Chenodeoxycholic acid (CDCA), also known as chenodiol, is a primary bile acid naturally synthesized in the human liver from cholesterol through a series of enzymatic steps.[1] It has the chemical formula C24H40O4 and a molecular weight of 392.572 g/mol, featuring a steroid nucleus with hydroxyl groups at the 3α and 7α positions.[1] As one of the two main primary bile acids—alongside cholic acid—CDCA is secreted into bile, where it acts as a physiological detergent to emulsify and facilitate the digestion and absorption of dietary fats, sterols, and fat-soluble vitamins in the small intestine.[1] It undergoes conjugation with glycine or taurine in the liver before release and is efficiently reabsorbed via the enterohepatic circulation, with about 95% recycled daily.[2]In its biological role, CDCA helps regulate cholesterolhomeostasis by promoting its excretion from the liver into bile and modulating bile flow and lipid secretion.[1] A portion of CDCA is metabolized by gut bacteria into the secondary bile acid lithocholic acid, which is largely excreted in feces, contributing to cholesterol elimination.[2] This bile acid also activates nuclear receptors such as the farnesoid X receptor (FXR), influencing genes involved in bile acid synthesis, transport, and inflammation.[2]Medically, chenodeoxycholic acid is approved for the dissolution of small, radiolucent cholesterol gallstones in patients with functioning gallbladders who are at high risk for surgery or elect non-surgical options.[3] Treatment typically involves oral doses of 13–16 mg/kg daily in divided doses for up to two years, achieving partial or complete gallstone dissolution in 15–30% of cases, though recurrence rates can reach 50% upon discontinuation.[3] It is also indicated for treating cerebrotendinous xanthomatosis (CTX), a rare autosomal recessive disorder caused by sterol 27-hydroxylase deficiency, where it replaces deficient bile acids, normalizes cholestanol levels, and inhibits overproduction of atypical bile acids by suppressing cholesterol 7α-hydroxylase (CYP7A1); it is approved by regulatory agencies such as the EMA and FDA (as of 2025 for adults in the US).[4][5] For CTX, dosing starts at 5–15 mg/kg/day in pediatrics and up to 750–1,000 mg/day in adults, with monitoring of plasma cholestanol and liver function.[4]The therapeutic mechanism of CDCA involves decreasing hepatic cholesterol synthesis via inhibition of HMG-CoA reductase and reducing biliary cholesterol saturation to promote gallstone dissolution.[2] However, it can cause dose-related diarrhea due to its cathartic effects and transient elevations in serum aminotransferases in up to 30% of patients, with rare instances of clinically apparent hepatotoxicity linked to its metabolite lithocholic acid.[3] Largely supplanted by ursodeoxycholic acid for gallstone therapy due to better tolerability, CDCA remains a key treatment for CTX and select bile acid deficiencies.[3]
Chemical properties
Molecular structure
Chenodeoxycholic acid (CDCA) is a primary bile acid with the molecular formula C24H40O4 and a molecular weight of 392.58 g/mol.[6] Its systematic IUPAC name is 3α,7α-dihydroxy-5β-cholan-24-oic acid, reflecting its classification as a dihydroxy derivative of the cholanoic acid backbone.[6]The molecule features a characteristic steroidnucleus composed of four fused rings: three six-membered rings (A, B, and C) and one five-membered ring (D), forming a curved core derived from cholesterol.[7] Attached to this nucleus are two hydroxyl groups oriented at the 3α and 7α positions on rings A and B, respectively, which contribute to its polarity, and an eight-carbon side chain at C-17 terminating in a carboxylic acid group at C-24.[6] These structural elements endow CDCA with amphipathic properties, where the hydrophobic steroid rings contrast with the hydrophilic hydroxyl and carboxyl moieties.[8]In comparison to cholic acid, another primary bile acid, CDCA lacks the additional hydroxyl group at the 12α position on ring C, resulting in only two hydroxyl groups instead of three.[9] This difference influences the molecule's overall hydrophobicity and solubility characteristics.The stereochemistry of CDCA is defined by the 5β configuration of the A/B ring junction, which imparts a cis fusion between rings A and B, along with α-orientation (axial, below the plane) of the hydroxyl groups at C-3 and C-7, and specific chiral centers at C-8 (β), C-9 (α), C-10 (β), C-13 (β), C-14 (α), C-17 (β), and C-20 (R).[6] In three-dimensional representations, this arrangement positions the polar groups on the concave α-face of the steroid nucleus, enhancing its detergent-like behavior in micelle formation and contributing to its biological solubility in aqueous environments.[10]
Physical and chemical characteristics
Chenodeoxycholic acid is a white crystalline powder that may absorb humidity under ambient conditions.[11]It exhibits low solubility in water, with a reported value of approximately 0.09 mg/mL, rendering it practically insoluble, but it dissolves readily in organic solvents such as ethanol (up to 20 mg/mL), methanol, acetone, and glacial acetic acid.[12][13]Solubility increases in alkaline solutions owing to the ionization of the carboxyl group.[11] It is weakly soluble in ether and ethyl acetate but insoluble in petrol ether and benzene.[11]The compound has a melting point of 165–167 °C.[14] The pKa of its carboxylic acid group is approximately 4.6.[2]Chenodeoxycholic acid demonstrates good chemical stability, remaining unchanged for up to 24 months when stored at room temperature without special packaging requirements; it is unaffected by exposure to light and resists degradation under acidic, basic, or oxidizing conditions, although thermal decomposition occurs above 130 °C.[11]Commercially, chenodeoxycholic acid is produced through extraction from animal bile sources, such as porcine by-products, or via semi-synthetic routes involving microbial transformation of precursor bile acids like cholic acid.[15]Total synthesis from cholesterol is possible but less common for large-scale production due to complexity.[16]
Chenodeoxycholic acid (CDCA) is a primary bile acid synthesized in hepatocytes from cholesterol primarily via the classic (neutral) pathway, which accounts for the majority of bile acid production in the liver. This pathway initiates with the rate-limiting step catalyzed by cholesterol 7α-hydroxylase (CYP7A1), a microsomal enzyme that hydroxylates cholesterol at the 7α position to form 7α-hydroxycholesterol. Subsequent enzymatic modifications oxidize this intermediate via 3β-hydroxy-Δ⁵-C₂₇-steroid dehydrogenase (HSD3B7) to 7α-hydroxy-4-cholesten-3-one, followed by stereospecific reduction of the Δ⁴ double bond by Δ⁴-3-oxosteroid-5β-reductase (AKR1D1) and oxidation at the 3α position by 3α-hydroxysteroid dehydrogenase (AKR1C4), yielding 3α,7α-dihydroxy-5β-cholestanoic acid (DHCA). The side chain is then oxidized at the 27 position by sterol 27-hydroxylase (CYP27A1), a mitochondrial and peroxisomal enzyme, leading to oxidative cleavage and formation of unconjugated CDCA. Unlike the parallel synthesis of cholic acid, CDCA production bypasses 12α-hydroxylation by sterol 12α-hydroxylase (CYP8B1).[17][18][19]The biosynthetic pathway is subject to negative feedback regulation primarily through the farnesoid X receptor (FXR), a nuclear receptor activated by bile acids such as CDCA. FXR activation in the liver induces small heterodimer partner (SHP), which represses CYP7A1 transcription by inhibiting liver receptor homolog-1 (LRH-1). Additionally, intestinal FXR activation by reabsorbed bile acids upregulates fibroblast growth factor 19 (FGF19), which travels to the liver and further inhibits CYP7A1 expression via signaling through fibroblast growth factor receptor 4 (FGFR4) and β-klotho, maintaining bile acid homeostasis.[17][18]In humans, CDCA comprises approximately 40% of the total bile acid pool, reflecting its significant contribution alongside cholic acid. A minor alternative (acidic) pathway, initiated extrahepatically by CYP27A1-mediated 27-hydroxylation of cholesterol to 27-hydroxycholesterol followed by 7α-hydroxylation via oxysterol 7α-hydroxylase (CYP7B1), converges with the classic pathway to produce CDCA but plays a limited role in overall synthesis, particularly in adults.[20][17][18]
Physiological functions
Chenodeoxycholic acid (CDCA), a primary bile acid synthesized in the liver, serves as a key component of bile salts that facilitate the emulsification and absorption of dietary lipids in the small intestine. By forming micelles with cholesterol, phospholipids, and monoglycerides, CDCA solubilizes these hydrophobic molecules, enabling their efficient uptake by enterocytes and preventing their precipitation in the aqueous environment of the intestinal lumen.[21] This process is essential for the digestion of fats and the absorption of fat-soluble vitamins, contributing to overall nutrient homeostasis.[22]CDCA participates in the enterohepatic circulation, a highly efficient recycling mechanism that conserves bile acids within the body. Approximately 95% of secreted bile acids, including CDCA, are reabsorbed in the terminal ileum via the apical sodium-dependent bile acid transporter (ASBT, also known as SLC10A2), which actively transports them into enterocytes against a concentration gradient using sodium co-transport.[23] From there, CDCA is shuttled to the portal vein and returned to the liver via basolateral transporters like OSTα/OSTβ, allowing it to be recycled 10-12 times daily to support repeated cycles of bile secretion and intestinal function.[24] This recirculation minimizes the need for de novo synthesis, with only about 5% of the bile acid pool lost in feces each day.[23]As a ligand for the nuclear receptor farnesoid X receptor (FXR), CDCA exerts regulatory effects on metabolic pathways. Upon binding to FXR in hepatocytes and enterocytes, CDCA induces the expression of small heterodimer partner (SHP), which suppresses transcription of the rate-limiting enzyme in bile acid synthesis, cholesterol 7α-hydroxylase (CYP7A1), thereby providing negative feedback to maintain bile acid homeostasis.[25] FXR activation by CDCA also promotes glucose and lipid metabolism by enhancing insulin sensitivity, inhibiting gluconeogenesis, and stimulating fatty acid oxidation in the liver.[26] Additionally, CDCA-mediated FXR signaling modulates inflammation by repressing pro-inflammatory cytokines such as TNF-α and IL-1β in immune cells and endothelial tissues.[17]CDCA activates the G protein-coupled membrane receptor TGR5 (also known as GPBAR1), which mediates diverse physiological responses independent of FXR. In enteroendocrine L cells, TGR5 stimulation by CDCA promotes the secretion of glucagon-like peptide-1 (GLP-1), an incretin hormone that enhances insulin release and suppresses glucagon, aiding postprandial glucose control.[27] In brown adipose tissue and skeletal muscle, CDCA-induced TGR5 signaling increases energy expenditure through cAMP-dependent activation of type 2 iodothyronine deiodinase (DIO2), which converts thyroxine to active triiodothyronine, thereby boosting thermogenesis.[28] Furthermore, TGR5 activation by CDCA exerts anti-inflammatory effects in macrophages by inhibiting NF-κB signaling and reducing the production of pro-inflammatory mediators like IL-6 and MCP-1.[29]Within the bile acid pool, CDCA contributes to compositional diversity and influences the overall hydrophobicity index, which affects detergent properties and toxicity. Compared to the more hydrophilic cholic acid, CDCA is relatively hydrophobic, enhancing the pool's ability to solubilize cholesterol while balancing cytotoxicity risks associated with excessive hydrophobicity.[22] This property supports cholesterolhomeostasis by promoting the biliary excretion of free cholesterol into bile, where it is solubilized for elimination via the feces, thus preventing hypercholesterolemia.[21] Through these mechanisms, CDCA helps regulate systemic lipid levels and maintains intestinal barrier integrity.[30]
Clinical uses
Gallstone dissolution
Chenodeoxycholic acid, marketed under the brand name Chenodiol, received FDA approval in 1983 for the oral dissolution of small, radiolucent, non-calcified cholesterol gallstones measuring less than 15 mm in diameter, specifically in patients with well-opacifying gallbladders for whom elective surgery would otherwise be considered.[31] This approval was based on clinical evidence demonstrating its ability to nonsurgically treat suitable gallstones by altering bile composition.[3]The therapeutic mechanism involves suppressing hepatic cholesterol synthesis and secretion into bile, which decreases the biliary cholesterol saturation index (SI) to below 1, rendering bile unsaturated and capable of solubilizing cholesterol from gallstones over time.[32] This desaturation promotes gradual dissolution, particularly effective for cholesterol-rich stones that appear radiolucent on imaging.Standard dosing regimen is 13 to 16 mg/kg body weight per day, administered orally in two divided doses (morning and evening), typically for 6 to 24 months depending on stone size and response.[33] Treatment success, defined as complete dissolution, ranges from 40% to 60% for small stones under 10 mm, though rates are lower for larger ones; for instance, the landmark National Cooperative Gallstone Study (NIH-NIDDK, 1980s) reported complete dissolution in 13.9% of patients after 2 years of therapy at 750 mg/day.[34][35]Appropriate patient selection is critical, favoring those with floating gallstones on oral cholecystography, which indicates a high cholesterol content and low density conducive to dissolution.[3]Therapy is contraindicated in cases of calcified (radiopaque) stones, stones exceeding 15 mm, nonvisualizing gallbladders, or conditions like hemolytic anemia that increase pigment stone risk.[36]Combination therapy with ursodeoxycholic acid (UDCA) at equimolar doses has shown improved efficacy, achieving higher dissolution rates (up to 70% in some trials) while minimizing chenodeoxycholic acid-associated side effects like diarrhea and hepatotoxicity.[37] This approach leverages the complementary actions of both bile acids to more effectively desaturate bile and inhibit stone formation.80004-3/fulltext)
Cerebrotendinous xanthomatosis treatment
Chenodeoxycholic acid (CDCA) is the standard treatment for cerebrotendinous xanthomatosis (CTX), a rare autosomal recessive disorder caused by mutations in the CYP27A1 gene, which encodes sterol 27-hydroxylase and leads to deficient production of chenodeoxycholic acid, resulting in accumulation of cholestanol in plasma and tissues.[38] In CTX, the lack of CDCA disrupts the normal negative feedback on bile acid synthesis via the farnesoid X receptor (FXR), promoting excessive production of abnormal bile alcohols and cholestanol through alternative pathways, including defective 26-hydroxylation.[5] CDCA therapy restores the deficient bile acid, activates FXR to downregulate cholesterol 7α-hydroxylase (CYP7A1), suppresses the overproduction of toxic bile alcohols, and reduces cholestanol accumulation.[5] CDCA received orphan drug designation in the United States in 2007 and in the European Union in 2014, with EU approval for CTX treatment in 2017 and US approval as chenodiol (Ctexli) in 2025.[39][40]Therapeutically, CDCA normalizes bile acid synthesis, reduces plasma and tissue cholestanol levels by 70-90%, and alleviates neurological and extraneurological symptoms in most patients.[41] It halts disease progression, shrinks xanthomas, and improves ataxia, cognition, pyramidal signs, and cerebellar function, with benefits emerging over months to years of lifelong therapy.[42] The recommended adult dose is 750 mg/day orally (250 mg three times daily), with adjustments for children based on body weight (typically 5-15 mg/kg/day divided into three doses); treatment is lifelong to maintain suppression of abnormal metabolites.[5][40]Pivotal evidence from a 1984 study of 17 CTX patients showed that CDCA (750 mg/day) reduced mean plasma cholestanol threefold, cleared dementia in 10 patients, resolved peripheral neuropathy in 6 of 7, and improved cerebellar and pyramidal signs in most, arresting or reversing progression.[42] Long-term follow-ups confirm sustained efficacy: a 2025 nationwide study of 86 patients reported 100% stabilization or improvement in those starting therapy before age 28, with reduced plasma cholestanol in all and halted progression of pyramidal and cerebellar symptoms, though psychiatric features like psychosis were less responsive.[43] Monitoring involves regular plasma cholestanol levels as a biomarker of efficacy (target: normalization) and MRI to assess brain lesions, alongside liver function tests.[5][44]
Other established applications
Chenodeoxycholic acid is authorized by the European Medicines Agency for the treatment of cerebrotendinous xanthomatosis (CTX), an inborn error of primary bile acid synthesis due to sterol 27-hydroxylase deficiency. It has also shown efficacy in other rare cholestatic disorders characterized by defective bile acid production, such as oxysterol 7α-hydroxylase deficiency, a condition presenting with progressive intrahepatic cholestasis, neurological impairment, and liver dysfunction in infancy. Oral chenodeoxycholic acid therapy has demonstrated efficacy in normalizing bile acid profiles, resolving cholestasis, and improving clinical outcomes, including liver enzyme levels and growth parameters, when initiated early. Doses typically range from 10 to 15 mg/kg/day, administered orally, with monitoring of liver function tests to ensure safety.[45]Historical investigations explored chenodeoxycholic acid for hypercholesterolemia based on its potential to induce cholesterol 7α-hydroxylase, the rate-limiting enzyme in bile acid synthesis, thereby promoting cholesterol conversion to bile acids and reducing low-density lipoprotein levels. However, clinical studies showed limited efficacy in lowering serum LDL cholesterol, with some reports indicating no change or even paradoxical increases due to reduced LDL clearance, leading to its diminished use in favor of more effective lipid-lowering agents.[46]Due to risks of hepatotoxicity, including elevated transaminases, chenodeoxycholic acid is contraindicated in patients with pre-existing cirrhosis or acute hepatitis, overlapping with precautions in cholestatic applications where baseline liver assessments are required.[3]
Pharmacology and safety
Mechanism of action
Chenodeoxycholic acid (CDCA) primarily exerts its pharmacological effects through activation of the farnesoid X receptor (FXR), a nuclear receptor that serves as a key regulator of bile acidhomeostasis. Upon binding to FXR in hepatocytes and enterocytes, CDCA induces the transcriptional upregulation of the small heterodimer partner (SHP), a nuclear receptor corepressor that inhibits liver receptor homolog-1 (LRH-1)-mediated activation of the cholesterol 7α-hydroxylase gene (CYP7A1). This repression of CYP7A1, the rate-limiting enzyme in the classic bile acid biosynthetic pathway, results in decreased hepatic bile acid synthesis and prevents bile acid overload.[47]In addition to FXR, CDCA acts as an agonist for the Takeda G-protein-coupled receptor 5 (TGR5, also known as GPBAR-1), a membrane-bound receptor expressed on enteroendocrine L-cells, macrophages, and adipocytes. TGR5 activation by CDCA elevates intracellular cyclic adenosine monophosphate (cAMP) levels via G-protein signaling, which in L-cells promotes the secretion of glucagon-like peptide-1 (GLP-1) to enhance insulin sensitivity and glucose homeostasis. In brown adipose tissue, this cAMP-mediated pathway stimulates type 2 iodothyronine deiodinase (DIO2) expression, increasing local thyroid hormone production and thereby promoting thermogenesis and energy expenditure.[48]CDCA also modulates cholesterol metabolism by inhibiting the enzyme 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMG-CoA reductase) and the intestinal cholesterol transporter Niemann-Pick C1-like 1 (NPC1L1), primarily through FXR-dependent mechanisms. FXR activation in the liver represses sterol regulatory element-binding protein-2 (SREBP-2), which downregulates HMG-CoA reductase expression and reduces de novo cholesterol synthesis, while in the intestine, FXR induces fibroblast growth factor 19 (FGF19) secretion to suppress NPC1L1-mediated cholesterol absorption. Furthermore, CDCA exhibits anti-inflammatory properties by suppressing the nuclear factor kappa B (NF-κB) pathway in hepatocytes and macrophages; FXR activation antagonizes NF-κB translocation and transcriptional activity, reducing pro-inflammatory cytokine production such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), while TGR5 contributes by inhibiting NF-κB and c-Jun N-terminal kinase (JNK) signaling in immune cells.[49][50][51]Compared to ursodeoxycholic acid (UDCA), CDCA demonstrates greater hydrophobicity, which enhances its potency in altering bile composition by more effectively reducing biliary cholesterol saturation and forming less toxic micelles at therapeutic doses. At pharmacological concentrations, which exceed physiological levels, CDCA amplifies FXR and TGR5 signaling pathways without the full engagement of endogenous negative feedback mechanisms, such as those mediated by FGF19, leading to sustained metabolic modulation.[7][52]
Pharmacokinetics
Chenodeoxycholic acid (CDCA) is administered orally and undergoes efficient absorption primarily in the proximal small intestine through a combination of passive diffusion and active transport via the apical sodium-dependent bile acid transporter (ASBT).[53]Absorption is nearly complete, ranging from 80% to 99% depending on the dose and site of infusion, with peak plasma concentrations achieved 1 to 2 hours after ingestion.[54] Food intake delays absorption but does not significantly alter overall bioavailability.[11]The systemic bioavailability of CDCA is approximately 70-80%, limited by extensive first-pass extraction in the liver following absorption.[53] Once absorbed, CDCA enters the portal circulation and is efficiently taken up by hepatocytes, contributing to its confinement within the enterohepatic system.CDCA exhibits high plasma protein binding, primarily to albumin at levels exceeding 95%, which facilitates its transport.[55] Its distribution is largely restricted to the enterohepatic circulation, involving the intestine, portal vein, liver, and biliary tract, with minimal entry into systemic circulation.[11] The endogenous pool size of CDCA in healthy individuals is typically 1-2 g, part of the total bile acid pool of 2-4 g, maintained through efficient reabsorption.[53]Metabolism of CDCA occurs primarily in the liver, where it undergoes conjugation with glycine or taurine to form more soluble bile salts, with a typical glycine:taurine ratio of about 3:1 in humans.[11][56] In the intestine, gut bacteria perform 7-dehydroxylation to form lithocholic acid, a secondary bile acid and potentially toxic metabolite.[57]Elimination of CDCA is predominantly fecal, with 80-90% excreted unchanged or as bacterial derivatives like lithocholic acid, while a small fraction (about 5%) enters the systemic circulation and undergoes minor urinary excretion.[53] Due to extensive enterohepatic recirculation, the biological half-life of CDCA is 3-5 days, reflecting the slow turnover of the bile acid pool.[4]In special populations, pharmacokinetics may vary. Liver impairment reduces hepatic uptake and biliary secretion of CDCA, leading to decreased clearance and potential accumulation in plasma.[53] In pediatrics, particularly for conditions like cerebrotendinous xanthomatosis, dosing is adjusted based on body weight (e.g., 15 mg/kg/day), though specific pharmacokinetic alterations are not well-characterized and require monitoring.[11]
Adverse effects and contraindications
Chenodeoxycholic acid therapy is commonly associated with gastrointestinal adverse effects, primarily diarrhea occurring in 30-40% of patients due to bile acid malabsorption in the colon, along with nausea, abdominal pain, bloating, cramps, and dyspepsia.[36] These effects are typically dose-related and manageable with dose reduction, though approximately 3% of patients discontinue treatment due to uncontrolled diarrhea.[36]Hepatic adverse effects include transient elevations in serum aminotransferases in 20-30% of patients, often appearing within the first few months of therapy and resolving with dose adjustment or discontinuation; rare cases of clinically apparent hepatitis have been reported, particularly in early gallstone dissolution trials.[3] A portion of this hepatotoxicity arises from bacterial conversion of chenodeoxycholic acid to lithocholic acid in the gut, which can accumulate and cause liver injury, though levels remain minor in humans and the risk is mitigated by co-administration of ursodeoxycholic acid, which reduces lithocholic acid formation.[3][58]Other reported adverse effects include headache and pruritus, as well as dose-related hypercholesterolemia in some patients, particularly at lower doses.[59] In early clinical trials for gallstone dissolution, overall discontinuation rates due to side effects ranged from 18-30%, though long-term data indicate that most hepatic changes are reversible upon cessation of therapy.[60]Contraindications for chenodeoxycholic acid include known hepatobiliary disorders such as biliary obstruction, cirrhosis, or intrahepatic cholestasis; non-functioning gallbladder; radiopaque or bile pigment gallstones; complications from gallstones requiring surgery; and hypersensitivity to bile acids.[36] It is also contraindicated in pregnancy (FDA Category X) due to evidence of fetal harm in animal studies at high doses, though human data are limited.[36][61]Monitoring recommendations include monthly liver function tests (aminotransferases, bilirubin, alkaline phosphatase) for the first three months, followed by assessments every three months thereafter, with discontinuation advised if aminotransferases exceed three times the upper limit of normal or if bilirubin rises above twice normal.[36] Periodic imaging, such as cholecystograms or ultrasonograms every 6-9 months, is also suggested to evaluate gallstone response and detect complications.[36]
Research developments
Role in cancer
Chenodeoxycholic acid (CDCA), a primary bile acid characterized by its hydrophobic properties, promotes colorectal cancer (CRC) development by inducing DNA damage in colon epithelial cells, as evidenced by its ability to trigger unscheduled DNA synthesis indicative of repair responses in human colon cell lines.[62] Furthermore, CDCA upregulates cyclooxygenase-2 (COX-2) expression, contributing to inflammatory processes that foster tumorigenesis in the colon.[63] In cells harboring APC mutations, CDCA activates β-catenin signaling, enhancing cell proliferation and leading to increased tumor formation in the intestinal tract of animal models.[64] Epidemiological studies have linked elevated fecal CDCA levels to higher CRC risk, with significantly greater excretion observed in patients with large bowel cancer compared to controls (weighted mean difference of 0.28 mg/g dry feces).[65]In hepatocellular carcinoma (HCC), CDCA exhibits a synergistic effect with sorafenib, enhancing its antitumor efficacy through FXR-mediated pathways that promote apoptosis; a 2022 preclinical study demonstrated a twofold increase in treatment efficacy in HepG2 cell models and xenografts when CDCA was combined with sorafenib.[66] This combination inhibits proliferation, migration, and invasion more effectively than sorafenib alone by modulating the PI3K/AKT/mTOR signaling pathway, underscoring CDCA's potential as an adjunct in HCC therapy.[67]CDCA also holds anticancer potential, particularly in pancreatic cancer, where it inhibits progression via FXR activation; administration of CDCA as an FXR agonist attenuated pancreatic intraepithelial neoplasia (PanIN) development in Kras-mutant mouse models.[68] A 2023 metabolomics analysis further identified CDCA levels as predictive of reduced pancreatic necrosis, with higher recovery-phase concentrations correlating with less tissue damage in acute pancreatitis, a precursor to pancreatic ductal adenocarcinoma.[69] Through gut microbiota modulation, CDCA reduces the production of secondary bile acid carcinogens like deoxycholic acid (DCA), which promotes inflammation and CRC, while CDCA itself suppresses tumor growth in certain contexts by altering microbial composition to favor anti-inflammatory metabolites.[70] This dual role highlights CDCA's context-dependent influence on carcinogenesis via microbiome interactions.[71]Preclinical evidence supports CDCA's delivery via nanoformulations for targeted HCC therapy, with lipoprotein-mimicking nanoparticles incorporating CDCA showing enhanced tumor accumulation and efficacy in liver cancer models.[72] Therapeutic trials exploring CDCA derivatives as FXR agonists, such as obeticholic acid, are ongoing, including phase II evaluations as adjuncts in HCC to improve outcomes in advanced disease.[73]
Emerging therapeutic applications
Chenodeoxycholic acid (CDCA) has shown promise in preclinical models of acute lung injury (ALI) by activating the farnesoid X receptor (FXR) to exert anti-inflammatory effects. In lipopolysaccharide-induced ALI mouse models, CDCA treatment significantly reduced pathological lungtissue damage and lowered levels of pro-inflammatory cytokines such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), highlighting its potential to mitigate inflammation through FXR-mediated pathways.[74]In lipid metabolism disorders, particularly non-alcoholic fatty liver disease (NAFLD), CDCA regulates dyslipidemia via multi-omics approaches, demonstrating therapeutic effects on fatty acid metabolism and lipidhomeostasis. A 2025 study using transcriptomics, proteomics, and metabolomics revealed that CDCA supplementation in high-fat diet-induced NAFLD models lowered triglyceride levels and improved lipid profiles by modulating key pathways in hepatic lipid synthesis and oxidation.[75]For obesity management, nanoparticle-based delivery of CDCA promotes adipocyte browning and enhances fat burning. A single subcutaneous injection of CDCA-loaded nanoparticles in obese mouse models reduced adipocyte size and fat mass by inducing mitochondrial function and thermogenesis in white adipose tissue, offering a targeted approach to combat obesity without systemic side effects.[76]CDCA has been identified as a potential therapeutic for pancreatic necrosis in acute pancreatitis through bile acid metabolomics profiling. In cerulein-induced acute pancreatitis models, CDCA levels decreased during the acute phase but rose in recovery, with supplementation improving outcomes by reducing acinar cell necrosis and inflammation, suggesting its role in modulating bile acid signaling to protect pancreatic tissue.[69]In type 1 diabetes, CDCA enhances the viability of islet cells for encapsulation and transplantation by providing anti-apoptotic protection. Preclinical studies demonstrated that incorporating CDCA into alginate-based microencapsulation matrices preserved primary islet function and reduced apoptosis during in vitro culture and transplantation, potentially improving graft survival and insulin production in diabetic models.[77]Beyond these applications, CDCA holds potential in metabolic syndrome through activation of the Takeda G protein-coupled receptor 5 (TGR5), which regulates energy expenditure and glucose homeostasis. A 2024 review emphasized TGR5 agonism by CDCA as a mechanism to increase thermogenesis and insulin sensitivity, addressing interconnected features of metabolic syndrome such as dyslipidemia and hyperglycemia in preclinical settings.[78]