treatmen’s mechanism of action or long-term side effects. Likewise, screening of donor stool is in its early stages in terms of protocol standardization. In this paper, we explore the regulatory and ethical concerns that arise from the need to balance access to a nascent but promising innovative treatment with the need for research into its efficacy, risk profile, and long-term impact.
Keywords: Gastrointestinal microbiome, C. difficile, Investigational drugs, Expanded access, Bioethics, Regulation
1. Introduction
The human microbiome, or community of microorganisms that reside on and within us, has become a rapidly growing area of study with exciting therapeutic prospects. With the human body hosting a nearly equal ratio of bacterial to human cells [1], understanding our cohabitants and the host-microbiome relationship could elucidate how microbes might cause, treat, modulate, or even prevent disease. One of the most well-known forms of microbiome-based therapy is fecal microbiota transplantation (FMT), in which a fecal suspension from a healthy donor is deposited into the gastrointestinal (GI) tract of a patient via endoscope, nasal tube, or capsule. The efficacy of FMT in combating C. difficile infection (CDI) has resulted in significant improvements in patients who have had either an inadequate response to traditional antibiotic therapies (i.e. refractory disease) or recurring infection after presumed successful treatment (i.e., recurrent disease) [2–5]. Both of these conditions are considered CDIs not responding to standard therapy, although most FMT research is focused on recurrent rather than refractory infections [2–4]. As interest in FMT and evidence of its ability to alter intestinal microbiome composition and/or function grows, enthusiasm about its applications beyond CDI is inevitable. However, FMT has not received marketing approval from the US Food and Drug Administration (FDA) for any indication; rather, the regulatory body has chosen to exercise “enforcement discretion” for FMTs performed for refractory and/or recurrent CDI. FMTs for other indications besides CDIs require that investigators obtain investigational new drug (IND) applications. A unique aspect of FMT, however, is that it can be performed outside of a professional medical facility as a “do-it-yourself’ (DIY) procedure [6]. Clinician reluctance to perform FMT in situations, whether based on lack of safety and efficacy data or due to the burden of undergoing an IND application, may have the unintended consequence of increasing the number of patients performing at-home variants of FMT without the guidance of a medical professional [6–8]. It is amidst this backdrop that we review FMT’s current applications within the United States (U.S.) and its therapeutic potential; its unanswered safety, efficacy, and regulatory concerns; and the ethical challenges raised by the procedure.
2. Current applications and therapeutic potential
Dating as far back as the times of ancient China, preparations of fecal solution were orally administered to treat gastrointestinal diseases [9, 10]. Stanley Falkow, the famed American microbiologist and father of molecular microbial pathogenesis, recalled working with an internist in 1957 to give patients capsules containing a formulation of their own pre-surgical stool in an attempt to combat antibiotic-associated diarrhea [11]. The following year, surgeon Ben Eiseman reported the first official use of fecal enemas in the U.S. as a successful treatment for pseudomembranous enterocolitis [12], now more commonly known as Clostridioides difficile (C. difficile) colitis.
To date, most FMT research has pertained to C. difficile, an anaerobic, spore-forming, Gram-positive bacillus, which is transmitted via the fecal-oral route. Individuals with altered GI microbiomes, often in association with antibiotic use, are particularly vulnerable to this infection [13]. Once established in the gut, C. difficile can cause enteric disease ranging from mild diarrhea to a life-threatening infection. CDI is furthermore a common cause of nosocomial infection [14–16]. Risk of disease recurrence is high, and patients with a history of 2 or more CDIs have a 65% risk of further episodes despite antibiotic therapy [17].
Despite the lack of any large, randomized control studies (akin to typical Phase 3 clinical trials needed for FDA approval of a treatment), multiple smaller-scale studies have shown FMT to be an effective alternative to standard antibiotics for patients with recurrent CDI [2,18–21]. In a randomized control trial (RCT) for subjects with recurrent CDI for whom standard antibiotic therapy had failed, vancomycin treatment followed by up to 2 FMT procedures led to disease resolution for at least 10 weeks in 98% of subjects compared to 31% of those treated with vancomycin alone [2]. Another double-blind RCT compared donor stool to autologous stool (i.e. inert comparator) for recurrent CDI, finding 90% resolution after donor stool—though, interestingly, 62% of the autologous treatment group were also cured [4]. Given mounting evidence pointing to FMT as a durable treatment for recurrent CDI, with typically only mild short-term adverse effects such as abdominal cramping [3,19], clinicians may be more willing to suggest FMT to treat CDI, even for pediatric patients [22].
Accordingly, academic studies of microbially-based therapies have grown rapidly. Published clinical trials using the term “fecal microbiota transplantation” first appeared in the PubMed database in 2014. Since then, 87 additional FMT-related clinical trials have been published: 78 within the last 5 years and 21 in 2019 alone. Non-CDI indications for which FMT has been considered span as widely as obesity, allergies, neurological and behavioral disorders, and multiple bowel and immune-mediated disorders [8,23–33].
3. Public perception
The rise of FMT comes at a time when public access to medical information is relatively unfiltered, of varying levels of scientific accuracy, and widely disseminated via the internet and social media. According to a 2017 survey by the Pew Research Center [34], a majority of respondents reported that they “do their own research in addition to seeking advice from a doctor or other health care provider” when making decisions about treatment for a serious health problem. Furthermore, about half of the respondents reported having tried some form of alternative medicine—for example, herbal remedies, acupuncture or chiropractic treatment, or energy therapies.
While reports of FMT success are understandably compelling, patient (and sometimes provider) perceptions are saturated with the idea of FMT as natural, safe, and somehow intangibly separate from conventional medicine [[6–9]]. The gut microbiome’s role in health and disease has captured mainstream interest, enjoying no small media popularity, with a subsequent boom of an entire industry focused on probiotics, prebiotics, medicinal foods, and other formulas promising to restore and/or improve one's microbiome [35].
In 2017, Park et al. [36] found that while only 12% of surveyed subjects initially knew of FMT as a therapeutic option for CDI, once informed, 77% would undergo the procedure if medically indicated. Studies since then have seen increasing proportions of both previous knowledge of and personal interest in the therapy [31–33]. Yet this favorable disposition to FMT, particularly when based on such sources as social media or personal anecdotes, may unintentionally serve to distract the public from long-term implications of the procedure, such as untoward or unintended changes in the gut microbiome [8,35,37].
Thus, increased research into FMT, its potential applications, long-term impact, and mechanism of action is both warranted and timely. Furthermore, the need for thoroughly vetted regulations and guidelines will have to be balanced with the reality that donor stool will always be obtainable to individuals willing to engage in DIY FMT, limiting the effectiveness of attempted “gatekeeping” on the part of either clinicians or regulators.
4. Unanswered questions
For all its utility in the treatment of recurrent CDI, there remain significant unanswered questions regarding FMT, including whether it works for other indications and its long-term physiological effects. With emerging evidence that gut microbes are able to dictate the induction of Th17 cells, which play a role in mucosal defense and the pathogenesis of autoimmune disease [38], the mechanisms by which gut microorganisms can impact health and disease are still largely unknown. It is furthermore debated whether the induction of an unedited donor sample is required for successful FMT [9] or whether a select consortium of bacteria, bacterial spores, and/or the degree of donor microbiome engraftment are the true determinants or modulators of FMT outcomes [39–42]. Similarly, advances in fecal preparation methods, including automated washing of fecal suspensions, have been shown to reduce the load of viruses and inflammatory cytokines which may be associated with FMT risks [43,44]. Microbial transfer technologies are growing in sophistication, with newer products tailoring the product delivered beyond donor stool. For example, the probiotic strain Escherichia coli Nissile has recently been engineered to act as targeted immunotherapy for treating certain cancers and for the congenital disorder phenylketonuria [45,46].
We also do not currently understand how to select for an ideal FMT recipient or how other underlying conditions might impact response [41, 42,47]. Similarly, little is known about what qualifies as an effective or safe donor, beyond the general idea of good health and screening for a select subset of transmissible pathogens. Finally, it must be acknowledged that questions regarding donor and recipient selection, mechanism of action, and best practices may be indication-specific.
4.1. Long-term risks
While information on long-term effects of FMT in humans is sparse, several animal models have shown that FMT can transfer disease phenotypes such as obesity and metabolic disorders [48–54]. Other animal studies demonstrate the impact of other gut microbiome alterations—such as antibiotic exposure or cecal content transfer—on disease phenotype, such as increasing the risk for colitis [50,55,56]. Gut bacteria have also been implicated in the risk of heart disease and colon cancer [53,57,58]. One well-publicized case study documented a patient who developed new-onset obesity after receiving stool from a healthy but overweight donor [59]. This transferability of obesity with, to-date, no biochemical marker for which to screen donor stool, raises concerns about what phenotypes may escape detection and subsequently be transmitted via FMT.
While the risk of acquiring conditions that do not manifest for many years may be largely irrelevant for an elderly population, FMT is also currently used in young adults and children. Such use is of concern because of the scarcity of data on potential long-term physiological, metabolic, and immunologic effects of FMT. Studies have already demonstrated that the gut microbiome in early life is both strongly susceptible to perturbations [60] and also a possible driving force in immune development [61]. Thus, a child undergoing FMT may be risking microbiome alterations that could influence the risk of disease in adulthood. In cases where no other therapy is effective, this may be a tradeoff that clinicians and patients, or their guardians, are willing to accept. However, there are increasing requests for FMT as a first-line therapy or for non-CDI indications [6,8]. For these indications, use of FMT is unjustified outside of a clinical trial, given the limited supporting data and the availability of other, better-researched treatment alternatives.
Additional work is needed to examine the potential of FMT to contribute to multifactorial diseases such as diabetes or cardiovascular disease, in which disease pathogenesis may take place over decades rather than the comparatively limited timescale of currently available FMT studies. In addition, given the high prevalence of these diseases, efforts to isolate a possible connection with FMT in a clinical context will require careful study design and data collection and will likely require a multicenter effort to enroll sufficient subjects; such a task will take significant time, funding, and collaboration to complete.
4.2. What constitutes thorough screening of donor stool
While there is no universally validated and adopted screening protocol for stool donors, most centers that conduct FMT require donors to undergo blood testing for HIV, hepatitis A, B and C, and stool testing for salmonella, parasites, and C. difficile, among other pathogenic organisms [62,63]. For one large public stool bank, only 8.5% of asymptomatic potential donor samples were ultimately accepted, often due to the incidental detection of possible gastrointestinal pathogens [64].
In addition, in 2019, there were two well-publicized cases of multi-drug resistant organism (MDRO) infections attributed to FMT, one of which led to the patienťs death [65]. These cases led the FDA to revise its guidance [66]—continuing the policy of enforcement discretion which allows the use of FMT for CDI indications, but mandating additional screening for MDROs. While these cases highlight the long-suspected potential for severe adverse effects related to FMT, they also highlight the tenuous access patients have to acceptable FMT product, particularly as some of the required screenings—such as for fecal extended spectrum beta-lactamase (ESBL)-producing organisms—do not have well-established clinical laboratory protocols, even at large academic medical centers.
Current screening protocols may be insufficient, however, and do not account for the possibility of gut microbiome perturbation through a mechanism other than a known infectious agent. In this respect, FMT today resembles the nascent stage of blood transfusions in the 1940s and 1950s, before testing for such blood-borne diseases as human immunodeficiency virus (HIV), human T-cell leukemia virus (HTLV), and hepatitis B and C was possible [67]. The inadvertent transmission of a harmful agent via a biological treatment was poignantly demonstrated in the transmission of HIV via contaminated Factor VIII used by patients with hemophilia [68]. Thus, FMT donor stool screening and quality control should reflect the gravity of these potential risks.
Furthermore, while medical institutions do screen donor stool prior to transplantation, DIY FMT may not involve any screening at all, or at best rely on unreliable claims of laboratory testing. In an appeal for donations to pay for FMT to be done by a medical provider at a hospital with standardized donor testing, a patient reported that she had “tried FMT from home in the past, but that was a nightmare as the only donor that met the criteria (and that I was paying money to) lied to me about their test results … [and] … had parasites” [69]. Such anecdotes cannot quantify the scale of the problem of insufficiently screened donor stool being used in DYI FMT attempts, but they do paint a picture of this very real concern.
4.3. Demand for Applications Beyond C. difficile.
A further question raised by FMT is whether its success in treating CDI can translate into success for other indications. Studies to determine FMT efficacy for a range of indications are in progress, but at this point, there are insufficient clinical or preclinical data to justify FMT in non-CDI contexts outside of clinical trials. Nevertheless, there is anecdotal evidence that such innovative therapeutic attempts are being made, with and without physician oversight.
A search of the GoFundMe crowdfunding platform for US-based medical campaigns that referenced “FMT” found it proposed as a therapy not only for CDI but also for ulcerative colitis; autism spectrum disorders; fluoroquinolone toxicity; mast cell activation syndrome and dysautonomia; chronic pain; chronic pouchitis; and combinations of complex medical disorders. Of these various campaigns, two specified that the FMT would be undertaken outside of the United States (Canada, Argentina), while one mentioned a DIY FMT attempt. Only one campaign referenced FMT in the context of a clinical trial [70]; none mentioned a clinician procuring the FDA-required IND. These cases represent only a small subset of all FMTs performed or considered, but they highlight the many complex issues surrounding the field, including cost and safety. Outside of clinical trial settings, the FDA permits use of FMT that is stricdy limited to select CDI indications; yet, as stool is readily available, it is impossible to control who is conducting FMT, for what indications, and with what safeguards.
4.4. Regulatory status
While FDA approvals of products are for certain indications (“on-label uses”), physicians have the prerogative to prescribe these products for “off-label use” however they feel fit, so long as they can defend themselves from potential legal charges that they deviated from the standard of care without cause. Thus, the FDA regulates when a new medical product may come to market, not the medical practice of its use. Given the ubiquitously available nature of stool, the agency's role in preventing access to this unapproved medical product is significantly undermined. The regulatory status of FMT is made even more problematic by the fact that the FDA determined that fecal microbiota falls into two classes: both that of “drug” and “biological product” [71].
In 2013, the FDA declared that a physician or a company must obtain an IND application to perform FMT. Investigators and clinicians alike pushed back, stating that clinical experience and early data for FMT already demonstrated its effectiveness as treatment for recurrent or refractory CDI, and that the regulatory burden of maintaining an IND may prevent many clinicians from providing a potentially life-saving therapy [72]. In acknowledgement of this concern, the FDA announced that it would exercise enforcement discretion for FMTs performed for CDI not responding to standard therapy [73]. However, the agency maintained that use of FMT for research or to treat conditions other than CDI would require an IND.
Typically, an IND is secured in order to conduct a clinical trial; less commonly, INDs (including for the treatment of single patients) may be obtained via FDA's Expanded Access (“compassionate use”) pathway [74]. An eligible patient would have to have no approved therapeutic options, be ineligible to participate in a clinical trial, be seriously or life-threateningly ill and deemed more likely to benefit than to be harmed from the unapproved treatment, and have a treating physician willing to seek a single patient IND from the FDA and approval from an institutional review board (IRB). The patient, or a legally-appropriate surrogate decision maker, would be required to provide informed consent, and any serious or unexpected adverse events deemed related to the FMT would need to be reported promptly to the FDA. It is preferable to enroll a patient in a clinical trial whenever possible, in order to accrue scientifically useful data; however, a clinician may seek a single-patient IND for patients without other options.
In March of 2016, the FDA drafted revised guidance regarding clinical use of FMT for CDI. This guidance, though ultimately never implemented, would have allowed for continued enforcement discretion, provided that clinicians obtain informed consent, the donor stool is obtained from a donor known to either the patient or the treating health care provider, and the donor and the donor stool are screened under the direction of the health care provider [71]. Following the aforementioned two cases of MDRO infections attributed to FMT in 2019, according to the FDA, all potential FMT donor stool must be tested for MDROs. Additionally, the donor screening and stool testing processes—as well as the potential risks of MDRO transmission and invasive infection—must be explicitly addressed in the informed consent process for all patients potentially undergoing FMT [75].
Regulatory violations regarding the use of FMT for indications other than CDI must be stemmed, either by enforcement of the IND requirement or by broadening permissibility of FMT outside of clinical trials. The lack of concordance between regulation and policy is not only a legal matter but also an ethical one, and may contribute to FMT recipients’ overestimating the safety of the procedure. However, regulating FMT to the point that clinicians are unwilling or unable to provide the procedure will likely lead to greater morbidity and mortality from CDI, as well as increased numbers of individuals pursuing DIY treatments [76].
5. Ethical concerns
As is often the case with new medical innovations, enthusiasm for FMT has outpaced available evidence of its efficacy and safety in treating a wide array of indications, thus raising ethical concerns [77–79]. The current popularity of FMT and the expansion—both in number and indication—of its use without FDA approval mirrors the initial and persistent popularity of many direct-to-consumer stem cell-based interventions. In both cases, treatment for some indications is evidence-based, while treatment for others reflects practitioners’ over-eagerness to treat a wide array of indications among credulous patients who are often desperate to believe these therapies offer cures to their ailments.
5.1. Informed consent
Informed, voluntary consent on the part of a patient is necessary for both research and clinical care. FMT exists in a no-man’s land between the two: Unless done in a clinical trial, it is performed as treatment that, although innovative, is considered non-standard of care. Even for CDI, FMT is not an FDA-approved therapy but rather merely permitted via the agency's discretion. As there is not a definitive consensus regarding the specific impact of gut microbiome perturbations on short-and long-term systemic health, and with uncertainties about long-term risks from stool transplantation, properly informing patients about the risks and benefits of FMT is challenging.
In a rapidly evolving field where clinical experience is not always in step with discrete understanding of therapeutic mechanisms, clinicians have the difficult task of educating patients about unresolved concepts in the context of scenarios that are sometimes urgent or dire. In the current setting of enforcement discretion by the FDA, the agency specifies that consent “should include, at a minimum, a statement that the use of FMT products to treat CDI is investigational and a discussion of its potential risks” [75]. When weighed against the possibility of rapid relief of CDI, a patient may discount both the likelihood and magnitude of theoretical risks. Yet this potential tradeoff should serve to caution against overzealousness in the use of FMT [80]. There is never an ethical obligation to perform an experimental intervention; however, it may be ethically permissible to do so, depending on possible risks, possible benefits, and other treatment options. The decision about whether to try an unapproved treatment that offers both a chance of relatively immediate benefit and a chance of unknown long-term consequences cannot be made properly without both the clinician and the patient (1) articulating their goals for the proposed FMT treatment and (2) ensuring concordance of understanding about what is sought and, based on current knowledge, what is likely and possible. This, of course, is more complicated in a pediatric setting, where the legal decision maker—a parent or guardian—is deciding whether to accept unknown delayed risks for someone else.
5.2. Balancing patient autonomy and risk of harm
If clinicians restrict FMT procedures only to those patients who demonstrate a comprehensive understanding of the theoretical risks or who, for reasons of age or severity of disease, may reasonably choose to accept the risks of long-term effects from the transplant, some patients denied the procedure may nevertheless choose to perform it themselves. Likewise, if a policy of barring pediatric patients from receiving FMT were imposed, caregivers may seek to conduct a “home-brew” version of FMT on the patient. It is always a strong possibility that DIY donor stool would be insufficiently screened. Given this reality, a clinician may arguably consider it a form of harm reduction to perform FMT on a patient who falls outside current treatment guidelines in lieu of the DIY version that would otherwise be pursued. We are not persuaded by this stance and hold that uses outside those covered by the FDA’s enforcement discretion policy should occur predominantly in trials, with one exception discussed in section 4.4. However, this raises the issue of whether clinicians should inquire about patients’ inclination for DIY FMT when this information is not volunteered.
5.3. Balancing patient autonomy and the common good
Another question arises when a patient expresses preference for FMT for CDI, but its application is, while likely effective, outside the scope of current guidelines. For instance, if a patient has been treated for CDI with antibiotics yet develops the infection a second time, the patient may ask to skip further antibiotic therapy in favor of FMT. While a clinician’s clinical preference would be to use antibiotics in this situation, she knows that the patient is statistically more likely to develop CDI again, that FMT would likely treat the patient’s infection, and that performing the transplant would align with her patient’s stated preference. In such a context, it is understandable that she may be tempted, and/or ultimately decide, to perform the FMT. However, it must be recognized that such a decision is not currently evidence-based and does not assist in answering the question of whether FMT should become a first-line treatment in recurrent CDI. While a patient is most likely seeking individual medical benefit, a clinician has the opportunity to try to advance both the patient’s welfare and the welfare of all current and future patients by limiting the conduct of innovative practice to within clinical trials that can help generate evidence on which to base treatment guidelines [81].
5.4. Equity of access
Though instructions for and information about performing DIY FMT has proven to be abundantly available on the internet, most patients would likely prefer to have this procedure performed by a medical professional. There are, however, tangible concerns about limited or unequal access due to financial barriers to treatment. With the growing promise of FMT as a potential treatment for conditions beyond CDI, pharmaceutical companies have demonstrated interest in reaping the potential financial benefits of bacteriotherapy. A key supplier of donor stool for FMT in the United States is the non-profit stool bank OpenBiome. From 2013 to 2018, OpenBiome provided material for over 42,643 FMT procedures at nearly 1192 clinical sites throughout the US, with 12,327 stool preparations shipped and 207 new clinical site partnerships established in 2018 alone [82]. When a virtual monopoly comers a market, potential arises for practices such as artificially-imposed shortages and/or price hikes, as well as increased vulnerability to errors in the supply chain. Furthermore, there is limited capability for additional FMT suppliers to become established, given that few facilities are currently capable of performing their own MDRO screening, as required by the 2019 FDA mandate for expanded scope of screening. Ominous signs have already begun to emerge: The price of a donor stool preparation from the company has skyrocketed from their original price of $250 up to $1695-$2050 USD per treatment [83]. Physicians and patients alike have expressed growing concern about growing costs of treatment as a consequence of “hyper-regulation” [84].
Yet patients might also experience a financial burden in the context of “under-regulation.” If FMT is neither FDA-approved nor obtained in a clinical trial, most insurance companies are unlikely to reimburse for it, in keeping with longstanding policies of not paying for experimental treatments [85]. Indeed, our survey of GoFundMe for US-base appeals concerning FMT demonstrates that some patients are already having to find ways to fund their procedures and its many associated costs [70]. For example, one appeal budgeted $5400 for a FMT, while another stated that “the hospital is demanding $1800 before going through with the fecal transplant.” Given the small number of appeals, it is impossible to discern the source of these variations in price. Individuals who cannot afford such expenses will have an additional, financial, incentive to consider pursuing a DIY FMT, with its attendant risks.
6. Moving forward
In keeping with the available data and FDA guidance, it is prudent at this time in the US to avoid FMT as a first-line treatment for CDI. Likewise, it would be appropriate to restrict the routine (i.e., non-trial) use of FMT to CDI indications. For seriously ill patients with conditions other than CDI, who have no other available treatments and no clinical trial options, and for whom a physician believes there is sufficient evidence to believe that FMT would be more likely to cause benefit than harm, the procedure should be conducted within the framework of the FDA’s Expanded Access pathway, which allows for single patient INDs [81]. Use of Expanded Access should be exceptional and reserved for seriously ill patients with no other approved or trial options. If there is a reasonable hypothesis that FMT will provide more benefit than harm for a particular condition, then a clinical trial should be established to test that hypothesis.
It may be that a patient values the possibility of benefit from FMT more than they are concerned about the possibility of latent harm. Patients are permitted to make such decisions with regard to unproven interventions in the context of research, but only with the oversight of the FDA and an IRB, entities which also monitor reporting of adverse events. FDA and IRB oversight occurs in Expanded Access; it does not occur in innovative practice. As such, clinicians should not use FMT without an IND, except for the conditions covered by the FDA’s enforcement discretion policy. Doing so is unethical, contrary to FDA regulation, and places a clinician’s license at risk.
There is currendy a need to develop uniform screening and testing requirements for donors and patients, to standardize treatment algorithms for FMT, and in particular to maintain records of its use in order to monitor short-term and long-term serious adverse reactions such as death or donor-derived infection. Understanding FMT as a subset of tissue donation might help clinicians to understand the vital necessity of these steps. While time-to-response for FMT is generally observed to be within a few days for CDI indications [3,20], there is currently a lack of a standardized, routine follow-up with FMT patients that could allow for the establishment of correlative data between FMT and late-onset adverse effects. The growing preponderance of evidence demonstrating efficacy of FMT at treating CDI may—or may not—be countered by the results of long-term follow-up, but only if such follow-up is systematically conducted. We encourage the delivery of FMTs at medical centers with specific expertise in this developing therapy. Such centers are more likely to employ clinicians and investigators who stay current with the rapidly evolving data and regulatory requirements of this treatment and who may be more vested in participating in FMT research and assessing longitudinally for longterm safety outcomes of this procedure. With more and better evidence, clinicians will be better able to decide whether to conduct FMT for specific indications.
Finally, there is a need for clinicians to strive to educate and to persuade patients not to pursue FMT as a do-it-yourself procedure any more than they would perform an organ transplant or blood transfusion at home. The relative ease of the procedure does not cancel out its risks of harm. As such, upon encountering a patient who mentions considering a “DIY FMT,” clinicians have an obligation to explain the real risks and to counsel against such a course of action. Less clear is whether clinicians should be the ones to initiate this conversation, seeking to learn if a patient is inclined to engage in such a practice, in order to counsel against it. Clinicians should not bow to patient wishes to perform unwarranted FMTs, even if the patient states that in the absence of a clinician-conducted procedure they will try a DIY approach.
7. Conclusion
At the frontier of medicine and innovation, it is difficult to balance theoretical long-term harms against immediate benefits. In the case of FMT, the situation is complicated by the reality that patients can attempt the procedure themselves with no clinical guidance or oversight. Ongoing trials and data accumulation will one day provide a clearer guide, but in the meantime, we should proceed with caution. Clinicians ought to conduct FMT only after a thorough informed consent process and only in line with the existing evidentiary base. If patients wish to pursue FMT for non-CDI indications that are not responding to standard therapies, it should be in the context of a clinical trial where individual outcomes will help to inform practice for the larger population. In exceptional cases, where the patient is severely ill and without alternative treatments (including clinical trials) and where the clinician judges that the procedure offers more possibility of benefit than of risk, FMT might be conducted under FDA and IRB oversight via FDA’s Expanded Access pathway.
We urge providers to monitor patients undergoing FMT carefully and longitudinally, and to contribute to efforts to collect long-term safety data to better understand the use of this powerful therapy for future patients. It is human nature to attribute successes to interventions while attributing failures to the underlying disease; as such, it is not surprising that FMT is viewed as a promising treatment for myriad conditions on the basis of anecdotes. We have an ethical obligation to our current patients and to future generations of patients to conduct the research needed to arrive at evidence-based treatment decisions.
Acknowledgements
This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
Conflict of interest disclosures
None for Bateman-House; Chen has received research funding from the American Gastroenterological Association to participate in FMT research. All authors have approved of the final article.
References
- [1].Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacterial cells in the body. PLoS Biol 2016;14:e1002533. 10.1371/journal.pbio.1002533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med 2013;368:407–15. 10.1056/NEJMoa1205037. [DOI] [PubMed] [Google Scholar]
- [3].Brandt LJ, Aroniadis OC, Mellow M, et al. Long-term follow-up of colonoscopic fecal microbiota transplant for recurrent Clostridium difficile infection. Am J Gastroenterol 2012;107:1079–87. 10.1038/ajg.2012.60. [DOI] [PubMed] [Google Scholar]
- [4].Kelly CR, Khoruts A, Staley C, Sadowsky MJ, Abd M, Alani M, et al. Effect of fecal microbiota transplantation on recurrence in multiply recurrent Clostridium difficile infection: a randomized trial. Ann Intern Med 2016;165:609–16. 10.7326/M16-0271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Staley C, Hamilton MJ, Vaughn BP, Graiziger CT, Newman KM, Kabage AJ, et al. Successful resolution of recurrent Clostridium difficile infection using freeze-dried, encapsulated fecal microbiota; pragmatic cohort study. Am J Gastroenterol 2017; 112:940–7. 10.1038/ajg.2017.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Ekekezie C, Perler BK, Wexler A, Duff C, Lillis CJ, Kelly CR. Understanding the scope of do-it-yourself fecal microbiota transplant. Am J Gastroenterol 2020;115:603–7. 10.14309/ajg.0000000000000499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Schmulson M, Bashashati M. Fecal microbiota transfer for bowel disorders: efficacy or hype? Curr Opin Pharmacol 2018;43:72–80. 10.1016/j.coph.2018.08.012. [DOI] [PubMed] [Google Scholar]
- [8].Eakin E The excrement experiment: treating disease with fecal transplants. The New Yorker: 2014;(December 1, 2014). In press, https://www.newyorker.com/magazine/2014/12/01/excrement-experiment. [Google Scholar]
- [9].Zhang F, Luo W, Shi Y, Fan Z, Ji G. Should we standardize the 1,700-year-old fecal microbiota transplantation? Am J Gastroenterol 2012;107:1755. 10.1038/ajg.2012.251. [DOI] [PubMed] [Google Scholar]
- [10].Li S Ben cao gang mu. Beijing: Huaxia Press; 2011. [Google Scholar]
- [11].Small Things Considered. Fecal transplants in the “good old days. https://schaechter.asmblog.org/schaechter/2013/05/fecal-transplants-in-the-good-old-days.html/. [Accessed 7 September 2020].
- [12].Eiseman B, Silen W, Bascom GS, Kauvar AJ. Fecal enema as an adjunct in the treatment of pseudomembranous enterocolitis. Surgery 1958;44:854–9. [PubMed] [Google Scholar]
- [13].de La Cochetiere, Durand T, Lalande V, Petit JC, Potel G, Beaugerie L. Effect of antibiotic therapy on human fecal microbiota and the relation to the development of Clostridium difficile. Microb Ecol 2008;56:395–402. 10.1007/S00248-007-9356-5. [DOI] [PubMed] [Google Scholar]
- [14].Loo VG, Poirier L, Miller MA, Oughton M, Libman MD, Michaud S, et al. A predominantly clonal multi-institutional outbreak of Clostridium difficile-associated diarrhea with high morbidity and mortality. N Engl J Med 2005;353: 2442–9. 10.1056/NEJMoa051639. [DOI] [PubMed] [Google Scholar]
- [15].Dubberke ER, Olsen MA. Burden of Clostridium difficile on the healthcare system. Clin Infect Dis 2012;55:S88–92. 10.1093/cid/cis335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Lessa FC, Mu Y, Bamberg WM, Beldavs ZG, Dumyati GK, Dunn JR, et al. Burden of Clostridium difficile infection in the United States. N Engl J Med 2015;372:825–34. 10.1056/NEJMoa1408913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].McFarland LV, Surawicz CM, Greenberg RN, Fekety R, Elmer GW, Moyer KA, et al. A randomized placebo-controlled trial of Saccharomyces boulardii in combination with standard antibiotics for Clostridium difficile disease. J Am Med Assoc 1994;271. 10.1001/jama.1994.03510480037031. [DOI] [PubMed] [Google Scholar]
- [18].Youngster I, Russell GH, Pindar C, Ziv-Baran T, Sauk J, Hohmann EL. Oral, capsulized, frozen fecal microbiota transplantation for relapsing Clostridium difficile infection. J Am Med Assoc 2014;312:1772–8. 10.1001/jama.2014.13875. [DOI] [PubMed] [Google Scholar]
- [19].Drekonja D, Reich J, Gezahegn S, Greer N, Shaukat A, MacDonald R, et al. Fecal microbiota transplantation for Clostridium difficile infection: a systematic review. Ann Intern Med 2015;162:630–8. 10.7326/M14-2693. [DOI] [PubMed] [Google Scholar]
- [20].Cammarota G, Masucci L, Ianiro G, Bibbò S, Dinoi G, Costamagna G, et al. Randomised clinical trial: faecal microbiota transplantation by colonoscopy vs. vancomycin for the treatment of recurrent Clostridium difficile infection. Aliment Pharmacol Ther 2015;41:835–43. 10.1111/apt.13144. [DOI] [PubMed] [Google Scholar]
- [21].Weingarden AR, Dosa PI, DeWinter E, Steer CJ, Shaughnessy MK, Johnson JR, et al. Changes in colonic bile acid composition following fecal microbiota transplantation are sufficient to control Clostridium difficile germination and growth. PloS One 2016; 11:e0147210. 10.1371/joumal.pone.0147210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Kronman MP, Nielson HJ, Adler AL, Giefer MJ, Wahbeh G, Singh N, et al. Fecal microbiota transplantation via nasogastric tube for recurrent Clostridium difficile infection in pediatric patients. J Pediatr Gastroenterol Nutr 2015;60:23–6. 10.1097/MPG.0000000000000545. [DOI] [PubMed] [Google Scholar]
- [23].Pinn DM, Aroniadis OC, Brandt LJ. Is fecal microbiota transplantation (FMT) an effective treatment for patients with functional gastrointestinal disorders (FGID)? Neuro Gastroenterol Motil 2015;27:19–29. 10.1038/ajg.2014.295. [DOI] [PubMed] [Google Scholar]
- [24].Colman RJ, Rubin DT. Fecal microbiota transplantation as therapy for inflammatory bowel disease: a systematic review and meta-analysis. J Crohns Colitis 2014;8: 1569–81. 10.1016/j.crohns.2014.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Moayyedi P, Surette MG, Kim PT, Libertucci J, Wolfe M, Onischi C, et al. Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trial. Gastroenterology 2015;149:102–9. 10.1053/j.gastro.2015.04.001. [DOI] [PubMed] [Google Scholar]
- [26].Wei Y, Zhu W, Gong J, Guo D, Gu L, Li N, et al. Fecal microbiota transplantation improves the quality of life in patients with inflammatory bowel disease. Gastroenterol Res Pract 2015;2015:517597. 10.1155/2015/517597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Scaldaferri F, Pecere S, Petito V, Zambrano D, Fiore L, Lopetuso LR, et al. Efficacy and mechanisms of action of fecal microbiota transplantation in ulcerative colitis: pitfalls and promises from a first meta-analysis. Transplant Proc 2016;48:402–7. 10.10l6/j.transproceed.2015.12.040. [DOI] [PubMed] [Google Scholar]
- [28].Borody TJ, Leis S, Campbell J, Campbell J, Torres M, Nowak A. Fecal microbiota transplantation (FMT) in multiple sclerosis (MS) [abstract]. Am J Gastroenterol 2011;106:S352. [Google Scholar]
- [29].Borody TJ, Campbell J, Torres M, Nowak A, Leis S. Reversal of idiopathic thrombocytopenic purpura (ITP) with fecal microbiota transplantation (FMT) [abstract]. Am J Gastroenterol 2011;106:S352. [Google Scholar]
- [30].Basson AR, Minh L, Cominelli F. Complementary and alternative medicine (CAM) and next-generation CAM (NG-CAM) strategies for therapeutic gut microbiota modulation in inflammatory bowel disease. Gastroenterol Clin N Am 2017;46: 689–729. 10.1016/j.gtc.2017.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Gundling F, Roggenbrod S, Schleifer S, Sohn M, Schepp W. Patient perception and approval of faecal microbiota transplantation (FMT) as an alternative treatment option for obesity. Obes Sci Pract 2018;5:68–74. 10.1002/osp4.302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Roggenbrod S, Schuler C, Haller B, Sohn M, Storr M, Schepp W, et al. Akzeptanz des fakalen Mikrobiota-Transfers (Stuhltransplantation) als alternative Therapie bei Patienten mit Colitis ulcerosa [Patient perception and approval of fecal microbiota transplantation (FMT) as an alternative treatment option for ulcerative colitis]. Z Gastroenterol 2019;57:296–303. 10.1055/a-0821-7166. [DOI] [PubMed] [Google Scholar]
- [33].Zeitz J, Bissig M, Barthel C, Biedermann L, Scharl S, Pohl D, et al. Patients’ views on fecal microbiota transplantation: an acceptable therapeutic option in inflammatory bowel disease? Eur J Gastroenterol Hepatol 2017;29:322–30. 10.1097/MEG.0000000000000783. [DOI] [PubMed] [Google Scholar]
- [34].Funk C, Kennedy B, Hefferon M. Vast majority of Americans say benefits of childhood vaccines outweigh risks. Pew Research Center; 2017. https://www.pewresearch.org/intemet/wp-content/uploads/sites/9/2017/02/PS_2017.02.02_Vaccines_FINAL.pdf. [Google Scholar]
- [35].Doré J, Multon MC, Béhier JM. Participants of Giens XXXII, Round Table No. 2. The human gut microbiome as source of innovation for health: which physiological and therapeutic outcomes could we expect? Therapie 2017;72:21–38. 10.1016/j.therap.2016.12.007. [DOI] [PubMed] [Google Scholar]
- [36].Park L, Mone A, Price JC, Tzimas D, Hirsh J, Poles MA, et al. Perceptions of fecal microbiota transplantation for Clostridium difficile infection: factors that predict acceptance. Ann Gastroenterol 2017;30:83–8. 10.20524/aog.2016.0098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Goloshchapov OV, Olekhnovich El, Sidorenko SV, Moiseev IS, Kucher MA, Fedorov DE, et al. Long-term impact of fecal transplantation in healthy volunteers. BMC Microbiol 2019;19:312. 10.1186/s12866-019-1689-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Yang Y, Torchinsky MB, Gobert M, Xiong H, Xu M, Linehan JL, et al. Focused specificity of intestinal TH17 cells towards commensal bacterial antigens. Nature 2014;510:152–6. 10.1038/nature13279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Staley C, Kaiser T, Vaughn BP, et al. Durable long-term bacterial engraftment following encapsulated fecal microbiota transplantation to treat Clostridium difficile infection. mBio 2019;10(4). 10.1128/mBio.01586-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Auchtung JM, Preisner EC, Collins J, Lerma AI, Britton RA. Identification of simplified microbial communities that inhibit Clostridioides difficile infection through dilution/extinction. mSphere 2020;5. 10.1128/mSphere.00387-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Khoruts A, Rank KM, Newman KM, Viskocil K, Vaughn BP, Hamilton MJ, et al. Inflammatory bowel disease affects the outcome of fecal microbiota transplantation for recurrent Clostridium difficile infection. Clin Gastroenterol Hepatol 2016;14: 1433–8. 10.1016/j.cgh.2016.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Khanna S, Pardi DS, Kelly CR, Kraft CS, Dhere T, Henn MR, et al. A novel microbiome therapeutic increases gut microbial diversity and prevents recurrent Clostridium difficile infection. J Infect Dis 2016;214:173–81. 10.1093/infdis/jiv766. [DOI] [PubMed] [Google Scholar]
- [43].Ting Zhang, et al. Washed microbiota transplantation vs. manual fecal microbiota transplantation: clinical findings, animal studies and in vitro screening. Protein & Cell; 2020. p. 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Fecal Microbiota Transplantation-standardization Study Group. Nanjing consensus on methodology of washed microbiota transplantation. Chinese Med J 2020; 133(19):2330–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Isabella VM, Ha BN, Castillo MJ, Lubkowicz DJ, Rowe SE, Millet YA, et al. Development of a synthetic live bacterial therapeutic for the human metabolic disease phenylketonuria. Nat Biotechnol 2018;36:857–64. 10.1038/nbt.4222. [DOI] [PubMed] [Google Scholar]
- [46].Leventhal DS, Sokolovska A, Li N, Plescia C, Kolodziej SA, Gallant CW, et al. Immunotherapy with engineered bacteria by targeting the STING pathway for antitumor immunity. Nat Commun 2020;11:2739. 10.1038/s41467-020-16602-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Newman KM, Rank KM, Vaughn BP, Khoruts A. Treatment of recurrent Clostridium difficile infection using fecal microbiota transplantation in patients with inflammatory bowel disease. Gut Microb 2017;8:303–9. 10.1080/19490976.2017.1279377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Vijay-Kumar M, Aitken JD, Carvalho FA, Cullender TC, Mwangi S, Srinivasan S, et al. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science 2010;328:228–31. 10.1126/science.1179721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Garrett WS, Lord GM, Punit S, Lugo-Villarino G, Mazmanian SK, Ito S, et al. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell 2007;131:33–45. 10.1016/j.cell.2007.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ, et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 2011; 145:745–57. 10.1016/j.cell.2011.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Cho I, Yamanishi S, Cox L, Methé BA, Zavadil J, Li K, et al. Antibiotics in early life alter the murine colonic microbiome and adiposity. Nature 2012;488:622–6. 10.1038/nature11400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 2013;341: 1241214. 10.l126/science.1241214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Wu S, Rhee K, Albesiano E, Rabizadeh S, Wu X, Yen H, et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat Med 2009;15:1016–22. 10.1038/nm.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Vrieze A, van Nood E, Holleman F, Salojärvi J, Kootte RS, Bartelsman JFWM, et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 2012;143:913–6. 10.1053/j.gastro.2012.06.031. [DOI] [PubMed] [Google Scholar]
- [55].Manichanh C, Reeder J, Gibert P, Varela E, Llopis M, Antolin M, et al. Reshaping the gut microbiome with bacterial transplantation and antibiotic intake. Genome Res 2010;20:1411–9. 10.1101/gr.107987.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Schulfer AF, Battaglia T, Alvarez Y, Bijnens L, Ruiz VE, Ho M, et al. Intergenerational transfer of antibiotic-perturbed microbiota enhances colitis in susceptible mice. Nat Microbiol 2017;3:234–42. 10.1038/s41564-017-0075-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nature 2013;19:576–85. 10.1038/nm.3145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Kostic AD, Gevers D, Pedamallu CS, Michaud M, Duke F, Earl AM, et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res 2012;22:292–8. 10.1101/gr.126573.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Alang N, Kelly CR. Weight gain after fecal microbiota transplantation. Open Forum Infect Dis 2015;2:ofv004. 10.1093/ofid/ofv004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Bokulich NA, Chung J, Battaglia T, Henderson N, Jay M, Li H, et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med 2016;8. 10.1126/scitranslmed.aad7121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Fujimura KE, Sitarik AR, Havstad S, Lin DL, Levan S, Fadrosh D, et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat Med 2016;22:1187–91. 10.1038/nm.4176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Owens C, Broussard E, Surawicz C. Fecal microbiota transplantation and donor standardization. Trends Microbiol 2013;21:443–5. 10.1016/j.tim.2013.07.003. [DOI] [PubMed] [Google Scholar]
- [63].Cammarota G, Ianiro G, Kelly CR, Mullish BH, Allegretti JA, Kassam Z, et al. International consensus conference on stool banking for faecal microbiota transplantation in clinical practice. Gut 2019;68:2111–21. 10.1136/gutjnl-2019-319548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Burns LJ, Dubois N, Smith MB, et al. Donor recruitment and eligibility for fecal microbiota transplantation: results from an international public stool bank [abstract]. Gastroenterology 2015;148:S96–7. 10.1016/S0016-5085(15)30331-0. [DOI] [Google Scholar]
- [65].DeFlipp Z, Bloom PP, Torres Soto M, Mansour MK, Sater MRA, Huntley MH, et al. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med 2019;381:2043–50. 10.1056/NEJMoa1910437. [DOI] [PubMed] [Google Scholar]
- [66].U.S. Food and Drug Administration. Important safety alert regarding use of fecal microbiota transplantation and risk of serious adverse reactions due to transmission of multi-drug resistant organisms, https://www.fda.gov/vaccines-blood-biologics/safety-availability-biologics/important-safety-alert-regarding-use-fecal-microbiota-transplantation-and-risk-serious-adverse/. [Accessed 7 September 2020]. [Google Scholar]
- [67].Alter HJ, Klein HG. The hazards of blood transfusion in historical perspective. Blood 2008;112:2617–26. 10.1182/blood-2008-07-077370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Evatt BL. The tragic history of AIDS in the hemophilia population, 1982–1984. J Thromb Haemostasis 2006;4. 10.1111/j.1538-7836.2006.02213.x. [DOI] [PubMed] [Google Scholar]
- [69].GoFundMe. Fundraiser by Georgianna Henry: please help Georgie get better, https://www.gofundme.com/f/Please-Help-Georgie-Heal/. [Accessed 7 September 2020],
- [70].https://www.gofundme.com/. [Accessed 7 September 2020],
- [71].U.S. Food and Drug Administration. Guidance for industry: enforcement policy regarding investigational new drug requirements for use of fecal microbiota for transplantation to treat Clostridium difficile infection not responsive to standard therapies, https://www.fda.gov/regulatory-information/search-fda-guidance-documents/enforcement-policy-regarding-investigational-new-drug-requirements-use-fecal-microbiota-0/. [Accessed 6 September 2020].
- [72].Hoffmann D, Palumbo F, Ravel J, Roghmann Rowthorn V, von Rosenvinge E. Improving regulation of microbiota transplants: policy should balance safety, efficacy, access, and research. Science 2017;358:1390–1. 10.1126/science.aaq0034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].U.S. Food and Drug Administration. Guidance for industry: enforcement policy regarding investigational new drug requirements for use of fecal microbiota for transplantation to treat Clostridium difficile infection not responsive to standard therapies, https://www.fda.gov/media/86440/download/. [Accessed 7 September 2020],
- [74].U.S. Food and Drug Administration. Expanded access. 7 September 2020, https://www.fda.gov/news-events/public-health-focus/expanded-access/;2020.
- [75].U.S. Food and Drug Administration. Fecal microbiota for transplantation: safety communication - risk of serious adverse reactions due to transmission of multi-drug resistant organisms. 2019. https://www.fda.gov/vaccines-blood-biologics/safety-availabihty-biologics/information-pertaining-additional-safety-protections-regarding-use-fecal-microbiota-transplantation. [Accessed 7 September 2020].
- [76].Sachs RE, Edelstein CA. Ensuring the safe and effective FDA regulation of fecal microbiota transplantation. J Law Biosci 2015;2:396–415. 10.1093/jlb/lsv032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Ma Y, Liu J, Rhodes C, Nie Y, Zhang F. Ethical issues in fecal microbiota transplantation in practice. Am J Bioeth 2017. May;17(5):34–45. 10.1080/15265161.2017.1299240. [DOI] [PubMed] [Google Scholar]
- [78].Murray TS, Herbst J. The Ethics of fecal microbiota transplant as a tool for antimicrobial stewardship programs. J Law Med Ethics 2019. December;47(4):541–54. 10.1177/1073110519897730. [DOI] [PubMed] [Google Scholar]
- [79].McGuire AL, Colgrove J, Whitney SN, Diaz CM, Bustillos D, Versalovic J. Ethical, legal, and social considerations in conducting the Human Microbiome Project. Genome Res 2008;18(12):1861–4. 10.1101/gr.081653.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Bunnik EM, Aarts N, Chen LA. Physicians must discuss potential long-term risks of fecal microbiota transplantation to ensure informed consent. Am J Bioeth 2017. May;17(5):61–3. 10.1080/15265161.2017.1299816. [DOI] [PubMed] [Google Scholar]
- [81].Hecht GA, Blaser MJ, Gordon J, Kaplan LM, Knight R, Laine L, Peek R, Sanders ME, Sartor B, Wu GD, Yang VW. What is the value of a food and drug administration investigational new drug application for fecal microbiota transplantation to treat Clostridium difficile Infection? Clin Gastroenterol Hepatol 2014. February;12(2):289–91. 10.1016/j.cgh.2013.10.009. Epub 2013 Oct 20.. [DOI] [PubMed] [Google Scholar]
- [82].OpenBiome. OpenBiome annual report, https://static1.squarespace.com/static/50e0c29ae4b0a05702af7e6a/t/5ea039c598b6d25d07138f20/158755886533l/2018+OpenBiome+Annual+Report.pdf. [Accessed 6 September 2020],
- [83].OpenBiome. Treatment information. https://www.openbiome.org/treatment-information/. [Accessed 8 September 2020].
- [84].Jacobs A Drug companies and doctors battle over the future of fecal transplants. NYT; 2019. [Google Scholar]
- [85].Folkers KM, Bateman-House A, Robertson C. Paying for unapproved medical products. Wake Forest J L & Pol’y; in press. [Google Scholar]