Research Collection

Review Article

Treatments of trimethylaminuria: where we are and where we might be heading

Author(s): Schmidt, Aaron C.; Leroux, Jean-Christophe

Publication Date: 2020-09-09

Permanent Link: https://doi.org/10.3929/ethz-b-000423705

Originally published in: Drug discovery today 25(9), http://doi.org/10.1016/j.drudis.2020.06.026

Rights / License: Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International

This page was generated automatically upon download from the ETH Zurich Research Collection. For more information please consult the Terms of use.

ETH Library

 

Drug Discovery Today Volume 00, Number 00 June 2020 REVIEWS

Treatments of trimethylaminuria:

POST SCREEN

where we are and where we might be 

heading Reviews

Aaron C. Schmidt and Jean-Christophe Leroux

Q2

Q1 Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland

Q3 (TMA) is a volatile, foul-smelling, diet-derived amine, primarily generated in the colon

and metabolized in the to its odorless N-oxide (TMAO). In primary trimethylaminuria (TMAU), an

Q4 inherited deficiency in flavin-containing monooxygenase 3 leads to elevated systemic TMA levels. The

excretion of elevated amounts of TMA in sweat, breath, urine and other bodily secretions gives

individuals affected by TMAU a smell resembling that of rotten fish. Although the disorder might not

seem an important health problem, its social and psychological burden can be devastating. To date, no

treatment modifying the disorder exists and only a few pharmacological therapies provide modest and

transient benefits. This review provides an overview of investigated TMAU treatments and outlines

promising new research directions.

Introduction Although the first case of TMAU was described in literature

Primary trimethylaminuria (TMAU) is a rare metabolic disorder around 1970 [3], to date, only a few hundred cases have been

where abnormally high levels of the aliphatic amine trimethyla- reported in the literature since [4,5]. A study on the incidence of

mine (TMA) are excreted through sweat, breath, urine and other TMAU revealed a rate of 1% of heterozygous carriers within the

bodily secretions, giving the patients a smell resembling that of white British population [6], whereas studies in other ethnic

rotting fish. TMAU has thus been referred to historically as fish groups, such as the New Guinean population, revealed a carrier

odor syndrome. TMA is a diet-derived amine that originates from rate of 11% [7]. Although passing one affected allele of the condi-

TMA N-oxide (TMAO) (which is present in marine fish), tion to the next generation, heterozygotes usually do not have any

and . Although at physiological pH most TMA is in its symptoms of TMAU, unless they are challenged with a TMA

protonated form (pKa 9.80) [1], it is in equilibrium with a small precursor overload [8].

fraction of the free base. The free base is highly volatile and readily TMAU usually manifests itself in childhood or early adult life

detected by the human olfactometric receptors in the ppb range but remains underdiagnosed. Online resources often attribute this

[2]. In unaffected individuals, the free base is metabolized in the to the fact that people having mild symptoms rarely tend to seek

liver to the odorless TMAO and excreted in the urine. Primary help. In addition, some physicians might be unaware of the

TMAU is a direct consequence of an impairment in this oxidation disorder, not recognizing the symptoms and potentially being

process. Secondary TMAU, however, is caused by an excess of unable to distinguish them from other conditions that result in

dietary precursors or other factors exacerbating the production an unpleasant (http://rarediseases.org). In fact, a study

of TMA, therefore causing substrate overload for the , including 353 patients complaining of idiopathic malodor

which is unable to oxidize the elevated burden of TMA. In both revealed that approximately one-third of them suffered from

forms of the disorder, the metabolite accumulates in bodily secre- TMAU [9]. Although the disease is considered benign, its psycho-

tions and gives them the characteristic odor. logical burden can be devastating. Studies have mentioned various

psychosocial reactions of sufferers, such as strong feelings of

shame, embarrassment, social isolation and even suicidal tenden-

Corresponding author: Leroux, J.-C. ([email protected]) cies, among others [8,10,11]. To date, the disease has no cure and

1359-6446/ã 2020 The Author(s). Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

https://doi.org/10.1016/j.drudis.2020.06.026 www.drugdiscoverytoday.com 1

Please cite this article in press as: Schmidt, A.C., Leroux, J.C. Treatments of trimethylaminuria: where we are and where we might be heading, Drug Discov Today (2020), https://doi.org/ 10.1016/j.drudis.2020.06.026

DRUDIS 2726 1–8

 

REVIEWS Drug Discovery Today Volume 00, Number 00 June 2020

Diet

Choline TMAO Oxidation Reviews L-carnitine TMA  OTSCREEN POST

Bacterial fermentation

TMAO Choline

TMA

Excretion L-carnitine

Drug Discovery Today

FIGURE 1

Dietary precursors and metabolism of TMA. The main precursors include choline, TMAO and L-carnitine, which are metabolized to TMA within the intestine. The

dietary breakdown of these precursors depends on a range of bacteria, among them: Actinobacteria, Firmicutes, Proteobacteria and Bacteroidetes [17]. TMA is

subsequently oxidized to TMAO by FMO3 in the liver and excreted through the kidneys.

only palliative measures exist, such as washing with acidic soap in broad spectrum of xenobiotics and dietary amines [20–22]. So far,

combination with dietary restriction of TMA precursors [12,13]. >40 variants of the have been associated with TMAU [23,24],

Other available pharmacological treatments aim to reduce the most of which are missense . Some of these are inacti-

generation and absorption of TMA or focus on improving oxida- vating mutations whereas other less severe ones reduce the thresh-

tion to the odorless TMAO. old of TMA oxidation of FMO3 [25]. The resulting impairment in

N-oxidation capacity accounts for most cases of TMAU reported in

Trimethylaminuria the literature.

TMA is produced in the gastrointestinal tract from dietary pre-

cursors such as choline, which is present in eggs, liver and poultry, Secondary TMAU

among others [14]. In the colon, choline is metabolized to TMA by Often referred to as acquired or sometimes transient TMAU, people

anaerobic microorganisms carrying the enzyme choline TMA lyase becoming afflicted with secondary TMAU might show prolonged

(Fig. 1). TMAO and carnitine, which are mainly present in marine symptoms. This form of the disorder can occur in a variety of

fish [15] and in red [16], respectively, are additional TMA different scenarios. Treatment with choline in Huntington’s and

precursors that are metabolized in the gut [17]. However, in the Alzheimer’s diseases has been associated with the development of

case of TMAO, the exact mechanism of conversion to TMA in a strong fishy body odor [26,27], and is a classic example of

humans remains unknown. TMA is readily absorbed by passive precursor overload where the enzyme is unable to completely

diffusion and enters enterohepatic circulation. It is subsequently oxidize the TMA burden. Liver failure and portosystemic shunting

oxidized in the liver to the non-odorous N-oxide form. The en- of the blood can also result in increased TMA levels, owing to

zyme, responsible for this oxidation process, is of the flavin-con- interference with first-pass metabolism [28,29]. Other factors that

taining monooxygenase (FMO) family [18,19]. The N-oxide is then have been reported to cause or exacerbate the condition (including

readily excreted in the urine. As mentioned previously, TMAU can primary TMAU) encompass menstruation [30,31], asymptomatic

be classified in two forms of the disorder, which are described viral [32] and testosterone treatment [33], and have been

below. reviewed elsewhere [4].

Primary TMAU Diagnosis

Primary TMAU is caused by a in the FMO3 gene, which is Diagnosis of TMAU is usually done by urinary analysis after oral

inherited in a Mendelian autosomal recessive fashion [18]. The precursor challenge using choline [9] or TMA [8,34]. TMAO has

affected enzyme has a widespread substrate specificity, including a also been indicated to drastically shift the total combined TMA

2 www.drugdiscoverytoday.com

Please cite this article in press as: Schmidt, A.C., Leroux, J.C. Treatments of trimethylaminuria: where we are and where we might be heading, Drug Discov Today (2020), https://doi.org/ 10.1016/j.drudis.2020.06.026

DRUDIS 2726 1–8

 

Drug Discovery Today Volume 00, Number 00 June 2020 REVIEWS

(TTMA) content (TMA + TMAO) toward TMA species after oral where degradation of carnitine to TMAO could be demonstrated

administration in the past, in affected individuals as well as [57]. A recent report on the mechanism of carnitine transforma-

heterozygous carriers of the disease [35,36]. However, it is not a tion to TMA identified a Rieske-type protein (a two-component

recognized diagnostic tool for TMAU. Upon precursor challenge, Rieske-type oxygenase/reductase) as the main protagonist [58,59].

the urinary samples are assayed for TMA as well as TMAO, and their L-carnitine is present in high levels in red meat, poultry and some

respective amounts are compared to the TTMA. Because neither dairy products, which is why TMAU sufferers are recommended to

TMA nor TMAO have a native chromophore, quantitative urine avoid these in their diet [60]. Interestingly, allicin, an organosulfur

1

analysis is performed using H NMR spectroscopy [37,38], head- compound obtained from garlic, lowered TMAO levels when

space MS [39,40] and electrospray ionization mass spectrometry supplemented in addition to L-carnitine in male mice [61], an

[41]. Affected individuals excrete 80% of TTMA in the form of animal model that is a natural knockout for FMO3, and in which

TMA, whereas heterozygous carriers as well as unaffected people FMO1 does not efficiently catalyze the transformation of TMA to

secrete roughly 4%. Interestingly, after oral challenge with TMA, TMAO [62]. The mechanism of allicin remains elusive but is POST SCREEN



this number increases to 25% in heterozygotes [42]. Genetic hypothesized to be connected to its potential role as a Rieske

analysis can be performed to confirm the diagnosis [43], provided protein inhibitor, which would reduce carnitine transformation

urinalysis indicates primary TMAU. to TMA by bacteria. However, the ability of allicin to decrease TMA

Reviews

levels remains to be demonstrated.

Treatment options

Unlike other metabolic disorders, TMAU appears to generally Protonation of TMA

attract little biomedical interest [4]. Consequently, research efforts Reducing the volatility of excreted TMA is a validated strategy for

for TMAU treatments have been modest. Existing treatment strat- the management of the fish-like body odor because TMA excretion

egies can be classified into the following groups: precursor intake in sweat is a significant source of it. Although only mentioned

limitation, protonation of TMA, targeting of the gut metabolism anecdotally in the scientific literature, frequent washing with low

and targeting of the enzyme FMO3. pH soaps in combination with dietary management constitutes

the state-of-the-art treatment for TMAU [13,63]. When applied on

Limiting precursor intake the skin on a regular basis, acidic body lotions and soaps fully

A common approach to lower systemic TMA levels is to limit TMA convert excreted TMA to the non-smelling conjugate acid, reduc-

precursor intake. Several food-derived compounds are metabo- ing the body odor. Recently, our group reported a topical odor-

lized to TMA in the colon by bacteria, a topic that has been quenching formulation based on polymeric vesicles of the diblock

discussed at length in the past [17]. Indeed, a study on dietary poly(isoprene)-b-poly(ethylene glycol). The acidic solution (pH 2)

precursors showed that a few of them could be linked to increased contained in the vesicles establishes a transmembrane pH gradient

levels in urinary TMA [44]. Specifically, choline, carnitine and across their bilayer membrane that constitutes the basis for the

TMAO have been shown to increase concentrations of urinary system’s mechanism. The TMA free base readily permeates

TMA and TMAO, whereas betaine, creatinine and have not through the vesicular bilayer membrane and becomes protonated

[44,45]. Metabolism of betaine to TMA has been reported in sheep once inside the acidic core. The positive charge traps the TMA in

rumen in the past [46] but other studies have failed to show a the vesicle core and prevents its diffusion (Fig. 2). In an in vitro

consistent effect in rats [47] or in humans [48,49]. setting, the polymeric vesicles readily sequestered TMA at skin pH

Choline is absorbed in the small intestine via carrier-mediated (pH 5.8) and successfully decreased the odor intensity of TMA

transport across the mucosal border. Ingestion of choline-rich perceived by human volunteers in an olfactory study [64]. The

food saturates this transport capacity, which is half saturated at system’s clinical efficacy is yet to be confirmed in clinical studies

concentrations of 200–300 mM [50,51], resulting in increased where the polymeric vesicles would be applied topically on TMAU

choline concentrations in the large bowel where intestinal bacteria patients.

metabolize it to TMA and dimethylamine, as was shown in rats Deodorizing TMA by masking it with another stronger smell has

[52]. Because choline is required for the biosynthesis of essential also been explored as a potential approach [65]. It was reported

phospholipids and the neurotransmitter acetylcholine, it is not that (E,E)-2,4-undecadienal, a naturally derived product from

possible to completely eliminate it from the diet [53]. In fact, coriander leaves, deodorized TMA levels of up to 1 mM. Although

healthy individuals on a choline-deficient diet develop signs of effective at extremely low levels (10 ppm), the compound was

incipient liver dysfunction after only 3 weeks. Consequently, shown to be toxic in human fibroblasts at 25 mM [66], which

intake of this nutrient can only be limited to a certain extent could represent a challenge when formulating topical or oral

[12,14], and personalized adjustments by a clinician are recom- formulations.

mended to prevent liver dysfunction and other side effects.

TMAO is an osmolyte found in marine organisms that counter- Targeting gut metabolism

acts the protein-destabilizing effects of hydrostatic and osmotic Most interventions reported in the literature target the production

pressures [15,54]. Ingestion of a fish-based meal leads to an in- of TMA in the colon and/or lower its subsequent absorption.

crease in urinary TMAO and subsequently TMA levels [55], so it is Inhibition of TMA precursor, inhibition of TMA metabolism

recommended to eliminate all seafood from the diet. Reports of L- and TMA sequestration have been investigated through different

carnitine and its effect on TMA levels are rather scarce. In vivo approaches. One aspect heavily intertwined with the metabolic

experiments have shown an increase in urinary TMAO after car- fate of TMA, is that of its N-oxide – TMAO. In recent years, TMAO

nitine challenge [56]. This was confirmed in healthy volunteers, has been the object of extensive research because it was identified

www.drugdiscoverytoday.com 3

Please cite this article in press as: Schmidt, A.C., Leroux, J.C. Treatments of trimethylaminuria: where we are and where we might be heading, Drug Discov Today (2020), https://doi.org/ 10.1016/j.drudis.2020.06.026

DRUDIS 2726 1–8

 

REVIEWS Drug Discovery Today Volume 00, Number 00 June 2020 (a) (b) Reviews

Archaebiotic cell  (d) OTSCREEN POST (c)

Choline

Drug Discovery Today

FIGURE 2

Clinical and investigational TMAU treatments. Targeting excreted TMA on patient skin by sequestration of its non-odorous protonated species with a

transmembrane pH-gradient polymersome-based topical formulation (a); restricting dietary intake of known TMA precursors, such as choline, carnitine and

TMAO (b); controlling TMA production in the colon with probiotics, such as methanogenic Archae to reduce TMA to methane via methyltransferase (Mtt) (c); or

with choline TMA-lyase inhibitors, such as DMB, FMC or iodomethylcholine (IMC) (d).

as a disease marker in cardiovascular disease (CVD). Further, it has is no medical emergency. Their extensive consumption could

been suggested to promote atherosclerosis in mice [67] and, in contribute to the emergence of antibiotic resistance at the indi-

addition, elevated TMAO plasma levels were associated with an vidual level, potentially compromising the treatment of bacterial

increased risk of cardiovascular events in patients following elec- infections [79,80]. In addition, antimicrobial treatment disrupts

tive coronary angiography [68]. Several studies have supported the microbiota composition and could be linked to other diseases

these findings in various patient populations [69–72]. Yet, litera- such as obesity [81,82].



ture findings on the topic remain controversial to this date. Several . The antibiotic metronidazole is thought to

studies indeed failed to show an association of TMAO levels with decrease the load of bacterial flora that produces TMA [83]. This

incident CVD [73]. However, because elevated plasma TMAO nitroimidazole that acts specifically on anaerobic bacteria is used

levels are a direct result of dietary breakdown of TMA precursors in the chronic treatment of methylmalonic and propionic

and their subsequent oxidation in the liver, treatments targeting acidemias to diminish toxic metabolites produced by anaerobes

the metabolic breakdown of these precursors are of growing inter- [84]. It has also been tested as a short-term treatment of HE but its

est as potential treatments in CVD, a strategy that could eventually ototoxicity, nephrotoxicity and neurotoxicity make it unsuitable

be applied to TMAU in the future. However, caution must be taken for prolonged use [85]. Reports of TMAU patients receiving the

while interpreting these data, because a change in TMAO levels antibiotic over a range of weeks showed variable results,

does not necessarily imply a change in TMA levels, wherefore decreasing urinary TMA by a factor of three in some patients

concrete experimental evidence needs to be provided in the un- but not showing any effect in others [83,86]. Improved outcomes

derlying experimental setup. Furthermore, studies suggest the were obtained when combining the treatment with dietary

involvement of TMAO in various important biological functions, restrictions [87] or with another antibiotic [43,55].



for instance acting as a protein stabilizer and as an osmotic agent Neomycin. One of the antibiotics that has been tested in

[74,75], which should be taken into account while aiming to lower combination with metronidazole is the poorly bioavailable

TMAO levels. aminoglycoside neomycin: This antimicrobial agent has been

Antibiotics used since the late 1960s when it was approved for treating

Because TMA production from dietary precursors is dependent on overt HE by the FDA [88]. Similarly to metronidazole, its major

its metabolism by intestinal microbiota [76,77], antibiotics have drawbacks are its ototoxicity and nephrotoxicity [89], the latter

been assessed as a treatment for TMAU. Antibiotics are used to affecting about one-third of the patient population [90]. Single

deplete intestinal bacteria in other diseases such as hepatic en- use of neomycin was reported to lower TMA levels in mice [76]

cephalopathy (HE) (a consequence of ), where and in small patient populations [91,92]. However, owing to a

they target urease-producing bacteria to decrease ammonia pro- non-permanent alteration of the microbiome, values quickly

duction [78]. Although antibiotics can alleviate the main symp- returned to baseline upon termination of the treatment and

tom of TMAU patients, their chronic use should be avoided if there subsequent proliferation of bacteria [92].

4 www.drugdiscoverytoday.com

Please cite this article in press as: Schmidt, A.C., Leroux, J.C. Treatments of trimethylaminuria: where we are and where we might be heading, Drug Discov Today (2020), https://doi.org/ 10.1016/j.drudis.2020.06.026

DRUDIS 2726 1–8

 

Drug Discovery Today Volume 00, Number 00 June 2020 REVIEWS



Rifaximin. Another antibiotic that has been explored for the especially E. limosum and members of the Clostridial clade XV

treatment of TMAU is rifaximin. It has been reported to lower family, claiming that they demethylated quaternary amines

TMA levels [93] and to be generally tolerated rather well [94]. that would otherwise form TMA and TMAO [107]. However, so

This poorly absorbed semisynthetic rifamycin derivative has a far this has only been shown in vitro observing the growth of gut

broad spectrum of antibacterial activity, including aerobes and microbes in the presence and absence of quaternary amines.

anaerobes, Gram-positive and Gram-negative bacteria, and was The main drawback of probiotics remains the difficulty to

approved by the FDA in 2010 for the treatment of HE. Unlike achieve significant shifts in the microbial community despite

systemically available antibiotics, rifaximin allows localized being administered continuously. The low survival of probiotics

targeting of enteric pathogens, which minimizes the risk of side in the GI tract presents a big hurdle to overcome, highlighting the

effects [95]. need for efficient colonic formulations.

Probiotics Miscellaneous: small molecules

Probiotics have recently emerged as a therapeutic alternative to In 2012, Hazen et al. patented the small molecule 3,3-dimethyl-1-

POST SCREEN

antibiotics for indications such as Clostridium difficile infection butanol (DMB) and similar compounds for their ability to lower 

[96,97]. Probiotics are bacteria that can alter the microbial metab- intestinal TMA in the treatment of CVD [108]. After observing

olism by implantation or colonization and exert various health- decreased TMA production from choline in mouse cecum in vitro,

Reviews

promoting functions such as stimulating immunity when present DMB was administered to C57BL/6J mice in drinking water, result-

in sufficient numbers [98]. Various mechanisms are associated ing in a reduction of urinary and plasma TMA levels. The proposed

with the beneficial effects of probiotics; for instance, production mechanism of action was the inhibition of several TMA lyases,

of inhibitory substances such as bacteriocins, competition with including choline TMA lyase, as determined in vitro [109]. The

pathogenic bacteria for nutrients, blockage of adhesion sites for patent was further extended to acetylsalicylic acid (ASA) and its

pathogenic bacteria, degradation of toxins and blockage of toxin derivatives, because ASA showed inhibition of TMA lyase activity

receptors, among others [99]. of Proteus mirabilis – a microbe with abundant TMA lyase activity in



Methanogenic archaebiotics. Natural methanogenic Archaea of an in vitro setup [110]. Subsequent studies in C57BL/6J mice treated

the human gut have been shown to reduce TMA with hydrogen with ASA-supplemented drinking water, and in human volunteers,

for methanogenesis (Fig. 2) [100]. Although the underlying following daily administration of 81 mg ASA for 1 month, showed

chemistry was described >40 years ago in the rumen of cows significant decreases in plasma TMAO levels but omitted to men-

[101], delivering sufficient amounts of these oxygen-sensitive tion the impact on plasma/urinary TMA levels. Further, it is to note

microorganisms to the gut remains an important hurdle. The that the influence of ASA on CVD is still controversial at this point

technique was patented, but so far has not been tested in [111]. In another study, the choline analogs iodomethylcholine

humans [102]. and fluoromethylcholine (FMC) were tested for their ability to



Fecal microbial transplantation (FMT). FMT refers to the inhibit choline TMA lyase in the above-mentioned in vitro setup,

transplantation of fecal matter from a healthy donor into the using Proteus mirabilis [112]. For both, a potency of about four

GI tract of a patient. It is used in the treatment of Clostridium orders of magnitude higher than that of DMB could be observed.

difficile infection and has been tested in several other When given as a single oral dose in C57BL/6J mice, both com-

indications [103]. A recent study with two individuals affected pounds induced an almost complete reduction in urinary TMA

with TMAU, receiving FMT after pre-treatment with metroni- levels, lasting for 2 days post treatment in the case of FMC.

dazole, reported a decrease in fish odor during the first 6 Inhibition of TMA production was shown to be dose-dependent,

months in one of the patients [104]. However, perceived odor whereas no signs of toxicity following chronic exposure could be

returned to baseline after one year, suggesting that recurrent observed [112].

treatment would be necessary. Similar findings were reported Besides TMA lyase inhibitors, a few other small molecules could

by Hazen et al. while studying FMT in the context of CVD [105]. prove useful in the treatment of TMAU. The anti-ischemic and

In this work, low and high TMAO-producing mouse strains antiatherosclerotic drug meldonium is known to work through an

were chosen as FMT donors for apolipoprotin E null mice in L-carnitine lowering effect [113] by competition for selected

which resident intestinal microbes had been suppressed by a 3- and transport proteins [114]. An in vivo study in rats

week course of antibiotics. Initially, plasma TMA and TMAO showed a decrease in microbiota-dependent TMA/TMAO from

levels were lower in the recipient mice transplanted with fecal carnitine, but not from choline [114]. In another study, the

matter from the low TMAO-producing strain. Yet, the naturally occurring phytoalexin resveratrol was shown to lower

differences with the mice that had the high TMAO strain as TMAO levels in mice by inhibition of commensal microbial TMA

the donor leveled out after 16–20 weeks. However, as production via gut microbiota remodeling [115]. Another small

mentioned, a reduction in TMAO levels might not necessarily molecule potentially providing therapeutic benefit for TMAU

translate into reduction in TMA. sufferers might be the drug desmopressin [116]. Although desmo-



Other probiotic treatments. Several other probiotics have been pressin has been linked to a decrease in odor in a TMAU sufferer,

tested for their ability to lower TMA levels, some of them resveratrol is mainly used in CVD and its use in TMAU has yet to be

showing promising results. Although screening various isolates investigated, especially regarding its efficacy to reduce TMA odor

of the Enterobacteriaceae family in healthy Chinese fecal and its risk:benefit ratio upon chronic use.

samples, E. aerogens ZDY01 was found to significantly decrease Miscellaneous: oral sequestering agents

serum TMAO and cecal TMA levels in choline-loaded mice Dietary charcoal has been used in gastrointestinal detoxification

[106]. Ferguson et al. patented the use of Eubacterium strains, for a long time given its physical adsorptive mechanism [117].

www.drugdiscoverytoday.com 5

Please cite this article in press as: Schmidt, A.C., Leroux, J.C. Treatments of trimethylaminuria: where we are and where we might be heading, Drug Discov Today (2020), https://doi.org/ 10.1016/j.drudis.2020.06.026

DRUDIS 2726 1–8

 

REVIEWS Drug Discovery Today Volume 00, Number 00 June 2020

Supplementation with 750 mg of activated charcoal twice daily approach is only mentioned in perspective articles [128] and

over the course of 10 days has been reported to reduce urinary free patents [129] for which scientific data is lacking. Therefore, TMAU

TMA in TMAU patients and shift the TMA:TMAO ratio to values sufferers must adhere to simpler approaches, all of which are

similar to those found in healthy patients during the administra- thought to influence residual enzyme activity. Dietary supplemen-

tion period [118]. This observation was confirmed in another case tation with riboflavin was reported to have a decreasing effect on

[119], although after termination of the treatment the values odor intensity as well as on TMA and TMAO levels in one patient

quickly returned to baseline. When using copper chlorophyllin [130], presumably owing to increasing FMO3 activity by acting as a

instead, the decrease in free urinary TMA could be extended to cofactor. However, additional studies are lacking at this point.

Reviews

several weeks post treatment [118]. Mechanistically, the decrease is Furthermore, affected individuals are advised to avoid drugs that

linked to complexation of TMA with copper chlorophyllin, which are metabolized by FMOs [21], such as clozapine, deprenyl and



OTSCREEN POST might open avenues for the exploration of similar compounds by ranitidine, among others [121], because these might exacerbate

chemical modification (i.e., screening of various heterocyclic Cu the condition by competing for residual FMO3 activity. Tannins

complexes and their ability to complex TMA) [120]. Activated and especially brussels sprouts have shown pronounced inhibition

charcoal and copper chlorophyllin are recommended dietary sup- of human FMO3 activity when being administered over longer

plements for TMAU sufferers [121]. Recently, Kellermann et al. periods and should therefore be avoided [131,132].

patented a technology based on zeolites, claiming their ability to

decrease TMA levels in therapeutically effective amounts [122]. Concluding remarks

However, data supporting their claims were not provided and the Owing to the low prevalence and medical awareness of TMAU,

patent application status was abandoned. One of the major draw- research efforts on its treatment remain scarce. Diagnosis is the

backs of sequestering agents is their non-specific mechanism of first step to helping affected individuals to manage the condition

action, which could result in the capture of other metabolites in and its social and psychological burden. Currently, the main

addition to the target [123]. treatment is dietary restriction of TMA precursors in combination

Miscellaneous: laxatives with frequent washing using acidic soaps. This approach might

The administration of the oligosaccharide lactulose, an osmotic prove sufficient for patients showing minor and/or temporary

laxative, has been reported in the treatment of TMAU [124]. By symptoms (i.e., those suffering from secondary TMAU) but pro-

altering the intraluminal pH, lactulose can modify the gut’s flora vide limited benefit for those with primary TMAU. Although

[125]. After oral treatment in two individuals suffering from TMAU numerous treatment strategies have been described in the litera-

for 14 days, no alteration in basal TMA excretion could be ob- ture, a limited number of clinical studies have been performed,

served. However, the treatment abolished rises in urinary TMA most of them with very few patients. Antibiotics that deplete TMA-

excretion after dietary provocation as well as the accompanying producing bacteria have been shown to decrease TMA levels in

increase in odor, probably caused by the laxative effect, making it several studies but their benefit:risk ratio might not be acceptable

convenient for occasional use but not long-term given the dehy- for chronic use given the potential side effects and risk of antibiotic

drating effects [124]. resistance promotion on an individual level, especially because

TMAU does not affect vital functions. Research into TMAO as a

Targeting FMO3 disease marker in CVD has drawn the attention of the scientific

TMAU is a genetic disorder related to variants of the enzyme community toward TMA and its metabolism, sparking novel

FMO3, which is responsible for the metabolism of TMA to the research directions such as the development of choline TMA lyase

non-smelling TMAO in the liver. Restoring FMO3 activity or inhibitors. The promising results that these novel molecules have

boosting its residual activity in cases of milder forms of TMAU, is shown in various in vivo models as well as in a limited number of

an interesting approach. Enzyme replacement therapy has pro- patients bring hope to those affected by TMAU, because they could

vided treatments for previously untreatable metabolic condi- potentially lead to an effective treatment for them in the near

tions [126] but so far there are no reports of its use in TMAU, future. Q5Q6

only publications naming it as a potential treatment option

[127]. Acknowledgments

Gene therapy aimed at restoring the oxidative function of The authors gratefully acknowledge funding from the Swiss

FMO3 might one day suppress TMAU, although at present this National Science Foundation (2-77082-16).

References

1 Chang, G.W. et al. (1976) Trimethylamine-specific electrode for fish quality 5 Messenger, J. et al. (2013) A review of trimethylaminuria (fish odor syndrome). J.

control. J. Food Sci. 41, 723–724 Clin. Aesthet. Dermatol. 6, 45–48

2 van Thriel, C. et al. (2006) From chemosensory thresholds to whole body 6 Mitchell, S.C. (2005) Trimethylaminuria (fish-odour syndrome) and oral

exposures-experimental approaches evaluating chemosensory effects of malodour. Oral Dis. 11, 10–13

chemicals. Int. Arch. Occup. Environ. Health. 79, 308–321 7 Mitchell, S.C. et al. (1997) Studies on the discontinuous N-oxidation of

3 Humbert, J.R. et al. (1970) Trimethylaminuria: the fish-odour syndrome. Lancet trimethylamine among Jordanian, Ecuadorian and New Guinean populations.

296, 770–771 Pharmacogenetics 7, 45–50

4 Mackay, R.J. et al. (2011) Trimethylaminuria: causes and diagnosis of a socially 8 Ayesh, R. et al. (1993) The fish odour syndrome: biochemical, familial, and clinical

distressing condition. Clin. Biochem. Rev. 32, 33–43 aspects. BMJ Br. Med. J. 307, 655–657

6 www.drugdiscoverytoday.com

Please cite this article in press as: Schmidt, A.C., Leroux, J.C. Treatments of trimethylaminuria: where we are and where we might be heading, Drug Discov Today (2020), https://doi.org/ 10.1016/j.drudis.2020.06.026

DRUDIS 2726 1–8

 

Drug Discovery Today Volume 00, Number 00 June 2020 REVIEWS

9 Wise, P.M. et al. (2011) Individuals reporting idiopathic malodor production: 41 Mamer, O.A. et al. (2010) Measurement of urinary trimethylamine and

demographics and incidence of trimethylaminuria. Am. J. Med. 124, 1058–1063 trimethylamime oxide by direct infusion electrospray quadrupole time-of-flight

10 Todd, W.A. (1979) Psychosocial problems as the major complication of an mass spectrometry. Anal. Biochem. 406, 80–82

adolescent with trimethylaminuria. J. Pediatr. 94, 936–937 42 Al-Waiz, M. et al. (1989) Trimethylaminuria: the detection of carriers using a

11 Khan, S. and Shagufta, K. (2014) A rare case of fish odor syndrome presenting as trimethylamine load test. J. Inherit. Metab. Dis. 12, 80–85

depression. Indian J. Psychiatry 56, 185–1870 43 Fraser-Andrews, E.A et al. (2003) Fish odour syndrome with features of both

12 Busby,M.G.etal.(2004)Choline-andbetaine-defineddietsforuseinclinicalresearch primary and secondary trimethylaminuria. Clin. Exp. Dermatol. 28, 203–205

and for the management of trimethylaminuria. J. Am. Diet. Assoc. 104, 1836–1845 44 Zhang, A.Q. et al. (1999) Dietary precursors of trimethylamine in man: a pilot

13 Wilcken, B. (1993) Acid soaps in the fish odour syndrome. BMJ Br. Med. J. 307, 1993 study. Food Chem. Toxicol. 37, 515–520

14 Zeisel, S.H. et al. (2003) Concentrations of choline-containing compounds and 45 Zeisel, S.H. et al. (1983) Formation of methylamines from ingested choline and

betaine in common foods. J. Nutr. 133, 1302–1307 lecithin. J. Pharmacol. Exp. Ther. 225, 320–324

15 Dyer, W.J. (1952) Trimethylamine oxide content of fish and marine invertebrates. 46 Mitchell, A.D. et al. (1979) Metabolism of betaine in the ruminant. J. Anim. Sci. 49,

J. Fish. Res. Board Canada 8c, 314–324 764–774

16 Koeth, R.A. et al. (2013) Intestinal microbiota metabolism of l-carnitine, a nutrient 47 Zeisel, S.H. et al. (1989) Formation of aliphatic amine precursors of N-

in red meat, promotes atherosclerosis. Nat. Med. 19, 576–585 nitrosodimethylamine after oral administration of choline and choline analogues

POST SCREEN

17 Fennema, D. et al. (2016) Trimethylamine and trimethylamine N-oxide, a flavin- in the rat. Food Chem. Toxicol. 27, 31–34



containing monooxygenase 3 (FMO3)-mediated host-microbiome metabolic axis 48 de la Huerge, J. and Popper, H. (1951) Urinary excretion of choline metabolites

implicated in health and disease. Drug Metab. Dispos. 44, 1839–1850 following choline administration in normals and patients with hepatobiliary

18 Dolphin, C.T. et al. (1997) in flavin-containing mono- diseases. J. Clin. Invest. 30, 463–470

Reviews

oxygenase 3 gene, FMO3, underlies fish-odour syndrome. Nat. Genet. 17, 491–494 49 Abdelmalek, M.F. et al. (2001) Betaine, a promising new agent for patients with

19 Lang, D.H. et al. (1998) Isoform specificity of trimethylamine N-oxygenation by nonalcoholic steatohepatitis: results of apilot study. Am. J. Gastroenterol. 96, 2711–2717

human flavin-containing monooxygenase (FMO) and P450 enzymes: selective 50 Ufnal, M. et al. (2015) TMAO: a small molecule of great expectations. Nutrition 31,

catalysis by fmo3. Biochem. Pharmacol. 56, 1005–1012 1317–1323

20 Hisamuddin, I.M. and Yang, V.W. (2007) Genetic polymorphisms of human 51 Kuczler, F.J. et al. (1977) Choline influx across the brush border of guinea pig

flavin-containing monooxygenase 3: implications for drug metabolism and jejunum. BBA Biomembr. 465, 131–137

clinical perspectives. Pharmacogenomics 8, 635–643 52 Zeisel, S.H. et al. (1989) Conversion of dietary choline to trimethylamine and

21 Philips, I.R. et al. (2007) The flavin-containing monoooxygenases (FMOs): genetic dimethylamine in rats: dose-response relationship. J. Nutr. 119, 800–804

variation and its consequences for the metabolism of therapeutic drugs. Curr. 53 Buchman, A.L. et al. (1995) Choline deficiency: a cause of hepatic steatosis during

Pharmacogenomics 5, 292–313 parenteral nutrition that can be reversed with intravenous choline

22 Cashman, J. et al. (2005) Biochemical and clinical aspects of the human flavin- supplementation. Hepatology 22, 1399–1403

containing monooxygenase form 3 (FMO3) related to trimethylaminuria. Curr. 54 Kelly, R.H. and Yancey, P.H. (1999) High contents of trimethylamine oxide

Drug Metab. 4, 151–170 correlating with depth in deep-sea teleost fishes, skates, and decapod crustaceans.

23 Hernandez, D. et al. (2003) Trimethylaminuria and a human FMO3 mutation Biol. Bull. 196, 18–25

database. Hum. Mutat. 22, 209–213 55 Chalmers, R.A. et al. (2006) Diagnosis and management of trimethylaminuria

24 Phillips, I.R. and Shephard, E.A. (2020) Flavin-containing monooxygenase 3 (FMO3 deficiency) in children. J. Inherit. Metab. Dis. 29, 162–172

(FMO3): genetic variants and their consequences for drug metabolism and disease. 56 Holmes, H.C. (1997) Choline and L-carnitine as precursors of trimethylamine.

Xenobiotica 50, 19–33 Biochem. Soc. Trans. 1997, 96S

25 Yamazaki, H. and Shimizu, M. (2013) Survey of variants of human flavin- 57 Rebouche, C.J. and Chenard, C.A. (1991) Metabolic fate of dietary carnitine in

containing monooxygenase 3 (FMO3) and their drug oxidation activities. Biochem. human adults: identification and quantification of urinary and fecal metabolites.

Pharmacol. 85, 1588–1593 J. Nutr. 121, 539–546

26 Growdon, J.H. et al. (1977) Huntington’s disease: clinical and chemical effects of 58 Zhu, Y. et al. (2014) Carnitine metabolism to trimethylamine by an unusual

choline administration. Ann. Neurol. 1, 418–422 Rieske-type oxygenase from human microbiota. Proc. Natl. Acad. Sci. U. S. A. 111,

27 Etienne, P. et al. (1978) Clinical effects of choline in Alzheimer’s disease. Lancet 4268–4273

311, 508–509 59 Koeth, R.A. et al. (2019) L-Carnitine in omnivorous diets induces an atherogenic

28 Mitchell, S. et al. (1999) Trimethylamine and foetor hepaticus. Scand. J. gut microbial pathway in humans. J. Clin. Invest. 129, 373–387

Gastroenterol. 34, 524–528 60 Seline, K.G. and Johein, H. (2007) The determination of L-carnitine in several food

29 Yamazaki, H. et al. (2005) Mild trimethylaminuria observed in a Japanese cohort samples. Food Chem. 105, 793–804

with liver damage. Am. J. Med. 118, 803–805 61 Wu, W-K. et al. (2015) Dietary allicin reduces transformation of L-carnitine to

30 Shimizu, M. et al. (2007) Transient trimethylaminuria related to menstruation. TMAO through impact on gut microbiota. J. Funct. Foods 15, 408–417

BMC Med. Genet. 8, 2 62 Falls, J.G. et al. (1995) Gender differences in hepatic expression of flavin-

31 Zhang, A.Q. et al. (1996) Exacerbation of symptoms of fish-odour syndrome during containing monooxygenase isoforms (FMO1, FMO3, and FMO5) in mice. J.

menstruation. Lancet 348, 1740–1741 Biochem. Toxicol. 10, 171–177

32 Ruocco, V. et al. (1989) An unusual case of trimethylaminuria. Br. J. Dermatol. 120, 63 Oliveira, A. et al. (2015) Fish malodour syndrome in a child. BMJ Case Rep. 2015,

459–461 bcr2014207002

33 Ayesh, R. et al. (1995) Dysfunctional N-oxidation of trimethylamine and the 64 Schmidt, A.C. et al. (2020) Engineered polymersomes for the treatment of fish odor

influence of testosterone treatment in man. Pharmacogenetics 5, 244–246 syndrome: a first randomized double blind olfactory study. Adv. Sci. 7, 1903697

34 Zhang, A.Q. et al. (1995) Fish odour syndrome: verification of carrier detection test. 65 Garg, P. et al. (2013) A pilot study of the effect of (E, E)-2,4-undecadienal on the

J. Inherit. Metab. Dis. 18, 669–674 offensive odour of trimethylamine. JIMD Rep. 8, 11–15

35 Al-Waiz, M. et al. (1987) Disclosure of the metabolic retroversion of 66 Kaneko, T. et al. (1987) Lethal effects of a linoleic acid hydroperoxide and its

trimethylamine N-oxide in humans: a pharmacogenetic approach. Clin. autoxidation products, unsaturated aliphatic aldehydes, on human diploid

Pharmacol. Ther. 42, 608–612 fibroblasts. Chem. Biol. Interact. 63, 127–137

36 Zeisel, S.H. and Da Costa, K.A. (1986) Increase in human exposure to methylamine 67 Wang, Z. et al. (2011) Gut flora metabolism of phosphatidylcholine promotes

precursors of N-nitrosamines after eating fish. Cancer Res. 46, 6136–6138 cardiovascular disease. Nature 472, 57–65

37 Podadera, P. et al. (2005) Diagnosis of suspected trimethylaminuria by NMR 68 Tang, W.H.W. et al. (2013) Intestinal microbial metabolism of

spectroscopy. Clin. Chim. Acta 351, 149–154 phosphatidylcholine and cardiovascular risk. N. Engl. J. Med. 368, 1575–1584

38 Maschke, S. et al. (1997) 1H-NMR analysis of trimethylamine in urine for the 69 Mente, A. et al. (2015) The relationship between trimethylamine-N-oxide and

diagnosis of fish-odour syndrome. Clin. Chim. Acta 263, 139–146 prevalent cardiovascular disease in a multiethnic population living in Canada.

39 Neyer, P. et al. (2020) Derivatization-free determination of short-chain volatile Can. J. Cardiol. 31, 1189–1194

amines in human plasma and urine by headspace gas chromatography-mass 70 Tang, W.H.W. et al. (2017) Increased trimethylamine N-oxide portends high

spectrometry. J. Clin. Lab. Anal. 34, e23062 mortality risk independent of glycemic control in patients with type 2 diabetes

40 Mills, G.A. et al. (1999) Quantitative determination of trimethylamine in urine by mellitus. Clin. Chem. 63, 297–306

solid-phase microextraction and gas chromatography-mass spectrometry. J. 71 Senthong, V. et al. (2016) Trimethylamine N-oxide and mortality risk in patients

Chromatogr. B Biomed. Sci. Appl. 723, 281–285 with peripheral artery disease. J. Am. Heart Assoc. 5, 2016

www.drugdiscoverytoday.com 7

Please cite this article in press as: Schmidt, A.C., Leroux, J.C. Treatments of trimethylaminuria: where we are and where we might be heading, Drug Discov Today (2020), https://doi.org/ 10.1016/j.drudis.2020.06.026

DRUDIS 2726 1–8

 

REVIEWS Drug Discovery Today Volume 00, Number 00 June 2020

72 Shafi, T. et al. (2017) Trimethylamine N-oxide and cardiovascular events in trimethylaminuria or of and the prevention of cardiovascular

hemodialysis patients. J. Am. Soc. Nephrol. 28, 321–331 diseases. WO2014082773A1.

73 Meyer, K. and Shea, J. (2017) Dietary choline and betaine and risk of CVD: a 103 Cohen, N.A. and Maharshak, N. (2017) Novel indications for fecal microbial

systematic review and meta-analysis of prospective studies. Nutrients 9, 711 transplantation: update and review of the literature. Dig. Dis. Sci. 62, 1131–1145

74 Gluick, T.C. and Yadav, S. (2003) Trimethylamine N-oxide stabilizes RNA tertiary 104 Lahtinen, P. et al. (2017) Faecal microbiota transplantation in patients with

structure and attenuates the denaturating effects of urea. J. Am. Chem. Soc. 125, Clostridium difficile and significant comorbidities as well as in patients with new

4418–4419 indications: a case series. World J. Gastroenterol. 23, 7174–7184

75 Ma, J. et al. (2014) Microscopic insights into the protein-stabilizing effect of 105 Gregory, J.C. et al. (2015) Transmission of atherosclerosis susceptibility with gut

trimethylamine N-oxide (TMAO). Proc. Natl. Acad. Sci. U. S. A. 111, 8476–8481 microbial transplantation. J. Biol. Chem. 290, 5647–5660

76 Al-Waiz, M. et al. (1992) The exogenous origin of trimethylamine in the mouse. 106 Qiu, L. et al. (2017) Enterobacter aerogenes ZDY01 attenuates choline-induced

Reviews

Metabolism 41, 135–136 trimethylamine N-oxide levels by remodeling gut microbiota in mice. J. Microbiol.

77 Seim, H. et al. (1985) Catabolic pathways for high-dosed L(-)- or D(+)-carnitine in Biotechnol. 27, 1491–1499

germ-free rats. Biol. Chem. 366, 1017–1021 107 Krzycki, J.A. et al. (2016) Eubacterium probiotics and methods of treating or 

OTSCREEN POST

78 Matoori, S. and Leroux, J-C. (2015) Recent advances in the treatment of preventing heart disease. WO2017218889A1.

hyperammonemia. Adv. Drug Deliv. Rev. 90, 55–68 108 Hazen, S.L. et al. (2013) Treatment and prevention of cardiovascular disease and

79 Bell, B.G. et al. (2014) A systematic review and meta-analysis of the effects of thrombosis. US9168233B2.

antibiotic consumption on antibiotic resistance. BMC Infect. Dis. 14, 13 109 Wang, Z. et al. (2015) Non-lethal inhibition of gut microbial trimethylamine

80 Costelloe, C. et al. (2010) Effect of antibiotic prescribing in primary care on production for the treatment of atherosclerosis. Cell 163, 1585–1595

antimicrobial resistance in individual patients: systematic review and meta- 110 Hazen, S.L. (2015) Treating and preventing disease with tma and tmao lowering

analysis. BMJ 340, 1120 agents US20160089386A1.

81 Blaser, M.J. (2016) Antibiotic use and its consequences for the normal microbiome. 111 Raber, I. et al. (2019) The rise and fall of aspirin in the primary prevention of

Science 352, 544–545 cardiovascular disease. Lancet 393, 2155–2167

82 Cox, L.M. and Blaser, M.J. (2015) Antibiotics in early life and obesity. Nat. Rev. 112 Roberts, A.B. et al. (2018) Development of a gut microbe-targeted nonlethal

Endocrinol. 11, 182–190 therapeutic to inhibit thrombosis potential. Nat. Med. 24, 1407–1417

83 Treacy, E. et al. (1995) Trimethylaminuria, fish odour syndrome: a new method of 113 Dambrova, M. et al. (2013) Meldonium decreases the diet-increased plasma levels

detection and response to treatment with metronidazole. J. Inherit. Metab. Dis. 18, of trimethylamine N-oxide, a metabolite associated with atherosclerosis. J. Clin.

306–312 Pharmacol. 53, 1095–1098

84 Baumgartner, M.R. et al. (2014) Proposed guidelines for the diagnosis and 114 Kuka, J. et al. (2014) Suppression of intestinal microbiota-dependent production of

management of methylmalonic and . Orphanet J. Rare Dis. 9, pro-atherogenic trimethylamine N-oxide by shifting L-carnitine microbial

130 degradation. Life Sci. 117, 84–92

85 Vilstrup, H. et al. (2014) Hepatic encephalopathy in chronic liver disease: 2014 115 Chen, M. et al. (2016) Atherosclerosis by regulating TMAO synthesis and bile acid

practice guideline by the American association for the study of liver diseases and metabolism via remodeling of the gut microbiota. MBio 7, 1–14

the European association for the study of the liver. Hepatology 60, 715–735 116 Werner, D. (2017) Die trimethylaminurie (Fischgeruchsyndrom) - hinweise auf

86 Ulman, C.A. et al. (2014) Fish odor syndrome: a case report of trimethylaminuria. eine neue therapiemo¨glichkeit mit desmopressin. Aktuelle Urol. 38, 406–407

Dermatol. Online J. 20, 2014 117 Bonanome, A. and Grundy, S.M. (1988) Gastrointestinal clearance of drugs with

87 Arseculeratne, G. et al. (2007) Trimethylaminuria (fish-odor syndrome). Arch. activated charcoal. N. Engl. J. Med. 307, 1244–1248

Dermatol. 143, 81–84 118 Yamazaki, H. et al. (2004) Effects of the dietary supplements, activated charcoal

88 Conn, H.O. et al. (1977) Comparison of lactulose and neomycin in the treatment and copper chlorophyllin, on urinary excretion of trimethylamine in Japanese

of chronic portal-systemic encephalopathy: a double blind controlled trial. trimethylaminuria patients. Life Sci. 74, 2739–2747

Gastroenterology 72, 573–583 119 Girdwichai, N. et al. (2015) Trimethylaminuria: report of two cases in Ramathibodi

89 Leise, M.D. et al. (2014) Management of hepatic encephalopathy in the hospital. hospital. Ann. Transl. Med. 3, AB134

Mayo Clin. Proc. 89, 241–253 120 Dashwood, R. et al. (1996) Study of the forces stabilizing complexes between

90 Last, P.M. and Sherlock, S. (1960) Systemic absorption of orally administered chlorophylls and heterocyclic amine mutagens. Environ. Mol. Mutagen. 27, 211–

neomycin in liver disease. N. Engl. J. Med. 262, 385–389 218

91 Danks, D.M. et al. (1976) Trimethylaminuria: diet does not always control the fishy 121 Shephard, E.A. et al. (2015) Clinical utility gene card for: trimethylaminuria –

odor. N. Engl. J. Med. 295, 962 update 2014. Eur. J. Hum. Genet. 23 1269–1269

92 Asatoor, A.M. et al. (1967) Metabolic effects of oral neomycin. Clin. Sci. 33, 111– 122 Kellermann, G.H. and Jirikowski, G.F. (2018) Compositions comprising a zeolite

124 and use thereof for the treatment of trimethylaminuria. US20180200291A1.

93 Stremmel, W. et al. (2017) Blood trimethylamine-N-oxide originates from 123 Neuvonen, P.J. and Olkkola, K.T. (1988) Oral activated charcoal in the treatment of

microbiota mediated breakdown of phosphatidylcholine and absorption from intoxications: role of single and repeated doses. Med. Toxicol. Adverse Drug Exp 3,

small intestine. PLoS One 12, e0170742 33–58

94 Descombe, J.J. et al. (1994) Pharmacokinetic study of rifaximin after oral 124 Pike, M.G. et al. (1989) Lactulose in trimethylaminuria, the fish-odour syndrome.

administration in healthy volunteers. Int. J. Clin. Pharmacol. Res. 14, 51–56 Helv. Paediatr. Acta 43, 345–348

95 Scarpignato, C. and Pelosini, I. (2005) Rifaximin, a poorly absorbed antibiotic: 125 Schumann, C. (2002) Medical, nutritional and technological properties of

pharmacology and clinical potential. Chemotherapy 51, 36–66 lactulose: an update. Eur. J. Nutr. 41, i17–i25

96 Fuller, R. (1989) Probiotics in man and animals. J. Appl. Bacteriol. 66, 365–378 126 Arnold, G.L. (2018) Inborn errors of metabolism in the 21st century: past to

97 Aureli, P. et al. (2011) Probiotics and health: an evidence-based review. Pharmacol. present. Ann. Transl. Med. 6 467–467

Res. 63, 366–376 127 Mitchell, S.C. and Smith, R.L. (2001) Trimethylaminuria: the fish malodor

98 Schrezenmeir, J. and de Vrese, M. (2001) Probiotics, prebiotics, and synbiotics- syndrome. Drug Metab. Dispos. 29, 517–521

approaching a definition. Am. J. Clin. Nutr. 73, 361–364 128 Mitchell, S.C. (1996) The fish-odor syndrome. Perspect. Biol. Med. 39, 514–526

99 Hill, C. et al. (2014) Expert consensus document: the international scientific 129 Patzel, V. (2016) Functional gene replacement therapy. WO2017018937A1.

association for probiotics and prebiotics consensus statement on the scope and 130 Manning, N.J. et al. (2011) Riboflavin-responsive trimethylaminuria in a patient

appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 11, 506–514 with on betaine therapy. JIMD Rep. 2011, 71–75

100 Bruge`re, J-F. et al. (2014) Archaebiotics. Gut Microbes 5, 5–10 131 Fenwick, G.R. et al. (1983) Are brassica vegetables aggravating factors in

101 Neill, A.R. et al. (1978) Conversion of choline methyl groups through trimethylaminuria (fish odour syndrome)? Lancet 322, 916

trimethylamine into methane in the rumen. Biochem. J. 170, 529–535 132 Cashman, J.R. et al. (1999) In vitro and in vivo inhibition of human flavin-

102 Brugere, J-F. et al. (2013) Use of microorganisms for reducing the level of containing monooxygenase form 3 (FMO3) in the presence of dietary indoles.

trimethylamine in a human body cavity, in particular for the treatment of Biochem. Pharmacol. 58, 1047–1055

8 www.drugdiscoverytoday.com

Please cite this article in press as: Schmidt, A.C., Leroux, J.C. Treatments of trimethylaminuria: where we are and where we might be heading, Drug Discov Today (2020), https://doi.org/ 10.1016/j.drudis.2020.06.026